CN108348580B - Peptide inhibitors of interleukin-23 receptor and their use to treat inflammatory diseases - Google Patents

Peptide inhibitors of interleukin-23 receptor and their use to treat inflammatory diseases Download PDF

Info

Publication number
CN108348580B
CN108348580B CN201680049562.2A CN201680049562A CN108348580B CN 108348580 B CN108348580 B CN 108348580B CN 201680049562 A CN201680049562 A CN 201680049562A CN 108348580 B CN108348580 B CN 108348580B
Authority
CN
China
Prior art keywords
phe
pen
nal
lys
amino acid
Prior art date
Legal status (The legal status 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 status listed.)
Active
Application number
CN201680049562.2A
Other languages
Chinese (zh)
Other versions
CN108348580A (en
Inventor
格雷戈里·伯恩
程晓力
布莱恩·特洛伊·弗莱德里克
张婕
迪奈施·V·帕特尔
刘永鸿
艾少克·伯翰德瑞
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Protagonist Therapeutics Inc
Original Assignee
Protagonist Therapeutics Inc
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 US14/800,627 external-priority patent/US9624268B2/en
Application filed by Protagonist Therapeutics Inc filed Critical Protagonist Therapeutics Inc
Publication of CN108348580A publication Critical patent/CN108348580A/en
Application granted granted Critical
Publication of CN108348580B publication Critical patent/CN108348580B/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/06Linear peptides containing only normal peptide links having 5 to 11 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/02Nasal agents, e.g. decongestants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/08Linear peptides containing only normal peptide links having 12 to 20 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Abstract

The present invention provides novel peptide inhibitors of the interleukin-23 receptor and related compositions and methods of using these peptide inhibitors to treat or prevent various diseases and disorders, including inflammatory bowel disease.

Description

Peptide inhibitors of interleukin-23 receptor and their use to treat inflammatory diseases
Cross Reference to Related Applications
This application claims priority to and is a continuation-in-part of U.S. application No. 14/800,627 filed on 7/15/2015, and also claims priority to international patent application PCT/US2015/040658 filed on 7/15/2015, U.S. provisional application No. 62/264,820 filed on 12/8/2015, and U.S. provisional application No. 62/281,123 filed on 1/20/2016, all of which are incorporated herein by reference in their entirety.
Sequence listing
This application contains a sequence listing that has been submitted electronically in ASCII format and is incorporated by reference herein in its entirety. The ASCII copy created on 26.12/2017 was named PRTH _002_03WO _ st25.txt and was 808KB in size.
Technical Field
The present invention relates to novel peptide inhibitors of the interleukin-23 receptor, and their use to treat or prevent a variety of diseases and disorders, including inflammatory bowel disease, crohn's disease, and psoriasis.
Background
Interleukin-23 (IL-23) cytokines have been thought to play a crucial role in the pathogenesis of autoimmune inflammation and related diseases and disorders such as multiple sclerosis, asthma, rheumatoid arthritis, psoriasis, and Inflammatory Bowel Disease (IBD), e.g., ulcerative colitis and crohn's disease. Studies in acute and chronic mouse models of IBD reveal important roles of IL-23R and downstream effector cytokines in the pathogenesis of disease. IL-23R is expressed on a variety of adaptive and innate immune cells, including Th17 cells, γ δ T cells, Natural Killer (NK) cells, dendritic cells, macrophages, and innate lymphocytes, which are abundant in the intestine. At the intestinal mucosal surface, gene expression and protein levels of IL-23R have been found to be elevated in IBD patients. IL-23 is thought to be pathogenic CD4 by promoting the production of IL-6, IL-17 and Tumor Necrosis Factor (TNF)+The development of a population of T cells mediates this effect.
The produced IL-23 is enriched in the intestine, which is thought to play a key role in regulating the balance between tolerance and immunity through the T cell dependent and T cell independent pathways of enteritis, through effects on helper T cell 1(Th1) and Th 17-related cytokines, as well as inhibiting regulatory T cell responses in the intestine, which contribute to inflammation. In addition, polymorphisms in the IL-23 receptor (IL-23R) are associated with susceptibility to IBD, further establishing the critical role of the IL-23 pathway in intestinal homeostasis.
Psoriasis (a chronic skin disease that affects approximately 2% -3% of the general population) has been shown to be mediated by T cell inflammatory response mechanisms in the body. IL-23 is one of several interleukins, believed to be a key role in the pathogenesis of psoriasis, and is said to maintain chronic autoimmune inflammation through induction of interleukin-17, regulation of memory T cells, and activation of macrophages. Expression of IL-23 and IL-23R has been shown to be increased in patient tissues with psoriasis, and antibodies that neutralize IL-23 show IL-23 dependent inhibition of psoriasis development in animal models of psoriasis.
IL-23 is a heterodimer composed of a unique p19 subunit and the p40 subunit of IL-12, which is a helper T cell 1 (T-cell) involved in the production of interferon-gamma (IFN-. gamma.)H1) The developed cytokine of (1). Although IL-23 and IL-12 both contain p40 subunit, they have different phenotypic characteristics. For example, IL-12 deficient animals are susceptible to inflammatory autoimmune disease, whereas IL-23 deficient animals are resistant, presumably due to IL-6, IL-17 and TNF-producing CD4 in the CNS of IL-23 deficient animals+The number of T cells decreased. IL-23 and IL-23R combined, the IL-23R is composed of IL-12R beta 1 and IL-23R subunit composed of heterodimeric receptors. Binding of IL-23 to IL-23R activates Jak-Stat signaling molecules Jak2, Tyk2 and Stat1, Stat3, Stat4 and Stat 5, although Stat4 is substantially less activated and forms a different DN Α binding Stat complex in response to IL-23 compared to IL-12. IL-23R binds constitutively to Jak2 and binds in a ligand-dependent manner to Stat 3. IL-23 preferentially acts on memory CD4(+) T cells compared to IL-12, which acts primarily on naive CD4(+) T cells.
Efforts have been made to identify therapeutic moieties that inhibit the IL-23 pathway for the treatment of IL-23 related diseases and disorders. A number of antibodies have been identified that bind to IL-23 or IL-23R, including Ultekumab (a humanized antibody that binds IL-23) which has been approved for the treatment of psoriasis. Recently, polypeptide inhibitors that bind to IL-23R and inhibit the binding of IL-23 to IL-23R have been identified (see, e.g., U.S. patent application publication No. US 2013/0029907). Clinical trials of crohn's disease or psoriasis with ustekinumab and brazinitumab (briakumab), which targets the common p40 subunit, as well as tiltrakizumab, gusekumab, MEDI2070 and BI-655066, which targets the unique p19 subunit of IL-23, highlight the potential of blockade of IL-23 signaling in the treatment of inflammatory diseases in humans. While these findings are promising, there remains a challenge with identifying stable and selective agents that preferentially target the IL-23 pathway in the intestine, which may be used to treat inflammatory bowel disease, such as bowel disease, including crohn's disease, ulcerative colitis, and related disorders.
Clearly, there remains a need in the art for new therapies targeting the IL-23 pathway that can be used to treat and prevent IL-23 related diseases, including diseases associated with autoimmune inflammation in the intestinal tract. In addition, compounds and methods that specifically target IL-23R from the luminal side of the intestine may provide therapeutic benefit to IBD patients suffering from local intestinal tissue inflammation. The present invention addresses these needs by providing novel peptide inhibitors that bind IL-23R to inhibit binding and signaling of IL-23 and are suitable for oral administration.
Summary of The Invention
The invention provides, inter alia, novel peptide inhibitors of IL-23R and related methods of use.
In a first aspect, the present invention provides a peptide inhibitor of the interleukin-23 receptor, or a pharmaceutically acceptable salt or solvate thereof, wherein the peptide inhibitor comprises an amino acid sequence of formula (Xa): X1-X2-X3-X4-X5-X6-X7-X8-X9-X10-X11-X12-X13-X14-X15-X16-X17-X18-X19-X20(Xa) (SEQ ID NO:1354),
wherein:
x1, X2, and X3 are any amino acid or absent;
x4 is any amino acid or chemical moiety capable of forming a bond with X9;
x5, X6, X7, and X8 are any amino acid;
x9 is any amino acid or chemical moiety capable of forming a bond with X4;
x10, X11, X12, X13, X14, and X15 are any amino acid; and
x16, X17, X18, X19, and X20 are any amino acid or absent;
wherein the peptide inhibitor is cyclized via a bond between X4 and X9, and
wherein the peptide inhibitor inhibits the binding of interleukin-23 (IL-23) to an IL-23 receptor.
In certain embodiments of Xa:
x1 is absent; x2 is absent; x3 is absent; x4 is Cys, Abu or Pen; x5 is Ala, α -MeOrn, α -MeSer, Cit, Dap, Dab, Dap (Ac), Gly, Lys, Asn, N-MeGln, N-MeArg, Orn, Gln, Arg, Ser, or Thr; x6 is Asp or Thr; x7 is Trp or 6-chloro-Trp; x8 is Glu, Gln, or Val; x9 is Cys, Abu or Pen; x10 is 2-Nal, a Phe analog, Tyr or Tyr analog; x11 is 1-Nal, 2-Nal, Phe (3, 4-dimethoxy), 5-hydroxy Trp, Phe (3, 4-Cl) 2) Trp or Tyr (3-tBu); x12 is 3-Pal, Acpc, Acbc, Acvc, Achc, Agp, Aib, α -diethylGly, α -MeLys (Ac), α -MeLeu, α -MeOrn, α -MeSer, α -MeVal, Cav, Cha, Cit, Cpa, D-Asn, Glu, His, hLeu, hArg, Lys, Leu, Octgly, Orn, 4-amino-4-carboxy-piperidine, Arg, Ser, Thr, or THP; x13 is Cit, Asp, Dab, Dap, Phe, His, Dap (Peg2-Ac), Dap (pyroglutamic acid), Glu, homo-Arg, Lys (Ac), Lys (benzoic acid), Lys (glutaric acid), Lys (IVA), Lys (Peg 4-isoGlu-Palm), Lys (pyroglutamic acid), Lys (succinic acid), Asn, Orn, Gln, Arg, Thr or Val; x14 is Asp, Dab (Ac), dap (Ac), Phe, His, Lys (Ac), Met, Asn (isobutyl), Gln, Arg, Tyr or Asp (1, 4-diaminobutane); and X15 is Ala, β Ala, Glu, Gly, Asn, Gln, Arg, or Ser.
In certain embodiments of Xa: x1 is absent; x2 is absent(ii) a X3 is absent; x4 is Cys, Abu or Pen; x5 is Ala, α -MeOrn, α -MeSer, Cit, Dap, Dab, Dap (Ac), Gly, Lys, Asn, Orn, Gln, Arg, Ser, or Thr; x6 is Asp or Thr; x7 is Trp or 6-chloro-Trp; x8 is Gln or Val; x9 is Cys, Abu or Pen; x10 is 2-Nal, a Phe analog, a Tyr or Tyr analog; x11 is 1-Nal, 2-Nal, Phe (3, 4-dimethoxy), 5-hydroxy Trp, Phe (3, 4-Cl) 2) Trp or Tyr (3-tBu); x12 is 3-Pal, Acpc, Acbc, Acvc, Achc, Agp, Aib, α -diethylGly, α -MeLys (Ac), α -MeLeu, α -MeOrn, α -MeSer, α -MeVal, Cav, Cha, Cit, Cpa, D-Asn, His, hLeu, hArg, Lys, Leu, Octgly, Orn, 4-amino-4-carboxy-piperidine or THP; x13 is Cit, Asp, Dab, Dap, Phe, His, Dap (Peg2-Ac), Dap (pyroglutamic acid), Glu, hArg, Lys (Ac), Lys (benzoic acid), Lys (glutaric acid), Lys (IVA), Lys (Peg 4-isoGlu-Palm), Lys (pyroglutamic acid), Lys- (succinic acid), Asn, Orn, Gln, Arg, Thr or Val; x14 is dab (Ac), dap (Ac), Phe, His, Lys (Ac), Met, Asn, Gln, Arg, or Tyr; and X15 is Ala, β Ala, Gly, Asn, Gln or Ser.
In certain embodiments of Xa: x1 is absent; x2 is absent; x3 is absent; x4 is Cys, Abu or Pen; x5 is Dap, Dap (Ac), Gly, Lys, Gln, Arg, Ser, Thr, or Asn; x6 is Thr; x7 is Trp or 6-chloro-Trp; x8 is Gln; x9 is Cys, Abu or Pen; x10 is 2-Nal, a Phe analog, Tyr or Tyr analog; x11 is 1-Nal, 2-Nal, Phe (3, 4-dimethoxy), Phe (3, 4-Cl)2) Or Trp; x12 is Acpc, Acbc, Acvc, Achc, Aib, α -diethylGly, α -MeLys (Ac), α -MeLeu, α -MeOrn, α -MeSer, α -MeVal, Cha, Cit, hLeu, Lys, Leu, Arg, or THP; x13 is Cit, Asp, Dap (Peg2-Ac), Dap (pyroglutamic acid), Glu, hArg, Lys (Ac), Lys (benzoic acid), Lys (glutaric acid), Lys (IVA), Lys (Peg 4-isoGlu-Palm), Lys (pyroglutamic acid), Lys- (succinic acid), Asn, Orn, Gln, Arg, or Val; x14 is dab (Ac), dap (Ac), His, Lys (Ac), Asn, Gln, or Tyr; and X15 is Ala, β Ala, Gly, Asn, Gln or Ser.
In certain embodiments of Xa: x1 is absent; x2 is absent; x3 is absent; x4 is Cys, Abu or Pen; x5 is Dap, Dap (Ac), Gln, Ser, Thr, or Asn; x6 is Thr; x7 is Trp; x8 is Gln; x9 is Cys, Abu or Pen; x10 is a Phe analog, Tyr or Tyr analog; x11 is 2-Nal or Trp; x12 is Acpc, Acbc, Acvc, Achc, Aib, α -diethylGly, α -MeLys (Ac), α -MeLeu, α -MeOrn, α -MeSer, α -MeVal, hLeu, Leu, or THP; x13 is Cit, Asp, Glu, Lys (Ac), Asn or Gln; x14 is dab (Ac), Asn or His; and X15 is Ala, β Ala, Gly, Asn or Gln.
In certain embodiments of Xa: x4 is Cys, Pen, hCys, D-Pen, D-Cys, D-hCys, Met, Glu, Asp, Lys, Orn, Dap, Dab, D-Dap, D-Dab, D-Asp, D-Glu, D-Lys, Sec, 2-chloromethylbenzoic acid, mercaptopropionic acid, mercaptobutyric acid, 2-chloro-acetic acid, 3-chloropropionic acid, 4-chlorobutyric acid, 3-chloroisobutyric acid, Abu, β -azido-Ala-OH, propargylglycine, 2- (3 '-butenyl) glycine, 2-allylglycine, 2- (3' -butenyl) glycine, 2- (4 '-pentenyl) glycine, 2- (5' -hexenyl) glycine or Abu; x7 is Trp, Glu, Gly, Ile, Asn, Pro, Arg, Thr, or OctGly or the corresponding α -methyl amino acid form of any one of the foregoing; x9 is Cys, Pen, hCys, D-Pen, D-Cys, D-hCys, Glu, Lys, Orn, Dap, Dab, D-Dap, D-Dab, D-Asp, D-Glu, D-Lys, Asp, Leu, Val, Phe or Ser, Sec, Abu, β -azido-Ala-OH, propargylglycine, 2-2-allylglycine, 2- (3 ' -butenyl) glycine, 2- (4 ' -pentenyl) glycine, Ala, hCys, Abu, Met, MeCys, (D) Tyr or 2- (5 ' -hexenyl) glycine; x10 is Tyr, Phe (4-OMe), 1-Nal, 2-Nal, Aic, alpha-MePhe, Bip, (D) Cys, Cha, DMT, (D) Tyr, Glu, His, hPhe (3, 4-dimethoxy), hTyr, N-Me-Tyr, Trp, Phe (4-CONH) 2) Phe (4-phenoxy), Thr, Tic, Tyr (3-tBu), Phe (4-CN), Phe (4-Br), Phe (4-NH)2)、Phe(4-F)、Phe(3,5-F2)、Phe(4-CH2CO2H)、Phe(5-F)、Phe(3,4-Cl2)、Phe(4-CF3)、Phe(4-OCH3) Bip, Cha, 4-pyridylalanine, beta hTyr, OctGly, Phe (4-N)3) Phe (4-Br), Phe [4- (2-aminoethoxy)]Or Phe, Phe analog, Tyr analog, or any of the foregoingThe alpha-methyl amino acid form of (a); x11 is 2-Nal, 1-Nal, 2, 4-dimethyl Phe, Bip, Phe (3, 4-Cl)2)、Phe(3,4-F2)、Phe(4-CO2H) Beta hPhe (4-F), alpha-Me-Trp, 4-phenylcyclohexyl, Phe (4-CF)3) alpha-MePhe, beta hNal, beta hPhe, beta hTyr, beta hUrp, Nva (5-phenyl), Phe, His, hPhe, Tic, Tqa, Trp, Tyr, Phe (4-OMe), Phe (4-Me), Trp (2,5, 7-tri-tert-butyl), Phe (4-O allyl), Tyr (3-tBu), Phe (4-guanidino, Phe (4-OBzl), Octgly, Glu (Bzl), 4-phenylbenzylalanine, Phe [4- (2-aminoethoxy)]5-hydroxy-Trp, 6-chloro-Trp, N-MeTrp, 1,2,3, 4-tetrahydro-norharman (norharman), Phe (4-CONH)2) Phe (3, 4-dimethoxy), Phe (2, 3-Cl)2)、Phe(2,3-F2) Phe (4-F), 4-phenylcyclohexylalanine, Bip, or the corresponding alpha-methyl amino acid form of any of the foregoing; x12 is His, Phe, Arg, N-Me-His, Val, Cav, Cpa, Leu, Cit, hLeu, 3-Pal, t-butyl-Ala, 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acvc, Acbc, Agp, Aib, α -diethylGly, α 7-MeLys, α 9-MeLys (Ac), α -Me-Leu, α 5-MeOrn, α 7-MeSer, α 9-MeVal, Aib, D-Ala, (D) Asn, (D) Asp, (D) Leu, (D) Phe, (D) Tyr, Aib, α -MeLeu, α -MeOrn, α 0-Aib, α 1-Ala, α 2hAla, α 3hArg, α 4hLeu, α 5 hLeu, α 6-spiro-pip, Glu, hArg, N-Leu, N-Arg, N-Megln, Thr, T-Gl, N-Arg, T-Met, t-L, L, t-butyl-Gly or the form of an alpha-methyl amino acid corresponding to any of the foregoing; x13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, Arg, Orn, Val, α 8hAla, Lys (Ac), (D) Asn, (D) Leu, (D) Phe, (D) Thr, Ala, α -MeLeu, Aib, α 0-Ala, α 1-Glu, α 2hLeu, α 3hVal, α 4-spiro-pip, Cha, Chg, Asp, Dab, Dap, α -diethylGly, hLeu, Asn, Ogl, Pro, Gln, Ser, α 6-spiro-pip, Thr, Tba, Tle or Aib, Cit, hA, Lys, Asn, Orn, Gln, or an α -methylamino form corresponding to any of the foregoing; x14 is Phe, Tyr, Glu, Gly, His, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Tic α 8hPhe, Arg, Lys (Ac), His; dap (ac), dab (ac), Asp, or an alpha-methyl amino acid form corresponding to any one of the preceding; x15 is Gly, Ser, Thr, Gln, Ala, (D) Asn, (D) Asp, (D) D) Leu, (D) Phe, (D) Thr, Aea, Asp, Asn, Glu, Phe, Gly, Lys, Leu, Pro, Arg, β -Ala, Sarc, or an α -methyl amino acid form corresponding to any of the foregoing; x16 is Asp, Glu, Ala, AEA, AEP, β hAla, Gaba, Gly, Ser, Pro, Asn, Thr or is absent, or is in the form of the corresponding α -methyl amino acid of any of the foregoing; and X17 is Leu, Lys, Arg, Glu, Ser, Gly, Gln or absent, or is in the form of an alpha-methyl amino acid corresponding to any of the foregoing.
In certain embodiments of the peptide inhibitor of Xa, the linkage is a disulfide linkage, a thioether linkage, a lactam linkage, a triazole ring, a selenoether linkage, a diselenide linkage, or an alkene linkage.
In a specific embodiment of the peptide inhibitor of Xa, X4 is Cys and X9 is Cys, and the bond is a disulfide bond. In a specific embodiment, X4 is Pen and X9 is Pen, and the bond is a disulfide bond. In certain embodiments: x7 is Trp; x10 is Phe, Tyr, a Phe analog, or a Tyr analog; x11 is Trp, 1-Nal or 2-Nal; and X12 is Aib, α -Me-Lys, α -Me-Leu, Achc, Acvc, Acpc, Acbc, or THP. In certain embodiments: x7 is Trp; x10 is Phe, Tyr, a Phe analog, or a Tyr analog; x11 is Trp, 1-Nal or 2-Nal; and X12 is Aib, α -Me-Lys or α -Me-Leu. In a specific embodiment, the peptide inhibitor comprises any one of the following amino acid sequences: Pen-Q-T-W-Q-Pen- [ Phe (4-OMe) ]-[2-Nal]-[α-Me-Lys]-E-N-G(SEQ ID NO:254);Pen-N-T-W-Q-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-N-N(SEQ ID NO:255);Pen-Q-T-W-Q-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[α-MeLeu]-[Lys(Ac)]-N-N (SEQ ID NO: 256); or Pen-Q-T-W-Q- [ Pen]-[Phe(4-CONH2)]-[2-Nal]-[α-MeLys]-[Lys(Ac)]N-N (SEQ ID NO:257), wherein the peptide inhibitor comprises a disulfide bond between two Pen amino acids.
In particular embodiments of peptide inhibitors of Xa, X4 is an amino acid, fatty acid, alicyclic acid, or modified 2-methyl aromatic acid having a carbon side chain capable of forming a thioether linkage with X9; x9 is a sulfur-containing amino acid capable of forming a thioether bond with X4, and the bond between X4 and X9 is a thioether bond. In certain embodiments, X4 is Abu, 2-chloromethylbenzoic acid, mercaptopropionic acid, mercaptobutyric acid, 2-chloro-acetic acid, 3-chloro-propionic acid, 4-chloro-butyric acid, 3-chloro-isobutyric acid; and X9 is Abu, Cys, Pen, hCys, D-Pen, D-Cys or D-hCys. In certain embodiments, X4 is Abu; and X9 is Cys. In certain embodiments, X7 is Trp; x10 is Phe, Tyr, a Phe analog, or a Tyr analog; x11 is Trp, 1-Nal or 2-Nal; and X12 is α -Me-Lys, α -Me-Leu, α -Me-Ser, α -Me-Val, Achc, Acvc, Acpc, Acbc, or [ 4-amino-4-carboxy-tetrahydropyran ]. In certain embodiments, X7 is Trp; x10 is Phe, Tyr, a Phe analog, or a Tyr analog; x11 is Trp, 1-Nal or 2-Nal; and X12 is alpha-Me-Lys or [ 4-amino-4-carboxy-tetrahydropyran ]. In a specific embodiment, the peptide inhibitor comprises any one of the following amino acid sequences: [ Abu ] -Q-T-W-Q-C- [ Phe (4-OMe) ] - [2-Nal ] - [ alpha-MeLys ] -E-N-G (SEQ ID NO: 258); [ Abu ] -Q-T-W-Q-C- [ Phe (4- (2-aminoethoxy)) ] -W- [ alpha-MeLys ] -E-N-G (SEQ ID NO: 259); or [ Abu ] -Q-T-W-Q-C- [ Phe [4- (2-aminoethoxy) ] ] - [2-Nal ] - [ 4-amino-4-carboxy-tetrahydropyran ] -E-N-N (SEQ ID NO:260), wherein the peptide inhibitor comprises a thioether bond between Abu and C.
In certain embodiments of the peptide inhibitor of Xa: x4 is Pen, Cys or hCys; x5 is any amino acid; x6 is any amino acid; x7 is Trp, Bip, Gln, His, Glu (Bzl), 4-phenylbenzylalanine, Tic, Phe [4- (2-aminoethoxy)]、Phe(3,4-Cl2) Phe (4-OMe), 5-hydroxy-Trp, 6-chloro-Trp, N-MeTrp, alpha-Me-Trp, 1,2,3, 4-tetrahydro-norharpagne, Phe (4-CO)2H)、Phe(4-CONH2) Phe (3, 4-dimethoxy), Phe (4-CF)3) Phe (4-tBu), β β -diphela, Glu, Gly, Ile, Asn, Pro, Arg, Thr, or Octgly, or an α -methyl amino acid form corresponding to any of the foregoing; x8 is any amino acid; x9 is Pen, Cys or hCys; x10 is 1-Nal, 2-Nal, Aic, Bip, (D) Cys, Cha, DMT, (D) Tyr, Glu, Phe, His, Trp, Thr, Tic, Tyr, 4-pyridyl Ala, Octgly, a Phe analog, or a Tyr analog (optionally, Phe (3, 4-F)2)、Phe(3,4-Cl2) F (3-Me), Phe [4- (2-aminoethoxy)]Phe [4- (2- (acetyl-aminoethoxy)]、Phe(4-Br)、Phe(4-CONH2) Phe (4-Cl), Phe (4-CN), Phe (4-guanidino), Phe(4-Me)、Phe(4-NH2)、Phe(4-N3) Phe (4-OMe) or Phe (4-OBzl)) or the corresponding alpha-methyl amino acid form of any of the foregoing; x11 is 2-Nal, 1-Nal, 2, 4-dimethyl Phe, Bip, Phe (3, 4-Cl)2)、Phe(3,4-F2)、Phe(4-CO2H) Beta hPhe (4-F), alpha-Me-Trp, 4-phenylcyclohexyl, Phe (4-CF) 3) alpha-MePhe, beta hNal, beta hPhe, beta hTyr, beta hUrp, Nva (5-phenyl), Phe, His, hPhe, Tic, Tqa, Trp, Tyr, Phe (4-OMe), Phe (4-Me), Trp (2,5, 7-tri-tert-butyl), Phe (4-O allyl), Tyr (3-tBu), Phe (4-guanidino, Phe (4-OBzl), Octgly, Glu (Bzl), 4-phenylbenzylalanine, Phe [4- (2-aminoethoxy)]5-hydroxy-Trp, 6-chloro-Trp, N-MeTrp, 1,2,3, 4-tetrahydro-norharman, Phe (4-CONH)2)、Phe(3,4-OMe2)Phe(2,3-Cl2)、Phe(2,3-F2) Phe (4-F), 4-phenylcyclohexylalanine or Bip or the corresponding alpha-methyl amino acid form of any of the foregoing; x12 is α -MeLys, α 1-MeOrn, α 2-MeLeu, α 3-MeVal, 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Acvc, MeLeu, Aib, (D) Ala, (D) Asn, (D) Leu, (D) Asp, (D) Phe, (D) Thr, 3-Pal, Aib, α 0-Ala, α 4hGlu, α 5hAla, α 6hLeu, α 7hVal, α 8-spiro-pip, Cha, Chg, Asp, Dab, Dap, α -diethylGly, Glu, Phe, hLeu, hArg, Ile, Lys, Leu, Asn, N-MeLeu, N-MeArg, Ogl, Orn, Pro, Gln, Arg, Ser, Thr or Tle, or any of the corresponding α 0-methyl amino acid form of the foregoing; x13 is Lys (Ac), (D) Asn, (D) Leu, (D) Thr, (D) Phe, Ala, Aib, α 1-MeLeu, α 9-Ala, β hGlu, β hAla, β hLeu, β hVal, β -spiro-pip, Cha, Chg, Asp, Lys, Arg, Orn, Dab, Dap, α 2-diethylGly, Glu, Phe, hLeu, Lys, Leu, Asn, Ogl, Pro, Gln, Asp, Arg, Ser, spiro-pip, Thr, Tba, Tlc, Val, or Tyr or an α -methyl amino acid form corresponding to any of the foregoing; x14 is Asn, Glu, Phe, Gly, His, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Tic or Tyr, Lys (Ac), Orn, or a form of any of the foregoing corresponding α -methyl amino acids; x15 represents Gly, (D) Ala, (D) Asn, (D) Asp, Asn, (D) Leu, (D) Phe, (D) Thr, Ala, AEA, Asp, Glu, Phe, Gly, Lys, Leu, Pro, G ln, Arg or Ser, beta-Ala, Arg or an alpha-methyl amino acid form corresponding to any one of the foregoing; x16 is absent, Gly, Ala, Asp, Ser, Pro, Asn, or Thr, or a corresponding α -methyl amino acid form of any of the foregoing; x17 is absent, is Glu, Ser, Gly, or gin, or the α -methyl amino acid form corresponding to any of the foregoing; x18 is absent or is any amino acid; x19 is absent or is any amino acid; and X20 is absent or is any amino acid. In a specific embodiment, the bond between X4 and X9 is a disulfide bond. In certain embodiments, X1, X2, and X3 are absent. In certain embodiments, X17, X19, and X20 are absent. In certain embodiments, one or both of X4 or X9 is Pen. In certain embodiments, both X4 and X9 are Pen. In a specific embodiment, X18 is (D) -Lys. In certain embodiments, the peptide inhibitor comprises one or more, two or more, three or more, or four of the following: x5 is Arg, Asn, Gln, Dap, Orn; x6 is Thr or Ser; x7 is Trp, 2-Nal, 1-Nal, Phe (4-O allyl), Tyr (3-tBu), Phe (4-guanidino), Phe (Bzl) or Phe (4-Me), 5-hydroxy-Trp, 6-chloro-Trp, N-MeTrp, α -MeTrp or 1,2,3, 4-tetrahydro-norharpagne; and X8 is Gln, Val, Phe, Glu, Lys. In certain embodiments, the peptide inhibitor comprises one or more, two or more, three or more, four or more, five or more, six or more, or seven of: x10 is Tyr, Phe (4-OBzl), Phe (4-OMe), Phe (4-CONH) 2)、Phe(3,4-Cl2)、Phe(4-tBu)、Phe(4-NH2)、Phe(4-Br)、Phe(4-CN)、Phe(4-CO2H) Phe (4- (2 aminoethoxy)) or Phe (4-guanidino); x11 is Trp, 2-Nal, 1-Nal, Phe (4-O allyl), Tyr (3-tBu), Phe (4-guanidino), Phe (Bzl) or Phe (4-Me), 5-hydroxy-Trp, 6-chloro-Trp, N-MeTrp, α -MeTrp or 1,2,3, 4-tetrahydro-norharpagne; x12 is Arg, α -MeLys, α -MeLeu, Aib, or α -MeOrn; x13 is Lys, Glu, or Lys (Ac); x14 is Phe or Asn; x15 is Gly, Sr, or Ala; and X16 is absent or is AEA. In certain embodiments, X4 and X9 are Pen; x5 is Gln; x6 is Thr; x7 is Trp; x8 is Gln; x10 is Tyr, Phe (4-OMe) or 2-Nal; x11 is Trp, 2-Nal or 1-Nal; x12 is Arg, α MeLys, or α -MeOrn; x13 is Lys, Glu, or Lys (Ac); x14 is Phe or Asn; x15 is Gly; and X16 is absent. In certain embodiments, one or more of X1, X2, and X3 is absent; and one or more, two or more, three or more, or four of X17, X18, X19, and X20 are absent.
In certain embodiments of the peptide inhibitor of Xa: x4 is Abu, Pen or Cys; x7 is Trp, Bip, Gln, His, Glu (Bzl), 4-phenylbenzylalanine, Tic, Phe [4- (2-aminoethoxy)]、Phe(3,4-Cl2) Phe (4-OMe), 5-hydroxy-Trp, 6-chloro-Trp, N-MeTrp, alpha-MeTrp, 1,2,3, 4-tetrahydro-norharpagne, Phe (4-CO) 2H)、Phe(4-CONH2) Phe (3, 4-dimethoxy), Phe (4-CF)3) β -dipheAla, Phe (4-tBu), Glu, Gly, Ile, Asn, Pro, Arg, Thr, or Octgly or any of the corresponding α -methyl amino acid forms thereof; x9 is Abu, Pen, or Cys; x10 is 1-Nal, 2-Nal, Aic, Bip, (D) Cys, Cha, DMT, (D) Tyr, Glu, Phe, His, Trp, Thr, Tic, Tyr, 4-pyridyl Ala, Octgly, a Phe analog, or a Tyr analog, or the corresponding α -methyl amino acid form of any of the foregoing; x11 is 2-Nal, 1-Nal, 2, 4-dimethyl Phe, Bip, 4-phenylcyclohexyl, Glu (Bzl), 4-phenylbenzylalanine, Tic, Phe [4- (2-aminoethoxy)]、Phe(3,4-Cl2)、Phe(3,4-F2) Beta hPhe (4-F), Phe (4-OMe), 5-hydroxy-Trp, 6-chloro-Trp, N-MeTrp, alpha-MeTrp, 1,2,3, 4-tetrahydro-norharpagne, Phe (4-CO)2H)、Phe(4-CONH2) Phe (3, 4-dimethoxy), Phe (4-CF)3)、Phe(2,3-Cl2)、Phe(2,3-F2) Phe (4-F), 4-phenylcyclohexylalanine,. alpha. -MePhe,. beta. -hNal,. beta.0 hPhe,. beta.1 hTyr,. beta.2 hTRp, Bip, Nva (5-phenyl), Phe, His, hPhe, Tqa, Trp, Tyr, Phe (4-Me), Trp (2,5, 7-tri-tert-butyl), Phe (4-O allyl), Tyr (3-tBu), Phe (4-guanidino), Phe (4-OBzl) or Octgly or any of the corresponding alpha-methylamino forms thereof; x12 represents α -MeLys, α -MeOrn, α -MeLeu, Aib, (D) Ala, (D) Asn, (D) Leu, (D) Asp, (D) Phe, (D) Thr, 3-Pal, Aib, β 3-Ala, β 4hGlu, β hAla, β hLeu, β hVal, β -spiro-pip, Cha, Chg, Asp, Dab, Dap, alpha-diethylGly, Glu, Phe, hLeu, hArg, hLeu, Ile, Lys, Leu, Asn, N-MeLeu, N-MeArg, Ogl, Orn, Pro, Gln, Arg, Ser, Thr or Tle or an alpha-methyl amino acid form corresponding to any of the foregoing; x13 is Lys (Ac), (D) Asn, (D) Leu, (D) Thr, (D) Phe, Ala, Aib, α 0-MeLeu, β Ala, β hGlu, β hLa, β hLeu, β hVal, β -spiro-pip, Cha, Chg, Asp, Arg, Orn, Dab, Dap, α 1-diethylGly, Glu, Phe, hLeu, Lys, Leu, Asn, Ogl, Pro, Gln, Asp, Arg, Ser, spiro-pip, Thr, Tba, Tlc, Val, or Tyr or the α 2-methylamino form corresponding to any of the foregoing; x14 is Asn, Glu, Phe, Gly, His, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Tic, or Tyr, or a α 3-methylamino form corresponding to any of the foregoing; x15 is Gly, (D) Ala, (D) Asn, (D) Asp, Asn, (D) Leu, (D) Phe, (D) Thr, Ala, AEA, Asp, Glu, Phe, Gly, Lys, Leu, Pro, Gln, Arg, or Ser, or an α 4-methylamino form corresponding to any of the foregoing, or X15 is Gly, (D) Ala, (D) Asn, (D) Asp, Asn, (D) Leu, (D) Phe, (D) Thr, Ala, Asn, Ser, AEA, Asp, Glu, Phe, Gly, Lys, Leu, Pro, gin, Arg, or Ser, or an α 5-methylamino form corresponding to any of the foregoing; x16 is absent, Gly, Ala, Asp, Ser, Pro, Asn or Thr or a corresponding α 6-methyl amino acid form of any of the foregoing; and X17 is absent, is Glu, Ser, Gly, or gin, or an alpha-methyl amino acid form corresponding to any of the foregoing. In a specific embodiment, the peptide inhibitor is cyclized via an intramolecular bond between X4 and X9. In certain embodiments, one or more of X1, X2, and X3 is absent. In certain embodiments, one or more of X17, X19, and X20 is absent. In certain embodiments, one of X4 or X9 is Abu and the other of X4 or X9 is not Abu. In certain embodiments, the peptide inhibitor comprises one or more, two or more, three or more, or four of the following: x5 is Arg, Gln, Dap, or Orn; x6 is Thr or Ser; x7 is Trp, 2-Nal, 1-Nal, Phe (4-O allyl), Tyr (3-tBu), Phe (4-guanidino), Phe (4-OBzl), Phe (4-Me), 5-hydroxy-Trp, 6-chloro-Trp, N-MeTrp or alpha-MeTrp, 1,2,3, 4-tetrahydro-nor Harbin man; and X8 is Gln, Val, Phe, Glu, or Lys. In certain embodiments, the peptide inhibitor comprises one or more, two or more, three or more, four or more, five or more, six or more, or seven of: x10 is Tyr, Phe (4-OBzl), Phe (4-OMe), Phe (4-CONH)2)、Phe(3,4-Cl2)、Phe(4-tBu)、Phe(4-NH2)、Phe(4-Br)、Phe(4-CN)、Phe(4-CO2H) Phe (4- (2 aminoethoxy)) or Phe (4-guanidino); x11 is Trp, 2-Nal, 1-Nal, Phe (4-O allyl), Tyr (3-tBu), Phe (4-guanidino), Phe (Bzl) or Phe (4-Me), 5-hydroxy-Trp, 6-chloro-Trp, N-MeTrp, α -MeTrp or 1,2,3, 4-tetrahydro-norharpagne; x12 is Arg, hLeu, (D) Asn, Aib, α -MeLys, α -MeLeu, or α -MeOrn; x13 is Lys, Glu, or Lys (Ac); x14 is Phe or Asn; x15 is Gly, Ser or Ala, or X15 is Asn, Gly, Ser, beta Ala or Ala; and X16 is absent or is AEA.
In a specific embodiment of any one of the peptide inhibitors, X4 and X9 are Pen. In particular embodiments, X4 and X9 form a disulfide bond.
In a specific embodiment, X4 is Abu and X9 is Cys. In particular embodiments, X4 and X9 form a thioether bond.
In a specific embodiment, the peptide inhibitor comprises SEQ ID NO: 365-. In particular embodiments, the peptide inhibitor is cyclized via the bond between X4 and X9, and the peptide inhibitor inhibits the binding of interleukin-23 (IL-23) to an IL-23 receptor.
In certain embodiments of the peptide inhibitor of Xa, the peptide inhibitor comprises an amino acid sequence represented by any one of formulas (V), (Va), (Vb), (Vc), (Vd), (Ve), (Vf), (Vg), and (Vh).
In certain embodiments of the peptide inhibitor of Xa, the peptide inhibitor comprises any one of the following amino acid sequences:
[Palm]- [ isoGlu]-[PEG4]-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-NNNH2(SEQ ID NO:1115);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-amino)Ethoxy group)]-[2-Nal]-[Aib]- [ Lys (PEG 4-isoGlu-Palm)]-NN-NH2(SEQ ID NO:1116);
Ac-[Pen]-QTWQ-[Pen]-Phe(4-CONH2)-[2-Nal]-[α-MeLys(Ac)]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1117);
[ octyl group]- [ isoGlu]-[PEG4]-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1118);
[ octyl group]-[PEG4]-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1119);
[Palm]-[PEG4]-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1120);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]- [ Lys (PEG 4-octyl)]-NN-NH2(SEQ ID NO:1121);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(PEG4-Palm)]-NN-NH2(SEQ ID NO:1122);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy) - (PEG4-Palm)]-[2-Nal]-[Aib]-[Lys(Ac)]NN-NH2(SEQ ID NO:1123);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy) - (PEG 4-lauryl)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1124);
Ac-[Pen]-QTWQ-[Pen]-Phe(4-CONH2)-[2-Nal]-[α-MeLys(PEG4-Palm)-[Lys(Ac)]-NN-NH2(SEQ ID NO:1125);
Ac-[Pen]-QTWQ-[Pen]-Phe(4-CONH2)-[2-Nal]- [ alpha-MeLys (PEG 4-lauryl)]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1126);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy) - (PEG 4-isoGlu-Palm)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1127);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy) - (PEG 4-isoGlu-lauryl)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1128);
Ac-[Pen]-QTWQ-[Pen]-Phe(4-CONH2)-[2-Nal]- [ alpha-MeLys (PEG 4-isoGlu-Palm)]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1129);
Ac-[Pen]-QTWQ-[Pen]-Phe(4-CONH2)-[2-Nal]- [ alpha-MeLys (PEG 4-isoGlu-lauryl)]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1130);
Ac-[Pen]-QTWQ-[Pen]-Phe(4-CONH2)-[2-Nal]-[α-MeLys(IVA)]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1131);
Ac-[Pen]-QTWQ-[Pen]-Phe(4-CONH2)-[2-Nal]- [ alpha-MeLys (biotin)]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1132);
Ac-[Pen]-QTWQ-[Pen]-Phe(4-CONH2)-[2-Nal]- [ alpha-MeLys (octyl)]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1133);
Ac-[Pen]-[Lys(IVA)]-TWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1134);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-[Lys(IVA)]-N-NH2(SEQ ID NO:1135);
Ac-[Pen]- [ Lys (biotin)]-TWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1136);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]- [ Lys (biotin)]-N-NH2(SEQ ID NO:1137);
Ac-[Pen]- [ Lys (octyl) ]-TWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1138);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]- [ Lys (octyl)]-N-NH2(SEQ ID NO:1139);
Ac-[Pen]-[Lys(Palm)]-TWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]--[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1140);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-Lys(Palm)]-N-NH2(SEQ ID NO:1141);
Ac-[Pen]-[Lys(PEG8)]-TWQ-[Pen]-[Phe[4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1142);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-[Lys(PEG8)]-N-NH2(SEQ ID NO:1143);
Ac-[Pen]-K(Peg11-Palm)TWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1144);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]--[Aib]-[Lys(Ac)]-[Lys(Peg11-palm)]-N-NH2(SEQ ID NO:1145);
Ac-[Pen]-[Cit]-TW-[Cit]-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]--[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1146);
Ac-[Pen]-[Lys(Ac)]-TW-[Cit]-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1147);
Ac-[Pen]-NT-[Phe(3,4-OCH3)2]-Q-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1148);
Ac-[Pen]-NT-[Phe(2,4-CH3)2]-Q-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1149);
Ac-[Pen]-NT-[Phe(3-CH3)]-Q-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1150);
Ac-[Pen]-NT-[Phe(4-CH3)]-Q-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1151);
Ac[(D)Arg]-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-N-[βAla]-NH2(SEQ ID NO:1152);
Ac-[(D)Tyr]-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-N-[βAla]-NH2(SEQ ID NO:1153);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-QN-NH2(SEQ ID NO:1154);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-[Lys(Ac)]-N-NH2(SEQ ID NO:1155);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-N-[Lys(Ac)]-NH2(SEQ ID NO:1156);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-QQ-NH2(SEQ ID NO:1157);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-Q-[βAla]-NH2(SEQ ID NO:1158);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-N-[Cit]-NH2(SEQ ID NO:1159);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-[Cit]-NNH2(SEQ ID NO:1160);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-[Cit]-Q-NH2(SEQ ID NO:1161);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-[Cit]-[Lys(Ac)]-NH2(SEQ ID NO:1162);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-[Lys(Ac)]-[Cit]-NH2(SEQ ID NO:1163);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-QN-[βAla]-NH2(SEQ ID NO:1164);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-E-[Cit]-Q-NH2(SEQ ID NO:1165);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Cit]-N-[Cit]-NH2(SEQ ID NO:1166);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Cit]-Q-[Cit]-NH2(SEQ ID NO:1167);
Ac-[Pen]-[Cit]-TWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1168);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1169);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-QNN-NH2(SEQ ID NO:1170);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-ENQ-NH2(SEQ ID NO:1171);
Ac-[Pen]-GPWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1172);
Ac-[Pen]-PGWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1173);
Ac-[Pen]-NTWN-[Pen]- [ Phe [4- (2-aminoethoxy) ]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1174);
Ac-[Pen]-NSWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1175);
Ac-[Pen]-N-[Aib]-WQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1176);
Ac-[Pen]-NTW-[Aib]-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]N-[Aib]-NH2(SEQ ID NO:1177);
Ac-[Pen]-QTW-[Lys(Ac)]-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1178);
Ac-[Pen]-[Lys(Ac)]-TWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]--[Aib]-[Lys(Ac)]NNNH2(SEQ ID NO:1179);
Ac-[Pen]-QVWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1180);
Ac-[Pen]-NT-[2-Nal]-Q-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1181);
Ac-[Pen]-NT-[1-Nal]-Q-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1182);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[α-MeLeu]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1183);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[α-MeLys]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1184);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1185);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[α-MeLeu]-[Lys(Ac)]-N-[βAla]-NH2(SEQ ID NO:1186);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[α-MeLys]-[Lys(Ac)]-N-[βAla]-NH2(SEQ ID NO:1187);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-[Lys(Ac)]-N-[βAla]-NH2(SEQ ID NO:1188);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-LN-NH2(SEQ ID NO:1189);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-GN-NH2(SEQ ID NO:1190);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-SN-NH2(SEQ ID NO:1191);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-[Aib]-N-NH2(SEQ ID NO:1192);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-FN-NH2(SEQ ID NO:1193);
Ac-[Pen]-NTW-[Cit]-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1194);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-[Tic]-[βAla]-NH2(SEQ ID NO:1195);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-[nLeu]-[βAla]-NH2(SEQ ID NO:1196);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-G-[βAla]-NH2(SEQ ID NO:1197);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-R-[βAla]-NH2(SEQ ID NO:1198);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]--[Aib]-[Lys(Ac)]-W-[βAla]-NH2(SEQ ID NO:1199);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]--[Aib]-[Lys(Ac)]-S-[βAla]-NH2(SEQ ID NO:1200);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]--[Aib]-[Lys(Ac)]-L-[βAla]-NH2(SEQ ID NO:1201);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]--[Aib]-[Lys(Ac)]-[AIB]-[βAla]-NH2(SEQ ID NO:1202);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]--[Aib]-[Lys(Ac)]-[N-MeAla]-[βAla]-NH2(SEQ ID NO:1203);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-[2-Nap]-[βAla]-NH2(SEQ ID NO:1204);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-F-[βAla]-NH2(SEQ ID NO:1205);
Ac-[(D)Arg]-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-[Lys(Ac)]NNNH2(SEQ ID NO:1206);
Biotin- [ PEG4]-cyclo [ [ Abu ]]-QTWQC]- [ Phe [4- (2-aminoethoxy) ]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN-NH2(SEQ ID NO:1269);
Ac-Ring [ [ Abu ]]-QTWQC]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1270);
Ac-[(D)Arg]-cyclo [ [ Abu ]]-QTWQC]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1271);
Ac-[(D)Arg]-cyclo [ [ Abu ]]-QTWQC]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1272);
Ac-E-[(D)Arg]-cyclo [ [ Abu ]]-QTWQC]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN-NH2(SEQ ID NO:1273);
Ac-[(D)Asp]-[(D)Arg]-cyclo [ [ Abu ]]-QTWQC]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN-NH2(SEQ ID NO:1274);
Ac-R-[(D)Arg]-cyclo [ [ Abu ]]-QTWQC]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN-NH2(SEQ ID NO:1275);
inoethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN-NH2(SEQ ID NO:1276);
Ac-F-[(D)Arg]-cyclo [ [ Abu ]]-QTWQC]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN-NH2(SEQ ID NO:1277);
Ac-[(D)Phe]-[(D)Arg]-cyclo [ [ Abu ]]-QTWQC]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN-NH2(SEQ ID NO:1278);
Ac-[2-Nal]-[(D)Arg]-cyclo [ [ Abu ]]-QTWQC]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN-NH2(SEQ ID NO:1279);
Ac-T-[(D)Arg]-cyclo [ [ Abu ]]-QTWQC]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN-NH2(SEQ ID NO:1280);
Ac-L-[(D)Arg]-cyclo [ [ Abu ]]-QTWQC]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN-NH2(SEQ ID NO:1281);
Ac-[(D)Gln]-[(D)Arg]-cyclo [ [ Abu ]]-QTWQC]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN-NH2(SEQ ID NO:1282);
Ac-[(D)Asn]-[(D)Arg]-cyclo [ [ Abu ]]-QTWQC]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN-NH2(SEQ ID NO:1283);
Ac-Ring [ [ Abu ]]-QTWQC]- [ Phe [4- (2-aminoethoxy) - (PEG4-Alexa488) ]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN-NH2(SEQ ID NO:1284);
[Alexa488]-[PEG4]-cyclo [ [ Abu ]]-QTWQC]- [ Phe [4- (2-aminoethoxy)]--[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN-NH2(SEQ ID NO:1285);
[Alexa647]-[PEG4]-cyclo [ [ Abu ]]-QTWQC]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN-NH2(SEQ ID NO:1286);
[Alexa-647]-[PEG4]-[(D)Arg]-cyclo [ [ Abu ]]-QTWQC]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1287);
[Alexa647]-[PEG12]-[(D)Arg]-cyclo [ [ Abu ]]-QTWQC]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-[Lys(Ac)]-NN-NH2(SEQ ID NO: 1288); and
[Alexa488]-[PEG4]-[(D)Arg]-cyclo [ [ Abu ]]-QTWQC]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1289),
Wherein the peptide inhibitor is cyclized via a disulfide bond between 2 Pen residues or by a thioether bond between an Abu and a Cys residue, and wherein the peptide inhibitor inhibits binding of interleukin-23 (IL-23) to an IL-23 receptor.
In particular embodiments, any of the peptide inhibitors described herein comprises one or more half-life extending moieties and/or one or more linker moieties conjugated to the peptide inhibitor. In particular embodiments, the half-life extending moiety is conjugated to the peptide inhibitor via one or more linker moieties.
In certain embodiments, any of the peptide inhibitors described herein further comprises a conjugated chemical substituent. In particular embodiments, the conjugated chemical substituent is a lipophilic substituent or a polymeric moiety, such as Ac, Palm, gamma Glu-Palm, isoGlu-Palm, PE G2-Ac, PEG 4-isoGlu-Palm, (PEG)5-Palm, succinic acid, glutaric acid, pyroglutamic acid, benzoic acid, IVA, octanoic acid, 1, 4-diaminobutane, isobutyl or biotin. In certain embodiments, the conjugated chemical substituent is polyethylene glycol having a molecular weight of 400Da to 40,000 Da.
In another aspect, the invention includes a peptide inhibitor comprising the structure of formula I:
R1-X-R2(I)
wherein
R1Is a bond, hydrogen, C1-C6 hydrocarbyl, C6-C12 aryl, C6-C12 aryl, C1-C6 hydrocarbyl, C1-C20 hydrocarboxyl, and includes pegylated forms of any of the foregoing either alone or as a spacer;
R2is a bond, OH or NH2(ii) a And
x is any peptide sequence described herein, e.g., Xa, I, Ia-It, II, IIa-IId, III, IIIa-IIIe, IV, IVa-IVb, V, or Va-Vh.
In a related aspect, the invention includes a peptide dimer inhibitor of an interleukin-23 receptor, wherein the peptide dimer inhibitor comprises two peptide monomer subunits connected via one or more linker moieties, wherein each peptide monomer subunit has a sequence or structure as set forth herein. In certain embodiments, one or both peptide monomer subunits are cyclized via an intramolecular bond between X4 and X9. In certain embodiments, one or both intramolecular bonds are a disulfide bond, a thioether bond, a lactam bond, a selenoether bond, a diselenide bond, or an alkene bond. In certain embodiments, the linker is any of those shown in table 2 or described herein. In certain embodiments, the linker moiety is a diethylene glycol linker, an iminodiacetic acid (IDA) linker, a β -Ala-iminodiacetic acid (β -Ala-IDA) linker, or a PEG linker. In particular embodiments, the N-termini of the individual peptide monomer subunits are joined by a linker moiety. In particular embodiments, the C-termini of the individual peptide monomer subunits are joined by a linker moiety. In certain embodiments, the linker links an internal amino acid residue in at least one peptide monomer subunit to an N-terminal, C-terminal, or internal amino acid residue of another peptide monomer subunit.
In other related aspects, the invention includes polynucleotides comprising sequences encoding one or both peptide monomer subunits of a peptide inhibitor of the invention or a peptide dimer inhibitor of the invention. The invention also includes vectors containing the polynucleotides.
In another aspect, the invention includes a pharmaceutical composition comprising a peptide inhibitor or peptide dimer inhibitor of the invention and a pharmaceutically acceptable carrier, excipient, or diluent. In a specific embodiment, the pharmaceutical composition comprises an enteric coating. In certain embodiments, the enteric coating protects and releases the pharmaceutical composition within the lower gastrointestinal system of the subject.
In another aspect, the invention includes a method of treating or preventing a disorder associated with IL-23 signaling in a subject, comprising providing to the subject an effective amount of a peptide inhibitor or pharmaceutical composition of the invention, including, but not limited to, Inflammatory Bowel Disease (IBD), ulcerative colitis, crohn's disease, celiac disease (non-tropical sprue), enteropathy associated with seronegative arthropathy, microscopic colitis, collagenous colitis, eosinophilic gastroenteritis, colitis associated with radiotherapy or chemotherapy, colitis associated with innate immune disorders (such as leukoadhesion deficiency type 1), chronic granulomatosis, colon storage disease type 1b, Hermansky-Pudlak syndrome, Chediak-Higashi syndrome and Wiskott-Aldrich syndrome, pouchitis, pouch inflammation after proctolectomy and ileoanal anastomosis, Gastrointestinal cancer, pancreatitis, insulin-dependent diabetes mellitus, mastitis, cholecystitis, cholangitis, peribiliary inflammation, chronic bronchitis, chronic sinusitis, asthma, psoriasis, or graft-versus-host disease. In certain embodiments, the inflammatory bowel disease is ulcerative colitis or crohn's disease. In particular embodiments, the peptide inhibitor or peptide dimer inhibitor inhibits the binding of interleukin-23 (IL-23) to an interleukin-23 receptor (IL-23R). In certain embodiments, the pharmaceutical composition is provided to the subject by an oral route of administration, an intravenous route of administration, a peritoneal route of administration, an intradermal route of administration, a subcutaneous route of administration, an intramuscular route of administration, an intrathecal route of administration, an inhalation route of administration, a vaporization route of administration, a nebulization route of administration, a sublingual route of administration, an oral route of administration, a parenteral route of administration, a rectal route of administration, an intraocular route of administration, an inhalation route of administration, a vaginal route of administration, or a topical route of administration. In particular embodiments, the pharmaceutical composition is provided orally for the treatment of Inflammatory Bowel Disease (IBD), ulcerative colitis, crohn's disease. In certain embodiments, the pharmaceutical composition is provided to the subject topically, parenterally, intravenously, subcutaneously, peritoneally, or intravenously for the treatment of psoriasis.
Brief Description of Drawings
FIG. 1 provides an example of a rat IL-23 dose response curve as measured by IL-17A levels in a rat splenocyte assay.
Figure 2 is a graph showing IL-12 dependent production of IFN γ from human PBMCs treated with the indicated amounts of compound a or compound B.
Fig. 3 shows the results of DAI values from day 7. Statistical significance analysis was determined using student's t-test (GraphPad Prism). Differences were recorded as significant, p <0.05, p <0.01, p <0.001, p < 0.0001.
FIG. 4 shows an alignment of the amino acid sequences of human IL23R, mouse IL-23R, rat IL23R, chimpanzee IL-23R, canine IL-23R, and bovine IL-23R, with highly conserved amino acid residues shaded. The region of mouse IL-23R that is not present in the other species IL-23R shown, as well as the region of IL23R that may be bound by certain peptide inhibitors of the invention, are indicated by dashed lines.
Figure 5 is a table summarizing the study design for TNBS-induced colitis in rats.
Fig. 6A-6D are graphs showing colon weight/length (fig. 6A), colon wall thickness (fig. 6B), colon macroscopic score (fig. 6C) or Myeloperoxidase (MPO) abundance in proximal colon extracts quantified by ELISA after sham treatment, vehicle treatment or treatment with the indicated amount of anti-IL 23p19 antibody or compound C. Values are shown as mean ± SD. Statistical significance was assessed by one-way ANOVA: 0.05; 0.01; p < 0.001; p < 0.0001; ns, not significant.
Figure 7 provides micrographs of colon lesions (showing transmural inflammation, presence of necrotic tissue, and crypts without mucosa) present in animals after sham treatment (top left panel), vehicle treatment (top right panel), anti-IL 23p19 antibody treatment (bottom left panel), or 160mg/kg/d compound C treatment (bottom right panel) (showing lesions limited to mucosa).
Fig. 8A-8E are graphs showing inflammation after vehicle treatment, treatment with anti-IL 23p19 antibody, or treatment with a specified amount of compound C (fig. 8A), mucosal necrosis (fig. 8B), gland loss (fig. 8C), colon wall thickness (fig. 8D), and histological score (fig. 8E).
Figure 9 shows the concentration of compound C in plasma and proximal colon determined one hour after the last PO dose (left panel), and the fold of IC75 greater than its activity as determined by rat splenocyte assay (middle panel) and rat IL23R ELISA assay (right panel).
Figure 10 provides a schematic depicting the structure of certain peptide inhibitors and illustrating representative types of bonds between X4 and X9.
FIGS. 11A-11E show pharmacokinetic data for IL-23R peptide inhibitor peptide 993(SEQ ID NO: 993). Figure 11A shows the concentration of peptide 993 (nM) in plasma measured at various time points up to 24 hours after oral administration of peptide 993. FIGS. 11B-11D show the concentration (nM) of peptide 993 in samples taken from Peyer's Patch (FIG. 11B), small intestine (FIG. 11C) and colon (FIG. 11D). The dotted line indicates 350 mM. Fig. 11E shows the amount of peptide 993 (% dose) detected in feces 24 hours after oral administration.
Figures 12A-12D summarize experiments comparing systemic treatment with prednisolone (prodnisolone) or anti-IL-23 p19 neutralizing antibodies to treatment by oral administration of peptide 993 in the TNBS model of acute colitis. Figure 12A shows the body weight change (percentage) from day 0 to day 7 in sham-operated, vehicle and peptide 993-treated rats. FIG. 12B shows the ratio of colon weight to colon length (in mg/cm) of the colon harvested from rats on day 7. Fig. 12C shows colon macroscopic scores of the colon harvested from rats on day 7. Figure 12D shows the sum of histopathological scores of the colon taken from sham-operated, vehicle and peptide 993-treated rats. For all experiments, statistical comparisons between groups were performed using one-way anova, followed by post-hoc tests: p < 0.05; p < 0.01; p < 0.001; p < 0.0001; ns, not significant.
Figures 13A-13C summarize experiments comparing systemic treatment with prednisolone or an anti-IL-23 p19 neutralizing antibody to treatment by oral administration of peptide 1185 in a TNBS model of acute colitis. Figure 13A shows the body weight change (in percent) from day 0 to day 7 for sham-operated, vehicle and peptide 1118-treated rats. FIG. 13B shows the ratio of colon weight to colon length (in mg/cm) of the colon harvested from rats on day 7. Fig. 13C shows colon macroscopic scores of colon harvested from rats on day 7. For all experiments, statistical comparisons between groups were performed with one-way anova followed by post-hoc tests: p < 0.05; p < 0.01; p < 0.001; p < 0.0001; ns, not significant.
Figures 14A-14D summarize experiments comparing systemic treatment with prednisolone or an anti-IL-23 p19 neutralizing antibody to treatment by oral administration of peptide 980 in the TNBS model of acute colitis. Figure 14A shows the body weight change (in percent) from day 0 to day 7 in sham-operated, vehicle and peptide 980-treated rats. FIG. 14B shows the ratio of colon weight to colon length (in mg/cm) of the colon harvested from rats on day 7. Fig. 14C shows colon macroscopic scores of the colon harvested from rats on day 7. Fig. 14D shows the sum of histopathological scores taken from the colon of sham-operated, vehicle and peptide 980-treated rats. For all experiments, statistical comparisons between groups were performed using one-way anova followed by post-hoc tests: p < 0.05; p < 0.01; p < 0.001; p < 0.0001; ns, not significant.
Figures 15A-15E show the levels of disease and biomarkers for IL-23 measured in the colon of rats in the sham (not TNBS exposed) experimental group or the TNBS exposed experimental group receiving vehicle or peptide 993 treatment. Data for MPO (FIG. 15A), IL-6 (FIG. 15B), IL-1 β (FIG. 15C), IL-22 (FIG. 15D), and IL-17A (FIG. 15E) are shown. For all experiments, statistical comparisons between groups were performed using one-way anova followed by post-hoc tests: p < 0.05; p < 0.01; p < 0.001; p < 0.0001; ns, not significant.
Figures 16A-16B show the levels of disease and biomarkers for IL-23 measured in the colon of rats in the sham (not TNBS exposed) experimental group or the TNBS exposed experimental group receiving vehicle or peptide 980 treatment. Data for MPO (FIG. 16A) and IL-22 (FIG. 16B) are shown. For all experiments, statistical comparisons between groups were performed using one-way anova followed by post-hoc tests: p < 0.05; p < 0.01; p < 0.001; p < 0.0001; ns, not significant.
FIGS. 17A-17D show Schild analysis of inhibitor peptides. Figure 17A shows a graph depicting the% Emax response as a function of increasing IL-23 concentration in the presence of peptide 993 at concentrations: 0nM (filled circles), 0.3nM (filled squares), 1nM (triangles), 3nM (inverted triangles), 10nM (diamonds), 30nM (open circles) or 100nM (open squares). The properties of the curves are shown in the graph. Figure 17B depicts results from the same set of experiments and shows that there is a Log (dose ratio) displayed on a logarithmic scale as a function of peptide 993 concentration (M)-1) A graph of (a). The properties of the linear function generated are shown below the graph. Fig. 17C shows a graph depicting SEQ ID NOs: graph of% Emax response as a function of increasing IL-23 concentration in the presence of peptide 1169: 0nM (filled circles), 0.3nM (filled squares), 1nM (triangles), 3nM (inverted triangles), 10nM (diamonds), 30nM (open circles) or 100nM (open squares). The properties of the curves are shown in the graph. Fig. 17D shows the sequences depicted with SEQ ID NOs: 1211 as a graph of% Emax response as a function of increasing IL-23 concentration in the presence of the peptide: 0nM (filled circles), 0.3nM (filled squares), 1nM (triangles), 3nM (inverted triangles), 10nM (diamonds), 30nM (open circles) or 100nM (open squares). The properties of the curves are shown in the graph.
FIGS. 18A-18B show pharmacokinetic data for IL-23R peptide inhibitor peptide 1185. Figure 18A shows the concentration of peptide 1185 in plasma and in samples taken from the small intestine and colon. Fig. 18B shows the amount of peptide 1185 (% dose) detected in urine and feces 24 hours after oral administration.
Fig. 19A and 19B show pharmacokinetic data for IL-23R peptide inhibitor peptide 980. Fig. 19A shows the concentration of peptide 980 in plasma and in samples taken from the small intestine and colon. Figure 19B shows the amount of peptide 980 detected in urine and feces (% dose) 24 hours after oral administration.
Detailed Description
Unless defined otherwise herein, scientific and technical terms used in the present application shall have the meanings that are commonly understood by those of ordinary skill in the art. Generally, the nomenclature used in connection with, and the techniques of, chemistry, molecular biology, cell and cancer biology, immunology, microbiology, pharmacology, and protein and nucleic acid chemistry described herein are those well known and commonly used in the art.
As used herein, the following terms have the meanings assigned to them unless otherwise indicated.
Throughout this specification the word "comprise", or variations such as "comprises" or "comprising", will be understood to imply the inclusion of a stated integer (or component) or group of integers (or components) but not the exclusion of any other integer (or component) or group of integers (or components).
The singular forms "a", "an" and "the" include plural referents unless the context clearly dictates otherwise.
The term "including" is used to mean "including but not limited to". "include" and "include but are not limited to" are used interchangeably.
The terms "patient," "subject," and "individual" are used interchangeably and refer to a human or non-human animal. These terms include mammals, such as humans, primates, livestock animals (e.g., bovines, porcines), companion animals (e.g., canines, felines), and rodents (e.g., mice and rats).
The term "peptide" as used herein refers broadly to a sequence of two or more amino acids linked together by peptide bonds. It is understood that the term does not imply a particular length of the amino acid polymer nor is it intended to imply or distinguish whether the polypeptide is produced using recombinant techniques, chemical synthesis or enzymatic synthesis, or is naturally occurring.
The expression "sequence identity", "percent homology" or, for example, a sequence comprising "identity with … 50% refers to the degree of identity over the comparison window based on the sequence of nucleotides to nucleotides or amino acids to amino acids. Thus, "percent sequence identity" can be calculated as follows: the two optimally aligned sequences are compared over a comparison window, the number of positions of the same nucleic acid base (e.g., A, T, C, G, I) or the same amino acid residue (e.g., Ala, Pro, Ser, Thr, Gly, Val, Leu, Ile, Phe, Tyr, Trp, Lys, Arg, His, Asp, Glu, Asn, Gln, Cys, and Met) that occur in both sequences is determined to yield the number of matched positions, the number of matched positions is divided by the total number of positions in the comparison window (i.e., the window size), and the result is multiplied by 100 to yield the percentage of sequence identity. Calculation of sequence similarity or sequence identity between sequences (the terms are used interchangeably herein) can be performed as follows. To determine the percent identity of two amino acid sequences or two nucleic acid sequences, the sequences can be aligned for optimal comparison purposes (e.g., gaps can be introduced in one or both of the first and second amino acid or nucleic acid sequences for optimal alignment; non-homologous sequences can be disregarded for comparison purposes). In certain embodiments, the length of a reference sequence aligned for comparison purposes is at least 30%, preferably at least 40%, more preferably at least 50%, 60%, and even more preferably at least 70%, 80%, 90%, 100% of the length of the reference sequence. The amino acid residues or nucleotides at the corresponding amino acid positions or nucleotide positions are then compared. When a position in the first sequence is occupied by the same amino acid residue or nucleotide as the corresponding position in the second sequence, then the molecules are identical at that position.
The percent identity between two sequences is a function of the number of identical positions that the sequences share (taking into account the number of gaps that must be introduced for optimal alignment of the two sequences and the length of each gap).
Sequence comparison and determination of percent identity between two sequences can be accomplished using mathematical algorithms. In some embodiments, the percent identity between two amino acid sequences is determined using the Needleman and Wunsch algorithm (1970, J.mol.biol.48: 444-. In yet another preferred embodiment, the percent identity between two nucleotide sequences is determined using the GAP program in the GCG software package using the nwsgapdna. cmp matrix with GAP weights of 40, 50, 60, 70 or 80 and length weights of 1, 2, 3, 4, 5 or 6. Another exemplary parameter set includes a Blossum 62 scoring matrix, a gap penalty of 12, a gap extension penalty of 4, and a frameshift gap penalty of 5. Percent identity between two amino acid or nucleotide sequences can also be determined using the e.meyers and w.miller algorithms (1989, cab, 4:11-17) that have been incorporated into the ALIGN program (version 2.0), using a PAM120 weight residue table, a gap length penalty of 12, and a gap penalty of 4. The peptide sequences described herein can be used as "query sequences" to search public databases, for example, to identify other family members or related sequences. Such searches can be performed using the NBLAST and XBLAST programs (version 2.0) of Altschul et al (1990, J.mol.biol., 215: 403-10). A BLAST nucleotide search can be performed using the NBLAST program with a score of 100 and a word length of 12 to obtain nucleotide sequences homologous to the nucleic acid molecules of the invention. BLAST protein searches can be performed using the XBLAST program with a score of 50 and a word length of 3 to obtain amino acid sequences homologous to the protein molecules of the invention. To obtain gap alignments for comparison purposes, Gapped BLAST as described in Altschul et al (Nucleic Acids Res.25:3389-3402,1997) can be used. When BLAST and Gapped BLAST programs are employed, the default parameters of the respective programs (e.g., XBLAST and NBLAST) can be used. The term "conservative substitution" as used herein denotes the replacement of one or more amino acids by another biologically similar residue. Examples include substitution of amino acid residues having similar properties, for example, small amino acids, acidic amino acids, polar amino acids, basic amino acids, hydrophobic amino acids, and aromatic amino acids. See, for example, the following table. In some embodiments of the invention, one or more Met residues are substituted with norleucine (Nle), which is a bioisostere of Met, but which, in contrast to Met, is not readily oxidized. Another example of a conservative substitution with a residue not normally found in endogenous mammalian peptides and proteins is the conservative substitution of Arg or Lys with, for example, ornithine, canavanine, aminoethylcysteine or another basic amino acid. In some embodiments, one or more cysteines in the peptide analogs of the present invention may be substituted with another residue, such as a serine. For additional information on phenotypically silent substitutions in peptides and proteins, see, e.g., Bowie et al, Science 247, 1306-. In the scheme below, conservative substitutions of amino acids are grouped by physicochemical properties. I: neutral, hydrophilic, II: acids and amides, III: basic, IV: hydrophobic, V: aromatic bulky amino acids.
I II III IV V
A N H M F
S D R L Y
T E K I W
P Q V
G C
In the scheme below, conservative substitutions of amino acids are grouped by physicochemical properties. VI: neutral or hydrophobic, VII: acidic, VIII: basic, IX: polar, X: is aromatic.
VI VII VIII IX X
A E H M F
L D R S Y
I K T W
P C
G N
V Q
The term "amino acid" or "any amino acid" as used herein refers to any and all amino acids, including naturally occurring amino acids (e.g., a-amino acids), unnatural amino acids, modified amino acids, and unnatural amino acids. Which include D-amino acids and L-amino acids. Natural amino acids include those occurring in nature, such as, for example, 23 amino acids that combine into a peptide chain to form a building block for a large number of proteins. These amino acids are mainly the L stereoisomers, although a few D-amino acids are present in the bacterial envelope and some antibiotics. The 20 "standard" natural amino acids are listed in the table above. The "nonstandard" natural amino acid is pyrrolysine (present in methanogenesisIn many non-eukaryotic organisms and in most eukaryotic organisms), selenocysteine (present in many non-eukaryotic organisms as well as in most eukaryotic organisms) and N-formylmethionine (encoded by the start codon AUG in bacteria, mitochondria and chloroplasts). An "Unnatural" or "non-natural" amino acid is a non-proteinogenic amino acid (i.e., one that is not naturally encoded or not present in the genetic code), either naturally occurring or chemically synthesized. Over 140 unnatural amino acids are known, and many thousands of more combinations are possible. Examples of "unnatural" amino acids include beta-amino acids (β) 3And beta2) Homo-amino acids, proline and pyruvate derivatives, 3-substituted alanine derivatives, glycine derivatives, ring-substituted phenylalanine and tyrosine derivatives, linear core amino acids, diamino acids, D-amino acids, alpha-methyl amino acids and N-methyl amino acids. Unnatural or unnatural amino acids also include modified amino acids. "modified" amino acids include amino acids that have been chemically modified to include groups or chemical moieties that are not naturally present on the amino acid (e.g., natural amino acids). According to certain embodiments, the peptide inhibitor comprises an intramolecular bond between two amino acid residues present in said peptide inhibitor. It is understood that the amino acid residues forming the bond are slightly altered when bonded to each other compared to when not bonded to each other. Reference to a particular amino acid is meant to include the amino acid in its non-bonded and bonded states. For example, when homoserine (hSer) or homoserine (Cl) of the amino acid residue in a non-bonded form participates in intramolecular linkage according to the present invention, 2-aminobutyric acid (Abu) form may be used. The invention includes peptide inhibitors that contain cross-links between X4 and X9, as well as peptide inhibitors that do not contain cross-links between X4 and X9, e.g., prior to cross-link formation. Thus, the names hSer and Abu are intended to represent the same amino acid and may be used interchangeably.
For the most part, the names of naturally occurring and non-naturally occurring aminoacyl residues as used herein follow the Nomenclature convention proposed by the IUPAC Commission on organic chemistry Nomenclature and the IUPAC-IUB Commission on Biochemistry Nomenclature, as described in "Nomenclature of α -Amino Acids (Nomenclature of α -Amino Acids) (Recommendations, 1974)" Biochemistry,14(2), (1975). If the names and degrees of abbreviations for amino acids and aminoacyl residues used in this specification and the appended claims are different from those indicated, it will be clear to the reader. Some abbreviations used to describe the invention are defined in table 1A below.
TABLE 1A. abbreviations for unnatural amino acids and chemical moieties (for amino acid derivatives, all L, unless specified)
Figure GDA0003395945150000301
Figure GDA0003395945150000311
Figure GDA0003395945150000321
Figure GDA0003395945150000331
Figure GDA0003395945150000341
Figure GDA0003395945150000351
Figure GDA0003395945150000361
Throughout the specification, unless naturally occurring amino acids are referred to by their full name (e.g., alanine, arginine, etc.), they are designated by conventional three-letter or one-letter abbreviations (e.g., Ala or a for alanine; Arg or R for arginine, etc.). Unless otherwise indicated, the three-letter and one-letter abbreviations for amino acids refer to the L-isomeric forms of the amino acids in question. The term "L-amino acid" as used herein refers to the "L" isomeric form of the peptide, and conversely, the term "D-amino acid" refers to the "D" isomeric form of the peptide (e.g., Dasp, (D) Asp or D-Asp; Dphe, (D) Phe or D-Phe). The amino acid residue of the D isomeric form may be substituted for any L-amino acid residue, as long as the peptide retains the desired function. When referred to using the single letter abbreviations, D-amino acids may be conventionally represented by lower case letters.
In the case of unusual amino acids or non-naturally occurring amino acids, unless they are referred to by their full name (e.g., sarcosine, ornithine, etc.), for their residues, the commonly used three-character code or four-character code is used, including, sar or Sarc (sarcosine, i.e., N-methylglycine), Aib (α -aminoisobutyric acid), Dab (2, 4-diaminobutyric acid), Dapa (2, 3-diaminopropionic acid), γ -Glu (γ -glutamic acid), Gaba (γ -aminobutyric acid), β -Pro (pyrrolidine-3-carboxylic acid) and 8Ado (8-amino-3, 6-dioxaoctanoic acid), Abu (2-aminobutyric acid), β hPro (β -homoproline), β hPhe (β -homophenylalanine) and Bip (β, β diphenylalanine) as well as Ida (iminodiacetic acid).
It will be clear to the skilled person that the peptide sequences disclosed herein are shown from left to right, with the left end of the sequence being the N-terminus of the peptide and the right end of the sequence being the C-terminus of the peptide. The sequences disclosed herein incorporate a "Hy-" moiety at the amino terminus (N-terminus) of the sequence and an "-OH" moiety or "-NH" moiety at the carboxy terminus (C-terminus) of the sequence2"partial sequence. In such cases, and unless otherwise indicated, the "Hy-" portion of the N-terminus of the sequence in question represents a hydrogen atom, corresponding to the presence of a free primary or secondary amino group at the N-terminus, while the "-OH" or "-NH" at the C-terminus of the sequence 2The "moieties" independently represent hydroxy or amino, corresponding to the C-terminal amido group (CONH)2) Is present. In each of the sequences of the present invention, a C-terminal "-OH" moiety may be substituted for the C-terminal "-NH moiety2"part of, and vice versa.
The term "DRP" as used herein refers to disulfide-rich peptides.
The term "dimer" as used herein broadly refers to a peptide comprising two or more monomeric subunits. Some dimers contain two DRPs. Dimers of the invention include homodimers and heterodimers. The monomeric subunits of the dimer may be linked at their C-terminus or N-terminus, or they may be linked via internal amino acid residues. The monomer subunits of the dimer may be linked by the same site, or each may be linked by a different site (e.g., C-terminal, N-terminal, or internal site).
The term "NH" as used herein2"may refer to a free amino group present at the amino terminus of a polypeptide. The term "OH" as used herein may refer to the free carboxyl group present at the carboxyl terminus of a peptide. In addition, the term "Ac" as used herein refers to acetyl protection formed by acylation of the C-terminus or N-terminus of the polypeptide. In certain of the peptides shown herein, NH2The C-terminus of the peptide represents an amino group.
The term "carboxy" as used herein refers to-CO2H。
The term "isostere substitute" as used herein refers to any amino acid or other analog moiety having similar chemical and/or structural properties as the specified amino acid.
The term "cyclization" as used herein refers to a reaction wherein: wherein a portion of a polypeptide molecule is linked to another portion of the polypeptide molecule to form a closed loop, such as by formation of a disulfide bridge or other similar bond.
The term "subunit" as used herein refers to one of a pair of polypeptide monomers that are joined to form a dimeric peptide composition.
The term "linker moiety" as used herein broadly refers to a chemical structure capable of linking or joining two peptide monomer subunits together to form a dimer.
The term "pharmaceutically acceptable salt" as used herein means a salt or zwitterionic form of the peptide or compound of the invention, which is water or oil soluble or dispersible, which is suitable for the treatment of disease without undue toxicity, irritation and allergic response; which is commensurate with a reasonable benefit/risk ratio, and which is effective for their intended use. The salts may be prepared during the final isolation and purification of the compounds or separately by reacting the amino group with a suitable acid. Representative acid addition salts include acetate, adipate, alginate, citrate, aspartate, benzoate, benzenesulfonate, bisulfate, butyrate, camphorate, camphorsulfonate, digluconate, glycerophosphate, hemisulfate, heptanoate, hexanoate, formate, fumarate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate (isethionate), lactate, maleate, mesitylenesulfonate, methanesulfonate, naphthalenesulfonate, nicotinate, 2-naphthalenesulfonate, oxalate, pamoate, pectate, persulfate, 3-phenylpropionate, picrate, pivalate, propionate, succinate, tartrate, trichloroacetate, trifluoroacetate, phosphate, glutamate, bicarbonate, trifluoroacetate, hydrochloride, or mixture thereof, P-toluenesulfonate and undecanoate. Also, the amino groups in the compounds of the present invention may be quaternized with: methyl, ethyl, propyl and butyl chlorides, bromides and iodides; dimethyl, diethyl, dibutyl and diamyl sulfates; chlorides, bromides and iodides of decyl, dodecyl, tetradecyl and sterol groups; and benzyl and phenethyl bromides. Examples of acids that may be used to form therapeutically acceptable addition salts include inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid and phosphoric acid, and organic acids such as oxalic acid, maleic acid, succinic acid and citric acid. The pharmaceutically acceptable salt may suitably be, for example, a salt selected from acid addition salts and base salts. Examples of acid addition salts include chloride salts, citrate salts, and acetate salts. Examples of basic salts include those wherein: wherein the cation is selected from alkali metal cations such as sodium or potassium, alkaline earth metal cations such as calcium or magnesium, and substituted ammonium ions, such as ions of the type N (R1) (R2) (R3) (R4) + wherein R1, R2, R3 and R4 are generally independently designated as hydrogen, optionally substituted C1-6-alkyl or optionally substituted C2-6-alkenyl. Examples of related C1-6-alkyl groups include methyl, ethyl, 1-propyl and 2-propyl. Examples of C2-6-alkenyl groups which may be relevant include ethenyl, 1-propenyl and 2-propenyl. Other examples of pharmaceutically acceptable salts are described in Remington's Pharmaceutical Sciences, 17 th edition, Alfonso R.Gennaro (Ed.), Mark Publishing Company, Easton, PA, USA,1985 (and later versions thereof), in encyclopedia of Pharmaceutical Technology, 3 rd edition, James Swarbrick (Ed.), Informata Healthcare USA (Inc.), NY, USA,2007, and in J.Pharm.Sci.66:2 (1977). Also, for an overview of suitable Salts, see Handbook of Pharmaceutical Salts, Properties, Selection, and Use, by Stahl and Wermuth (Wiley-VCH, 2002). Other suitable base salts are made from bases which form non-toxic salts. Representative examples include aluminum, arginine, benzathine, calcium, choline, diethylamine, diethanolamine, glycinate, lysine, magnesium, meglumine, ethanolamine, potassium, sodium, tromethamine and zinc salts. Hemisalts of acids and bases, for example, hemisulfate and hemicalcium salts, may also be formed.
The term "N (α) methylation" as used herein describes the methylation of the α amine of an amino acid, also commonly referred to as N-methylation.
The term "symmetric methylation" or "Arg-Me-sym" as used herein describes the symmetric methylation of the two nitrogens of the guanidino group of arginine. In addition, the term "asymmetric methylation" or "Arg-Me-asym" describes the methylation of a single nitrogen of the guanidino group of arginine.
The term "acylated organic compound" as used herein refers to a variety of compounds having a carboxylic acid functional group that are used to acylate the N-terminus of an amino acid or a monomer or dimer (e.g., a monomer subunit prior to formation of a C-terminal dimer). Non-limiting examples of acylated organic compounds include cyclopropaneacetic acid, 4-fluorobenzoic acid, 4-fluorobenzeneacetic acid, 3-phenylpropionic acid, succinic acid, glutaric acid, cyclopentanecarboxylic acid, 3,3, 3-trifluoropropionic acid, 3-fluoromethylbutyric acid, tetrahydro-2H-pyran-4-carboxylic acid.
The term "hydrocarbon group" includes straight or branched, acyclic or cyclic saturated aliphatic hydrocarbons containing from 1 to 24 carbon atoms. Representative saturated straight chain hydrocarbyl groups include, but are not limited to, methyl, ethyl, n-propyl, n-butyl, n-pentyl, n-hexyl, and the like, while saturated branched chain hydrocarbyl groups include, but are not limited to, isopropyl, sec-butyl, isobutyl, tert-butyl, isopentyl, and the like. Representative saturated cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like, while unsaturated cycloalkyl groups include, but are not limited to, cyclopentenyl, cyclohexenyl, and the like.
The term "mammal" refers to any mammalian species, such as human, mouse, rat, dog, cat, hamster, guinea pig, rabbit, livestock, and the like.
As used herein, a "therapeutically effective amount" of a peptide inhibitor of the present invention is intended to describe a sufficient amount of a peptide inhibitor to treat an IL-23/IL-23R-associated disease (e.g., to reduce inflammation associated with IBD), including, but not limited to, any of the diseases and disorders described herein. In particular embodiments, a therapeutically effective amount will achieve the desired benefit/risk ratio applicable to any medical treatment.
An "analogue" of an amino acid, for example a "Phe analogue" or a "Tyr analogue", means an analogue of the amino acid in question. A variety of amino acid analogs are known and available in the art, including Phe and Tyr analogs. In certain embodiments, an amino acid analog, e.g., a Phe analog or a Tyr analog, comprises one, two, three, four, or five substitutions relative to Phe or Tyr, respectively. In certain embodiments, the substitution is in a side chain of the amino acid. In certain embodiments, the Phe analog has the structure Phe (R)2) Wherein R is2Is Hy, OH, CH3、CO2H、CONH2、CONH2OCH2CH2NH2、t-Bu、OCH2CH2NH2Phenoxy, OCH 3O allyl, Br, Cl, F, NH2N3 or guanidino. In certain embodiments, R2Is CONH2OCH2CH2NH2、OCH3、CONH2、OCH3Or CO2H. Examples of Phe analogs include, but are not limited to: hPhe, Phe (4-OM)e) alpha-Me-Phe, hPhe (3, 4-dimethoxy), Phe (4-CONH)2) Phe (4-phenoxy), Phe (4-guanidino), Phe (4-tBu), Phe (4-CN), Phe (4-Br), Phe (4-OBzl), Phe (4-NH)2)、BhPhe(4-F)、Phe(4-F)、Phe(3,5DiF)、Phe(CH2CO2H)、Phe(5-F)、Phe(3,4-Cl2)、Phe(3,4-F2)、Phe(4-CF3) Beta-bis PheAla, Phe (4-N)3) Phe [4- (2-aminoethoxy)]4-Phenylbenzylalanine, Phe (4-CONH)2) Phe (3, 4-dimethoxy), Phe (4-CF)3)、Phe(2,3-Cl2) And Phe (2, 3-F)2). Examples of Tyr analogs include, but are not limited to: hTyr, N-Me-Tyr, Tyr (3-tBu), Tyr (4-N)3) And β hTyr.
Peptide inhibitors of IL-23R
Genome-wide association studies (GWAS) have demonstrated an important association of the IL-23 receptor (IL-23R) gene with Inflammatory Bowel Disease (IBD), suggesting that perturbation of IL-23 signaling may be involved in the pathogenesis of the disease. The present invention provides compositions and methods for modulating the IL-23 pathway by selectively antagonizing IL-23R via oral treatment with peptides that are stable and localized to Gastrointestinal (GI) tissue. Novel inhibitory peptides that are uniquely resistant to oxidation/reduction conditions and proteolytic degradation in a variety of assays that mimic multiple compartments of the GI environment were identified. Functionally, these peptides efficiently neutralize IL-23 mediated signal transduction in transformed human cell lines and human primary cells. Binding to IL-23R is selective, because the peptide does not block the IL-6 and IL-6R interaction or antagonize IL-12 signal transduction pathway. Moreover, these orally delivered peptides were effective in attenuating colitis in a rat model of 2,4, 6-trinitrobenzenesulfonic acid (TNBS) induced acute IBD, as shown by the significant reduction in colon weight-to-length ratio, colon macroscopic score, neutrophil infiltration, and histopathology, which was comparable to the control anti-IL-23 p19 mAb.
The present invention relates generally to peptides having IL-23R antagonist activity, including peptide monomers and peptide dimers. In certain embodiments, the invention demonstrates a novel paradigm for treating IBD and other diseases and disorders by oral delivery of antagonists of IL-23. IBD represents local inflammation of intestinal tissue; thus, when compared to systemic methods, beneficial therapeutic agents will start acting from the luminal side of the intestine, resulting in high drug concentrations in the diseased tissue, minimizing systemic availability and resulting in improved efficacy and safety. Oral administration of the compounds of the present invention is expected to maximize drug levels in diseased intestinal tissue while limiting circulating drug concentrations, thereby providing effective, safe and long-lasting delivery for life-long treatment of IBD and other diseases and disorders.
In certain embodiments, the invention relates to various peptides or peptide dimers comprising heteromonomers or homomonomelic subunits that form cyclized structures via disulfide or other bonds. In certain embodiments, the disulfide or other bond is an intramolecular bond. The cyclized structures of the monomer subunits of peptide monomer inhibitors and peptide dimer inhibitors have been shown to increase the potency and selectivity of the peptide inhibitors. In certain embodiments, the peptide dimer inhibitor may comprise one or more intermolecular bonds within the peptide dimer inhibitor that connect two monomeric peptide subunits, e.g., an intermolecular bridge between two cysteine residues (one in each peptide monomeric subunit).
The present invention provides peptide inhibitors that bind to IL-23R, which may be monomeric or dimeric. In a specific embodiment, the peptide inhibitor inhibits the binding of IL-23 to IL-23R. In certain embodiments, IL-23R is human IL-23R, and IL-23 is human IL-23. In certain embodiments, the peptide inhibitors of the invention reduce binding of IL-23 to IL-23R by at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, or at least 90% as compared to a negative control peptide. Methods of determining binding are known in the art and include ELISA assays, as described in the accompanying examples.
In certain embodiments, for example, for inhibiting binding of IL-23 to IL-23R (e.g., human IL-23 and human IL-23R), the IC50 of the peptide inhibitors of the invention is >1mM, <1nM, 500nM to 1000nM, <500nM, <250nM, <100nM, <50nM, <25nM, <10nM, <5nM, <2nM, <1nM, or <5 mM. Methods of determining activity are known in the art and include any of those described in the accompanying examples.
In certain embodiments, the peptide inhibitors of the invention have increased stability, increased gastrointestinal stability, or increased stability in Simulated Intestinal Fluid (SIF) or Simulated Gastric Fluid (SGF) and/or under redox conditions (e.g., DTT) relative to control peptides. In certain embodiments, the control peptide is an unrelated peptide of the same or similar length. In particular embodiments, the control peptide is a peptide having the same or highly related amino acid sequence (e.g., > 90% sequence identity) as the peptide inhibitor. In particular embodiments, the control peptide is a peptide having the same or highly related amino acid sequence (e.g., > 90% sequence identity) as the peptide inhibitor, but which does not have a cyclized structure, e.g., formed by an intramolecular bond between two amino acid residues therein, or which is not dimerized, or which does not comprise a conjugate for stabilization. In particular embodiments, the only difference between the peptide inhibitor and the control peptide is that the peptide inhibitor comprises a substitution of one or more amino acids that introduces one or more amino acid residues into the peptide inhibitor, wherein the introduced amino residue forms an intra-sulfide disulfide bond or a thioether bond with another amino acid residue in the peptide inhibitor. An example of a control for a peptide dimer inhibitor is a monomer having the same sequence as one of the monomer subunits present in the peptide dimer inhibitor. An example of a control comprising a peptide inhibitor of a conjugate is a peptide having the same sequence but not comprising a conjugate moiety. In certain embodiments, the control peptide is a peptide (e.g., a naturally occurring peptide) corresponding to a region of IL-23 that binds to IL-23R.
Methods for determining the stability of peptides are known in the art. In certain embodiments, the stability of the peptide inhibitor is determined using SIF analysis, e.g., as described in example 3. In certain embodiments, the stability of a peptide inhibitor is determined using an SGF assay, e.g., as described in example 3. In particular embodiments, when the peptide inhibitor is exposed to SIF or SGF or DTT, it has a half-life (e.g., in SIF or SGF or DTT) of greater than 1 minute, greater than 10 minutes, greater than 20 minutes, greater than 30 minutes, greater than 60 minutes, greater than 90 minutes, greater than 120 minutes, greater than 3 hours, or greater than 4 hours under a given set of conditions (e.g., temperature). In certain embodiments, the temperature is about 25 ℃, about 4 ℃ or about 37 ℃, and the pH is physiological pH, or the pH is about 7.4.
In some embodiments, the half-life is measured in vitro using any suitable method known in the art, e.g., in some embodiments, the stability of a peptide of the invention is determined by incubating the peptide with pre-warmed human serum (Sigma) at 37 ℃. Samples were taken at different time points, typically up to 24 hours, and the stability of the samples was analyzed by separating the peptides or peptide dimers from the serum proteins, and then analyzing for the presence of the peptide or peptide dimer of interest using LC-MS.
In some embodiments, the peptide inhibitors of the invention exhibit improved solubility or improved aggregation properties compared to control peptides. Solubility can be determined via any suitable method known in the art. In some embodiments, suitable methods known in the art for determining solubility include incubating the peptide in various buffers (acetate pH4.0, acetate pH5.0, phosphate/citrate pH5.0, phosphate citrate pH6.0, phosphate pH 7.0, phosphate pH7.5, strong PBS pH7.5, Tris pH 8.0, glycine pH 9.0), water, acetic acid (pH 5.0 and others known in the art) and testing for aggregation or solubility using standard techniques. These standard techniques include, but are not limited to, for example, visualization of precipitation, dynamic light scattering, circular dichroism, and fluorescent dyes to measure surface hydrophobicity, and to detect aggregation or fibrillation. In some embodiments, improved solubility means that the peptide is more soluble in a given liquid than a control peptide. In some embodiments, improved aggregability means that the peptide has less aggregability in a given liquid than a control peptide under a given set of conditions.
In certain embodiments, when delivered orally, the peptide inhibitors of the invention are stable in the Gastrointestinal (GI) environment, facilitating achievement of high levels in intestinal tissueCompound concentration. Proteolytic metabolism in the GI tract is driven by enzymes (including pepsin, trypsin, chymotrypsin, elastase, aminopeptidase and carboxypeptidase a/B) that are secreted from the pancreas into the lumen or produced as brush border enzymes. Proteases typically cleave peptides and proteins in an extended conformation. Under the reducing environment of intestinal fluids, disulfide bonds can be broken, resulting in linear peptides and rapid proteolysis. The redox environment of the chamber is determined primarily by the Cys/CySS redox cycle. In intestinal cells, the relevant activities involve a number of digestive enzymes, such as CYP450 and UDP-glucuronosyl-transferase. Finally, by 1010-1012Bacteria present in the large intestine at concentrations of CFU/ml constitute another metabolic barrier. In certain embodiments, the peptide inhibitor is stable to a variety of pH's ranging from strongly acidic in the stomach (pH 1.5-1.9), more alkaline in the small intestine (pH 6-7.5), and less acidic in the colon (pH 5-7). Such peptide inhibitors are stable during their passage through multiple GI compartments (a process estimated to take 3-4 hours in the intestine and 6-48 hours in the colon).
In some embodiments, a peptide inhibitor of the invention, for example, has less degradation (i.e., greater degradation stability) over a period of time, e.g., greater than or about 10% less degradation, greater than or about 20% less degradation, greater than or about 30% less degradation, greater than or about 40% less degradation, or greater than or about 50% less degradation, as compared to a control peptide. In some embodiments, degradation stability is determined via any suitable method known in the art. In some embodiments, the degradation is enzymatic degradation. For example, in certain embodiments, the peptide inhibitor has reduced sensitivity to degradation by trypsin, chymotrypsin, or elastase. In some embodiments, suitable methods known in the art for determining degradation stability include the methods described in Hawe et al, J Pharm Sci, VOL.101, No.3,2012, p 895-913, which are incorporated herein in their entirety. In some embodiments, such methods are used to select effective peptide sequences with enhanced half-lives. In particular embodiments, the stability of the peptide is determined using a SIF assay or SGF assay as described herein.
In certain embodiments, the peptide inhibitors of the invention inhibit or reduce IL-23 mediated inflammation. In related embodiments, the peptide inhibitors of the invention inhibit or reduce IL-23-mediated secretion of one or more cytokines, for example, by binding to IL-23R on the surface of a cell, thereby inhibiting the binding of IL-23 to the cell. In particular embodiments, the peptide inhibitors of the invention inhibit or reduce IL-23 mediated activation of Jak2, Tyk2, Stat1, Stat3, Stat4, or Stat 5. Methods of measuring inhibition of cytokine secretion and inhibition of signal transduction molecules are known in the art. For example, inhibition of IL-23/IL-23R signaling can be determined by measuring inhibition of phosphorylation-Stat 3 levels in cell lysates, as described in the accompanying examples (including example 2).
In certain embodiments, the peptide inhibitors of the invention inhibit or reduce IL-23 mediated inflammation. In related embodiments, the peptide inhibitors of the invention inhibit or reduce IL-23 mediated secretion of one or more cytokines, for example, by binding to IL-23R on the surface of a cell, thereby inhibiting IL-23 binding to the cell. In particular embodiments, the peptide inhibitors of the invention inhibit or reduce IL-23 mediated activation of Jak2, Tyk2, Stat1, Stat3, Stat4, or Stat 5. Methods of measuring inhibition of cytokine secretion and inhibition of signal transduction molecules are known in the art. For example, inhibition of IL-23/IL-23R signaling can be determined by measuring inhibition of phosphorylation-Stat 3 levels in cell lysates, as described in the accompanying examples (including example 2).
In certain embodiments, the redox stability of the peptide inhibitor is increased compared to a control peptide. Various assays that can be used to determine redox stability are known and available in the art. Any of these can be used to determine the redox stability of the peptide inhibitors of the present invention.
In certain embodiments, the invention provides a plurality of peptide inhibitors that bind or associate with IL-23R in vitro or in vivo to disrupt or block the binding between IL-23 and IL-23R. In certain embodiments, the peptide inhibitor binds to and/or inhibits human IL-23R. In certain embodiments, the peptide inhibitors bind to and/or inhibit both human and rodent IL-23R. In certain embodiments, the peptide inhibitor binds to and/or inhibits IL-23R in both human and rat. In particular embodiments, the peptide inhibitors inhibit rat IL-23R by at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, or at least 95%, and they bind or inhibit human IL-23R, e.g., as determined by the assays described herein. In certain embodiments, the peptide inhibitor preferentially binds to and/or inhibits human and/or rat IL-23R as compared to mouse IL-23R. In a specific embodiment, the peptide inhibitor binds preferentially to rat IL-23R as compared to mouse IL-23R. In particular embodiments, the peptide inhibitor binds preferentially to human IL-23R as compared to mouse IL-23R. In certain embodiments, less than 75%, less than 50%, less than 40%, less than 30%, less than 20%, or less than 10% of the same peptide inhibitor binds to mouse IL-23R and/or rat IL-23R. In certain embodiments of the peptide inhibitor that preferentially binds to and/or inhibits human IL-23R and/or rat IL-23R as compared to mouse IL-23R, the peptide inhibitor binds to a region of IL-23R that is disrupted by the presence of additional amino acids present in mouse IL-23R but not in human IL-23R or rat IL-23. In one embodiment, the additional amino acids present in mouse IL-23R are in a region corresponding to about amino acid residue 315 to about amino acid residue 340 of the mouse IL-23R protein, e.g., amino acid region NWQPWSSPFVHQTSQETGKR (SEQ ID NO:1355) (see, e.g., FIG. 4). In a specific embodiment, the peptide inhibitor binds to the region of human IL-23R from about amino acid 230 to about amino acid residue 370.
In certain embodiments, the peptide inhibitor exhibits GI restriction localization after oral administration. In particular embodiments, greater than 50%, greater than 60%, greater than 70%, greater than 80%, or greater than 90% of the orally administered peptide inhibitor is localized to gastrointestinal organs and tissues. In specific embodiments, the plasma level of orally administered peptide inhibitor is less than 20%, less than 10%, less than 5%, less than 2%, less than 1%, or less than 0.5% of the level of peptide inhibitor present in the small intestinal mucosa, colonic mucosa, or proximal colon.
The various peptide inhibitors of the present invention may be composed of only natural amino acids. Alternatively, the peptide inhibitor may comprise unnatural amino acids, including but not limited to modified amino acids. In certain embodiments, a modified amino acid includes a natural amino acid that has been chemically modified to include a group or chemical moiety that does not naturally occur on the amino acid. The peptide inhibitors of the invention may additionally comprise one or more D-amino acids. Still further, the peptide inhibitors of the present invention may comprise amino acid analogs.
In certain embodiments, the peptide inhibitors of the invention comprise one or more modified or unnatural amino acids. For example, in certain embodiments, the peptide inhibitor comprises one or more of the following: dab, Dap, Pen, Sarc, Cit, Cav, hLeu, 2-Nal, D-1-Nal, D-2-Nal, Phe (4-OMe), β hTRp, α -MePhe, α -MeTyr, α -MeTrp, β -HPhe, Phe (4-CF) 3) 2-2-indane, 1-1-indane, cyclobutyl, beta-hPhe, Gla, Phe (4-NH)2) hPhe, 1-Nal, Nle, homo-amino acids, D-amino acids, 4' -biphenylalanine (Bip), cyclobutyl-Ala, hCHa, beta hPhe, beta Glu, Phe (4-guanidino), Phe [4- (2-aminoethoxy)]Phe [4- (2-acetamidoethoxy)]、Phe(4-CONH2) Phe (4-Me), Tyr (Bzl) or Tyr (Me), Phe (3, 4-di F2)、Phe(3,4-Cl2) Phe (3-Me), Phe [4- (2-aminoethoxy)]Phe [4- (2-acetamidoethoxy)]、Phe(Br)、Phe(4-CONH2) Phe (Cl), Phe (4-CN), Phe (4-guanidino), Phe (4-Me), Phe (4-NH)2) Phe (4-N3), Tyr (Bzl) or Tyr (Me), Phe (3, 4-dimethoxy), 5-hydroxy Trp, Phe (3, 4-Cl)2) Tyr (3-tBu), and various N-methylated amino acids and alpha-methyl amino acids. In some embodiments of the invention, the peptide inhibitor comprises one or more unnatural amino acids shown in table 1A. Those skilled in the art will appreciate that additional modified or unnatural amino acids, as well as numerous other substitutions of natural amino acids with modified or unnatural amino acids, can achieve similar desired results, and that such substitutions are within the teachings and spirit of the invention. In certain embodiments, the peptide inhibitors of the present invention include any of those described herein, including but not limited to any of the peptide inhibitors comprising an amino acid sequence or peptide inhibitor structure shown in any of the tables, accompanying sequence listings, or figures herein, wherein one or more residues are substituted with a modified or non-natural amino acid.
The invention also includes any of the peptide inhibitors described herein in free or salt form. Accordingly, embodiments of any of the peptide inhibitors described herein (and related methods of use thereof) include pharmaceutically acceptable salts of the peptide inhibitors.
The invention also includes variants of any of the peptide inhibitors described herein, including but not limited to any of those comprising the sequences shown in any of the tables, accompanying sequence listings or figures herein, wherein one or more L-amino acid residues are substituted with a D-isomeric form of the amino acid residue, e.g., L-Ala is substituted with D-Ala.
In particular embodiments of the peptide inhibitors described herein, they comprise one or more unnatural or unnatural amino acid residue.
The invention also includes any of the peptide monomer inhibitors described herein attached to a linker moiety comprising any of the specific linker moieties described herein. In particular embodiments, the linker is attached to the N-terminal or C-terminal amino acid, while in other embodiments, the linker is attached to an internal amino acid. In particular embodiments, the linker is linked to two internal amino acids, e.g., an internal amino acid in each of the two monomeric subunits that form a dimer. In some embodiments of the invention, the peptide inhibitor is attached to one or more linker moieties as indicated.
The invention also includes peptides and peptide dimers comprising a peptide having at least 90%, at least 95%, at least 98%, or at least 99% sequence identity to the peptide sequence of a peptide inhibitor described herein. In particular embodiments, the peptide inhibitors of the invention comprise a core peptide sequence and one or more N-terminal and/or C-terminal modifications (e.g., Ac and NH)2) And/or one or moreA conjugated linker moiety and/or a half-life extending moiety. As used herein, a core peptide sequence is an amino acid sequence of a peptide in the absence of such modifications and conjugates. For example, for peptide inhibitors: [ Palm ] film]- [ isoGlu]-[PEG4]-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1115) and the core peptide sequence is: [ Pen ]]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN(SEQ ID NO:1115)。
In certain embodiments, the peptide inhibitor or monomeric subunit of a peptide inhibitor of the present invention comprises, consists essentially of, or consists of: 7 to 35 amino acid residues, 8 to 35 amino acid residues, 9 to 35 amino acid residues, 10 to 35 amino acid residues, 7 to 25 amino acid residues, 8 to 25 amino acid residues, 9 to 25 amino acid residues, 10 to 25 amino acid residues, 7 to 20 amino acid residues, 8 to 20 amino acid residues, 9 to 20 amino acid residues, 10 to 20 amino acid residues, 7 to 18 amino acid residues, 8 to 18 amino acid residues, 9 to 18 amino acid residues or 10 to 18 amino acid residues, and optionally, one or more further non-amino acid moieties, such as conjugated chemical moieties, e.g. PEG or linker moieties. In particular embodiments, the peptide inhibitors (or monomeric subunits thereof) of the invention (including but not limited to those of any embodiment of formula X, formula I, formula II, formula III, formula IV, or formula V) are greater than 10 amino acids, greater than 12 amino acids, greater than 15 amino acids, greater than 20 amino acids, greater than 25 amino acids, greater than 30 amino acids, or greater than 35 amino acids, e.g., from 35 to 50 amino acids. In certain embodiments, the peptide inhibitor (or monomeric subunit thereof) is less than 50 amino acids, less than 35 amino acids, less than 30 amino acids, less than 25 amino acids, less than 20 amino acids, less than 15 amino acids, less than 12 amino acids, or less than 10 amino acids. In particular embodiments, the monomeric subunit of a peptide inhibitor (or peptide monomeric inhibitor) comprises or consists of: 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, or 35 amino acid residues. In particular embodiments, the monomeric subunits of the peptide inhibitors of the present invention comprise or consist of: 10 to 18 amino acid residues and, optionally, one or more additional non-amino acid moieties such as conjugated chemical moieties, e.g., PEG or linker moieties. In various embodiments, the monomeric subunit comprises or consists of: 7 to 35 amino acid residues, 7 to 20 amino acid residues, 8 to 20 amino acid residues, 9 to 20 amino acid residues, 10 to 20 amino acid residues, 8 to 18 amino acid residues, 8 to 19 amino acid residues, 8 to 18 amino acid residues, 9 to 18 amino acid residues, or 10 to 18 amino acid residues. In a particular embodiment of any of the formulae described herein, X comprises or consists of: 7 to 35 amino acid residues, 8 to 35 amino acid residues, 9 to 35 amino acid residues, 10 to 35 amino acid residues, 7 to 25 amino acid residues, 8 to 25 amino acid residues, 9 to 25 amino acid residues, 10 to 25 amino acid residues, 7 to 18 amino acid residues, 8 to 18 amino acid residues, 9 to 18 amino acid residues, or 10 to 18 amino acid residues.
Certain exemplary peptide inhibitors described herein comprise 12 or more amino acid residues. However, the invention also includes peptide inhibitors comprising fragments of any of the peptide sequences described herein, including peptide inhibitors having 7, 8, 9, 10 or 11 amino acid residues. For example, peptide inhibitors of the invention include peptides comprising or consisting of: X4-X9, X4-X10, X4-X11, X4-X12, X4-X13, X4-X14, X4-X15 or X4-X16. In particular embodiments, the invention includes peptide inhibitors having any of the sequences described herein, including but not limited to those shown in any of the formulae described herein, the sequence listing, or any table provided herein, wherein one or more of X10, X11, X12, X13, X14, X15, or X16 is absent. In particular embodiments, one or more of X13, X14, X15, or X16 is absent.
In a particular embodiment of the invention, the peptide inhibitor or the X region thereof is not present in the antibody. In the detailed descriptionIn (2), the peptide inhibitor or X region thereof is not present in V of the antibodyHOr VLWithin a zone.
In particular embodiments of the peptide inhibitors described herein, they comprise one or more unnatural or unnatural amino acid residue.
In particular embodiments, the peptide inhibitors of the invention are cyclized via a cyclic amide bond, a disulfide bond, or a thioether bond. In a specific embodiment, the bond is an intramolecular bond between two amino acid residues within the peptide inhibitor or a monomeric subunit thereof.
Peptide inhibitors
The peptide inhibitors of the invention include peptides having any of the amino acid sequences described herein, compounds having any of the structures described herein (including compounds comprising any of the peptide sequences described herein), and dimers of any such peptides and compounds. Peptide inhibitors of the invention include peptides that do not have a bond between X4 and X9 and peptides that do have a bond between X4 and X9, e.g., before and after cross-linking is introduced between X4 and X9. Exemplary peptides of the invention comprise the amino acid sequences or structures depicted in any of the accompanying tables, examples, figures, and sequence listing.
In certain embodiments, the invention encompasses a peptide inhibitor of the interleukin-23 receptor, or a pharmaceutically acceptable salt or solvate thereof, wherein the peptide inhibitor comprises an amino acid sequence of formula (Xa):
X1-X2-X3-X4-X5-X6-X7-X8-X9-X10-X11-X12-X13-X14-X15-X16-X17-X18-X19-X20(Xa)(SEQ ID NO:1354)
wherein
X1 is any amino acid or absent;
x2 is any amino acid or is absent;
X3 is any amino acid or absent;
x4 is any amino acid or chemical moiety capable of forming a bond with X9;
x5 is any amino acid;
x6 is any amino acid;
x7 is any amino acid;
x8 is any amino acid;
x9 is any amino acid or chemical moiety capable of forming a bond with X4;
x10 is any amino acid;
x11 is any amino acid;
x12 is any amino acid;
x13 is any amino acid;
x14 is any amino acid;
x15 is any amino acid selected from the group consisting of,
x16 is any amino acid or absent;
x17 is any amino acid or absent;
x18 is any amino acid or absent;
x19 is any amino acid or absent; and
x20 is any amino acid or is absent,
wherein X4 and X9 are capable of forming a bond with each other. In particular embodiments, the bond is a disulfide bond, a thioether bond, a lactam bond, a triazole ring, a selenoether bond, a diselenide bond, or an alkene bond. In particular embodiments, the linkage is a disulfide linkage or a thioether linkage. In certain embodiments, the peptide inhibitor is cyclized via a bond between X4 and X9. In certain embodiments, the peptide inhibitor inhibits binding of interleukin-23 (IL-23) to an IL-23 receptor. In particular embodiments, when X4 is not an amino acid, then X1, X2, and X3 are absent. In certain embodiments, X1 is a D-amino acid or is absent. In certain embodiments, X2 is a D-amino acid or is absent. In certain embodiments, X3 is a D-amino acid or is absent. In certain embodiments, X16 is a D-amino acid or is absent. In certain embodiments, X17 is a D-amino acid or is absent. In certain embodiments, X18 is a D-amino acid or is absent. In certain embodiments, X19 is a D-amino acid or is absent. In certain embodiments, X20 is a D-amino acid or is absent.
In one embodiment of the peptide inhibitor of formula Xa,
x1 is absent;
x2 is absent;
x3 is Glu, D-Glu, Arg, (D) Arg, Phe, (D) Phe, 2-Nal, Thr, Leu, (D) Gln, or deleted;
x4 is Cys, Abu or Pen;
x5 is Ala, α -MeOrn, α -MeSer, Cit, Dap, Dab, Dap (Ac), Gly, Lys, Asn, N-MeGln, N-MeArg, Orn, Gln, Arg, Ser, or Thr;
x6 is Asp or Thr;
x7 is Trp or 6-chloro-Trp;
x8 is Glu, Gln, or Val;
x9 is Cys, Abu or Pen;
x10 is 2-Nal, a Phe analog, a Tyr or Tyr analog, wherein in particular embodiments, X10 is 2-Nal, Phe (3, 4-di F)2)、Phe(3,4-Cl2) Phe (3-Me), Phe [4- (2-aminoethoxy)]Phe [4- (2-acetamidoethoxy)]、Phe(Br)、Phe(4-CONH2) Phe (Cl), Phe (4-CN), Phe (4-guanidino), Phe (4-Me), Phe (4-NH)2) Phe (4-N3), Tyr (Bzl) or Tyr (Me);
x11 is 1-Nal, 2-Nal, Phe (3, 4-dimethoxy), 5-hydroxy Trp, Phe (3, 4-Cl)2) Trp or Tyr (3-tBu);
x12 is 3-Pal, Acpc, Acbc, Acvc, Achc, Agp, Aib, α -diethylGly, α -MeLys (Ac), α -MeLeu, a- α -MeOrn, α -MeSer, α -MeVal, Cav, Cha, Cit, Cpa, D-Asn, Glu, His, hLeu, hArg, Lys, Leu, Octgly, Orn, piperidine, Arg, Ser, Thr, or THP;
X13 is Cit, Asp, Dab, Dap, Phe, His, Dap (Peg2-Ac), Dap (pyroglutamic acid), Glu, hArg, Lys (Ac), Lys (benzoic acid), Lys (glutaric acid), Lys (IVA), Lys (Peg 4-isoGlu-Palm), Lys (pyroglutamic acid), Lys-succinic acid, Asn, Orn, Gln, Arg, Thr or Val;
x14 is Asp, Dab (Ac), dap (Ac), Phe, His, Lys (Ac), Met, Asn (isobutyl), Gln, Arg, Tyr or Asp (1, 4-diaminobutane);
x15 is Ala, β Ala, Glu, Gly, Asn, Gln, Arg or Ser,
x16 is any amino acid or absent;
x17 is any amino acid or absent;
x18 is any amino acid or absent;
x19 is any amino acid or absent; and
x20 is any amino acid or is absent.
In certain embodiments, X3 is absent. In particular embodiments, X16, X17, X18, X19, and X20 are absent. In particular embodiments, X4 and X9 are both Cys, and X4 and X9 are linked via a disulfide bond. In a specific embodiment, X4 is Abu and X9 is Pen, and X4 and X9 are linked via a thioether bond. In a specific embodiment, X4 is Abu and X9 is Cys, and X4 and X9 are linked via a thioether bond.
In another embodiment of the peptide inhibitor of formula Xa,
X1 is absent;
x2 is absent;
x3 is Glu, D-Glu, Arg, (D) Arg, Phe, (D) Phe, 2-Nal, Thr, Leu, (D) Gln, or deleted;
x4 is Cys, Abu or Pen;
x5 is Ala, α -MeOrn, α -MeSer, Cit, Dap, Dab, Dap (Ac), Gly, Lys, Asn, Orn, Gln, Arg, Ser, or Thr;
x6 is Asp or Thr;
x7 is Trp or 6-chloro-Trp;
x8 is Gln or Val;
x9 is Cys, Abu or Pen;
x10 is 2-Nal, a Phe analog, a Tyr or Tyr analog, wherein in particular embodiments, X10 is 2-Nal, Phe (3, 4-di F)2) Phe (3-Me), Phe [4- (2-aminoethoxy)]Phe [4- (2-acetamidoethoxy)]、Phe(Br)、Phe(4-CONH2) Phe (4-Cl), Phe (4-CN), Phe (4-guanidino), Phe (4-Me), Phe (4-NH)2)、Phe(4-N3) Tyr, Tyr (Bzl) or Tyr (Me);
x11 is 1-Nal, 2-Nal, Phe (3, 4-dimethoxy), 5-hydroxy Trp, Phe (3, 4-Cl)2) Trp or Tyr (3-tBu);
x12 is 3-Pal, Acpc, Acbc, Acvc, Achc, Agp, Aib, α -diethylGly, α -MeLys (Ac), α -MeLeu, α -MeOrn, α -MeSer, α -MeVal, Cav, Cha, Cit, Cpa, D-Asn, His, hLeu, hArg, Lys, Leu, Octgly, Orn, 4-amino-4-carboxy-piperidine or THP;
x13 is Cit, Asp, Dab, Dap, Phe, His, Dap (Peg2-Ac), Dap (pyroglutamic acid), Glu, hArg, Lys (Ac), Lys (benzoic acid), Lys (glutaric acid), Lys (IVA), Lys (Peg 4-isoGlu-Palm), Lys (pyroglutamic acid), Lys-succinic acid, Asn, Orn, Gln, Arg, Thr or Val;
X14 is dab (Ac), dap (Ac), Phe, His, Lys (Ac), Met, Asn, Gln, Arg, or Tyr;
x15 is Ala, β Ala, Gly, Asn, Gln or Ser,
x16 is any amino acid or absent;
x17 is any amino acid or absent;
x18 is any amino acid or absent;
x19 is any amino acid or absent; and
x20 is any amino acid or absent.
In some embodiments, X3 is absent. In particular embodiments, X16, X17, X18, X19, and X20 are absent. In particular embodiments, X4 and X9 are Cys, and X4 and X9 are linked via a disulfide bond. In a specific embodiment, X4 is Abu and X9 is Pen, and X4 and X9 are linked via a thioether bond. In a specific embodiment, X4 is Abu and X9 is Cys, and X4 and X9 are linked via a thioether bond.
In another embodiment of the peptide inhibitor of formula Xa,
x1 is absent;
x2 is absent;
x3 is Glu, D-Glu, Arg, (D) Arg, Phe, (D) Phe, 2-Nal, Thr, Leu, (D) Gln, or deleted;
x4 is Cys, Abu or Pen;
x5 is Dap, Dap (Ac), Gly, Lys, Gln, Arg, Ser, Thr, or Asn;
x6 is Thr;
x7 is Trp or 6-chloro-Trp;
x8 is Gln;
x9 is Cys, Abu or Pen;
X10 is 2-Nal, a Phe analog, a Tyr or Tyr analog, wherein in particular embodiments, X10 is 2-Nal, Phe (3-Me), Phe [4- (2-aminoethoxy)]Phe [4- (2-acetamidoethoxy)]、Phe(4-CONH2)、Phe(4-Me)、Phe(4-NH2) Tyr, Tyr (Bzl) or Tyr (Me);
x11 is 1-Nal, 2-Nal, Phe (3, 4-dimethoxy), Phe (3, 4-Cl)2) Or Trp;
x12 is Acpc, Acbc, Acvc, Achc, Aib, α -diethylGly, α -MeLys (Ac), α -MeLeu, α -MeOrn, α -MeSer, α -MeVal, Cha, Cit, hLeu, Lys, Leu, Arg, or THP;
x13 is Cit, Asp, Dap (Peg2-Ac), Dap (pyroglutamic acid), Glu, hArg, Lys (Ac), Lys (benzoic acid), Lys (glutaric acid), Lys (IVA), Lys (Peg 4-isoGlu-Palm), Lys (pyroglutamic acid), Lys (succinic acid), Asn, Orn, Gln, Arg, or Val;
x14 is dab (Ac), dap (Ac), His, Lys (Ac), Asn, Gln, or Tyr;
x15 is Ala, β Ala, Gly, Asn, Gln or Ser,
x16 is any amino acid or absent;
x17 is any amino acid or absent;
x18 is any amino acid or absent;
x19 is any amino acid or absent; and
x20 is any amino acid or absent.
In some embodiments, X3 is absent. In particular embodiments, X16, X17, X18, X19, and X20 are absent. In particular embodiments, X4 and X9 are both Cys, and X4 and X9 are linked via a disulfide bond. In a specific embodiment, X4 is Abu and X9 is Pen, and X4 and X9 are linked via a thioether bond. In a specific embodiment, X4 is Abu and X9 is Cys, and X4 and X9 are linked via a thioether bond.
In another embodiment of the peptide inhibitor of formula Xa,
x1 is absent;
x2 is absent;
x3 is Glu, D-Glu, Arg, (D) Arg, Phe, (D) Phe, 2-Nal, Thr, Leu, (D) Gln, or deleted;
x4 is Cys, Abu or Pen;
x5 is Dap, Dap (Ac), Gln, Ser, Thr, or Asn;
x6 is Thr;
x7 is Trp;
x8 is Gln;
x9 is Cys, Abu or Pen;
x10 is a Phe analog, Tyr or Tyr analog, wherein in particular embodiments, X10 is Phe [4- (2-aminoethoxy)]Phe [4- (2-acetamidoethoxy)]、Phe(4-CONH2) Phe (4-Me), Tyr (Bzl) or Tyr (Me);
x11 is 2-Nal or Trp;
x12 is Acpc, Acbc, Acvc, Achc, Aib, α -diethylGly, α -MeLys (Ac), α -MeLeu, α -MeOrn, α -MeSer, α -MeVal, hLeu, Leu, or THP;
x13 is Cit, Asp, Glu, Lys (Ac), Asn or Gln;
x14 is dab (Ac), Asn or His;
x15 is Ala, β Ala, Gly, Asn or Gln;
x16 is any amino acid or absent;
x17 is any amino acid or absent;
x18 is any amino acid or absent;
x19 is any amino acid or absent; and
x20 is any amino acid or absent.
In some embodiments, X3 is absent. In particular embodiments, X16, X17, X18, X19, and X20 are absent. In particular embodiments, X4 and X9 are both Cys, and X4 and X9 are linked via a disulfide bond. In a specific embodiment, X4 is Abu and X9 is Pen, and X4 and X9 are linked via a thioether bond. In a specific embodiment, X4 is Abu, and X9 is Cys, and X4 and X9 are linked via a thioether bond.
In a specific embodiment, the peptide inhibitor comprises an amino acid sequence of any one of the formulae described herein, e.g., shown in Ia-It, IIa-IId, IIIa-IIIe, or IV.
In certain embodiments, the present invention includes a peptide inhibitor of the interleukin-23 receptor, or a pharmaceutically acceptable salt or solvate thereof, wherein the peptide inhibitor has the structure of formula I:
R1-X-R2(I)
wherein R is1Is a bond, hydrogen, C1-C6 hydrocarbyl, C6-C12 aryl, C6-C12 aryl C1-C6 hydrocarbyl, C1-C20 hydrocarboxyl, and includes pegylated forms of any of the foregoing either alone or as a spacer;
R2is a bond, OH or NH2(ii) a And
x is an amino acid sequence, e.g., an amino acid comprising 7 to 35 amino acid residues. In certain embodiments, R2Is OH or NH2
In certain embodiments, X comprises a sequence of formula Xa.
In particular embodiments of formula (I), X comprises a sequence of formula Ia:
X1-X2-X3-X4-X5-X6-W-X8-X9-X10-X11-X12-X13-X14-X15-X16-X17-X18-X19-X20(Ia)(SEQ ID NO:1356)
wherein
X1 is any amino acid or absent;
x2 is any amino acid or absent;
x3 is any amino acid or is absent;
x4 is Cys, Pen, hCys, D-Pen, D-Cys, D-hCys, Met, Glu, Asp, Lys, Orn, Dap, Dab, D-Dap, D-Dab, D-Asp, D-Glu, D-Lys, Sec, 2-chloromethylbenzoic acid, mercaptopropionic acid, mercaptobutyric acid, 2-chloro-acetic acid, 3-chloro-propionic acid, 4-chloro-butyric acid, 3-chloro-isobutyric acid, Abu, β -azido-Ala-OH, propargylglycine, 2- (3 '-butenyl) glycine, 2-allylglycine, 2- (3' -butenyl) glycine, 2- (4 '-pentenyl) glycine, 2- (5' -hexenyl) glycine or deleted;
X5 is Ala, Arg, Glu, Phe, Leu, Thr, Ser, Aib, Sarc, D-Ala, D-Arg, D-Glu, D-Phe, D-Leu, D-Thr, D-Ser, α -MeOrn, α -MeSer, CitDap, Dab, Dap (Ac), Gly, Lys, Asn, N-Me-Gln, N-Me-Arg, Orn, or Gln,
x6 is Asp, Thr, Asn, Phe, D-Asp, D-Thr, D-Asn or D-Phe;
x8 is Val, Gln, Glu, or Lys;
x9 is Cys, Pen, hCys, D-Pen, D-Cys, D-hCys, Glu, Lys, Orn, Dap, Dab, D-Dap, D-Dab, D-Asp, D-Glu, D-Lys, Asp, Leu, Val, Phe, Ser, Sec, Abu, β -azido-Ala-OH, propargylglycine, 2-2-allylglycine, 2- (3 ' -butenyl) glycine, 2- (4 ' -pentenyl) glycine or 2- (5 ' -hexenyl) glycine;
x10 is Tyr, Phe (3, 4-F)2)、Phe(3,4-Cl2) F (3-Me), Phe [4- (2-aminoethoxy)]Phe [4- (2- (acetyl-aminoethoxy)]、Phe(4-Br)、Phe(4-CONH2) Phe (4-Cl), Phe (4-CN), Phe (4-guanidino), Phe (4-Me), Phe (4-NH)2)、Phe(4-N3) Phe (4-OMe), Phe (4-OBzl) or Tyr;
x11 is Trp, 1-Nal, 2-Nal, Phe (3, 4-OMe)2) 5-hydroxy-Trp, Phe (3, 4-Cl)2) Or Tyr (3-t-Bu)
X12 is His, Phe, Arg, N-Me-His or Val, Cav, Cpa, Leu, Cit, hLeu, 3-Pal, t-butyl-Ala, t-butyl-Gly, 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Acvc, Agp, Aib, α -diethylGly, α -MeLys (Ac), α -Me-Leu, α -MeOrn, α -MeSer, α -MeVal, Cha, Cit, Cpa, (D) Asn, Glu, hArg, or Lys;
X13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, Val, β hAla, Aib, Lys (Ac), Cit, Asp, Dab, Dap, Glu, hArg, Lys, Asn, Orn, or Gln;
x14 is Phe, Tyr,. beta.hPhe, Asn, Arg, Qln, Lys (Ac), His; dap (Ac), dab (Ac), or Asp;
x15 is Gly, Ser, Thr, Gln, Ala, Sarc, β -Ala, Glu, Arg, or Asn;
x16 is any amino acid or absent;
x17 is any amino acid or absent;
x18 is any amino acid or absent;
x19 is any amino acid or absent; and
x20 is any amino acid or absent.
In a specific embodiment of Ia: x5 is Ala, Arg, Glu, Phe, Leu, Thr, Ser, Aib, Sarc, D-Ala, D-Arg, D-Glu, D-Phe, D-Leu, D-Thr, D-Ser, D-Aib, or D-Sarc; x10 is Tyr or Phe; x11 is Trp, 1-Nal or 2-Nal; x12 is His, Phe, Arg, N-Me-His or Val, Cav, Cpa, Leu, Cit, hLeu, 3-Pal, tert-butyl-Ala or tert-butyl-Gly; x13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, Val, β hAla, or Aib; x14 is Phe, Tyr or beta hPhe; x15 is Gly, Ser, Thr, Gln, Ala or Sarc; x16 is Asp, Glu, Ala, AEA, AEP,. beta.hAla, Gaba or deleted; and X17 is Leu, Lys, Arg or deleted.
In particular embodiments, X4 is present.
In certain embodiments, the peptide inhibitor is cyclized.
In certain embodiments, the peptide inhibitor is linear or not cyclized.
In certain embodiments, the peptide inhibitor is cyclized or contains an intramolecular bond between X4 and X9.
In certain embodiments of formula I, X comprises a sequence of formula Ib:
X1-X2-X3-X4-X5-X6-W-X8-X9-X10-X11-X12-X13-X14-X15-X16-X17-X18-X19-X20(Ib)(SEQ ID NO:1357),
wherein:
x1 is any amino acid or absent;
x2 is any amino acid or absent;
x3 is any amino acid or absent;
x4 is Cys, Pen, hCys, D-Pen, D-Cys, D-hCys, Glu, Asp, Lys, Orn, Dap, Dab, D-Dap, D-Dab, D-Asp, D-Glu, D-Lys, Sec, 2-chloromethylbenzoic acid, mercaptopropionic acid, mercaptobutyric acid, 2-chloro-acetic acid, 3-chloro-propionic acid, 4-chloro-butyric acid, 3-chloro-isobutyric acid, Abu, β -azido-Ala-OH, propargylglycine, 2- (3 '-butenyl) glycine, 2-2-allylglycine, 2- (3' -butenyl) glycine, 2- (4 '-pentenyl) glycine, 2- (5' -hexenyl) glycine or deleted;
x5 is Ala, Arg, Glu, Phe, Leu, Thr, Ser, Aib, Sarc, D-Ala, D-Arg, D-Glu, D-Phe, D-Leu, D-Thr, D-Ser, α -MeOrn, α -MeSer, CitDap, Dab, Dap (Ac), Gly, Lys, Asn, N-Me-Gln, N-Me-Arg, Orn, or Gln;
X6 is Asp, Thr, Asn, Phe, D-Asp, D-Thr, D-Asn or D-Phe;
x8 is Val, Gln, Glu, or Lys;
x9 is Cys, Pen, hCys, D-Pen, D-Cys, D-hCys, Glu, Lys, Orn, Dap, Dab, D-Dap, D-Dab, D-Asp, D-Glu, D-Lys, Asp, Sec, Abu, β -azido-Ala-OH, propargylglycine, 2-allylglycine, 2- (3 ' -butenyl) glycine, 2- (4 ' -pentenyl) glycine or 2- (5 ' -hexenyl) glycine;
x10 is Tyr, Phe (3, 4-F)2)、Phe(3,4-Cl2) F (3-Me), Phe [4- (2-aminoethoxy)]Phe [4- (2- (acetyl-aminoethoxy)]、Phe(4-Br)、Phe(4-CONH2) Phe (4-Cl), Phe (4-CN), Phe (4-guanidino), Phe (4-Me), Phe (4-NH)2)、Phe(4-N3) Phe (4-OMe), Phe (4-OBzl) or Tyr;
x11 is Trp, 1-Nal, 2-Nal, Phe (3, 4-OMe)2) 5-hydroxy-Trp, Phe (3, 4-Cl)2)、Tyr(3-t-Bu);
X12 is His, Phe, Arg, N-Me-His, Val, Cav, Cpa, Leu, Cit, hLeu, 3-Pal, t-butyl-Ala, t-butyl-Gly 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Acvc, Agp, Aib, α -diethylGly, α -MeLys (Ac), α -Me-Leu, α -MeOrn, α -MeSer, α -MeVal, Cha, Cit, Cpa, (D) Asn, Glu, hArg, or Lys;
x13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, Val, β hAlaAib, Lys (Ac), Cit, Asp, Dab, Dap, Glu, hArg, Lys, Asn, Orn, or Gln;
X14 is Phe, Tyr or beta hPhe, Asn, Arg, Qln, Lys (Ac), His; dap (Ac), dab (Ac), or Asp;
x15 is Gly, Ser, Thr, Gln, Ala or Sarc, β -Ala, Glu, Arg or Asn;
x16 is any amino acid or absent;
x17 is any amino acid or absent;
x18 is any amino acid or absent;
x19 is any amino acid or absent; and
x20 is any amino acid or absent.
In a specific embodiment of Ib: x5 is Ala, Arg, Glu, Phe, Leu, Thr, Ser, Aib, Sarc, D-Ala, D-Arg, D-Glu, D-Phe, D-Leu, D-Thr, D-Ser, D-Aib, or D-Sarc; x10 is Tyr or Phe; x11 is Trp, 1-Nal or 2-Nal; x12 is His, Phe, Arg, N-Me-His, Val, Cav, Cpa, Leu, Cit, hLeu, 3-Pal, t-butyl-Ala or t-butyl-Gly; x13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, Val, β hAla, or Aib; x14 is Phe, Tyr or beta hPhe; x15 is Gly, Ser, Thr, Gln, Ala, or Sarc; x16 is Asp, Glu, Ala, AEA, AEP,. beta.hAla, Gaba or deleted; and X17 is Leu, Lys, Arg or deleted.
In particular embodiments, X4 is present.
In certain embodiments, the peptide inhibitor is cyclized.
In certain embodiments, the peptide inhibitor is linear or not cyclized.
In certain embodiments, the peptide inhibitor is cyclized or contains an intramolecular bond between X4 and X9.
In certain embodiments of formula I, X comprises a sequence of formula Ic:
X1-X2-X3-X4-X5-X6-W-X8-X9-Y-X11-X12-X13-X14-X15-X16-X17-X18-X19-X20
(Ic)(SEQ ID NO:1358)
wherein
X1 is any amino acid or absent;
x2 is any amino acid or absent;
x3 is any amino acid or absent;
x4 is Cys, Pen, hCys, D-Pen, D-Cys, D-hCys, Met, Glu, Asp, Lys, Orn, Dap, Dab, D-Dap, D-Dab, D-Asp, D-Glu, D-Lys, Sec, 2-chloromethylbenzoic acid, mercaptopropionic acid, mercaptobutyric acid, 2-chloro-acetic acid, 3-chloro-propionic acid, 4-chloro-butyric acid, 3-chloro-isobutyric acid, Abu, β -azido-Ala-OH, propargylglycine, 2-allylglycine, 2- (3 ' -butenyl) glycine, 2- (4 ' -pentenyl) glycine, 2- (5 ' -hexenyl) glycine or absent;
x5 is Ala, Arg, Glu, Phe, Leu, Thr, Ser, Aib, Sarc, D-Ala, D-Arg, D-Glu, D-Phe, D-Leu, D-Thr, D-Ser, α -MeOrn, α -MeSer, CitDap, Dab, Dap (Ac), Gly, Lys, Asn, N-Me-Gln, N-Me-Arg, Orn, or Gln;
x6 is Asp, Thr, Asn, Phe, D-Asp, D-Thr, D-Asn or D-Phe;
X8 is Val, Gln, Glu, or Lys;
x9 is Cys, Pen, hCys, D-Pen, D-Cys, D-hCys, Glu, Lys, Orn, Dap, Dab, D-Dap, D-Dab, D-Asp, D-Glu, D-Lys, Asp, Sec, Abu, β -azido-Ala-OH, propargylglycine, 2-allylglycine, 2- (3 ' -butenyl) glycine, 2- (4 ' -pentenyl) glycine or 2- (5 ' -hexenyl) glycine;
x11 is Trp, 1-Nal, 2-Nal, Phe (3, 4-OMe)2) 5-hydroxy-Trp, Phe (3, 4-Cl)2) Or Tyr (3-t-Bu);
x12 is His, Phe, Arg, N-Me-His, Val, Cav, Cpa, Leu, Cit, hLeu, 3-Pal, t-butyl-Ala, t-butyl-Gly; 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Acvc, Agp, Aib, α -diethylgly, α -MeLys (ac), α -Me-Leu, α -MeOrn, α -mesr, α -MeVal, Cha, Cit, Cpa, (D) Asn, Glu, hArg, or Lys;
x13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, Val, β hAla or Aib, Lys (Ac), Cit, Asp, Dab, Dap, Glu, hArg, Lys, Asn, Orn, or Gln;
x14 is Phe, Tyr,. beta.hPhe, Asn, Arg, Qln, Lys (Ac), His; dap (Ac), dab (Ac), or Asp;
x15 is Gly, Ser, Thr, Gln, Ala, Sarc, β -Ala, Glu, Arg, or Asn;
X16 is any amino acid or is absent;
x17 is any amino acid or absent;
x18 is any amino acid or absent;
x19 is any amino acid or absent; and
x20 is any amino acid or absent.
In a specific embodiment of Ic, X5 is Ala, Arg, Glu, Phe, Leu, Thr, Ser, Aib, Sarc, D-Ala, D-Arg, D-Glu, D-Phe, D-Leu, D-Thr, D-Ser, D-Aib, or D-Sarc; x11 is Trp, 1-Nal or 2-Nal; x12 is His, Phe, Arg, N-Me-His, Val, Cav, Cpa, Leu, Cit, hLeu, 3-Pal, t-butyl-Ala or t-butyl-Gly; x13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, Val, β hAla, or Aib; x14 is Phe, Tyr or beta hPhe; x15 is Gly, Ser, Thr, Gln, Ala, or Sarc; x16 is Asp, Glu, Ala, AEA, AEP,. beta.hAla, Gaba or deleted; and X17 is Leu, Lys, Arg or deleted.
In particular embodiments, X4 is present.
In certain embodiments, the peptide inhibitor is cyclized.
In certain embodiments, the peptide inhibitor is linear or not cyclized.
In certain embodiments, the peptide inhibitor is cyclized or contains an intramolecular bond between X4 and X9.
In certain embodiments of formula I, X comprises a sequence of formula Id:
X1-X2-X3-C-X5-X6-W-X8-C-X10-X11-X12-X13-X14-X15-X16-X17-X18-X19-X20
(Id)(SEQ ID NO:1359)
wherein
X1 is any amino acid or absent;
x2 is any amino acid or absent;
x3 is any amino acid or absent;
x5 is Ala, Arg, Glu, Phe, Leu, Thr, Ser, Aib, Sarc, D-Ala, D-Arg, D-Glu, D-Phe, D-Leu, D-Thr, D-Ser, α -MeOrn, α -MeSer, CitDap, Dab, Dap (Ac), Gly, Lys, Asn, N-Me-Gln, N-Me-Arg, Orn, or Gln;
x6 is Asp, Thr, Asn, Phe, D-Asp, D-Thr, D-Asn or D-Phe;
x8 is Val, Gln, Glu, or Lys;
x10 is Tyr, Phe (3, 4-F)2)、Phe(3,4-Cl2) F (3-Me), Phe [4- (2-aminoethoxy)]Phe [4- (2- (acetyl-aminoethoxy)]、Phe(4-Br)、Phe(4-CONH2) Phe (4-Cl), Phe (4-CN), Phe (4-guanidino), Phe (4-Me), Phe (4-NH)2)、Phe(4-N3) Phe (4-OMe), Phe (4-OBzl) or Tyr;
x11 is Trp, 1-Nal, 2-Nal, Phe (3, 4-OMe)2) 5-hydroxy-Trp, Phe (3, 4-Cl)2)、Tyr(3-t-Bu);
X12 is His, Phe, Arg, N-Me-His, Val, Cav, Cpa, Leu, Cit, hLeu, 3-Pal, t-butyl-Ala, t-butyl-Gly, 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Acvc, Agp, Aib, α -diethylGly, α -MeLys (Ac), α -Me-Leu, α -MeOrn, α -MeSer, α -MeVal, Cha, Cit, Cpa, (D) Asn, Glu, hArg, or Lys;
X13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, Val, β hAla, Aib, Lys (Ac), Cit, Asp, Dab, Dap, Glu, hArg, Lys, Asn, Orn, or Gln;
x14 is Phe, Tyr, β hPhe, Asn, Arg, Qln, Lys (Ac), His, Dap (Ac), dab (Ac), or Asp;
x15 is Gly, Ser, Thr, Gln, Ala, Sarc, β -Ala, Glu, Arg, or Asn;
x16 is any amino acid or absent;
x17 is any amino acid or absent;
x18 is any amino acid or absent;
x19 is any amino acid or absent; and
x20 is any amino acid or is absent,
wherein X4 and X9 are optionally connected by intramolecular disulfide bridges.
In certain embodiments of Id: x5 is Ala, Arg, Glu, Phe, Leu, Thr, Ser, Aib, Sarc, D-Ala, D-Arg, D-Glu, D-Phe, D-Leu, D-Thr, D-Ser, D-Aib, or D-Sarc; x10 is Tyr or Phe; x11 is Trp, 1-Nal or 2-Nal; x12 is His, Phe, Arg, N-Me-His, Val, Cav, Cpa, Leu, Cit, hLeu, 3-Pal, t-butyl-Ala or t-butyl-Gly; x13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, Val, β hAla, or Aib; x14 is Phe, Tyr or beta hPhe; x15 is Gly, Ser, Thr, Gln, Ala, or Sarc; x16 is Asp, Glu, Ala, AEA, AEP,. beta.hAla, Gaba or deleted; and X17 is Leu, Lys, Arg or deleted.
In certain embodiments of formula I, X comprises a sequence of formula Ie:
X1-X2-X3-X4-X5-X6-W-X8-X9-X10-X11-X12-X13-X14-X15-X16-X17-X18-X19-X20
(Ie)(SEQ ID NO:1360)
wherein
X1 is any amino acid or absent;
x2 is any amino acid or absent;
x3 is any amino acid or absent;
x4 is Pen, hCys, D-Pen, D-Cys or D-hCys;
x5 is Ala, Arg, Glu, Phe, Leu, Thr, Ser, Aib, Sarc, D-Ala, D-Arg, D-Glu, D-Phe, D-Leu, D-Thr, D-Ser, α -MeOrn, α -MeSer, CitDap, Dab, Dap (Ac), Gly, Lys, Asn, N-Me-Gln, N-Me-Arg, Orn, or Gln;
x6 is Asp, Thr, Asn, Phe, D-Asp, D-Thr, D-Asn or D-Phe;
x8 is Val, Gln, Glu, or Lys;
x9 is Pen, hCys, D-Pen, D-Cys, D-hCys;
x10 is Tyr, Phe (3, 4-F)2)、Phe(3,4-Cl2) F (3-Me), Phe [4- (2-aminoethoxy)]Phe [4- (2- (acetyl-aminoethoxy)]、Phe(4-Br)、Phe(4-CONH2) Phe (4-Cl), Phe (4-CN), Phe (4-guanidino), Phe (4-Me), Phe (4-NH)2)、Phe(4-N3) Phe (4-OMe), Phe (4-OBzl) or Tyr;
x11 is Trp, 1-Nal, 2-Nal, Phe (3, 4-OMe)2) 5-hydroxy-Trp, Phe (3, 4-Cl)2) Or Tyr (3-t-Bu);
x12 is His, Phe, Arg, N-Me-His, Val, Cav, Cpa, Leu, Cit, hLeu, 3-Pal, t-butyl-Ala, t-butyl-Gly, 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Acvc, Agp, Aib, α -diethylGly, α -MeLys (Ac), α -Me-Leu, α -MeOrn, α -MeSer, α -MeVal, Cha, Cit, Cpa, (D) Asn, Glu, hArg, or Lys;
X13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, Val, β hAla, Aib, Lys (Ac), Cit, Asp, Dab, Dap, Glu, hArg, Lys, Asn, Orn, or Gln;
x14 is Phe, Tyr,. beta.hPhe, Asn, Arg, Qln, Lys (Ac), His; dap (Ac), dab (Ac) or Asp;
x15 is Gly, Ser, Thr, Gln, Ala, Sarc, β -Ala, Glu, Arg, or Asn;
x16 is any amino acid or absent;
x17 is any amino acid or absent;
x18 is any amino acid or absent;
x19 is any amino acid or absent; and
x20 is any amino acid or is absent,
wherein X4 and X9 are optionally connected by intramolecular disulfide bridges.
In certain embodiments of Ie: x5 is Ala, Arg, Glu, Phe, Leu, Thr, Ser, Aib, Sarc, D-Ala, D-Arg, D-Glu, D-Phe, D-Leu, D-Thr, D-Ser, D-Aib, or D-Sarc; x10 is Tyr or Phe; x11 is Trp, 1-Nal or 2-Nal; x12 is His, Phe, Arg, N-Me-His, Val, Cav, Cpa, Leu, Cit, hLeu, 3-Pal, t-butyl-Ala or t-butyl-Gly; x13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, Val, β hAla, or Aib; x14 is Phe, Tyr or beta hPhe; x15 is Gly, Ser, Thr, Gln, Ala, or Sarc; x16 is Asp, Glu, Ala, AEA, AEP,. beta.hAla, Gaba or deleted; and X17 is Leu, Lys, Arg or deleted.
In particular embodiments, X4 is present.
In certain embodiments, the peptide inhibitor is cyclized.
In certain embodiments, the peptide inhibitor is linear or not cyclized.
In certain embodiments, the peptide inhibitor is cyclized or contains an intramolecular bond between X4 and X9.
In particular embodiments, both X4 and X9 are Pen.
In certain embodiments of formula I, X comprises a sequence of formula If:
X1-X2-X3-X4-X5-X6-W-X8-X9-X10-X11-X12-X13-X14-X15-X16-X17-X18-X19-X20
(If)(SEQ ID NO:1361)
wherein
X1 is any amino acid or absent;
x2 is any amino acid or absent;
x3 is any amino acid or absent;
x4 is Glu, Lys, Orn, Dap, Dab, D-Dap, D-Dab, D-Asp, D-Glu, D-Lys or Asp;
x5 is Ala, Arg, Glu, Phe, Leu, Thr, Ser, Aib, Sarc, D-Ala, D-Arg, D-Glu, D-Phe, D-Leu, D-Thr, D-Ser, α -MeOrn, α -MeSer, Cit, Dap, Dab, Dap (Ac), Gly, Lys, Asn, N-Me-Gln, N-Me-Arg, Orn, or Gln;
x6 is Asp, Thr, Asn, Phe, D-Asp, D-Thr, D-Asn or D-Phe;
x8 is Val, Gln, Glu, or Lys;
x9 is Glu, Lys, Orn, Dap, Dab, D-Dap, D-Dab, D-Asp, D-Glu, D-Lys or Asp;
x10 is Tyr, Phe (3,4-F2), Phe (3,4-Cl2), F (3-Me), Phe [4- (2-aminoethoxy) ]Phe [4- (2- (acetyl-aminoethoxy)]Phe (4-Br), Phe (4-CONH2), Phe (4-Cl), Phe (4-CN), Phe (4-guanidino), Phe (4-Me), Phe (4-NH2), Phe (4-N)3) Phe (4-OMe), Phe (4-OBzl) orTyr;
X11 is Trp, 1-Nal, 2-Nal, Phe (3, 4-OMe)2) 5-hydroxy-Trp, Phe (3, 4-Cl)2) Or Tyr (3-t-Bu);
x12 is His, Phe, Arg, N-Me-His, Val, Cav, Cpa, Leu, Cit, hLeu, 3-Pal, t-butyl-Ala or t-butyl-Gly, 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Acvc, Agp, Aib, α -diethylGly, α -MeLys (Ac), α -Me-Leu, α -MeOrn, α -MeSer, α -MeVal, Cha, Cit, Cpa, (D) Asn, Glu, hArg or Lys;
x13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, Val, β hAla, Aib, Lys (Ac), Cit, Asp, Dab, Dap, Glu, hArg, Lys, Asn, Orn, or Gln;
x14 is Phe, Tyr,. beta.hPhe, Asn, Arg, Qln, Lys (Ac), His; dap (Ac), dab (Ac), or Asp;
x15 is Gly, Ser, Thr, Gln, Ala, Sarc, β -Ala, Glu, Arg, or Asn;
x16 is any amino acid or absent;
x17 is any amino acid or absent;
x18 is any amino acid or absent;
x19 is any amino acid or absent; and
X20 is any amino acid or is absent,
wherein X4 and X9 are optionally cyclized by intramolecular bond.
In certain embodiments of If: x5 is Ala, Arg, Glu, Phe, Leu, Thr, Ser, Aib, Sarc, D-Ala, D-Arg, D-Glu, D-Phe, D-Leu, D-Thr, D-Ser, D-Aib, or D-Sarc; x6 is Asp, Thr, Asn, Phe, D-Asp, D-Thr, D-Asn or D-Phe; x8 is Val, Gln, Glu, or Lys; x9 is Glu, Lys, Orn, Dap, Dab, D-Dap, D-Dab, D-Asp, D-Glu, D-Lys or Asp; x10 is Tyr or Phe; x11 is Trp, 1-Nal or 2-Nal; x12 is His, Phe, Arg, N-Me-His, Val, Cav, Cpa, Leu, Cit, hLeu, 3-Pal, t-butyl-Ala or t-butyl-Gly; x13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, Val, β hAla, or Aib; x14 is Phe, Tyr or beta hPhe; x15 is Gly, Ser, Thr, Gln, Ala, or Sarc; x16 is Asp, Glu, Ala, AEA, AEP,. beta.hAla, Gaba or deleted; and X17 is Leu, Lys, Arg or deleted.
In certain embodiments, the intramolecular bond is a lactam bond.
In certain embodiments of formula I, X comprises a sequence of formula Ig:
X1-X2-X3-X4-X5-X6-W-X8-X9-X10-X11-X12-X13-X14-X15-X16-X17-X18-X19-X20
(Ig)(SEQ ID NO:1362)
wherein
X1 is any amino acid or absent;
x2 is any amino acid or absent;
X3 is any amino acid or is absent;
x4 is β -azido-Ala-OH or propargylglycine;
x5 is Ala, Arg, Glu, Phe, Leu, Thr, Ser, Aib, Sarc, D-Ala, D-Arg, D-Glu, D-Phe, D-Leu, D-Thr, D-Ser, α -MeOrn, α -MeSer, Cit, Dap, Dab, Dap (Ac), Gly, Lys, Asn, N-MeGln, N-MeArg, Orn, or Gln;
x6 is Asp, Thr, Asn, Phe, D-Asp, D-Thr, D-Asn or D-Phe;
x8 is Val, Gln, Glu, or Lys;
x9 is β -azido-Ala-OH or propargylglycine;
x10 is Tyr, Phe (3, 4-F)2)、Phe(3,4-Cl2) F (3-Me), Phe [4- (2-aminoethoxy)]Phe [4- (2- (acetyl-aminoethoxy)]、Phe(4-Br)、Phe(4-CONH2) Phe (4-Cl), Phe (4-CN), Phe (4-guanidino), Phe (4-Me), Phe (4-NH)2)、Phe(4-N3) Phe (4-OMe), Phe (4-OBzl) or Tyr;
x11 is Trp, 1-Nal, 2-Nal, Phe (3, 4-OMe)2) 5-hydroxy-Trp, Phe (3, 4-Cl)2) Or Tyr (3-t-Bu);
x12 is His, Phe, Arg, N-Me-His, Val, Cav, Cpa, Leu, Cit, hLeu, 3-Pal, t-butyl-Ala or t-butyl-Gly, 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Acvc, Agp, Aib, α -diethylGly, α -MeLys (Ac), α -Me-Leu, α -MeOrn, α -MeSer, α -MeVal, Cha, Cit, Cpa, (D) Asn, Glu, hArg or Lys;
X13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, Val, β hAla, Aib, Lys (Ac), Cit, Asp, Dab, Dap, Glu, hArg, Lys, Asn, Orn, or Gln;
x14 is Phe, Tyr,. beta.hPhe, Asn, Arg, Qln, Lys (Ac), His; dap (Ac), dab (Ac), or Asp;
x15 is Gly, Ser, Thr, Gln, Ala or Sarc, β -Ala, Glu, Arg or Asn;
x16 is any amino acid or absent;
x17 is any amino acid or absent;
x18 is any amino acid or absent;
x19 is any amino acid or absent; and
x20 is any amino acid or is absent,
wherein X4 and X9 are optionally cyclized through an intramolecular triazole ring.
In a specific embodiment of Ig: x5 is Ala, Arg, Glu, Phe, Leu, Thr, Ser, Aib, Sarc, D-Ala, D-Arg, D-Glu, D-Phe, D-Leu, D-Thr, D-Ser, D-Aib, or D-Sarc; x6 is Asp, Thr, Asn, Phe, D-Asp, D-Thr, D-Asn or D-Phe; x8 is Val, Gln, Glu, or Lys; x9 is β -azido-Ala-OH or propargylglycine; x10 is Tyr or Phe; x11 is Trp, 1-Nal or 2-Nal; x12 is His, Phe, Arg, N-Me-His, Val, Cav, Cpa, Leu, Cit, hLeu, 3-Pal, t-butyl-Ala or t-butyl-Gly; x13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, Val, β hAla, or Aib; x14 is Phe, Tyr or beta hPhe; x15 is Gly, Ser, Thr, Gln, Ala, or Sarc; x16 is Asp, Glu, Ala, AEA, AEP,. beta.hAla, Gaba or deleted; and X17 is Leu, Lys, Arg or deleted.
In certain embodiments of formula I, X comprises a sequence of formula Ih:
X1-X2-X3-C-X5-X6-W-X8-C-Y-X11-H-X13-F-X15-X16-X17-X18-X19-X20
(Ih)(SEQ ID NO:1363)
wherein
X1 is any amino acid or absent;
x2 is any amino acid or absent;
x3 is any amino acid or absent;
x4 is 2-allylglycine, 2- (3 ' -butenyl) glycine, 2- (4 ' -pentenyl) glycine or 2- (5 ' -hexenyl) glycine;
x5 is Ala, Arg, Sarc, α -MeOrn, α -MeSer, Cit, Dap, Dab, Dap (Ac), Gly, Lys, Asn, N-MeGln, N-MeArg, Orn, or Gln;
x6 is Asp, Thr, or Asn;
x8 is Val, Gln, or Glu;
x9 is 2-allylglycine, 2- (3 ' -butenyl) glycine, 2- (4 ' -pentenyl) glycine or 2- (5 ' -hexenyl) glycine;
x11 is Trp, 1-Nal, 2-Nal, Phe (3, 4-OMe)2) 5-hydroxy-Trp, Phe (3, 4-Cl)2) Or Tyr (3-t-Bu);
x13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, β hAla, Val, Aib, Lys (Ac), Cit, Asp, Dab, Dap, Glu, hArg, Lys, Asn, Orn, or Gln;
x15 is Gly, Ser, Thr, Gln, Ala, Sarc, β -Ala, Glu, Arg, or Asn;
x16 is any amino acid or absent;
x17 is any amino acid or absent;
x18 is any amino acid or absent;
x19 is any amino acid or absent; and
X20 is any amino acid or is absent,
wherein X4 and X9 are optionally cyclized via intramolecular ring closing metathesis (ring closing metathesis) to produce the corresponding alkene.
In a specific embodiment of Ih: x5 is Ala, Arg, or Sarc; x6 is Asp, Thr, or Asn; x11 is Trp, 1-Nal or 2-Nal; x13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, β hAla, Val, or Aib; x15 is Gly, Ser, Thr, Gln, Ala, or Sarc; x16 is Asp, Glu, Ala, AEA, AEP,. beta.hAla, Gaba or deleted; and X17 is Leu, Lys, Arg or deleted.
In certain embodiments of formula I, X comprises the sequence of formula Ii:
X1-X2-X3-X4-X5-X6-W-X8-X9-X10-X11-X12-X13-X14-X15-X16-X17-X18-X19-X20(Ii)(SEQ ID NO:1364),
wherein:
x1 is any amino acid or absent;
x2 is any amino acid or absent;
x3 is any amino acid or absent;
x4 is Cys, Pen, hCys, D-Pen, D-Cys, D-hCys, 2-chloromethylbenzoic acid, mercaptopropionic acid, mercaptobutyric acid, 2-chloro-acetic acid, 3-chloro-propionic acid, 4-chloro-butyric acid or 3-chloro-isobutyric acid;
x5 is Ala, Arg, Glu, Phe, Leu, Thr, Ser, Aib, Sarc, D-Ala, D-Arg, D-Glu, D-Phe, D-Leu, D-Thr, D-Ser, D-Aib or D-Sarc, α -MeOrn, α -MeSer, Cit, Dap, Dab (Ac), Dap (Ac), Gly, Lys, Asn, N-MeGln, N-MeArg, Orn or Gln;
X6 is Asp, Thr, Asn, Phe, D-Asp, D-Thr, D-Asn or D-Phe;
x8 is Val, Gln, Glu, or Lys;
x9 is Cys, Pen, hCys, D-Pen, D-Cys, D-hCys or Abu;
x10 is Tyr, Phe (3, 4-F)2)、Phe(3,4-Cl2) F (3-Me), Phe [4- (2-aminoethoxy)]Phe [4- (2- (acetyl-aminoethoxy)]、Phe(4-Br)、Phe(4-CONH2) Phe (4-Cl), Phe (4-CN), Phe (4-guanidino), Phe (4-Me), Phe (4-NH)2)、Phe(4-N3) Phe (4-OMe), Phe (4-OBzl) or Tyr;
x11 is Trp, 1-Nal, 2-Nal, Phe (3, 4-OMe)2) 5-hydroxy-Trp, Phe (3, 4-Cl)2) Or Tyr (3-t-Bu);
x12 is His, Phe, Arg, N-Me-His, Val, Cav, Cpa, Leu, Cit, hLeu, 3-Pal, t-butyl-Ala, t-butyl-Gly, 4-amino-4-carboxy-tetrahydropyran, Achc Acpc, Acbc, Acvc, Agp, Aib, α -diethyl Gly, α -MeLys (Ac), α -Me-Leu, α -MeOrn, α -MeSer, α -MeVal, Cha, Cit, Cpa, (D) Asn, Glu, hArg, or Lys;
x13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, Val, β hAla, Aib, Lys (Ac), Cit, Asp, Dab, Dap, Glu, hArg, Lys, Asn, Orn, or Gln;
x14 is Phe, Tyr, beta hPhe, Asn, Arg, Qln, Lys (Ac), His; dap (Ac), dab (Ac), or Asp;
x15 is Gly, Ser, Thr, Gln, Ala, Sarc, β -Ala, Glu, Arg, or Asn;
X16 is any amino acid or absent;
x17 is any amino acid or absent;
x18 is any amino acid or absent;
x19 is any amino acid or absent; and
x20 is any amino acid or is absent,
wherein X4 and X9 are optionally cyclized via an intramolecular thioether bond.
In a specific embodiment of Ii: x5 is Ala, Arg, Glu, Phe, Leu, Thr, Ser, Aib, Sarc, D-Ala, D-Arg, D-Glu, D-Phe, D-Leu, D-Thr, D-Ser, D-Aib, or D-Sarc; x6 is Asp, Thr, Asn, Phe, D-Asp, D-Thr, D-Asn or D-Phe; x10 is Tyr or Phe; x11 is Trp, 1-Nal or 2-Nal; x12 is His, Phe, Arg, N-Me-His, Val, Cav, Cpa, Leu, Cit, hLeu, 3-Pal, t-butyl-Ala or t-butyl-Gly; x13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, Val, β hAla, or Aib; x14 is Phe, Tyr or beta hPhe; x15 is Gly, Ser, Thr, Gln, Ala, or Sarc; x16 is Asp, Glu, Ala, AEA, AEP,. beta.hAla, Gaba or deleted; and X17 is Leu, Lys, Arg or deleted.
In certain embodiments of formula I, X comprises a sequence of formula Ij:
X1-X2-X3-X4-X5-X6-W-X8-X9-X10-X11-X12-X13-X14-X15-X16-X17-X18-X19-X20(Ij)(SEQ ID NO:1365),
wherein:
x1 is any amino acid or absent;
x2 is any amino acid or absent;
X3 is any amino acid or is absent;
x4 is Sec, 2-chloromethylbenzoic acid, 3-chloro-propionic acid, 4-chloro-butyric acid, 3-chloro-isobutyric acid or Abu;
x5 is Ala, Arg, Glu, Phe, Leu, Thr, Ser, Aib, Sarc, D-Ala, D-Arg, D-Glu, D-Phe, D-Leu, D-Thr, D-Ser, α -MeOrn, α -MeSer, Cit, Dap, Dab, Dap (Ac), Gly, Lys, Asn, N-MeGln, N-MeArg, Orn, or Gln;
x6 is Asp, Thr, Asn, Phe, D-Asp, D-Thr, D-Asn or D-Phe;
x8 is Val, Gln, Glu or Lys;
x9 is Sec or Abu;
x10 is Tyr, Phe (3, 4-F)2)、Phe(3,4-Cl2) F (3-Me), Phe [4- (2-aminoethoxy)]Phe [4- (2-aminoethoxy), Phe [4- (2- (acetyl-aminoethoxy)]、Phe(4-Br)、Phe(4-CONH2) Phe (4-Cl), Phe (4-CN), Phe (4-guanidino), Phe (4-Me), Phe (4-NH)2)、Phe(4-N3) Phe (4-OMe), Phe (4-OBzl) or Tyr;
x11 is Trp, 1-Nal, 2-Nal, Phe (3, 4-OMe)2) 5-hydroxy-Trp, Phe (3, 4-Cl)2) Or Tyr (3-t-Bu);
x12 is His, Phe, Arg, N-Me-His, Val, Cav, Cpa, Leu, Cit, hLeu, 3-Pal, t-butyl-Ala, t-butyl-Gly, 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Agp, Aib, α -diethylGly, α -MeLys (Ac), α -Me-Leu, α -MeOrn, α -MeSer, α -MeVal, Cha, Cit, Cpa, (D) Asn, Glu, hArg, or Lys;
X13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, Val, β hAla, Aib, Lys (Ac), Cit, Asp, Dab, Dap, Glu, hArg, Lys, Asn, Orn, or Gln;
x14 is Phe, Tyr,. beta.hPhe, Asn, Arg, Qln, Lys (Ac), His; dap (Ac), dab (Ac), or Asp;
x15 is Gly, Ser, Thr, Gln, Ala, Sarc, β -Ala, Glu, Arg, or Asn;
x16 is any amino acid or absent;
x17 is any amino acid or absent;
x18 is any amino acid or absent;
x19 is any amino acid or absent; and
x20 is any amino acid or is absent,
wherein X4 and X9 are optionally cyclized via an intramolecular selenosulfonic or diselenic bond.
In specific embodiments of Ij: x5 is Ala, Arg, Glu, Phe, Leu, Thr, Ser, Aib, Sarc, D-Ala, D-Arg, D-Glu, D-Phe, D-Leu, D-Thr, D-Ser, D-Aib, or D-Sarc; x10 is Tyr or Phe; x11 is Trp, 1-Nal or 2-Nal; x12 is His, Phe, Arg, N-Me-His, Val, Cav, Cpa, Leu, Cit, hLeu, 3-Pal, t-butyl-Ala or t-butyl-Gly; x13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, Val, β hAla, or Aib; x14 is Phe, Tyr or beta hPhe; x15 is Gly, Ser, Thr, Gln, Ala, or Sarc; x16 is Asp, Glu, Ala, AEA, AEP,. beta.hAla, Gaba or deleted; and X17 is Leu, Lys, Arg or deleted.
In certain embodiments of formula I, X comprises a sequence of formula Ik:
X1-X2-X3-X4-X5-X6-W-X8-X9-X10-X11-X12-X13-X14-X15-X16-X17-X18-X19-X20(Ik)(SEQ ID NO:1366),
wherein
X1 is any amino acid or absent;
x2 is any amino acid or absent;
x3 is any amino acid or absent;
x4 is Cys, Pen, hCys, D-Pen, D-Cys, D-hCys, Met, Glu, Asp, Lys, Orn, Dap, Dab, D-Dap, D-Dab, D-Asp, D-Glu, D-Lys or deleted;
x5 is Ala, Arg, Glu, Phe, Leu, Thr, Ser, Aib, Sarc, D-Ala, D-Arg, D-Glu, D-Phe, D-Leu, D-Thr, D-Ser, α -MeOrn, α -MeSer, Cit, Dap, Dab, Dap (Ac), Gly, Lys, Asn, N-MeGln, N-MeArg, Orn, or Gln;
x6 is Asp, Thr, Asn, Phe, D-Asp, D-Thr, D-Asn or D-Phe;
x8 is Val, Gln, Glu, or Lys;
x9 is Cys, Pen, hCys, D-Pen, D-Cys, D-hCys, Glu, Lys, Orn, Dap, Dab, D-Dap, D-Dab, D-Asp, D-Glu, D-Lys, Asp, Leu, Val, Phe, or Ser;
x10 is Tyr, Phe (3, 4-F)2)、Phe(3,4-Cl2) F (3-Me), Phe [4- (2-aminoethoxy)]Phe [4- (2- (acetyl-aminoethoxy)]、Phe(4-Br)、Phe(4-CONH2) Phe (4-Cl), Phe (4-CN), Phe (4-guanidino), Phe (4-Me), Phe (4-NH)2)、Phe(4-N3) Phe (4-OMe), Phe (4-OBzl) or Tyr;
x11 is Trp, 1-Nal, 2-Nal, Phe (3, 4-OMe) 2) 5-hydroxy-Trp, Phe (3, 4-Cl)2) Or Tyr (3-t-Bu);
x12 is His, Phe, Arg, N-Me-His, Val, D-His, Cav, Cpa, Leu, Cit, hLeu, 3-Pal, t-butyl-Ala, t-butyl-Gly, 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Acvc, Agp, Aib, α -diethylGly, α -MeLys (Ac), α -Me-Leu, α -MeOrn, α -MeSer, α -MeVal, Cha, Cit, Cpa, (D) Asn, Glu, hArg, or Lys;
x13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, Val, β hAla, Aib, Lys (Ac), Cit, Asp, Dab, Dap, Glu, hArg, Lys, Asn, Orn, or Gln;
x14 is Phe, Tyr,. beta.hPhe, Asn, Arg, Qln, Lys (Ac), His; dap (Ac), dab (Ac) or Asp, or is absent;
x15 is Gly, Ser, Thr, Gln, Ala, Sarc, β -Ala, Glu, Arg, or Asn, or absent;
x16 is any amino acid or absent;
x17 is any amino acid or absent;
x18 is any amino acid or is absent;
x19 is any amino acid or absent; and
x20 is any amino acid or absent.
In a specific embodiment of Ik: x5 is Ala, Arg, Glu, Phe, Leu, Thr, Ser, Aib, Sarc, D-Ala, D-Arg, D-Glu, D-Phe, D-Leu, D-Thr, D-Ser, D-Aib, or D-Sarc; x10 is Tyr or Phe; x11 is Trp, 1-Nal or 2-Nal; x12 is His, Phe, Arg, N-Me-His, Val, D-His, Cav, Cpa, Leu, Cit, hLeu, 3-Pal, t-butyl-Ala or t-butyl-Gly; x13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, Val, β hAla, Aib, or deleted; x14 is Phe, Tyr, β hPhe or deleted; x15 is Gly, Ser, Thr, Gln, Ala, Sarc, or deleted; x16 is Asp, Glu, Ala, AEA, AEP,. beta.hAla, Gaba, Leu or deleted; and X17 is Leu, Lys, Arg or deleted.
In certain embodiments of formula I, X comprises or consists of the sequence of formula Il:
X1-X2-X3-X4-X5-X6-W-X8-X9-X10-X11-X12-X13-X14-X15-X16-X17-X18-X19-X20(Il)(SEQ ID NO:1367),
wherein
X1 is any amino acid or absent;
x2 is any amino acid or absent;
x3 is any amino acid or absent;
x4 is Cys, Pen, hCys, D-Pen, D-Cys, D-hCys, Met, Glu, Asp, Lys, Orn, Dap, Dab, D-Dap, D-Dab, D-Asp, D-Glu, D-Lys or deleted;
x5 is Ala, Arg, Glu, Phe, Leu, Thr, Ser, Aib, Sarc, α -MeOrn, α -MeSer, Cit, Dap, Dab, Dap (Ac), Gly, Lys, Asn, N-MeGln, N-MeArg, Orn, or Gln;
x6 is Asp, Thr, Asn or Phe;
x8 is Val, Gln, Glu, or Lys;
x9 is Cys, Pen, hCys, D-Pen, D-Cys, D-hCys, Glu, Lys, Orn, Dap, Dab, D-Dap, D-Dab, D-Asp, D-Glu, D-Lys, Asp, Leu, Val, Phe, or Ser;
x10 is Tyr, Phe (3, 4-F)2)、Phe(3,4-Cl2) F (3-Me), Phe [4- (2-aminoethoxy)]Phe [4- (2- (acetyl-aminoethoxy)]、Phe(4-Br)、Phe(4-CONH2) Phe (4-Cl), Phe (4-CN), Phe (4-guanidino), Phe (4-Me), Phe (4-NH)2)、Phe(4-N3) Phe (4-OMe), Phe (4-OBzl) or Tyr;
X11is Trp, 1-Nal, 2-Nal, Phe (3, 4-OMe)2) 5-hydroxy-Trp, Phe (3, 4-Cl)2) Or Tyr (3-t-Bu);
x12 is His, Phe, Arg, N-Me-His, Val, D-His, Cav, Cpa, Leu, Cit, hLeu, 3-Pal, t-butyl-Ala, t-butyl-Gly, 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Acvc, Agp, Aib, α -diethylGly, α -MeLys (Ac), α -Me-Leu, α -MeOrn, α -MeSer, α -MeVal, Cha, Cit, Cpa, (D) Asn, Glu, hArg, or Lys;
X13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, Val, β hAla, Aib, Lys (Ac), Cit, Asp, Dab, Dap, Glu, hArg, Lys, Asn, Orn, or Gln or deleted;
x14 is Phe, Tyr,. beta.hPhe, Asn, Arg, Qln, Lys (Ac), His; dap (Ac), dab (Ac) or Asp, or is absent;
x15 is Gly, Ser, Thr, Gln, Ala, Sarc, β -Ala, Glu, Arg, or Asn, or absent;
x16 is any amino acid or absent;
x17 is any amino acid or absent;
x18 is any amino acid or absent;
x19 is any amino acid or absent; and
x20 is any amino acid or absent.
In a specific embodiment of Il: x5 is Ala, Arg, Glu, Phe, Leu, Thr, Ser, Aib, or Sarc; x10 is Tyr or Phe; x11 is Trp, 1-Nal or 2-Nal; x12 is His, Phe, Arg, N-Me-His, Val, D-His, Cav, Cpa, Leu, Cit, hLeu, 3-Pal, t-butyl-Ala or t-butyl-Gly; x13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, Val, β hAla, Aib, or deleted; x14 is Phe, Tyr, β hPhe or deleted; x15 is Gly, Ser, Thr, Gln, Ala, Sarc, or deleted; x16 is Asp, Glu, Ala, AEA, AEP,. beta.hAla, Gaba, Leu or deleted; and X17 is Leu, Lys, Arg or deleted.
In certain embodiments, X13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, β hAla, Aib, Lys (Ac), Cit, Asp, Dab, Dap, Glu, hArg, Lys, Asn, Orn, or Gln. In certain embodiments, X13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, β hAla, or Aib.
In certain embodiments, X14 is Phe, Tyr, β hPhe, Asn, Arg, Qln, Lys (Ac), His; dap (Ac), dab (Ac), or Asp. In certain embodiments, X14 is Phe, Tyr, or β hPhe.
In certain embodiments, X15 is Gly, Ser, Thr, Gln, Ala, Sarc, β -Ala, Glu, Arg, or Asn. In certain embodiments, X15 is Gly, Ser, Thr, Gln, Ala, or Sarc.
In certain embodiments, X12 is an alpha amino acid, e.g., 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Aib, alpha-MeGly (diethyl), alpha-MeLys (Ac), alpha-Me-Leu, alpha-MeOrn, alpha-MeSer, alpha-MeVal.
In certain embodiments, X13 is present.
In certain embodiments, X13 and X14 are present.
In certain embodiments, X13, X14, and X15 are present.
In particular embodiments, X4 is present.
In certain embodiments, the peptide inhibitor is cyclized.
In certain embodiments, the peptide inhibitor is linear or not cyclized.
In certain embodiments, the peptide inhibitor is cyclized or contains an intramolecular bond between X4 and X9.
In certain embodiments of the peptide inhibitor of formula I, X comprises or consists of the sequence of formula Im:
X1-X2-X3-X4-X5-X6-W-X8-X9-Y-X11-X12-X13-X14-X15-X16-X17-X18-X19-X20(Im)(SEQ ID NO:1368),
wherein
X1 is any amino acid or absent;
x2 is any amino acid or absent;
x3 is any amino acid or absent;
x4 is Cys, Pen, hCys, D-Pen, D-Cys, D-hCys, Met, Glu, Asp, Lys, Orn, Dap, Dab, D-Dap, D-Dab, D-Asp, D-Glu, D-Lys or deleted;
x5 is Ala, Arg, Glu, Phe, Leu, Thr, Ser, Aib, Sarc, α -MeOrn, α -MeSer, Cit, Dap, Dab, Dap (Ac), Gly, Lys, Asn, N-MeGln, N-MeArg, Orn, or Gln;
x6 is Asp, Thr, Asn or Phe;
x8 is Val, Gln, Glu, or Lys;
x9 is Cys, Pen, hCys, D-Pen, D-Cys, D-hCys, Glu, Lys, Orn, Dap, Dab, D-Dap, D-Dab, D-Asp, D-Glu, D-Lys, Asp, Leu, Val, Phe, or Ser;
x11 is Trp, 1-Nal, 2-Nal, Phe (3, 4-OMe)2) (ii) a 5-hydroxy-Trp, Phe (3, 4-Cl)2) Or Tyr (3-t-Bu);
x12 is His, Phe, Arg, N-Me-His, Val, Cav, Cpa, Leu, Cit, hLeu, 3-Pal, t-butyl-Ala or t-butyl-Gly, 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Acvc, Agp, Aib, α -diethylGly, α -MeLys (Ac), α -Me-Leu, α -MeOrn, α -MeSer, α -MeVal, Cha, Cit, Cpa, (D) Asn, Glu, hArg or Lys;
X13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, β hAla, Val, Aib, Lys (Ac), Cit, Asp, Dab, Dap, Glu, hArg, Lys, Asn, Orn, or Gln or deleted;
x14 is Phe, Tyr,. beta.hPhe, Asn, Arg, Qln, Lys (Ac), His; dap (ac), dab (ac), or Asp or absent;
x15 is Gly, Ser, Thr, Gln, Ala, Sarc, β -Ala, Glu, Arg, or Asn, or absent;
x16 is any amino acid or absent;
x17 is any amino acid or absent;
x18 is any amino acid or absent;
x19 is any amino acid or absent; and
x20 is any amino acid or absent.
In certain embodiments of Im: x5 is Ala, Arg, Glu, Phe, Leu, Thr, Ser, Aib, or Sarc; x11 is Trp, 1-Nal or 2-Nal; x12 is His, Phe, Arg, N-Me-His, Val, Cav, Cpa, Leu, Cit, hLeu, 3-Pal, t-butyl-Ala or t-butyl-Gly; x13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, β hAla, Val, Aib, or deleted; x14 is Phe, Tyr, β hPhe or deleted; x15 is Gly, Ser, Thr, Gln, Ala, Sarc, or deleted; x16 is Asp, Glu, Ala, AEA, AEP,. beta.hAla, Gaba or deleted; and X17 is Leu, Lys, Arg or deleted.
In certain embodiments, X13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, β hAla, or Aib. In certain embodiments, X13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, β hAla, Aib, Lys (Ac), Cit, Asp, Dab, Dap, Glu, hArg, Lys, Asn, Orn, or Gln.
In certain embodiments, X14 is Phe, Tyr, β hPhe, Asn, Arg, Qln, Lys (Ac), His; dap (Ac), dab (Ac) or Asp. In certain embodiments, X14 is Phe, Tyr, or β hPhe.
In certain embodiments, X15 is Gly, Ser, Thr, Gln, Ala or Sarc, β -Ala, Glu, Arg, or Asn. In certain embodiments, X15 is Gly, Ser, Thr, Gln, Ala, or Sarc.
In certain embodiments, X12 is an alpha amino acid, e.g., 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Aib, alpha-MeGly (diethyl), alpha-MeLys (Ac), alpha-Me-Leu, alpha-MeOrn, alpha-MeSer, alpha-MeVal.
In certain embodiments, X13 is present.
In certain embodiments, X13 and X14 are present.
In certain embodiments, X13, X14, and X15 are present.
In particular embodiments, X14 is present.
In certain embodiments, the peptide inhibitor is cyclized.
In certain embodiments, the peptide inhibitor is linear or not cyclized.
In certain embodiments, the peptide inhibitor is cyclized or contains an intramolecular bond between X4 and X9.
In certain embodiments of the peptide inhibitor of formula I, X comprises or consists of a sequence of formula In:
X1-X2-X3-C-X5-X6-W-X8-C-X10-X11-X12-X13-X14-X15-X16-X17-X18-X19-X20(In)(SEQ ID NO:1369)
wherein
X1 is any amino acid or is absent;
x2 is any amino acid or absent;
x3 is any amino acid or absent;
x5 is Ala, Arg, Glu, Phe, Leu, Thr, Ser, Aib, Sarc, α -MeOrn, α -MeSer, Cit, Dap, Dab, Dap (Ac), Gly, Lys, Asn, N-MeGln, N-MeArg, Orn, or Gln;
x6 is Asp, Thr, Asn or Phe;
x8 is Val, Gln, Glu, or Lys;
x10 is Tyr, Phe (3, 4-F)2)、Phe(3,4-Cl2) F (3-Me), Phe [4- (2-aminoethoxy)]Phe [4- (2- (acetyl-aminoethoxy)]、Phe(4-Br)、Phe(4-CONH2) Phe (4-Cl), Phe (4-CN), Phe (4-guanidino), Phe (4-Me), Phe (4-NH)2)、Phe(4-N3) Phe (4-OMe), Phe (4-OBzl) or Tyr;
x11 is Trp, 1-Nal, 2-Nal, Phe (3, 4-OMe)2) 5-hydroxy-Trp, Phe (3, 4-Cl)2) Or Tyr (3-t-Bu);
x12 is His, Phe, Arg, N-Me-His, Val, Cav, Cpa, Leu, Cit, hLeu, 3-Pal, t-butyl-Ala or t-butyl-Gly, 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Acvc, Agp, Aib, α -diethylGly, α -MeLys (Ac), α -Me-Leu, α -MeOrn, α -MeSer, α -MeVal, Cha, Cit, Cpa, (D) Asn, Glu, hArg or Lys;
X13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, β hAla, Val, Aib, Lys (Ac), Cit, Asp, Dab, Dap, Glu, hArg, Lys, Asn, Orn, or Gln or deleted;
x14 is Phe, Tyr,. beta.hPhe, Asn, Arg, Qln, Lys (Ac), His; dap (Ac), dab (Ac) or Asp, or is absent;
x15 is Gly, Ser, Thr, Gln, Ala, Sarc, β -Ala, Glu, Arg, or Asn, or absent;
x16 is any amino acid or absent;
x17 is any amino acid or is absent;
x18 is any amino acid or absent;
x19 is any amino acid or absent; and
x20 is any amino acid or is absent,
wherein the Cys at X4 and the Cys at X9 are optionally linked by a disulfide bridge.
In certain embodiments of In: x5 is Ala, Arg, Glu, Phe, Leu, Thr, Ser, Aib, Sarc, α -MeOrn, α -MeSer, Cit, Dap, Dab, Dap (Ac), Gly, Lys, Asn, N-MeGln, N-MeArg, Orn, or Gln; x10 is Tyr, Phe (3, 4-F)2)、Phe(3,4-Cl2) F (3-Me), Phe [4- (2-aminoethoxy)]Phe [4- (2- (acetyl-aminoethoxy)]、Phe(4-Br)、Phe(4-CONH2) Phe (4-Cl), Phe (4-CN), Phe (4-guanidino), Phe (4-Me), Phe (4-NH)2)、Phe(4-N3) Phe (4-OMe), Phe (4-OBzl) or Tyr; x11 is Trp, 1-Nal, 2-Nal, Phe (3, 4-OMe) 2) 5-hydroxy-Trp, Phe (3, 4-Cl)2) Or Tyr (3-t-Bu); x12 is His, Phe, Arg, N-Me-His, Val, Cav, Cpa, Leu, Cit, hLeu, 3-Pal, t-butyl-Ala or t-butyl-Gly, 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Acvc, Agp, Aib, α -diethylGly, α -MeLys (Ac), α -Me-Leu, α -MeOrn, α -MeSer, α -MeVal, Cha, Cit, Cpa, (D) Asn, Glu, hArg or Lys; x13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, α 0hAla, Val, Aib, Lys (Ac), Cit, Asp, Dab, Dap, Glu, hArg, Lys, Asn, Orn, or Gln or deleted; x14 is Phe, Tyr,. beta.hPhe, Asn, Arg, Qln, Lys (Ac), His; dap (Ac), dab (Ac) or Asp, or is absent; x15 is Gly, Ser, Thr, Gln, Ala, Sarc, β -Ala, Glu, Arg, or Asn, or absent; x16 is Asp, Glu, Ala, AEA, AEP,. beta.hAla, Gaba or deleted; and X17 is Leu, Lys, Arg or deleted.
In certain embodiments, X13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, β hAla, Aib, Lys (Ac), Cit, Asp, Dab, Dap, Glu, hArg, Lys, Asn, Orn, or Gln. In certain embodiments, X13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, β hAla, or Aib.
In certain embodiments, X14 is Phe, Tyr, β hPhe, Asn, Arg, Qln, Lys (Ac), His; dap (Ac), dab (Ac), or Asp. In certain embodiments, X14 is Phe, Tyr, or β hPhe.
In certain embodiments, X15 is Gly, Ser, Thr, Gln, Ala, Sarc, β -Ala, Glu, Arg, or Asn. In certain embodiments, X15 is Gly, Ser, Thr, Gln, Ala, or Sarc.
In certain embodiments, X12 is an alpha amino acid, e.g., 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Acvc, Aib, alpha-diethylGly, alpha-MeLys (Ac), alpha-Me-Leu, alpha-MeOrn, alpha-MeSer, alpha-MeVal.
In certain embodiments, X13 is present.
In certain embodiments, X13 and X14 are present.
In certain embodiments, X13, X14, and X15 are present.
In certain embodiments of the peptide inhibitor of formula I, X comprises or consists of a sequence of formula Io:
X1-X2-X3-C-X5-X6-W-X8-C-Y-X11-H-X13-X14-X15-X16-X17-X18-X19-X20(Io)(SEQ ID NO:1370)
wherein
X1 is any amino acid or absent;
x2 is any amino acid or absent;
x3 is any amino acid or absent;
x5 is Ala, Arg, Glu, Phe, Leu, Thr, Ser, Aib, Sarc, α -MeOrn, α -MeSer, Cit, Dap, Dab, Dap (Ac), Gly, Lys, Asn, N-MeGln, N-MeArg, Orn, or Gln;
X6 is Asp, Thr, Asn or Phe;
x8 is Val, Gln, Glu, or Lys;
x11 is Trp, 1-Nal, 2-Nal、Phe(3,4-OMe2) 5-hydroxy-Trp, Phe (3, 4-Cl)2) Or Tyr (3-t-Bu);
x13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, β hAla, Val, Aib, Lys (Ac), Cit, Asp, Dab, Dap, Glu, hArg, Lys, Asn, Orn, or Gln or deleted;
x14 is Phe, Tyr, Asn, Arg, Qln, Lys (Ac), His; dap (Ac), dab (Ac) or Asp, or is absent;
x15 is Gly, Ser, Thr, Gln, Ala, Sarc, β -Ala, Glu, Arg, or Asn, or absent;
x16 is any amino acid or absent;
x17 is any amino acid or absent;
x18 is any amino acid or absent;
x19 is any amino acid or absent; and
x20 is any amino acid or is absent,
wherein the Cys at X4 and the Cys at X9 are optionally linked by a disulfide bridge.
In certain embodiments of Io: x5 is Ala, Arg, Glu, Phe, Leu, Thr, Ser, Aib, Sarc, α -MeOrn, α -MeSer, Cit, Dap, Dab, Dap (Ac), Gly, Lys, Asn, N-MeGln, N-MeArg, Orn, or Gln; x11 is Trp, 1-Nal, 2-Nal, Phe (3, 4-OMe)2) 5-hydroxy-Trp, Phe (3, 4-Cl)2) Or Tyr (3-t-Bu); x13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, β hAla, Val, Aib, Lys (Ac), Cit, Asp, Dab, Dap, Glu, hArg, Lys, Asn, Orn, Gln, or deleted; x14 is Phe, Tyr, Asn, Arg, Qln, Lys (Ac), His; dap (ac), dab (ac), Asp, or absent; x15 is Gly, Ser, Thr, Gln, Ala, Sarc, β -Ala, Glu, Arg, or Asn, or absent; x16 is Asp, Glu, Ala, AEA, AEP,. beta.hAla, Gaba or deleted; and X17 is Leu, Lys, Arg or deleted.
In certain embodiments, X12 is an alpha amino acid, e.g., 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Acvc, Aib, alpha-diethylGly, alpha-MeLys (Ac), alpha-Me-Leu, alpha-MeOrn, alpha-MeSer, alpha-MeVal.
In certain embodiments, X13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, β hAla, Aib, Lys (Ac), Cit, Asp, Dab, Dap, Glu, hArg, Lys, Asn, Orn, or Gln. In certain embodiments, X13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, β hAla, or Aib.
In certain embodiments, X14 is Phe, Tyr, Asn, Arg, Qln, Lys (Ac), His; dap (Ac), dab (Ac), or Asp. In certain embodiments, X14 is Phe or Tyr.
In certain embodiments, X15 is Gly, Ser, Thr, Gln, Ala, Sarc, β -Ala, Glu, Arg, or Asn. In certain embodiments, X15 is Gly, Ser, Thr, Gln, Ala, or Sarc.
In certain embodiments, X13 is present.
In certain embodiments, X13 and X14 are present.
In certain embodiments, X13, X14, and X15 are present.
In certain embodiments of the peptide inhibitor of formula I, X comprises or consists of a sequence of formula Ip:
X1-X2-X3-C-X5-X6-W-X8-C-Y-X11-H-X13-F-X15-X16-X17-X18-X19-X20(Ip)(SEQ ID NO:1371)
Wherein
X1 is any amino acid or absent;
x2 is any amino acid or absent;
x3 is any amino acid or absent;
x5 is Ala, Arg, Sarc, α -MeOrn, α -MeSer, Cit, Dap, Dab, Dap (Ac), Gly, Lys, Asn, N-MeGln, N-MeArg, Orn, or Gln;
x6 is Asp, Thr, or Asn;
x8 is Val, Gln, or Glu;
x11 is Trp, 1-Nal, 2-Nal, Phe (3, 4-OMe)2) 5-hydroxy-Trp, Phe (3, 4-Cl)2) Or Tyr (3-t-Bu);
x13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, β hAla, Val, Aib, Lys (Ac), Cit, Asp, Dab, Dap, Glu, hArg, Lys, Asn, Orn, Gln, or deleted;
x15 is Gly, Ser, Thr, Gln, Ala, Sarc, β -Ala, Glu, Arg, Asn, or deleted;
x16 is any amino acid or absent;
x17 is any amino acid or absent;
x18 is any amino acid or absent;
x19 is any amino acid or absent; and
x20 is any amino acid or is absent,
wherein the Cys at X4 and the Cys at X9 are optionally linked by a disulfide bridge.
In certain embodiments of Ip: x5 is Ala, Arg, or Sarc; x11 is Trp, 1-Nal or 2-Nal; x13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, β hAla, Val, Aib, or deleted; x15 is Gly, Ser, Thr, Gln, Ala, Sarc, or deleted; x16 is Asp, Glu, Ala, AEA, AEP,. beta.hAla, Gaba or deleted; and X17 is Leu, Lys, Arg or deleted.
In certain embodiments, X13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, β hAla, Aib, Lys (Ac), Cit, Asp, Dab, Dap, Glu, hArg, Lys, Asn, Orn, or Gln. In certain embodiments, X13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, β hAla, or Aib.
In certain embodiments, X15 is Gly, Ser, Thr, Gln, Ala or Sarc, β -Ala, Glu, Arg, or Asn. In certain embodiments, X15 is Gly, Ser, Thr, Gln, Ala, or Sarc.
In certain embodiments, X13 is present.
In certain embodiments, X13 and X14 are present.
In certain embodiments, X13, X14, and X15 are present.
In certain embodiments of the peptide inhibitor of formula I, X comprises or consists of the sequence of formula Iq:
X1-X2-X3-C-X5-X6-W-X8-C-X10-X11-X12-X13-X14-X15-X16-X17-X18-X19-X20(Iq)(SEQ ID NO:1372),
wherein
X1 is any amino acid or absent;
x2 is any amino acid or absent;
x3 is any amino acid or absent;
x5 is Ala, Arg, Glu, Phe, Leu, Thr, Ser, Aib, Sarc, D-Ala, D-Arg, D-Glu, D-Phe, D-Leu, D-Thr, D-Ser, D-Aib, D-Sarc, α -MeOrn, α -MeSer, Cit, Dap, Dab (Ac), Dap (Ac), Gly, Lys, Asn, N-MeGln, N-MeArg, Orn, or Gln;
X6 is Asp, Thr, Asn, Phe, D-Asp, D-Thr, D-Asn or D-Phe;
x8 is Val, Gln, Glu, or Lys;
x10 is Tyr, Phe (3, 4-F)2)、Phe(3,4-Cl2) F (3-Me), Phe [4- (2-aminoethoxy)]Phe [4- (2- (acetyl-aminoethoxy)]、Phe(4-Br)、Phe(4-CONH2) Phe (4-Cl), Phe (4-CN), Phe (4-guanidino), Phe (4-Me), Phe (4-NH)2)、Phe(4-N3) Phe (4-OMe), Phe (4-OBzl) or Tyr;
x11 is Trp, 1-Nal, 2-Nal, Phe (3, 4-OMe)2) 5-hydroxy-Trp, Phe (3, 4-Cl)2) Or Tyr (3-t-Bu);
x12 is His, Phe, Arg, N-Me-His, Val or D-His, Cav, Cpa, Leu, Cit, hLeu, 3-Pal, t-butyl-Ala, t-butyl-Gly, 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Acvc, Agp, Aib, α -diethylGly, α -MeLys (Ac), α -Me-Leu, α -MeOrn, α -MeSer, α -MeVal, Cha, Cit, Cpa, (D) Asn, Glu, hArg or Lys;
x13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, β hAla, Val, Aib, Lys (Ac), Cit, Asp, Dab, Dap, Glu, hArg, Lys, Asn, Orn, Gln, or deleted;
x14 is Phe, Tyr,. beta.hPhe, Asn, Arg, Qln, Lys (Ac), His; dap (ac), dab (ac), Asp, or absent;
x15 is Gly, Ser, Thr, Gln, Ala, Sarc, β -Ala, Glu, Arg, Asn, or deleted;
X16 is any amino acid or is absent;
x17 is any amino acid or absent;
x18 is any amino acid or absent;
x19 is any amino acid or is absent; and
x20 is any amino acid or is absent,
wherein the Cys at position X4 and the Cys at position X9 are optionally linked.
In certain embodiments of Iq: x5 is Ala, Arg, Glu, Phe, Leu, Thr, Ser, Aib, Sarc, D-Ala, D-Arg, D-Glu, D-Phe, D-Leu, D-Thr, D-Ser, D-Aib, or D-Sarc; x10 is Tyr or Phe; x11 is Trp, 1-Nal or 2-Nal; x12 is His, Phe, Arg, N-Me-His, Val or D-His, Cav, Cpa, Leu, Cit, hLeu, 3-Pal, t-butyl-Ala or t-butyl-Gly; x13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, β hAla, Val, Aib, or deleted; x14 is Phe, Tyr, β hPhe or deleted; x15 is Gly, Ser, Thr, Gln, Ala, Sarc, or deleted; x16 is Asp, Glu, Ala, AEA, AEP,. beta.hAla, Gaba, Leu or deleted; and X17 is Leu, Lys, Arg or deleted.
In certain embodiments, X12 is an alpha amino acid, e.g., 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Acvc, Aib, alpha-diethylGly, alpha-MeLys (Ac), alpha-Me-Leu, alpha-MeOrn, alpha-MeSer, alpha-MeVal.
In certain embodiments, X13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, β hAla, Aib, Lys (Ac), Cit, Asp, Dab, Dap, Glu, hArg, Lys, Asn, Orn, or Gln. In certain embodiments, X13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, β hAla, or Aib.
In certain embodiments, X14 is Phe, Tyr, β hPhe, Asn, Arg, Qln, Lys (Ac), His; dap (Ac), dab (Ac), or Asp. In certain embodiments, X14 is Phe, Tyr, or β hPhe.
In certain embodiments, X15 is Gly, Ser, Thr, Gln, Ala, Sarc, β -Ala, Glu, Arg, or Asn. In certain embodiments, X15 is Gly, Ser, Thr, Gln, Ala, or Sarc.
In certain embodiments, X13 is present.
In certain embodiments, X13 and X14 are present.
In certain embodiments, X13, X14, and X15 are present.
In certain embodiments, Iq comprises or consists of a sequence of formula Iq':
X1-X2-X3-C-X5-X6-W-X8-C-X10-X11-X12-X13-X14-X15(Iq’)(SEQ ID NO:1373),
wherein X1-X14 have the definitions provided for Iq, and
wherein Cys at position X4 and Cys at position X9 are optionally linked.
In certain embodiments of Iq': x5 is Ala, Arg, Glu, Phe, Leu, Thr, Ser, Aib, Sarc, D-Ala, D-Arg, D-Glu, D-Phe, D-Leu, D-Thr, D-Ser, D-Aib, or D-Sarc; x10 is Tyr or Phe; x11 is Trp, 1-Nal or 2-Nal; x12 is His, Phe, Arg, N-Me-His, Val or D-His, Cav, Cpa, Leu, Cit, hLeu, 3-Pal, t-butyl-Ala or t-butyl-Gly; x13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, β hAla, Val, Aib, or deleted; x14 is Phe, Tyr, β hPhe or deleted; x15 is Gly, Ser, Thr, Gln, Ala, Sarc, or deleted; x16 is Asp, Glu, Ala, AEA, AEP,. beta.hAla, Gaba, Leu or deleted; and X17 is Leu, Lys, Arg or deleted.
In certain embodiments, X13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, β hAla, Aib, Lys (Ac), Cit, Asp, Dab, Dap, Glu, hArg, Lys, Asn, Orn, or Gln. In certain embodiments, X13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, β hAla, or Aib.
In certain embodiments, X14 is Phe, Tyr, β hPhe, Asn, Arg, Qln, Lys (Ac), His; dap (Ac), dab (Ac), or Asp. In certain embodiments, X14 is Phe, Tyr, or β hPhe.
In certain embodiments, X15 is Gly, Ser, Thr, Gln, Ala or Sarc, β -Ala, Glu, Arg, or Asn. In certain embodiments, X14 is Phe, Tyr, or β hPhe.
In certain embodiments, X13 is present.
In certain embodiments, X13 and X14 are present.
In certain embodiments, X13, X14, and X15 are present.
In certain embodiments of the peptide inhibitor of formula I, X comprises or consists of a sequence of formula Ir:
X1-X2-X3-X4-X5-X6-X7-X8-X9-X10-X11-X12-X13-X14-X15-X16-X17-X18-X19-X20(Ir)(SEQ ID NO:1374)
wherein
X1 is any amino acid or absent;
x2 is any amino acid or absent;
x3 is any amino acid or absent;
x4 is Cys, Pen, hCys, D-Pen, D-Cys, D-hCys, Met, Glu, Asp, Lys, Orn, Dap, Dab, D-Dap, D-Dab, D-Asp, D-Glu, D-Lys, Sec, 2-chloromethylbenzoic acid, mercaptopropionic acid, mercaptobutyric acid, 2-chloro-acetic acid, 3-chloro-propionic acid, 4-chloro-butyric acid, 3-chloro-isobutyric acid, Abu, β -azido-Ala-OH, propargylglycine, 2- (3 '-butenyl) glycine, 2-allylglycine, 2- (3' -butenyl) glycine, 2- (4 '-pentenyl) glycine, 2- (5' -hexenyl) glycine, Abu or deleted;
X5 is any amino acid;
x6 is any amino acid;
x7 is Trp, Glu, Gly, Ile, Asn, Pro, Arg, Thr, or OctGly or any of the corresponding α -methyl amino acid forms;
x8 is any amino acid;
x9 is Cys, Pen, hCys, D-Pen, D-Cys, D-hCys, Glu, Lys, Orn, Dap, Dab, D-Dap, D-Dab, D-Asp, D-Glu, D-Lys, Asp, Leu, Val, Phe or Ser, Sec, Abu, β -azido-Ala-OH, propargylglycine, 2-2-allylglycine, 2- (3 ' -butenyl) glycine, 2- (4 ' -pentenyl) glycine, Ala, hCys, Abu, Met, MeCys, (D) Tyr or 2- (5 ' -hexenyl) glycine;
x10 is Tyr, Phe (4-OMe), 1-Nal, 2-Nal, Aic, alpha-MePhe, Bip, (D) Cys, Cha, DMT, (D) Tyr, Glu, His, hPhe (3, 4-dimethoxy), hTyr, N-Me-Tyr, Trp, Phe (4-CONH)2) Phe (4-phenoxy), Thr、Tic、Tyr(3-tBu)、Phe(4-tBu)、Phe(4-CN)、Phe(4-Br)、Phe(4-NH2)、Phe(4-F)、Phe(3,5-F2)、Phe(4-CH2CO2H)、Phe(5-F)、Phe(3,4-Cl2)、Phe(4-CF3) Bip, Cha, 4-pyridylalanine, beta hTyr, OctGly, Phe (4-N)3) Phe (4-Br), Phe [4- (2-aminoethoxy)]Or a Phe, Phe analog, Tyr analog, or any of the foregoing corresponding alpha-methyl amino acid forms;
x11 is 2-Nal, 1-Nal, 2, 4-dimethyl Phe, Bip, Phe (3, 4-Cl)2)、Phe(3,4-F2)、Phe(4-CO2H) Beta hPhe (4-F), alpha-Me-Trp, 4-phenylcyclohexyl, Phe (4-CF) 3)、、Phe(3,4-OMe2) alpha-MePhe, beta hNal, beta hPhe, beta hTyr, beta hTrp, Nva (5-phenyl), Phe, His, hPhe, Tic, Tqa, Trp, Tyr, Phe (4-OMe), Phe (4-Me), Trp (2,5, 7-tri-t-butyl), Phe (4-O allyl), Tyr (3-tBu), Phe (4-guanidino, Phe (4-OBzl), Octgly, Glu (Bzl), 4-phenylbenzylalanine, Phe [4- (2-aminoethoxy)]5-hydroxy-Trp, 6-chloro-Trp, N-MeTrp, 1,2,3, 4-tetrahydro-norharman, Phe (4-CONH)2) Phe (3, 4-dimethoxy), Phe (2, 3-Cl)2)、Phe(2,3-F2) Phe (4-F), 4-phenylcyclohexylalanine or Bip or any of the corresponding alpha-methyl amino acid forms described above;
x12 is His, Phe, Arg, N-Me-His or Val, Cav, Cpa, Leu, Cit, hLeu, 3-Pal, t-butyl-Ala, α -MeLys, D-Ala, (D) Asn, (D) Asp, (D) Leu, (D) Phe, (D) Tyr, Aib, α -MeLeu, α -MeOrn, β -Aib, β -Ala, β -hAla, β -hArg, β -hLeu, β -hVal, β -spiro-pip, Glu, hArg, Ile, Lys, N-MeLeu, N-MeArg, og, Orn, Pro, Gln, Ser, Thr, Tle or t-butyl-Gly, 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Acvc, Agp, Ser, Aib, α -diethylGly, α -MeAc, (α -Lys, α -Me, α -Val, α -Orn, MeVal, α -MeAc, and, Cha, Cit, Cpa, (D) Asn, Glu, hArg, or Lys, or any of the corresponding α -methyl amino acid forms described above;
X13 is Thr, Sarc, Glu, Phe, Arg, Leu, Asn, Cit, Lys, Arg, Orn, Val, β hAla, Lys (Ac), (D) Asn, (D) Leu, (D) Phe, (D) Thr, Ala, α -MeLeu, Aib, β -Ala, β -Glu, β hLeu, β hVal, β -spiro-pip, Cha, Chg, Asp, Dab, Dap, α -diethylGly, hLeu, Asn, Ogl, Pro, Gln, Ser, β -spiro-pip, Thr, Tba, Tle, or Aib, or any of the corresponding α -methyl amino acid forms described above;
x14 is Phe, Tyr, Glu, Gly, His, Lys, Leu, Met, Asn, Lys (Ac), dap (Ac), Asp, Pro, Gln, Arg, Ser, Thr, Tic, or β hPhe, or any of the corresponding α -methyl amino acid forms;
x15 is Gly, Ser, Thr, Gln, Ala, (D) Asn, (D) Asp, (D) Leu, (D) Phe, (D) Thr, Aea, Asp, Asn, Glu, Phe, Gly, Lys, Leu, Pro, Arg, β -Ala or Sarc or any of the corresponding α -methyl amino acid forms;
x16 is any amino acid or is absent;
x17 is any amino acid or absent;
x18 is any amino acid or absent;
x19 is any amino acid or absent; and
x20 is any amino acid or absent.
In particular embodiments, the peptide is cyclized via X4 and X9.
In a specific embodiment, X3 is Glu, D-Glu, Arg, (D) Arg, Phe, (D) Phe, 2-Nal, Thr, Leu, (D) Gln.
In certain embodiments of Ir: x11 is 2-Nal, 1-Nal, 2, 4-dimethyl Phe, Bip, Phe (3, 4-Cl)2)、Phe(3,4-F2)、Phe(4-CO2H) Beta hPhe (4-F), alpha-Me-Trp, 4-phenylcyclohexyl, Phe (4-CF)3) alpha-MePhe, beta hNal, beta hPhe, beta hTyr, beta hUrp, Nva (5-phenyl), Phe, His, hPhe, Tic, Tqa, Trp, Tyr, Phe (4-OMe), Phe (4-Me), Trp (2,5, 7-tri-tert-butyl), Phe (4-O allyl), Tyr (3-tBu), Phe (4-guanidino, Phe (4-OBzl), Octgly, Glu (Bzl), 4-phenylbenzylalanine, Phe [4- (2-aminoethoxy)]5-hydroxy-Trp, 6-chloro-Trp, N-MeTrp, 1,2,3, 4-tetrahydro-norhaben, Phe (4-CONH)2) Phe (3, 4-dimethoxy))、Phe(2,3-Cl2)、Phe(2,3-F2) Phe (4-F), 4-phenylcyclohexylalanine or Bip or any of the corresponding alpha-methyl amino acid forms described above; x12 is His, Phe, Arg, N-Me-His or Val, Cav, Cpa, Leu, Cit, hLeu, 3-Pal, t-butyl-Ala, α -MeLys, D-Ala, (D) Asn, (D) Asp, (D) Leu, (D) Phe, (D) Tyr, Aib, α 1-MeLeu, α 3-MeOrn, α 0-Aib, α 2-Ala, α 4hAla, α 5hArg, α 6hLeu, α 7hVal, α 8-spiro-pip, Glu, hArg, Ile, Lys, N-MeLeu, N-MeArg, Ogl, Orn, Pro, Gln, Ser, Thr, Tle, or t-butyl-Gly or any of the corresponding α 9-methylamino forms described above; x13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, Arg, Orn, Val, α 0hAla, Lys (Ac), (D) Asn, (D) Leu, (D) Phe, (D) Thr, Ala, α -MeLeu, Aib, α 2-Ala, α 5-Glu, β hLeu, β hVal, β -spiro-pip, Cha, Chg, Asp, Dab, Dap, α 1-diethylGly, hLeu, Asn, Ogl, Pro, Gln, Ser, β -spiro-pip, Thr, Tba, Tle or Aib, or any of the corresponding α 3-methylamino forms described above; x14 is Phe, Tyr, Glu, Gly, His, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Tic, or β hPhe, or any of the foregoing corresponding α 4-methyl amino acid forms; x15 is Gly, Ser, Thr, Gln, Ala, (D) Asn, (D) Asp, (D) Leu, (D) Phe, (D) Thr, Aea, Asp, Asn, Glu, Phe, Gly, Lys, Leu, Pro, Arg, or Sarc, or any of the corresponding α 6-methyl amino acid forms; x16 is Asp, Glu, Ala, AEA, AEP,. beta.hAla, Gaba, Gly, Ser, Pro, Asn, Thr or the absence or any of the foregoing corresponding α 7-methyl amino acid forms; and X17 is Leu, Lys, Arg, Glu, Ser, Gly, Gln or absent or any of the corresponding α -methyl amino acid forms.
In certain embodiments, both X4 and X9 are Pen. In particular embodiments, X4 and X9 are cyclized via a disulfide bond.
In certain embodiments, X4 is Abu and X9 is Cys. In certain embodiments, X4 and X9 are cyclized via a thioether bond.
In a specific embodiment, X5 is Ala, Arg, Glu, Phe, Leu, Thr, Ser, Aib, Sarc, D-Ala, D-Arg, D-Glu, D-Phe, D-Leu, D-Thr, D-Ser, D-Aib, Cys, Cit, Asp, Dab, Dap, Gly, His, hCys, Lys, Met, Asn, N-Me-Ala, N-Me-Asn, N-Me-Lys, N-Me-Gln, Orn, Pro, Pen, Gln, Val, α Me-Lys, α Me-Orn, or D-Sarc, α -OrMen, α -MeSer, Cit, Dap, Dab, Dap (Gly), Ac, Lys, Asn, N-MeGln, N-Arg, or Gln. In certain embodiments, X5 is gin or Asn. In a specific embodiment, X5 is Ala, Arg, Glu, Phe, Leu, Thr, Ser, Aib, Sarc, D-Ala, D-Arg, D-Glu, D-Phe, D-Leu, D-Thr, D-Ser, D-Aib, Cys, Cit, Asp, Dab, Dap, Gly, His, hCys, Lys, Met, Asn, N-Me-Ala, N-Me-Asn, N-Me-Lys, N-Me-Gln, N-Me-Arg, Orn, Pro, Pen, Gln, Val, α Me-Lys, α Me-Orn, or D-Sarc. In certain embodiments, X5 is Gln.
In particular embodiments, X6 is Asp, Thr, Asn, Phe, D-Asp, D-Thr, D-Asn, Glu, Arg, Ser or D-Phe. In a specific embodiment, X6 is Thr.
In a specific embodiment, X7 is Trp.
In particular embodiments, X8 is Val, Gln, Glu, Phe, Asn, Pro, Arg, Thr, Trp, or Lys. In particular embodiments, X8 is Gln.
In particular embodiments, X1, X2, and X3 are absent.
In certain embodiments, X11 is a Trp analog.
In a specific embodiment, X10 is a Phe analog. In a specific embodiment, X10 is Phe (4-OMe), Phe (4-CONH)2) Or Phe [4- (2-aminoethoxy)](also referred to herein as Phe [4-2ae ]]). In a particular embodiment, X10 is Phe (4-OMe) or Phe [4- (2-aminoethoxy)](also referred to herein as Phe [4-2ae ]])。
In particular embodiments, X11 is 2-Nal or 1-Nal. In certain embodiments, X11 is 2-Nal.
In certain embodiments, X12 is α -MeLys, 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Acvc, Agp, Aib, α -diethylGly, α -MeLys (Ac), α -Me-Leu, α -MeOrn, α -MeSer, or α -MeVal. In certain embodiments, X12 is α -MeLys.
In certain embodiments, X13 is Glu or lys (ac). In certain embodiments, X13 is Glu.
In certain embodiments, X14 is Asn.
In certain embodiments, X15 is Gly or Asn. In certain embodiments, X15 is Gly.
In certain embodiments, one or more, two or more, three or more, or four or more of X16, X17, X18, X19, and X20 are absent. In particular embodiments, X16, X17, X18, X19, and X20 are absent.
In a specific embodiment of Ir, X4 and X9 are Cys, X7 is Trp, and X18 is [ (D) Lys ]. In a specific embodiment of Ir, X4 and X9 are Cys, X7 is Trp, X10 is Tyr, and X18 is [ (D) Lys ]. In specific embodiments of Ir, X4 and X9 are Cys, X7 is Trp, X1, X2, and X3 are absent, X17 is absent, X18 is [ (D) Lys ], and X19 and X20 are absent. In a specific embodiment of Ir, X4 and X9 are Cys, X7 and X11 are Trp, X10 is Tyr, and X18 is [ (D) Lys. In certain embodiments, X1, X2, and X3 are absent; and in certain embodiments, X17 is absent.
In a specific embodiment of Ir, X4 and X9 are Pen, and X12 is α -MeLys. In specific embodiments of Ir, X4 and X9 are Pen, X12 is α -MeLys, and X16, X17, X18, X19, and X20 are absent. In a specific embodiment of Ir, X4 and X9 are Pen, X12 is α -MeLys, 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Acvc, Agp, Aib, α -diethylGly, α -MeLys (Ac), α -Me-Leu, α -MeOrn, α -MeSer, α -MeVal, X16, X17, X18, X19 and X20 are absent, and X7 is Trp. In a specific embodiment of Ir, X4 and X9 are Pen, X12 is α -MeLys, X16, X17, X18, X19 and X20 are absent, and X7 is Trp. In a specific embodiment of Ir, X4 and X9 are Pen, X7 is Trp, and X12 is α -MeLys. In certain embodiments, X1, X2, and X3 are absent. In particular embodiments, a disulfide bond exists between X4 and X9.
In a specific embodiment of Ir, X4 is Abu, X9 is Cys, and X12 is 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Acvc, Agp, Aib, α -diethylGly, α -MeLys or α -MeLys (Ac), α -Me-Leu, α -MeOrn, α -MeSer or α -MeVal. In a specific embodiment of Ir, X4 is Abu, X9 is Cys, and X12 is α -MeLys. In a specific embodiment of Ir, X4 is Abu, X9 is Cys, X12 is α -MeLys, 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Acvc, Agp, Aib, α -diethylGly, α -MeLys or α -MeLys (Ac), α -Me-Leu, α -MeOrn, α -MeSer or α -MeVal, and X16, X17, X18, X19 and X20 are absent. In a specific embodiment of Ir, X4 is Abu, X9 is Cys, X12 is α -MeLys, and X16, X17, X18, X19, and X20 are absent. In a specific embodiment of Ir, X4 is Abu, X9 is Cys, X12 is α -MeLys, 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Acvc, Agp, Aib, α -diethylGly, α -MeLys or α -MeLys (Ac), α -Me-Leu, α -MeOrn, α -MeSer or α -MeVal, X16, X17, X18, X19 and X20 are absent, and X7 is Trp. In a specific embodiment of Ir, X4 is Abu, X9 is Cys, X12 is α -MeLys, X16, X17, X18, X19 and X20 are absent, and X7 is Trp. In a specific embodiment of Ir, X4 is Abu, X9 is Cys, X7 is Trp, and X12 is α -MeLys. In certain embodiments, X1, X2, and X3 are absent. In particular embodiments, a thioether linkage is present between X4 and X9.
In certain embodiments of the peptide inhibitor of formula I, X comprises or consists of a sequence of formula Is:
X1-X2-X3-C-X5-X6-W-X8-C-X10-X11-X12-X13-X14-G-X16-X17-X18-X19-X20(Is)(SEQ ID NO:1375)
wherein
X1 is any amino acid or absent;
x2 is any amino acid or absent;
x3 is any amino acid or absent;
x5 is any amino acid;
x6 is any amino acid;
x8 is any amino acid;
x10 is Tyr, 1-Nal, 2-Nal, Phe (3, 4-F)2)、Phe(3,4-Cl2) F (3-Me), Phe [4- (2-aminoethoxy)]Phe [4- (2- (acetyl-aminoethoxy)]、Phe(4-Br)、Phe(4-CONH2) Phe (4-Cl), Phe (4-CN), Phe (4-guanidino), Phe (4-Me), Phe (4-NH)2)、Phe(4-N3) Phe (4-OMe), Phe (4-OBzl) or Tyr;
x11 is Trp, 1-Nal, Phe (3, 4-OMe)2) 5-hydroxy-Trp, Phe (3, 4-Cl)2) Or Tyr (3-t-Bu);
x12 is Arg, Lys, His, hArg, Cit, Orn, 1-Nal, D-Ala, D-Leu, D-Phe, D-Asn, D-Asp, Agp, Leu, β hLeu, Aib, β hAla, β hVal, β hArg, hLeu, Dap, 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Acvc, Agp, Aib, α -diethylGly, α -MeLys (Ac), α -Me-Leu, α -MeOrn, α -MeSer, α -MeVal, Cha, Cit, Cpa, (D) Asn, Glu, hArg, or Lys;
x13 is Cha, Ogl, Aib, Leu, Val, Dab, Glu, Lys, β hLeu, β hAla, β hVal, β Glu, Lys (Ac), Cit, Asp, Dab, Dap, Glu, hArg, Lys, Asn, Orn, Lys (Ac), or Gln;
X14 is Phe, Tic, Asn, Tyr, Asn, Arg, Qln, Lys (Ac), His; dap (Ac), dab (Ac), or Asp;
x16 is any amino acid;
x17 is absent;
x18 is D-Lys;
x19 is any amino acid or absent; and
x20 is any amino acid or absent.
In specific embodiments of Is: x10 is Tyr, 1-Nal or 2-Nal; x11 is Trp or 1-Nal; x12 is Arg, Lys, His, hArg, Cit, Orn, 1-Nal, D-Ala, D-Leu, D-Phe, D-Asn, D-Asp, Agp, Leu, β hLeu, Aib, β hAla, β hVal, β hArg, hLeu, or Dap; x13 is Cha, Ogl, Aib, Leu, Val, Dab, Glu, Lys, β hLeu, β hAla, β hVal, or β GLu; x14 is Phe, Tic, Asn or Tyr; and X16 is AEA, Ala or β Ala.
In a specific embodiment, X5 is Glu, Arg, Ala, N-Me-Arg, N-Me-Ala, N-Me-Gln, Orn, N-Me-Asn, N-Me-Lys, Ser, Gln, Orn, Asn, or Dap. In a specific embodiment, X5 is Glu, Arg, Ala, N-Me-Arg, N-Me-Ala, N-Me-Gln, Orn, N-Me-Asn, N-Me-Lys, Ser, Asn, or Dap.
In a specific embodiment, X6 is Asp or Thr.
In particular embodiments, X8 is Gln or Val.
In a specific embodiment, the peptide of Is cyclized via a disulfide bond between X4 and X9.
In certain embodiments of the peptide inhibitor of formula I, X comprises or consists of the sequence of formula It:
X1-X2-X3-C-X5-X6-W-X8-C-X10-X11-X12-X13-X14-X15-X16-X17-X18-X19-X20(It)(SEQ ID NO:1376)
wherein
X1 is any amino acid or absent;
x2 is any amino acid or absent;
x3 is any amino acid or absent;
x5 is any amino acid;
x6 is any amino acid;
x8 is any amino acid;
x10 is Tyr, 1-Nal, 2-Nal, Phe [4- (2-aminoethoxy)]、Phe(4-CONH2)、Phe(4-OMe);
X11 is Trp, 1-Nal, 2-Nal, Bip, Phe (3, 4-OMe)2) 5-hydroxy-Trp;
x12 is Arg, His, 3-Pal, Leu, Thr, Gln, Asn, Glu, Ile, Phe, Ser, Lys, hLeu, α -MeLeu, D-Leu, D-Asn, h-Leu, 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Acvc, Agp, Aib, α -diethylGly, α -MeLys (Ac), α -Me-Leu, α -MeOrn, α -MeSer, or α -MeVal;
x13 is Thr, Glu, Tyr, Lys, Gln, Asn, Lys (Ac), Asp, Arg, Ala, Ser, Leu;
x14 is Phe, Tyr, Asn, Gly, Ser, Met, Arg, His, Lys, Leu, or Gln;
x15 is Gly, Ser, Arg, Leu, Asp, Ala, β -Ala, Glu, Arg, or Asn;
x16 is absent or is any amino acid;
x17 is absent or is any amino acid;
x18 is any amino acid or absent;
X19 is any amino acid or absent; and
x20 is any amino acid or absent.
In certain embodiments of It: x10 is Tyr, 1-Nal or 2-Nal; x11 is Trp, 1-Nal, 2-Nal or Bip; x12 is Arg, His, 3-Pal, Leu, Thr, Gln, Asn, Glu, Ile, Phe, Ser, Lys, hLeu, α -MeLeu, D-Leu, D-Asn, or h-Leu; x13 is Thr, Glu, Tyr, Lys, Gln, Asn, Lys, Asp, Arg, Ala, Ser, Leu; x15 is Gly, Ser, Arg, Leu, Asp, or Ala; x16 is absent or is Asn, Glu, Phe, Ala, Gly, Pro, Asp, Gln, Ser, Thr, D-Glu, or Lys; and X17 is absent or is Pro, Arg, Glu, Asp, Ser, Gly or Gln.
In particular embodiments, X5 is Ser, Asp, Asn, Gln, Ala, Met, Arg, His, or Gly. In particular embodiments, X5 is Ser, Asp, Gln, Ala, Met, Arg, His, or Gly.
In particular embodiments, X6 is any Asp, Ser or Thr.
In particular embodiments, X8 is Gln, Glu, or Thr.
In a specific embodiment, the peptide of It is cyclized via a disulfide bond between X4 and X9.
In another embodiment, the invention includes a peptide inhibitor of the interleukin-23 receptor, or a pharmaceutically acceptable salt or solvate thereof, wherein the peptide inhibitor comprises an amino acid sequence of formula (Va):
(X1-X2-X3-X4-X5-X6-X7-X8-X9-X10-X11-X12-X13-X14-X15-X16-X17-X18-X19-X20(Va)(SEQ ID NO:1377)
Wherein the content of the first and second substances,
x1 is any amino acid or absent;
x2 is any amino acid or absent;
x3 is any D-amino acid or is absent;
x4 is Cys, hCys, Pen, hPen, Abu, Ser, hSer or a chemical moiety capable of forming a bond with X9;
x5 is Ala, α -MeOrn, α -MeSer, Cit, Dap, Dab, Dap (Ac), Gly, Lys, Asn, N-MeGln, N-MeArg, Orn, Gln, Arg, Ser or Thr.
X6 is Thr, Ser, Asp, Ile or any amino acid;
x7 is Trp, 6-chloro-Trp, 1-Nap or 2-Nap;
x8 is Glu, Gln, Asn, Lys (Ac), Cit, Cav, Lys (N-e- (N-a-palmitoyl-L-y-glutamyl)) or Lys (N-e-palmitoyl);
x9 is Cys, hCys, Pen, hPen, Abu or a chemical moiety capable of forming a bond with X4;
x10 is 2-Nal, a Phe analog, Tyr or Tyr analog;
x11 is 1-Nal, 2-Nal, Phe (3, 4-dimethoxy), 5-hydroxy Trp, Phe (3, 4-Cl)2) Trp or Tyr (3-tBu);
x12 is Aib, 4-amino-4-carboxy-tetrahydropyran, any alpha-methyl amino acid, alpha-ethyl-amino acid, Achc, Acvc, Acbc, Acpc, 4-amino-4-carboxy-piperidine, 3-Pal, Agp, alpha-diethylGly, alpha-MeLys (Ac), alpha-MeLeu, a-alpha-MeOrn, alpha-MeSer, alpha-MeVal, Cav, Cha, Cit, Cpa, D-Asn, Glu, His, hLeu, hArg, Lys, Leu, Octgly, Orn, piperidine, Arg, Ser, Thr, or THP;
X13 is Lys (Ac), Gln, Cit, Glu or any amino acid;
x14 is Asn, Gln, Lys (Ac), Cit, Cav, Lys (N-e- (N- α -palmitoyl-L- γ -glutamyl)), Lys (N-e-palmitoyl), or any amino acid;
x15 is β -Ala, Asn, Gly, Gln, Ala, Ser, Aib, or Cit;
x16 is any amino acid or absent;
x17 is any amino acid or absent;
x18 is any amino acid or absent;
x19 is any amino acid or absent; and
x20 is any amino acid or is absent,
wherein X4 and X9 are capable of forming a bond with each other. In particular embodiments, the linkage is an ether linkage, a disulfide linkage, or a thioether linkage. In certain embodiments, the peptide inhibitor is cyclized via a bond between X4 and X9.
In certain embodiments, X1 is a D-amino acid or is absent. In certain embodiments, X2 is a D-amino acid or is absent.
In certain embodiments, X16 is a D-amino acid or is absent. In certain embodiments, X17 is a D-amino acid or is absent. In certain embodiments, X18 is a D-amino acid or is absent. In certain embodiments, X19 is a D-amino acid or is absent. In certain embodiments, X20 is a D-amino acid or is absent.
In a particular embodiment of formula (Ia), X10 is 2-Nal, Phe (3, 4-dif F)2)、Phe(3,4-Cl2) Phe (3-Me), Phe [4- (2-aminoethoxy)]Phe [4- (2-acetylaminoethoxy)]Phe (Br), Phe (4-CONH2), Phe (Cl), Phe (4-CN), Phe (4-guanidino), Phe (4-Me), Phe (4-NH)2)、Phe(4-N3) Tyr, Tyr (Bzl) or Tyr (Me). In certain embodiments of formula (Ia), X10 is Phe (4-ZR), Phe (3-ZR), or Phe (2-ZR), wherein R ═ CH2(CH2)nY and n is 1-25, Z is NH, O, CO, CONH or CH2And Y ═ NH2、CO2H. OH or CH3
In a further related embodiment, the present invention includes a peptide inhibitor of the interleukin-23 receptor, or a pharmaceutically acceptable salt or solvate thereof, wherein the peptide inhibitor comprises an amino acid sequence of formula (Vb):
X1-X2-X3-X4-X5-X6-X7-X8-X9-X10-X11-X12-X13-X14-X15-X16-X17-X18-X19-X20(Vb)(SEQ ID NO:1378)
wherein the content of the first and second substances,
x1 is any amino acid or absent;
x2 is any amino acid or absent;
x3 is D-Arg, D-Phe, any D amino acid, or absent;
x4 is Cys, hCys, Pen, hPen, Abu or a chemical moiety capable of forming a bond with X9;
x5 is Gln, Asn, Lys (Ac), Cit, Cav, Lys (N-. epsilon. - (N-. alpha. -palmitoyl-L-. gamma. -glutamyl)) or Lys (N-. epsilon. -palmitoyl).
X6 is Thr, Ser, Asp, Ile or any amino acid;
x7 is Trp, 1-Nap or 2-Nap;
X8 is Gln, Asn, Lys (Ac), Cit, Cav, Lys (N-e- (N-a-palmitoyl-L-y-glutamyl)) or Lys (N-e-palmitoyl);
x9 is Cys, hCys, Pen, hPen, Abu, any amino acid or chemical moiety capable of forming a bond with X4;
x10 is Phe [4- (2-aminoethoxy)]Phe [4- (2-acetylaminoethoxy)]、Phe(4-CONH2) Phe (4-guanidino), Phe (4-NH)2) Tyr (Me) or Phe (4-ZR), wherein R is CH2(CH2)nY; n is 1-25; z ═ O, CO, NH, CONH, or CH2(ii) a And Y ═ NH2、CO2H. OH or CH3
X11 is 2-Nal or Trp;
x12 is Aib, 4-amino-4-carboxy-tetrahydropyran, α -diethylGly, α -MeLys (Ac), α -MeLeu, α -MeOrn, α -MeSer, α -MeVal, acid, Achc, Acvc, Acbc Acpc, or 4-amino-4-carboxy-piperidine;
x13 is Lys (Ac), Gln, Cit, Glu, or any amino acid;
x14 is Asn, Gln, Lys (Ac), Cit, Cav, Lys (N-epsilon- (N-alpha-palmitoyl-L-gamma-glutamyl)), Lys (N-epsilon-palmitoyl) or any amino acid;
x15 is β -Ala, Asn, Gln, Ala, Ser or Aib;
x16 is any amino acid or absent;
x17 is any amino acid or absent;
x18 is any amino acid or absent;
x19 is any amino acid or absent; and
X20 is any amino acid or is absent,
wherein X4 and X9 are capable of forming a bond with each other. In particular embodiments, the linkage is a disulfide linkage or a thioether linkage. In certain embodiments, the peptide inhibitor is cyclized via a bond between X4 and X9.
In certain embodiments, X1 is a D-amino acid or is absent. In certain embodiments, X2 is a D-amino acid or is absent.
In certain embodiments, X16 is a D-amino acid or is absent. In certain embodiments, X17 is a D-amino acid or is absent. In certain embodiments, X18 is a D-amino acid or is absent. In certain embodiments, X19 is a D-amino acid or is absent. In certain embodiments, X20 is a D-amino acid or is absent.
In another related embodiment, the invention encompasses a peptide inhibitor of the interleukin-23 receptor, or a pharmaceutically acceptable salt or solvate thereof, wherein the peptide inhibitor comprises an amino acid sequence of formula (Vc):
X1-X2-X3-X4-X5-X6-X7-X8-X9-X10-X11-X12-X13-X14-X15-X16-X17-X18-X19-X20(Vc)(SEQ ID NO:1379)
wherein the content of the first and second substances,
x1 is absent;
x2 is absent;
x3 is D-Arg or absent;
x4 is Cys, Pen, Abu or a chemical moiety capable of forming a bond with X9;
x5 is Gln, Asn, Lys (Ac), Cit, or Cav;
x6 is Thr or Ser;
x7 is Trp, 1-Nap or 2-Nap;
X8 is Gln, Asn, Lys (Ac), Cit or Cav;
x9 is Cys, hCys, Pen, hPen, Abu, or any amino acid or chemical moiety capable of forming a bond with X4;
x10 is Phe [4- (2-aminoethoxy)]Phe (4-CONH2) OR Phe (4-OR), wherein R ═ CH2(CH2)nY; n is 1-25; and Y ═ NH2、CO2H. OH or CH3
X11 is Trp or 2-Nal;
x12 is Aib, 4-amino-4-carboxy-tetrahydropyran, α -MeLys (Ac), α -MeLeu, Achc, Acvc, Acbc, or Acpc;
x13 is Lys (Ac) or Glu;
x14 is Asn, Gln, Lys (Ac), Lys (N-. epsilon. - (N-. alpha. -palmitoyl-L-. gamma. -glutamyl)) or Lys (N-. epsilon. -palmitoyl).
X15 is Gly, β -Ala, Asn, Gln, Ala, Ser or Aib;
x16 is absent;
x17 is absent;
x18 is absent;
x19 is absent; and
the absence of X20 is not present,
wherein X4 and X9 are capable of forming a bond with each other. In particular embodiments, the linkage is a disulfide linkage or a thioether linkage. In certain embodiments, the peptide inhibitor is cyclized via a bond between X4 and X9.
In certain embodiments, X1 is a D-amino acid or is absent. In certain embodiments, X2 is a D-amino acid or is absent.
In certain embodiments, X16 is a D-amino acid or is absent. In certain embodiments, X17 is a D-amino acid or is absent. In certain embodiments, X18 is a D-amino acid or is absent. In certain embodiments, X19 is a D-amino acid or is absent. In certain embodiments, X20 is a D-amino acid or is absent.
In another related embodiment, the present invention includes a peptide inhibitor of the interleukin-23 receptor, or a pharmaceutically acceptable salt or solvate thereof, wherein the peptide inhibitor comprises an amino acid sequence of formula (Vd):
X1-X2-X3-X4-X5-X6-X7-X8-X9-X10-X11-X12-X13-X14-X15-X16-X17-X18-X19-X20(Vd)(SEQ ID NO:1380)
wherein, the first and the second end of the pipe are connected with each other,
x1 is absent;
x2 is absent;
x3 is absent;
x4 is Pen or Abu;
x5 is Gln or Asn;
x6 is Thr or Ser;
x7 is Trp;
x8 is Gln or Asn;
x9 is Pen or Cys;
x10 is Phe [4- (2-aminoethoxy)]Or Phe (4-CONH)2);
X11 is Trp or 2-Nal;
x12 is Aib, 4-amino-4-carboxy-tetrahydropyran, α -MeLys, α -MeLeu, or Achc;
x13 is Lys (Ac) or Glu;
x14 is Asn, Gln, or Lys (Ac);
x15 is Gly, Ala, Ser, β -Ala, Asn or Gln;
x16 is absent;
x17 is absent;
x18 is absent;
x19 is absent; and
the absence of X20 is not present,
wherein X4 and X9 are capable of forming a bond with each other. In particular embodiments, the linkage is a disulfide linkage or a thioether linkage. In certain embodiments, the peptide inhibitor is cyclized via a bond between X4 and X9.
Any peptide inhibitor of the invention (e.g., any of those of formula I (e.g., Ix, Ia-It)) can be further defined, for example, as described below. It will be appreciated that each of the further defining features described herein may be applied to any peptide inhibitor in which the specified amino acid at a particular position allows for the presence of the further defining feature.
In certain embodiments, any Phe [4- (2-aminoethoxy) ] residue present in the peptide inhibitor may be substituted with Phe [4- (2-acetylaminoethoxy) ].
In certain embodiments, X1-X20 is any amino acid shown at the corresponding position relative to the cyclized Pen-Pen or cyclized Abu-Cys residue of an illustrative peptide inhibitor set forth in tables 2-5.
In certain embodiments, any of the peptide inhibitors described herein, including but not limited to formula (X), (Va), (Vb), (Vc), (Vd), (Ve), (Vf), (Vg), or (Vh), further comprises a linker or spacer portion between any 2 amino acid residues of the peptide. In particular embodiments, the linker or spacer moiety is a PEG moiety.
In certain embodiments, the peptide inhibitor is cyclized via a disulfide bridge.
In certain embodiments, X10 is Tyr, Phe [4- (2-aminoethoxy)]、Phe(4-CONH2) Or Phe (4-OMe). In certain embodiments, X10 is Tyr.
In certain embodiments, X11 is 2-Nal, Trp, or 5-hydroxy-Trp. In certain embodiments, X11 is Trp.
In certain embodiments, X10 is Tyr or Phe [4- (2-aminoethoxy) ], and X11 is Trp or 2-Nal. In certain embodiments, X10 is Tyr and X11 is Trp.
In particular embodiments, both X4 and X9 are Cys.
In particular embodiments, X4 is Cys, Pen, hCys, or absent.
In particular embodiments, X7 and X11 are not both W.
In particular embodiments, both X7 and X11 are W.
In particular embodiments, X7 and X11 are both W, X10 is Y, and X4 and X9 are both Cys.
In particular embodiments, X15 is Gly, Asn, β -ala, or Ser. In a specific embodiment, X15 is Gly or Ser.
In particular embodiments, X16 is AEA or AEP.
In specific embodiments, X10 is Tyr, Phe or Phe [4- (2-aminoethoxy). In particular embodiments, X10 is Tyr or Phe.
In particular embodiments, X11 is Trp or 2-Nal. In a specific embodiment, X11 is Trp.
In particular embodiments, X1, X2, and X3 are absent.
In particular embodiments, X18, X19, and X20 are absent.
In particular embodiments, X1, X2, X3, X18, X19, and X20 are absent.
In particular embodiments, one or more of X1, X2, or X3 is present.
In a specific embodiment of any of Ix, I α -Ir, one of X1, X2, and X3 is present, and the remaining two are absent. In one embodiment, X1, X2, or X3 present is Ala.
In certain embodiments, X3 is present. In a specific embodiment, X3 is Glu, (D) Glu, Arg, (D) Arg, Phe, (D) Phe, 2-Nal, Thr, Leu, (D) Gln. In certain embodiments, X3 is (D) Arg or (D) Phe. In particular embodiments, X1 and X2 are absent, and X3 is present.
In particular embodiments, two of X1, X2, and X3 are present, and the remaining one is absent. In certain embodiments, two present consist of SG, NK, DA, PE, QV, or DR.
In particular embodiments, X1, X2, and X3 are present. In certain embodiments, X1, X2, and X3 consist of ADQ, KEN, VQE, GEE, DGF, NAD, ERN, RVG, KAN, or YED.
In certain embodiments, the peptide comprises an AEP residue. In particular embodiments, any one of X15, X16, X17, X18, X19, or X20 is AEP.
In certain embodiments of any of the peptide inhibitors or peptide monomer subunits, X13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys (Ac), β hAla, or Aib. In certain embodiments of any of the peptide inhibitors or peptide monomer subunits, X13 is Thr, Sarc, Glu, Phe, Arg, Leu, Lys, β hAla, or Aib. In certain embodiments, X14 is Phe, Asn, Tyr, or β hPhe. In certain embodiments, X14 is Phe, Tyr, or β hPhe. In certain embodiments, X15 is Gly, Asn, Ser, Thr, Gln, Ala, or Sarc. In certain embodiments, X15 is Gly, Ser, Thr, Gln, Ala, or Sarc. In certain embodiments, X12 is an alpha amino acid, e.g., 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Acvc, Aib, alpha-diethylGly, alpha-MeLys (Ac), alpha-Me-Leu, alpha-MeOrn, alpha-MeSer, or alpha-MeVal.
In certain embodiments, X13 is present.
In certain embodiments, X13 and X14 are present.
In certain embodiments, X13, X14, and X15 are present.
In a specific embodiment of any one of Ia-It, one or more of X16-X20 is present. In particular embodiments, two or more or three or more of X16-X20 are present. In a specific embodiment, X18 is [ (D) Lys ]. In a specific embodiment, X17 is absent, and X18 is [ (D) Lys ]. In certain embodiments wherein X4 and X9 are optionally Cys, X4 and X9 are Cys, X7 is Trp, and X18 is [ (D) Lys ]. In particular embodiments where X4 and X9 are optionally Cys, X4 and X9 are Cys, X7 is Trp, X10 is Tyr or Phe [4- (2-aminoethoxy) ], and X18 is [ (D) Lys ]. In particular embodiments where X4 and X9 are optionally Cys, X4 and X9 are Cys, X7 is Trp, X10 is Tyr, and X18 is [ (D) Lys ]. In particular embodiments where X4 and X9 are optionally Cys, X4 and X9 are Cys, X7 is Trp, X1, X2 and X3 are absent, X17 is absent, X18 is [ (D) Lys ], and X19 and X20 are absent. In a specific embodiment of Ir, X4 and X9 are Cys, X7 and X11 are Trp, X10 is Tyr, and X18 is [ (D) Lys. In certain embodiments, X1, X2, and X3 are absent; and in certain embodiments, X17 is absent.
In certain embodiments, any of the peptide inhibitors (or monomeric subunits) described herein are cyclized. In particular embodiments, the peptide inhibitor is cyclized via a bond between two or more internal amino acids of the peptide inhibitor. In particular embodiments, the cyclized peptide inhibitor is not cyclized via a bond between the N-terminal and C-terminal amino acids of the peptide inhibitor. In certain embodiments, one of the amino acid residues involved in the intramolecular bond cyclizing the peptide is the amino-terminal amino acid residue. In certain embodiments, any peptide inhibitor is cyclized via a peptide bond between its N-terminal amino acid and its C-terminal amino acid.
In certain embodiments of any peptide inhibitor or one or both of its monomeric subunits, the peptide inhibitor (or one or both of its monomeric subunits) is cyclized via an intramolecular bond between X4 and X9 or through a triazole ring. In particular embodiments, the intramolecular bond is any of a disulfide bond, a thioether bond, a lactam bond, a triazole bond, a selenoether bond, a diselenide bond, or an olefin bond.
In one embodiment, X4 and X9 in the peptide inhibitor (or one or both monomeric subunits thereof) are Cys, Pen, hCys, D-Pen, D-Cys or D-hCys, and the intramolecular bond is a disulfide bond. In certain embodiments, X4 and X9 are both Cys, or X4 and X9 are both Pen, and the intramolecular bond is a disulfide bond.
In one embodiment, X4 and X9 in the peptide inhibitor (or one or both monomeric subunits thereof) are Glu, Asp, Lys, Orn, Dap, Dab, D-Dap, D-Dab, D-Asp, D-Glu or D-Lys, and the intramolecular bond is a lactam bond.
In one embodiment, X4 is Abu, 2-chloromethylbenzoic acid, mercaptopropionic acid, mercaptobutyric acid, 2-chloro-acetic acid, 3-chloro-propionic acid, 4-chloro-butyric acid, or 3-chloro-isobutyric acid; x9 is Abu, Cys, Pen, hCys, D-Pen, D-Cys or D-hCys; and the intramolecular bond is a thioether bond. In certain embodiments, X4 is Abu, and X9 is Pen, and the intramolecular linkage is a thioether linkage. In a particular embodiment, X4 is a 2-methylbenzoyl moiety capable of forming a thioether bond with X9, and X9 is selected from Cys, N-Me-Cys, D-Cys, hCys, Pen, and D-Pen. In a specific embodiment, X4 is Abu, and X9 is Cys, and the intramolecular bond is a thioether bond. In specific examples of peptide monomers, dimers, or subunits thereof of any of the formulae and peptides described herein, X4 is selected from the group consisting of modified Ser, modified hSer (e.g., high-Ser-Cl), suitable isosteres, and the corresponding D-amino acid. In other examples, X4 is a fatty acid having one to four carbons and forming a thioether bond with X9. In some examples, X4 is a five or six membered alicyclic acid having a modified 2-methyl group that forms a thioether bond with X9. In some embodiments, X4 is a 2-methylbenzoyl moiety. In certain embodiments, X4 is selected from the group consisting of Cys, hCys, Pen, and 2-methylbenzoyl moieties. In certain embodiments, X4 is selected from the group consisting of modified Ser, modified hSer, a suitable isostere, and a corresponding D-amino acid. In one embodiment, X4 is hSerCl (prior to forming a thioether bond with X9, thereby removing Cl) or a hSer precursor (e.g., high Ser (O-TBDMS). in other examples, X4 is a fatty acid having one to four carbons and forming a thioether bond with X9. in some examples, X4 is a five-or six-membered cycloaliphatic acid having a modified 2-methyl group that forms a thioether bond with X9. in some examples, X4 is a 2-methylbenzoyl moiety. in certain embodiments where X4 is not an amino acid but is a chemical moiety that binds to X9, X1, X2, and X3 are absent, and X4 is conjugated or bound to X5. in some embodiments, an amino acid that is directly carboxylated with X9 is an aromatic amino acid. in certain embodiments, X4 is an amino acid, while in other embodiments, X4 is an amino acid that is capable of binding to X9, e.g., in another embodiment, x4 is another chemical moiety selected from any of the non-amino acid moieties described herein for X4. In particular embodiments wherein X4 is another chemical moiety, X1, X2, and X3 are absent, and the other chemical moiety is bound or conjugated to X5. In certain embodiments, X4 is defined as a chemical moiety that includes a group such as chloride, for example, 2-chloromethylbenzoic acid, 2-chloro-acetic acid, 3-chloropropionic acid, 4-chlorobutyric acid, 3-chloroisobutyric acid. However, the skilled person understands that once the peptide has undergone ring closing cyclization to form a thioether bond between X4 and X9, the chloride group is no longer present. Thus, the description of a chemical moiety at X4 that contains a reactant group such as chloride means a group with a chloride and a group without a chloride (i.e., after forming a bond with X9). The invention also includes peptides comprising the same structure as shown in any other formula or table described herein, but wherein the thioether linkages are in the opposite orientation. In such embodiments of the invention, it may generally be said that the amino acid residue or other chemical moiety represented by X4 is instead present at X9, and the amino acid residue represented by X9 is instead present at X4, i.e., the amino acid residue containing the resulting thioether bond sulfur is located at X4 rather than X9, and the amino acid residue or other moiety having a carbon side chain capable of forming a thioether bond with X4 is located at X9. However, in this reverse orientation, the amino acid or chemical moiety at X9 is an amino acid or chemical moiety comprising a free amine. For example, in a specific embodiment, the amino acid at X9 is protected homoserine, such as, for example, homoserine (OTBDMS). Thus, in a particular counter-oriented embodiment of a peptide inhibitor of any of the formulae described herein, X9 is an amino acid residue having a side chain containing one or two carbons and forming a thioether bond with X4, and X4 is selected from Cys, N-Me-Cys, D-Cys, HCys, Pen, and D-Pen. Specific examples of amino acid residues and other chemical moieties present at corresponding positions in other formulae and tables are described herein.
One skilled in the art understands that certain amino acids and other chemical moieties are modified when combined with another molecule. For example, an amino acid side chain may be modified when it forms an intramolecular bridge with another amino acid side chain, e.g., one or more hydrogens may be removed or replaced by a bond. Furthermore, when hSer-Cl is bound to an amino acid such as Cys or Pen via a thioether bond, the Cl moiety is released. Thus, as used herein, reference to an amino acid or modified amino acid (such as hSer-Cl) (e.g., at position X4 or X9) present in a peptide dimer of the invention is meant to include the form of such amino acid or modified amino acid present in the peptide before and after the formation of intramolecular bonds.
In certain embodiments, the peptide inhibitors of the invention (or one or both of its monomeric subunits) are cyclized through the triazole ring. In certain embodiments, the peptide inhibitors of the invention (or one or both of its monomeric subunits) are linear or not cyclized. In certain embodiments of any of the peptide inhibitors described herein (including monomeric and dimeric peptide inhibitors), one (or both) monomeric subunit of the peptide comprises or consists of a cyclized peptide having the structure or sequence shown In any one of Ix, Ia, Ib, Ic, Id, Ie, If, Ig, Ih, Ii, Ij, Ik, Il, Im, In, Io, Ip, Iq', Ir, Is, or It, IIa-IId, IIIa-IIIe, Iva, or IVb.
In certain embodiments of any peptide inhibitor or monomeric subunit, both X7 and X11 are W.
In certain embodiments of any peptide inhibitor or monomeric subunit, X7 and X11 are not both W. In particular embodiments, X7 is W, and X11 is not W.
In certain embodiments of any peptide inhibitor or monomer subunit, X4 and X9 are amino acid residues capable of forming an intramolecular bond with each other, said intramolecular bond being a thioether, lactam, triazole, selenoether, diselenide, or alkene bond.
In certain embodiments, X7 and X11 are both W and X10 is Y, Phe [4- (2-aminoethoxy) or Phe (CONH)2) And X4 and X9 are amino acid residues capable of forming an intramolecular bond with each other, the intramolecular bond being a thioether bond, a lactam bond, a triazole bond, a selenoether bond, a diselenide bond, or an olefin bond. In certain embodiments, X7 and X11 are both W, X10 is Y, and X4 and X9 are amino acid residues capable of forming an intramolecular bond with each other, said intramolecular bond being a thioether, lactam, triazole, selenoether, diselenide, or alkene bond.
In certain embodiments, X7 and X11 are both W, X10 is Y, and X4 and X9 are both C.
In certain embodiments, X4 and X9 are each Cys, Pen, hCys, D-Pen, D-Cys, or D-hCys, and the intramolecular bond is a disulfide bond.
In certain embodiments, X4 and X9 are each Glu, Asp, Lys, Orn, Dap, Dab, D-Dap, D-Dab, D-Asp, D-Glu, or D-Lys, and the intramolecular bond is a lactam bond.
In certain embodiments, X4 and X9 are each β -azido-Ala-OH or propargylglycine, and the peptide inhibitor (or monomeric subunit) is cyclized through the triazole ring.
In certain embodiments, X4 and X9 are each 2-allylglycine, 2- (3 ' -butenyl) glycine, 2- (4 ' -pentenyl) glycine, or 2- (5 ' -hexenyl) glycine, and the peptide inhibitor (or monomeric subunit) is cyclized via a ring closing metathesis reaction to produce the corresponding alkene/"stapled peptide".
In certain embodiments, X4 is 2-chloromethylbenzoic acid, mercaptopropionic acid, mercaptobutyric acid, 2-chloro-acetic acid, 3-chloro-propionic acid, 4-chloro-butyric acid, 3-chloro-isobutyric acid, or hser (cl); x9 is hSer (Cl), Cys, Pen, hCys, D-Pen, D-Cys or D-hCys; and the intramolecular bonds are thioether bonds. In certain embodiments, X4 is 2-chloromethylbenzoic acid or hser (cl); x9 is Cys or Pen, and the intramolecular bond is a thioether bond. In certain embodiments, X4 is Abu and X9 is Cys or Pen.
In certain embodiments, X4 is 2-chloromethylbenzoic acid, 2-chloro-acetic acid, 3-chloro-propionic acid, 4-chloro-butyric acid, 3-chloro-isobutyric acid, Abu, or Sec; x9 is Abu or Sec; and the intramolecular bond is a selenoether bond.
In certain embodiments, the intramolecular bond between X4 and X9 is a diselenide bond.
In certain embodiments of any of the peptide inhibitors described herein that contain two amino acid residues (e.g., cysteine residues) linked by an intramolecular bond (e.g., a disulfide bond), the two amino acid residues involved in the intramolecular bond are not both located at the N-terminal or C-terminal position of the peptide inhibitor. In certain embodiments, neither of the two amino acid residues (e.g., cysteines) involved in intramolecular bonds are located at the N-or C-terminal position of the peptide inhibitor. In other words, in certain embodiments, at least one or both of the two amino acid residues (e.g., cysteines) involved in the intramolecular bond are internal amino acid residues of the peptide inhibitor. In certain embodiments, neither of the two amino acid residues (e.g., cysteines) involved in intramolecular bonds are located at the C-terminal position of the peptide inhibitor. In one embodiment, the two amino acid residues involved in the intramolecular linkage are Cys, Pen, hCys, D-Pen, D-Cys or D-hCys residues. In certain embodiments, the two amino acid residues involved in intramolecular bonds are located at X4 and X9. In one embodiment, a disulfide bond exists between the amino acid residues at X4 and X9 (e.g., cysteine or Pen residues). In particular embodiments, both X4 and X9 are Pen. In certain embodiments, one or both peptide monomer subunits in the peptide inhibitor are cyclized via a disulfide bond between, for example, two Pen residues at positions X4 and X9.
In particular embodiments of any of the peptide inhibitors described herein, one or both peptide monomer subunits present in the peptide inhibitor (whether it be a monomer or a dimer) are cyclized or cyclized, e.g., via an intramolecular bond (such as a disulfide bond) between two cysteines present in the peptide monomer or peptide monomer subunit. In certain embodiments, the peptide inhibitor comprises two or more cysteine residues. In some embodiments, the peptide inhibitor is cyclized via an intramolecular disulfide bond between two cysteines. In a specific embodiment of a peptide inhibitor having any of the formulae described herein, two cysteines are present at positions X4 and X9. In other embodiments, one or both peptide monomer subunits in the peptide inhibitor are cyclized via a disulfide bond between, for example, two Pen residues at positions X4 and X9.
In some embodiments, the peptide inhibitor has the structure of any formula described herein (e.g., formula I) and comprises a disulfide bond, e.g., an intramolecular disulfide bond or a thioether bond. Illustrative examples of such peptide inhibitors are shown in table E3 a-E3H and table E4A, table E4B, table E9, or table E11. Such disulfide-bonded peptides may have particular advantages in that the disulfide bonds enhance structural stability and may improve the biological activity of many biologically active peptides. However, in some cases, these bonds are unstable to reducing agents. Those skilled in the art understand that disulfides are suitable for simple isosteric replacement. Illustrative examples of such substitutions include, but are not limited to, the groups of thioethers, disulfides, selenoethers, diselenides, triazoles, lactams, alkanes, and alkenes. Thus, in certain embodiments of any of the peptide inhibitors described herein, one, two or more cysteine residues are substituted, for example, with groups of thioether, disulfide, selenide, diselenide, triazole, lactam, alkane, and alkene (including but not limited to any of those shown below or described herein). In particular embodiments, two of these substituted groups form a bond (e.g., an intramolecular bond), thereby cyclizing the peptide inhibitor or one or both monomeric subunits thereof.
In certain embodiments, the peptide inhibitors of the invention comprise or consist of: herein, an amino acid sequence as shown, for example, in any one of table E3 a-E3H, table E4A, table E4B, table E5 a-E5C, table E6, table E7, table E8, table E9, table E10, table E11, table E12, table E13, or table 14. In certain embodiments, a peptide inhibitor of the invention has a structure shown herein, e.g., in any one of table E3 a-E3H, table E4A, table E4B, table E5 a-E5C, table E6, table E7, table E8, table E9, table E10, table E11, table E12, table E13, or table 14.
In certain embodiments, the invention includes a peptide inhibitor comprising a core consensus sequence (shown in an N-terminal to C-terminal direction) selected from one of:
X1X2X2WX2X1X2W(SEQ ID NO:1381);
X1X2X2WX2X1X2(1-Nal)(SEQ ID NO:1382);
X1X2X2WX2X1X2(2-Nal)(SEQ ID NO:1383);
X1X2X2WX2X1YW(SEQ ID NO:1384);
X1X2X2WX2X1Y(1-Nal)(SEQ ID NO:1385);
X1X2X2WX2X1Y(2-Nal)(SEQ ID NO:1386);
X1X2X2WX2X1X2X2(SEQ ID NO:1387);
X1X2X2WX2X1X2X2X2X2X2-[(D)Lys](SEQ ID NO:1388);
X1X2X2WX2X1X3X2(SEQ ID NO:1389);
X1X2X2WX2X1X3(1-Nal) (SEQ ID NO: 1390); and
X1X2X2WX2X1X3(2-Nal)(SEQ ID NO:1391)。
wherein W is tryptophan, Y is tyrosine, and the two X1 residues are each amino acids or other chemical moieties capable of forming intramolecular bonds with each other; each X2 is independently selected from all amino acids including, for example, natural amino acids, L-amino acids, D-amino acids, unnatural amino acids, and unnatural amino acids; and X3 is any amino acid. In particular embodiments, X3 is Phe, a Phe analog (e.g. Phe [4- (2-aminoethoxy)]Or Phe (4-CONH)2) Tyr or Tyr analogs (e.g., Tyr (Me)). In a specific embodiment, each X1 is selected from Cys, Pen, and Abu. In particular embodiments, each X1 is Cys. In certain embodiments, each X1 is Pen. In certain embodiments, one X1 is Cys and the other X1 is Abu. In a specific embodiment, the N-terminal X1 is Abu and the C-terminal X1 is Cys. In a specific embodiment, the N-terminal X1 is Cys and the C-terminal X1 is Abu. In particular embodiments, the residues between the two X1 residues are Gln, Thr, Trp, and Gln. In particular embodiments, each X1 is selected from Cys, Pen, and Abu; and X3 is Phe, an analog of Phe (e.g., Phe [4- (2-aminoethoxy)]Or Phe (4-urea)), Tyr or Tyr analogs (e.g., Tyr (Me)). In a specific embodiment, X3 is a Phe analog.
In certain embodiments, a peptide inhibitor of the invention comprises any of the following consensus sequences, wherein X1, X2, X3, X4, X5, X6, X7, X8, X9, X10, X11, X12, X13, X14, and X15 are defined as shown in any of the formulae or peptide inhibitors described herein:
X1-X2-X3-Pen-X5-X6-W-X8-Pen-X10-X11-X12-X13-X14-X15(SEQ ID NO:1392);
Pen-X5-X6-W-Q-Pen(SEQ ID NO:1393);
Pen-X5-X6-W-X8-Pen(SEQ ID NO:1394);
Pen-X5-X6-W-X8-Pen-[Phe(4-CONH2)](SEQ ID NO:1395);
Pen-X5-X6-W-X8-Pen- [ Phe [4- (2-aminoethoxy) ] ] (SEQ ID NO: 1396);
X1-X2-X3-Abu-X5-X6-W-X8-C-X9-X10-X11-X12-X13-X14-X15(SEQ ID NO:1397);
Abu-X5-X6-W-Q-C(SEQ ID NO:1398);
Abu-X5-X6-W-X8-C(SEQ ID NO:1399);
Abu-X5-X6-W-X8-C- [ Phe (4-CONH2) ] (SEQ ID NO: 1400); or
Abu-X5-X6-W-X8-C- [ Phe [4- (2-aminoethoxy) ] ] (SEQ ID NO: 1401).
In certain embodiments of any peptide inhibitor or monomeric subunit, both X7 and X11 are W. In certain embodiments of any of the peptide inhibitors, X7 and X11 are both W, and X10 is Y. In certain embodiments, X7 and X11 are both W, and X10 is Phe [4- (2-aminoethoxy) ] or Phe (4-OMe).
In certain embodiments of any peptide inhibitor or monomeric subunit, X7 and X11 are not both W.
In certain embodiments of the peptide inhibitor of formula I, X4 and X9 are each Pen and the intramolecular bond is a disulfide bond.
In certain embodiments, the peptide inhibitors of the present invention comprise or consist of an amino acid sequence as set forth in any one of the tables, sequence listings, or figures herein.
In certain embodiments of any of the peptide inhibitors described herein that contain two amino acid residues (e.g., Pen residues) linked by an intramolecular bond (e.g., a disulfide bond), one or both of the two amino acid residues involved in the intramolecular bond are not located at the N-terminal or C-terminal position of the peptide inhibitor. In certain embodiments, neither of the two amino acid residues (e.g., Pen) involved in the intramolecular bond are located at the N-or C-terminal position of the peptide inhibitor. In other words, in certain embodiments, at least one or both of the two amino acid residues (e.g., Pen) involved in the intramolecular bond are internal amino acid residues of the peptide inhibitor. In certain embodiments, neither of the two amino acid residues (e.g., Pen) involved in the intramolecular bond are located at the C-terminal position of the peptide inhibitor.
In some embodiments, wherein the peptide of the invention is conjugated to an acidic compound such as, for example, isovalerate, isobutyrate, valerate, and the like, the presence of such conjugation is referred to in the acid form. Thus, for example, but not limiting in any way, in some embodiments, the present application mentions such conjugation as isovaleric acid- [ Pen]-QTWQ[Pen]-[Phe(4-OMe)]-[2-Nal]-[α-MeL ys]-[Lys(Ac)]-NG-NH2(SEQ ID NO:262) instead of indicating the conjugation of isovaleric acid to a peptide by reference to isovaleryl, e.g., isovaleryl- [ Pen]-QTWQ[Pen]-[Phe(4-OMe)]-[2-Nal]-[α-MeLys]-[Lys(Ac)]-NG-NH2(SEQ ID NO:262)。
The invention also includes peptide inhibitors that selectively bind to an epitope or binding domain present within amino acid residues 230-349 of the human IL23R protein. In a specific embodiment, the peptide inhibitor binds to human IL23R but not to mouse IL-23R. In certain embodiments, the peptide inhibitor also binds to rat IL-23R.
In certain embodiments of the peptide inhibitors of formula I, X4 is Abu; x9 is Cys, Pen, high Cys, and the intramolecular bond is a thioether bond.
In certain embodiments, the peptide inhibitor does not include a compound disclosed in PCT application No. PCT/US2014/030352 or PCT application No. PCT/US 2015/038370.
Exemplary peptide inhibitors comprising a Pen-Pen disulfide bond
In certain embodiments, the present invention includes a peptide inhibitor of the interleukin-23 receptor, or a pharmaceutically acceptable salt or solvate thereof, wherein the peptide inhibitor has the structure of formula II:
R1-X-R2(II)
Wherein R is1Is a bond, hydrogen, C1-C6 hydrocarbyl, C6-C12 aryl, C6-C12 aryl, C1-C6 hydrocarbyl, C1-C20 hydrocarboxyl, hydrocarbyl sulfonate, acid, γ -Glu, or pGlu (added to the N-terminus), and includes a pegylated form of any of the foregoing alone or as a spacer (e.g., 200Da to 60,000 Da);
R2is a bond, OH or NH2(ii) a And
x is an amino acid sequence of 8 to 20 amino acids or 8 to 35 amino acids.
In a particular embodiment of the peptide inhibitor of formula II, X comprises or consists of the sequence of formula IIa:
X1-X2-X3-X4-X5-X6-X7-X8-X9-X10-X11-X12-X13-X14-X15-X16-X17-X18-X19-X20(IIa)(SEQ ID NO:1402)
wherein
X1 is absent or is any amino acid;
x2 is absent or is any amino acid;
x3 is absent or is any amino acid;
x4 is Pen, Cys or homo-Cys;
x5 is any amino acid;
x6 is any amino acid;
x7 is Trp, Bip, Gln, His, Glu (Bzl), 4-phenylbenzylalanine, Tic, Phe [4- (2-aminoethoxy)]、Phe(3,4-Cl2) Phe (4-OMe), 5-hydroxy-Trp, 6-chloro-Trp, N-MeTrp, alpha-Me-Trp, 1,2,3, 4-tetrahydro-norharpagne, Phe (4-CO)2H)、Phe(4-CONH2) Phe (3, 4-dimethoxy), Phe (4-CF)3) Phe (4-tBu), β β -diphela, Glu, Gly, Ile, Asn, Pro, Arg, Thr, or Octgly, or any of the corresponding α -methyl amino acid forms described above;
X8 is any amino acid;
x9 is Pen, Cys or hCys;
x10 is 1-Nal, 2-Nal, Aic, Bip, (D) Cys, Cha, DMT, (D) Tyr, Glu, Phe, His, Trp, Thr, Tic, Tyr, 4-pyridyl Ala, Octgly, a Phe analog, or a Tyr analog (optionally, Phe (3, 4-F)2)、Phe(3,4-Cl2) F (3-Me), Phe [4- (2-aminoethoxy)]Phe [4- (2- (acetyl-aminoethoxy)]、Phe(4-Br)、Phe(4-CONH2) Phe (4-Cl), Phe (4-CN), Phe (4-guanidino), Phe (4-Me), Phe (4-NH)2)、Phe(4-N3) Phe (4-OMe) or Phe (4-OBzl)) or any of the corresponding alpha-methyl amino acid forms described above;
x11 is 2-Nal, 1-Nal, 2, 4-dimethyl Phe, Bip, Phe (3, 4-Cl)2)、Phe(3,4-F2)、Phe(4-CO2H) Beta hPhe (4-F), alpha-Me-Trp, 4-phenylcyclohexyl, Phe (4-CF)3) alpha-MePhe, beta hNal, beta hPhe, beta hTyr, beta hUrp, Nva (5-phenyl), Phe, His, hPhe, Tic, Tqa, Trp, Tyr, Phe (4-OMe), Phe (4-Me), Trp (2,5, 7-tri-tert-butyl), Phe (4-O allyl), Tyr (3-tBu), Phe (4-guanidino, Phe (4-OBzl), Octgly, Glu (Bzl), 4-phenylbenzylalanine, Phe [4- (2-aminoethoxy)]5-hydroxy-Trp, 6-chloro-Trp, N-MeTrp, 1,2,3, 4-tetrahydro-norhaben, Phe (4-CONH)2)、Phe(3,4-OMe2)、Phe(2,3-Cl2)、Phe(2,3-F2) Phe (4-F), 4-phenylcyclohexylalanine or Bip or any of the corresponding alpha-methyl amino acid forms described above;
X12 is α -MeLys, α -MeOrn, α -MeLeu, α -MeVal, 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Acvc, MeLeu, Aib, (D) Ala, (D) Asn, (D) Leu, (D) Asp, (D) Phe, (D) Thr, 3-Pal, Aib, β -Ala, β hGlu, β hAla, β hLeu, β hVal, β -spiro-pip, Cha, Chg, Asp, Dab, Dap, α -diethylGly, Glu, Phe, hLeu, hArg, hLeu, Ile, Lys, Leu, Asn, N-MeLeu, N-MeArg, Ogl, Orn, Pro, gin, Arg, Ser, Thr, or Tle, or any of the corresponding α -methyl amino acid forms;
x13 is Lys (Ac), (D) Asn, (D) Leu, (D) Thr, (D) Phe, Ala, Aib, alpha-MeLeu, beta-Ala, beta hGlu, beta hAla, beta hLeu, beta hVal, beta-spiro-pip, Cha, Chg, Asp, Lys, Arg, Orn, Dab, Dap, alpha-diethylGly, Glu, Phe, hLeu, Lys, Leu, Asn, Ogl, Pro, Gln, Asp, Arg, Ser, spiro-pip, Thr, Tba, Tlc, Val, or Tyr or any of the corresponding alpha-methyl amino acid forms;
x14 is Asn, Glu, Phe, Gly, His, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Tic, or Tyr, Lys (Ac), Orn, or any of the corresponding α -methyl amino acid forms of the foregoing;
x15 is Gly, (D) Ala, (D) Asn, (D) Asp, Asn, (D) Leu, (D) Phe, (D) Thr, Ala, AEA, Asp, Glu, Phe, Gly, Lys, Leu, Pro, Gln, Arg or Ser, β -Ala, Arg or any of the corresponding α -methyl amino acid forms;
X16 is absent and is Gly, Ala, Asp, Ser, Pro, Asn or Thr or any of the foregoing corresponding α -methyl amino acid forms;
x17 is absent and is Glu, Ser, Gly, or Gln or any of the corresponding α -methyl amino acid forms described above;
x18 is absent or is any amino acid;
x19 is absent or is any amino acid; and
x20 is absent or is any amino acid.
In certain embodiments of IIa: x10 is 1-Nal, 2-Nal, Aic, Bip, (D) Cys, Cha, DMT, (D) Tyr, Glu, Phe, His, Trp, Thr, Tic, Tyr, 4-pyridyl Ala, Octgly, Y,A Phe analog or Tyr analog or any of the foregoing corresponding alpha-methyl amino acid forms; x11 is 2-Nal, 1-Nal, 2, 4-dimethyl Phe, Bip, Phe (3, 4-Cl)2)、Phe(3,4-F2)、Phe(4-CO2H) Beta hPhe (4-F), alpha-Me-Trp, 4-phenylcyclohexyl, Phe (4-CF)3) alpha-MePhe, beta hNal, beta hPhe, beta hTyr, beta hTrp, Nva (5-phenyl), Phe, His, hPhe, Tic, Tqa, Trp, Tyr, Phe (4-OMe), Phe (4-Me), Trp (2,5, 7-tri-t-butyl), Phe (4-O allyl), Tyr (3-tBu), Phe (4-guanidino, Phe (4-OBzl), Octgly, Glu (Bzl), 4-phenylbenzylalanine, Phe [4- (2-aminoethoxy)]5-hydroxy-Trp, 6-chloro-Trp, N-MeTrp, 1,2,3, 4-tetrahydro-norhaben, Phe (4-CONH) 2) Phe (3, 4-dimethoxy), Phe (2, 3-Cl)2)、Phe(2,3-F2) Phe (4-F), 4-phenylcyclohexylalanine or Bip or any of the corresponding alpha-methyl amino acid forms described above; x12 is α -MeLys, α 0-MeOrn, α 1-MeLeu, Aib, (D) Ala, (D) Asn, (D) Leu, (D) Asp, (D) Phe, (D) Thr, 3-Pal, Aib, α 3-Ala, α 4hGlu, α 5hAla, α 6hLeu, α 7hVal, α 8-spiro-pip, Cha, Chg, Asp, Dab, Dap, α 2-diethylGly, Glu, Phe, hLeu, hArg, hLeu, Ile, Lys, Leu, Asn, N-MeLeu, N-MeArg, Ogl, Orn, Pro, Gln, Arg, Ser, Thr, or Tle, or any of the corresponding α 9-methylamino forms; x13 is Lys (Ac), (D) Asn, (D) Leu, (D) Thr, (D) Phe, Ala, Aib, α -MeLeu, β -Ala, β hGlu, β hLa, β hLeu, β hVal, β -spiro-pip, Cha, Chg, Asp, Lys, Arg, Orn, Dab, Dap, α 0-diethylGly, Glu, Phe, hLeu, Lys, Leu, Asn, Ogl, Pro, Gln, Asp, Arg, Ser, spiro-pip, Thr, Tba, Tlc, Val, or Tyr or any of the corresponding α 1-methyl amino acid forms; x14 is Asn, Glu, Phe, Gly, His, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Tic, or Tyr, or any of the corresponding α 2-methyl amino acid forms described above; and X15 is Gly, (D) Ala, (D) Asn, (D) Asp, Asn, (D) Leu, (D) Phe, (D) Thr, Ala, AEA, Asp, Glu, Phe, Gly, Lys, Leu, Pro, Gln, Arg, or Ser, or any of the corresponding α -methyl amino acid forms.
In certain embodiments, X3 is present. In a specific embodiment, X3 is Glu, (D) Glu, Arg, (D) Arg, Phe, (D) Phe, 2-Nal, Thr, Leu, (D) Gln. In certain embodiments, X3 is (D) Arg or (D) Phe. In particular embodiments, X1 and X2 are absent, and X3 is present.
In certain embodiments, X5 is Gln, Ala, Cit, Asp, Dab, Dap, Cit, Glu, Phe, Gly, His, hCys, Lys, Leu, Met, Asn, N-Me-Ala, N-Me-Asn, N-Me-Lys, α Me-Orn, N-Me-Gln, N-Me-Arg, α -MeSer, Orn, Pro, Arg, Ser, Thr, or Val. In certain embodiments, X5 is Gln, Ala, Cit, Asp, Dab, Dap, Glu, Phe, Gly, His, hCys, Lys, Leu, Met, Asn, N-Me-Ala, N-Me-Asn, N-Me-Lys, α -Me-Orn, N-Me-Gln, N-Me-Arg, Orn, Pro, Arg, Ser, Thr, or Val. In certain embodiments, X5 is gin or Asn.
In certain embodiments, X6 is Thr, Asp, Glu, Phe, Asn, Pro, Arg, or Ser.
In certain embodiments, X7 is Trp.
In certain embodiments, X8 is Gln, Glu, Phe, Lys, Asn, Pro, Arg, Val, Thr, or Trp.
In certain embodiments, X10 is a Tyr analog or a Phe analog. In a specific embodiment, X10 is a Phe analog.
In certain embodiments wherein X10 is a Phe analog, X10 is selected from hPhe, Phe (4-OMe), α -Me-Phe, hPhe (3, 4-dimethoxy), Phe (4-CONH)2) Phe (4-phenoxy), Phe (4-guanidino), Phe (4-tBu), Phe (4-CN), Phe (4-Br), Phe (4-OBzl), Phe (4-NH)2) Phe (4-F), Phe (3, 5-dif), Phe (CH)2CO2H)、Phe(5-F)、Phe(3,4-Cl2)、Phe(4-CF3) beta-bis-PheAla, Phe (4-N)3) And Phe [4- (2-aminoethoxy)]. In a particular embodiment, X10 is Phe (4-OMe) or Phe [4- (2-aminoethoxy)]. In a specific embodiment, X10 is Phe (4-OMe), Phe (4-CONH)2) Or Phe [4- (2-aminoethoxy)]. In certain embodiments wherein X10 is a Phe analog, X10 is selected from the group consisting of hPhe, Phe (4-OMe), α -Me-Phe, hPhe (3, 4-dimethoxy), Phe (4)-CONH2) Phe (4-phenoxy), Phe (4-guanidino), Phe (4-tBu), Phe (4-CN), Phe (4-Br), Phe (4-OBzl), Phe (4-NH)2) Phe (4-F), Phe (3, 5-di-F), Phe (CH)2CO2H)、Phe(5-F)、Phe(3,4-Cl2)、Phe(4-CF3) beta-bis-PheAla, Phe (4-N)3) And Phe [4- (2-aminoethoxy)]. In a specific embodiment, X10 is Phe (4-OMe).
In certain embodiments wherein X10 is a Tyr analog, X10 is selected from the group consisting of hTyr, α -MeTyr, N-Me-Tyr, Tyr (3-tBu), Phe (4-CONH)2) Phe [4- (2-aminoethoxy)]And β hTyr. In certain embodiments wherein X10 is a Tyr analog, X10 is selected from the group consisting of hTyr, α -MeTyr, N-Me-Tyr, Tyr (3-tBu), and bhTyr.
In certain embodiments, X10 is Tyr, Phe (4-OMe), Phe [4- (2-aminoethoxy)]、Phe(4-CONH2) Or 2-Nal. In certain embodiments, X10 is Phe (4-OMe) or Phe [4- (2-aminoethoxy)]. In certain embodiments, X10 is not Tyr.
In certain embodiments, X11 is a Trp analog. In particular embodiments, X11 is 2-Nal or 1-Nal. In certain embodiments, X11 is 2-Nal.
In certain embodiments, X12 is Aib, α -MeLys, or α -MeLeu.
In particular embodiments of the peptide inhibitor of formula II, one or both of X4 or X9 is Pen. In particular embodiments, both X4 and X9 are Pen.
In certain embodiments, the peptide inhibitor of formula II is cyclized. In a specific embodiment, the peptide inhibitor of formula II is cyclized via an intramolecular bond between X4 and X9. In a specific embodiment, the intramolecular bond is a disulfide bond. In particular embodiments, both X4 and X9 are Pen.
In certain embodiments, the peptide inhibitor of formula II is linear or not cyclized. In particular embodiments of the linear peptide inhibitors of formula I, X4 and/or X9 are any amino acid.
In particular embodiments of the peptide inhibitor of formula II, one or more, two or more, or all three of X1, X2, and X3 are absent. In certain embodiments, X1 is absent. In certain embodiments, X1 and X2 are absent. In certain embodiments, X1, X2, and X3 are absent.
In particular embodiments of the peptide inhibitor of formula II, one or more, two or more, three or more, four or more, or all of X16, X17, X18, X19, and X20 are absent. In particular embodiments of the peptide inhibitors of formula I, one or more, two or more, three or more, or all of X17, X18, X19, and X20 are absent. In certain embodiments, one or more, two or more, or all three of X17, X19, and X20 are absent. In certain embodiments, one or more of X1, X2, and X3 is absent; and one or more, two or more, three or more, or none of X17, X18, X19, and X20 are present.
In a specific embodiment of the peptide inhibitor of formula II, X18 is (D) -Lys. In certain embodiments, X18 is (D) -Lys, and X17 is absent.
In particular embodiments of the peptide inhibitor of formula II, the peptide inhibitor comprises one or more, two or more, three or more, or four of the following characteristics: x5 is Asn, Arg or Gln; x6 is Thr; x7 is Trp; and X8 is Gln. In particular embodiments of the peptide inhibitor of formula I, X4 is Pen; x5 is Gln, Asn, or Arg; x6 is Thr; x7 is Trp, 5-hydroxy-Trp, 6-chloro-Trp, N-MeTrp, α -Me-Trp, or 1,2,3, 4-tetrahydro-norhaben; x8 is Gln; and X9 is Pen. In particular embodiments, X5 is Gln. In certain embodiments, X1, X2, and X3 are absent. In particular embodiments, both X4 and X9 are Pen.
In particular embodiments of the peptide inhibitor of formula II, the peptide inhibitor comprises one or more, two or more, three or more, four or more, five or more, six or more, or seven of the following characteristics: x10 is Tyr, Phe analog, Tyr analog or 2-Nal; x11 is Trp, 5-hydroxy-Trp, 6-chloro-Trp, N-MeTrp, α -Me-Trp, 1,2,3, 4-tetrahydro-norharman, 2-Nal or 1-Nal; x12 is Aib, alpha-MeLys, alpha-MeOrn and alpha-MeLeu; x13 is Lys, Glu, or Lys (Ac); x14 is Phe or Asn; x15 is Gly, Ser or Ala; and X16 is absent or is AEA. In certain embodiments, X10 is Tyr, Phe (4-OMe), Phe [4- (2-aminoethoxy)]、Phe(CONH2) Or 2-Nal. In certain embodiments, X11 is 2-Nal or 1-Nal. In certain embodiments, X10 is not Tyr. In certain embodiments, X1, X2, and X3 are absent. In particular embodiments, both X4 and X9 are Pen.
In particular embodiments of the peptide inhibitor of formula II, the peptide inhibitor comprises one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, or eleven of the following features: x5 is Arg or Gln; x6 is Thr; x7 is Trp; x8 is Gln; x10 is a Phe analog; x11 is Trp, 2-Nal or 1-Nal; x12 is Aib, α -MeLys or α -MeOrn; x13 is Lys, Glu, or Lys (Ac); x14 is Asn; x15 is Gly, Ser or Ala; and X16 is absent or is AEA. In certain embodiments, X10 is Phe (4-OMe) or Phe [4- (2-aminoethoxy) ]. In certain embodiments, X11 is 2-Nal or 1-Nal. In certain embodiments, X1, X2, and X3 are absent. In particular embodiments, both X4 and X9 are Pen.
In particular embodiments of the peptide inhibitor of formula II, the peptide is cyclized via X4 and X9; x4 and X9 are Pen; x5 is Gln; x6 is Thr; x7 is Trp; x8 is Gln; x10 is Tyr, Phe analog, or 2-Nal; x11 is Trp, 2-Nal or 1-Nal; x12 is Arg, α -MeLys, α -MeOrn, or α -MeLeu; x13 is Lys, Glu, or Lys (Ac); x14 is Phe or Asn; x15 is Gly, Ser or Ala; and X16 is absent. In certain embodiments, X10 is Tyr, Phe (4-OMe), Phe [4- (2-aminoethoxy) ], Phe (4-OMe), or 2-Nal. In certain embodiments, X10 is Phe (4-OMe). In certain embodiments, X10 is not Tyr. In certain embodiments, X11 is 2-Nal or 1-Nal. In certain embodiments, X1, X2, and X3 are absent.
In particular embodiments of the peptide inhibitor of formula II, the peptide is cyclized via X4 and X9; x4 and X9 are Pen; x5 is Gln; x6 is Thr; x7 is Trp; x8 is Gln; x10 is Tyr, Phe (4-OMe) or 2-Nal; x11 is Trp, 2-Nal or 1-Nal; x12 is Arg, α -MeLys, or α -MeOrn; x13 is Lys, Glu, or Lys (Ac); x14 is Phe or Asn; x15 is Gly; and X16 is absent. In certain embodiments, X10 is Phe (4-OMe). In certain embodiments, X11 is 2-Nal or 1-Nal. In certain embodiments, X1, X2, and X3 are absent.
In particular embodiments of the peptide inhibitor of formula II, the peptide is cyclized via X4 and X9; x4 and X9 are Pen; x5 is Gln; x6 is Thr; x7 is Trp; x8 is Gln; x10 is Phe (4-OMe) or Phe [4- (2-aminoethoxy) ]; x11 is Trp, 2-Nal or 1-Nal; x12 is α -MeLys, α -MeOrn, or α -MeLeu; x13 is Lys, Glu, or Lys (Ac); x14 is Asn; x15 is Gly, Ser or Ala; and X16 is absent. In certain embodiments, X10 is Phe (4-OMe). In certain embodiments, X11 is 2-Nal or 1-Nal. In certain embodiments, X1, X2, and X3 are absent.
In a specific embodiment of the peptide inhibitor of formula II, X10 is not Tyr.
In certain embodiments, the invention includes optionally cyclized peptides of optionally 8 to 35, 8 to 20, 8 to 16, or 8 to 12 amino acids in length comprising or consisting of a core sequence of formula lib:
Pen-Xaa5-Xaa6-Trp-Xaa8-Pen-Xaa10-[(2-Nal)](IIb)(SEQ ID NO:1403)
wherein Xaa5, Xaa6 and Xaa8 are any amino acid residue; and Xaa10 is a Phe analog, wherein the peptide inhibits binding of IL-23 to IL-23R. In a specific embodiment, X10 is a Phe analog selected from the group consisting of α -Me-Phe, Phe (4-OMe), Phe (4-OBzl), Phe (4-OMe), Phe (4-CONH)2)、Phe(3,4-Cl2)、Phe(4-tBu)、Phe(4-NH2)、Phe(4-Br)、Phe(4-CN)、Phe(4-CO2H) Phe [4- (2-aminoethoxy)]Or Phe (4-guanidino). In specific embodiments, Xaa10 is Phe (4-OMe) or Phe [4- (2-aminoethoxy) ]. In one embodiment, Xaa10 is Phe (4-OMe). In certain embodiments, the peptide is cyclized via an intramolecular bond between Pen of Xaa4 and Pen of Xaa 9. In particular embodiments, the peptide is a peptide inhibitor of formula II, and wherein in certain embodiments, X1, X2, and X3 are absent.In particular embodiments, the peptide inhibits the binding of IL-23 to IL-23R. In certain embodiments, the peptide of formula IIb further comprises an amino acid bound to the N-terminal Pen residue. In particular embodiments, the amino acid bound is Glu, (D) Glu, Arg, (D) Arg, Phe, (D) Phe, 2-Nal, Thr, Leu, or (D) Gln. In certain embodiments, it is (D) Arg or (D) Phe.
In certain embodiments, the invention includes optionally cyclized peptides of optionally 8 to 35, 8 to 20, 8 to 16, or 8 to 12 amino acids in length comprising or consisting of a core sequence of formula IIc:
Pen-Xaa5-Xaa6-Trp-Xaa8-Pen-Xaa10-[(2-Nal)](IIc)(SEQ ID NO:1404)
wherein Xaa5, Xaa6 and Xaa8 are any amino acid residue; and Xaa10 is Tyr, Phe analog, α -Me-Tyr, α -Me-Trp, or 2-Nal, wherein the peptide inhibits the binding of IL-23 to IL-23R. In certain embodiments, X10 is Tyr, Phe (4-OMe), Phe [4- (2-aminoethoxy) ]alpha-Me-Tyr, alpha-Me-Phe, alpha-Me-Trp or 2-Nal. In certain embodiments, Xaa10 is Tyr, Phe (4-OMe), Phe (CONH)2) Phe [4- (2-aminoethoxy)]Or 2-Nal. In certain embodiments, Xaa10 is Tyr, Phe (4-OMe), Phe [4- (2-aminoethoxy)]Or 2-Nal. In specific embodiments, Xaa10 is Phe (4-OMe) or Phe [4- (2-aminoethoxy)]. In one embodiment, Xaa10 is Phe [4- (2-aminoethoxy)]Or Phe (CONH)2). In specific embodiments, Xaa10 is Phe (4-OMe) or Phe [4- (2-aminoethoxy)]. In one embodiment, Xaa10 is Phe [4- (2-aminoethoxy)]. In certain embodiments, Xaa10 is not Tyr. In certain embodiments, the peptide is cyclized via an intramolecular bond between Pen of Xaa4 and Pen of Xaa 9. In particular embodiments, the peptide is a peptide inhibitor of formula II, and wherein in certain embodiments, X1, X2, and X3 are absent. In particular embodiments, the peptide inhibits the binding of IL-23 to IL-23R. In certain embodiments, the peptide of formula IIc further comprises an amino acid bound to the N-terminal Pen residue. In particular embodiments, the amino acid bound is Glu, (D) Glu, Arg, (D) Arg, Phe, (D) Phe, 2-Nal, Thr, Leu, or (D) Gln. In some embodiments of the present invention, the substrate is, It is (D) Arg or (D) Phe.
In certain embodiments, the invention includes optionally cyclized peptides of optionally 8 to 35, 8 to 20, 8 to 16, or 8 to 12 amino acids in length comprising or consisting of a core sequence of formula IId:
Pen-Xaa5-Xaa6-Trp-Xaa8-Pen-Phe [4- (2-aminoethoxy) ] - [2-Nal ] (IId) (SEQ ID NO:1405)
Wherein Xaa5, Xaa6 and Xaa8 are any amino acid residue. In certain embodiments, the peptide comprises a disulfide bond between Xaa4 and Xaa 9. In certain embodiments, the peptide is a peptide inhibitor of formula I, and wherein in certain embodiments, X1, X2, and X3 are absent. In particular embodiments, the peptide inhibits the binding of IL-23 to IL-23R. In certain embodiments, the peptide of formula IId further comprises an amino acid bound to the N-terminal Pen residue. In particular embodiments, the amino acid bound is Glu, (D) Glu, Arg, (D) Arg, Phe, (D) Phe, 2-Nal, Thr, Leu, or (D) Gln. In certain embodiments, it is (D) Arg or (D) Phe.
In particular embodiments of the peptide inhibitor of formula II, the peptide inhibitor has any of the structures shown in table 2, 3, E4A, table E4B, table E8, or table E11 or comprises an amino acid sequence shown in table 2, table 3, table E4A, table E4B, table E8, or table E11.
TABLE 2 exemplary Di-Pen inhibitors
Figure GDA0003395945150001211
Figure GDA0003395945150001221
Figure GDA0003395945150001231
Figure GDA0003395945150001241
TABLE 3 exemplary peptides containing Ac- [ Pen ] -XXWX- [ Pen ] -XXXXXXXX motif (SEQ ID NO:1406) and analogs thereof
Figure GDA0003395945150001251
Figure GDA0003395945150001261
Figure GDA0003395945150001271
Exemplary peptide inhibitors containing a thioether bond
In certain embodiments, the present invention includes a peptide inhibitor of the interleukin-23 receptor, or a pharmaceutically acceptable salt or solvate thereof, wherein the peptide inhibitor has the structure of formula III:
R1-X-R2(III)
wherein R is1Is a bond, hydrogen, C1-C6 hydrocarbyl, C6-C12 aryl, C6-C12 aryl, C1-C6 hydrocarbyl, C1-C20 hydrocarboxyl, hydrocarbyl sulfonate, acid, γ -Glu, or pGlu (added to the N-terminus), and includes a pegylated form of any of the foregoing alone or as a spacer (e.g., 200Da to 60,000 Da);
R2is a bond, OH or NH2(ii) a And
x is an amino acid sequence of 8 to 20 amino acids or 8 to 35 amino acids,
in particular embodiments of the peptide inhibitor of formula III, X comprises or consists of the sequence of formula IIIa:
X1-X2-X3-X4-X5-X6-X7-X8-X9-X10-X11-X12-X13-X14-X15-X16-X17-X18-X19-X20(IIIa)(SEQ ID NO:1407)
wherein
X1 is absent or is any amino acid;
x2 is absent or is any amino acid;
x3 is absent or is any amino acid;
x4 is Abu, Pen or Cys;
x5 is any amino acid;
x6 is any amino acid;
x7 is Trp, Bip, Gln, His, Glu (Bzl), 4-phenylbenzylalanine, Tic, Phe [4- (2-aminoethoxy) ]、Phe(3,4-Cl2) Phe (4-OMe), 5-hydroxy-Trp, 6-chloro-Trp, N-MeTrp, alpha-MeTrp, 1,2,3, 4-tetrahydro-norharpagne, Phe (4-CO)2H)、Phe(4-CONH2)、Phe(3,4-(OCH3)2)、Phe(4-CF3) β -dipheAla, Phe (4-tBu), Glu, Gly, Ile, Asn, Pro, Arg, Thr, or Octgly or any of the corresponding α -methyl amino acid forms described above;
x8 is any amino acid;
x9 is Abu, Pen or Cys;
x10 is 1-Nal, 2-Nal, Aic, Bip, (D) Cys, Cha, DMT, (D) Tyr, Glu, Phe, His, Trp, Thr, Tic, Tyr, 4-pyridyl Ala, Octgly, a Phe analog, or a Tyr analog (optionally, Phe (3, 4-F)2)、Phe(3,4-Cl2) F (3-Me), Phe [4- (2-aminoethoxy)]Phe [4- (2- (acetyl-aminoethoxy)]、Phe(4-Br)、Phe(4-CONH2) Phe (4-Cl), Phe (4-CN), Phe (4-guanidino), Phe (4-Me), Phe (4-NH)2)、Phe(4-N3) Phe (4-OMe), Phe (4-OBzl)), or any of the foregoing corresponding alpha-methyl amino acid forms;
x11 is 2-Nal, 1-Nal, 2, 4-dimethyl Phe, Bip, 4-phenylcyclohexyl, Glu (Bzl), 4-phenylbenzylalanine, Tic, Phe [4- (2-aminoethoxy)]、Phe(3,4-Cl2)、Phe(3,4-F2) Beta hPhe (4-F), Phe (4-OMe), 5-hydroxy-Trp, 6-chloro-Trp, N-MeTrp, alpha-MeTrp, 1,2,3, 4-tetrahydro-norharpagne, Phe (4-CO)2H)、Phe(4-CONH2) Phe (3, 4-dimethoxy), Phe (4-CF)3)、Phe(2,3-Cl2)、Phe(3,4-Cl2)、Phe(2,3-F2) Phe (4-F), 4-phenylcyclohexylalanine, alpha-MePhe, beta-hNal, beta-hPhe, and beta-hPhe, β hTyr, β hTrp, Bip, Nva (5-phenyl), Phe, His, hPhe, Tqa, Trp, Tyr, Phe (4-Me), Trp (2,5, 7-tri-tert-butyl), Phe (4-O allyl), Tyr (3-tBu), Phe (4-guanidino), Phe (4-OBzl), or Octgly or any of the corresponding α -methyl amino acid forms described above;
x12 is α -MeLys, α -MeOrn, α -MeLeu, Aib, (D) Ala, (D) Asn, (D) Leu, (D) Asp, (D) Phe, (D) Thr, 3-Pal, Aib, β -Ala, β hGlu, β hAla, β hLeu, β hVal, β -spiro-pip, Cha, Chg, Asp, Dab, Dap, α -diethylGly, Glu, Phe, hLeu, Ile, Lys, Leu, Asn, N-MeLeu, N-MeArg, og, Orn, Pro, Gln, Arg, Ser, Thr or Tle, amino-4-carboxy-Tetrahydropyran (THP), Achc, Acpc, Acbc, Acvc, Aib, or any of the corresponding α -methyl amino acid forms;
x13 is Lys, Lys (Ac), (D) Asn, (D) Leu, (D) Thr, (D) Phe, Ala, Aib, α -MeLeu, β Ala, β hGlu, β hAla, β hLeu, β hVal, β -spiro-pip, Cha, Chg, Asp, Arg, Orn, Dab, Dap, α -diethylGly, Glu, Phe, hLeu, Lys, Leu, Asn, og, Pro, Gln, Asp, Arg, Ser, spiro-pip, Thr, Tba, Tlc, Val or Tyr or any of the corresponding α -methylamino forms described above;
X14 is Asn, Glu, Phe, Gly, His, Lys (Ac), Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Tic, Asp, or Tyr or any of the corresponding α -methyl amino acid forms of the foregoing;
x15 is Gly, (D) Ala, (D) Asn, (D) Asp, Asn, (D) Leu, (D) Phe, (D) Thr, Ala, AEA, Asp, Glu, Phe, Gly, Lys, Leu, Pro, Gln, Arg, β -Ala or Ser, or any of the corresponding α -methyl amino acid forms;
x16 is absent and is Gly, Ala, Asp, Ser, Pro, Asn or Thr or any of the foregoing corresponding α -methyl amino acid forms;
x17 is absent and is Glu, Ser, Gly, or Gln or any of the corresponding α -methyl amino acid forms described above;
x18 is absent or is any amino acid;
x19 is absent or is any amino acid; and
x20 is absent or is any amino acid.
In certain embodiments, X14 is Asn, Glu, Phe, Gly, His, Lys (Ac), Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Tic, or Tyr, or any of the corresponding α -methyl amino acid forms described above.
In certain embodiments of IIIa: x7 is Trp, Bip, Gln, His, Glu (Bzl), 4-phenylbenzylalanine, Tic, Phe [4- (2-aminoethoxy) ]、Phe(3,4-Cl2) Phe (4-OMe), 5-hydroxy-Trp, 6-chloro-Trp, N-MeTrp, alpha-MeTrp, 1,2,3, 4-tetrahydro-norharpagne, Phe (4-CO)2H)、Phe(4-CONH2) Phe (3, 4-dimethoxy), Phe (4-CF)3) β -dipheAla, Phe (4-tBu), Glu, Gly, Ile, Asn, Pro, Arg, Thr, or Octgly or any of the corresponding α -methyl amino acid forms described above; x10 is 1-Nal, 2-Nal, Aic, Bip, (D) Cys, Cha, DMT, (D) Tyr, Glu, Phe, His, Trp, Thr, Tic, Tyr, 4-pyridyl Ala, Octgly, a Phe analog, or a Tyr analog, or any of the foregoing corresponding alpha-methyl amino acid forms; x11 is 2-Nal, 1-Nal, 2, 4-dimethyl Phe, Bip, 4-phenylcyclohexyl, Glu (Bzl), 4-phenylbenzylalanine, Tic, Phe [4- (2-aminoethoxy)]、Phe(3,4-Cl2)、Phe(3,4-F2) Beta hPhe (4-F), Phe (4-OMe), 5-hydroxy-Trp, 6-chloro-Trp, N-MeTrp, alpha-MeTrp, 1,2,3, 4-tetrahydro-norharpagne, Phe (4-CO)2H)、Phe(4-CONH2) Phe (3, 4-dimethoxy), Phe (4-CF)3)、Phe(2,3-Cl2)、Phe(2,3-F2) Phe (4-F), 4-phenylcyclohexylalanine,. alpha. -MePhe,. beta.hNal,. beta.0 hPhe,. beta.1 hTyr,. beta.2 hTRp, Bip, Nva (5-phenyl), Phe, His, hPhe, Tqa, Trp, Tyr, Phe (4-Me), Trp (2,5, 7-tri-tert-butyl), Phe (4-O allyl), Tyr (3-tBu), Phe (4-guanidino), Phe (4-OBzl) or Octgly or any of the corresponding β 5-methylamino forms described above; x12 is α -MeLys, α -MeOrn, α -MeLeu, Aib, (D) Ala, (D) Asn, (D) Leu, (D) Asp, (D) Phe, (D) Thr, 3-Pal, Aib, β 3-Ala, β 4hGlu, β hAla, β hLeu, β hVal, β -spiro-pip, Cha, Chg, Asp, Dab, Dap, α -diethylGly, Glu, Phe, hLeu, hArg, h Leu, Ile, Lys, Leu, Asn, N-MeLeu, N-MeArg, Ogl, Orn, Pro, Gln, Arg, Ser, Thr, or Tle, or any of the corresponding alpha-methyl amino acid forms; x13 is Lys (Ac), (D) Asn, (D) Leu, (D) Thr, (D) Phe, Ala, Aib, α -MeLeu, β Ala, β hGlu, β hAla, β hLeu, β hVal, β -spiro-pip, Cha, Chg, Asp, Arg, Orn, Dab, Dap, α 0-diethylGly, Glu, Phe, hLeu, Lys, Leu, Asn, Ogl, Pro, Gln, Asp, Arg, Ser, spiro-pip, Thr, Tba, Tlc, Val, or Tyr or any of the corresponding α 1-methyl amino acid forms; x14 is Asn, Glu, Phe, Gly, His, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Tic, or Tyr or any of the corresponding α -methyl amino acid forms; and X15 is Gly, (D) Ala, (D) Asn, (D) Asp, Asn, (D) Leu, (D) Phe, (D) Thr, Ala, AEA, Asp, Glu, Phe, Gly, Lys, Leu, Pro, Gln, Arg, or Ser or any of the corresponding α -methyl amino acid forms described above.
In certain embodiments, X3 is present. In particular embodiments, X3 is Glu, (D) Glu, Arg, (D) Arg, Phe, (D) Phe, 2-Nal, Thr, Leu, or (D) Gln. In certain embodiments, it is (D) Arg or (D) Phe.
In a specific embodiment, X5 is Gln, Ala, Cys, Cit, Asp, Dab, Dap, Glu, Phe, Gly, His, hCys, Lys, Leu, Met, Asn, N-Me-Ala, N-M-Asn, N-Me-Lys, N-Me-Gln, N-Me-Arg, Orn, Pro, Pen, Gln, Arg, Ser, Thr, or Val.
In particular embodiments, X6 is Thr, Asp, Glu, Phe, Asn, Pro, Arg, Ser, or Thr.
In a specific embodiment, X8 is Gln, Glu, Phe, Lys, Asn, Pro, Arg, Val, Thr, or Trp.
In certain embodiments, X10 is a Tyr analog or a Phe analog. In a particular embodiment, X10 is Phe (4-OMe), Phe (CONH)2) Or Phe [4- (2-aminoethoxy)]. In certain embodiments, X10 is a Tyr analog or a Phe analog. In a particular embodiment, X10 is Phe (4-OMe) or Phe [4- (2-aminoethoxy)]。
In certain embodiments wherein X10 is a Phe analog, X10 is selected fromhPhe, Phe (4-OMe), alpha-MePhe, hPhe (3, 4-dimethoxy), Phe (4-CONH)2) Phe (4-O-Bzl)), Phe (4-guanidino), Phe (4-tBu), Phe (4-CN), Phe (4-Br), Phe (4-NH)2) Phe (4-F), Phe (3, 5-di-F), Phe (CH)2CO2H)、Phe(5-F)、Phe(3,4-Cl2) Phe (4-CF3), beta-dipheAla, Phe (4-N)3) And Phe [4- (2-aminoethoxy)]. In a specific embodiment, X10 is Phe [4- (2-aminoethoxy) ]Or Phe (CONH)2). In a specific embodiment, X10 is Phe [4- (2-aminoethoxy)]。
In certain embodiments, wherein X10 is a Tyr analog, X10 is selected from the group consisting of hTyr, N-Me-Tyr, Tyr (3-tBu), Phe (4-OMe), and bhTyr. In a specific embodiment, X10 is Phe (4-OMe).
In a specific embodiment, X10 is Tyr, Phe (4-OMe), Phe (4-OBzl), Phe (4-OMe), Phe (4-CONH)2)、Phe(3,4-Cl2) Phe (4-tBu), Phe (4-NH2), Phe (4-Br), Phe (4-CN), Phe (4-carboxy), Phe [4- (2-aminoethoxy)]Or Phe (4-guanidino). In a specific embodiment, X10 is not Tyr.
In certain embodiments, X11 is Trp or an analog of Trp. In particular embodiments, X11 is 2-Nal or 1-Nal.
In a specific embodiment, the peptide inhibitor of formula III is cyclized. In certain embodiments, the peptide inhibitor is cyclized via an intramolecular bond between X4 and X9. In certain embodiments, the intramolecular linkage is a thioether linkage.
In certain embodiments, the peptide inhibitor of formula III is linear or not cyclized. In particular embodiments of the linear peptide inhibitors of formula III, X4 and/or X9 are any amino acid.
In particular embodiments of the peptide inhibitors of formula III, one or more, two or more, or all three of X1, X2, and X3 are absent. In certain embodiments, X1 is absent. In certain embodiments, X1 and X2 are absent. In certain embodiments, X1, X2, and X3 are absent.
In particular embodiments of the peptide inhibitors of formula III, one or more, two or more, three or more, four or more, or all of X16, X17, X18, X19, and X20 are absent. In particular embodiments of the peptide inhibitors of formula III, one or more, two or more, three or more, or all of X17, X18, X19, and X20 are absent. In certain embodiments, one or more, two or more, or all three of X17, X19, and X20 are absent. In certain embodiments, one or more of X1, X2, and X3 is absent; and one or more, two or more, three or more, or none of X17, X18, X19, and X20 are present.
In particular embodiments of the peptide inhibitor of formula III, one of X4 or X9 is Abu and the other of X4 or X9 is not Abu. In certain embodiments, X4 is Abu and X9 is Cys.
In particular embodiments, the peptide inhibitor of formula III comprises one or more, two or more, three or more, or four of the following features: x5 is Arg or Gln; x6 is Thr; x7 is Trp; and X8 is Gln. In particular embodiments, X5 is Gln, X6 is Thr, X7 is Trp, and X8 is Gln. In certain embodiments, X5 is Gln. In certain embodiments, X1, X2, and X3 are absent. In certain embodiments, X4 is Abu and X9 is Cys.
In particular embodiments, the peptide inhibitor of formula III comprises one or more, two or more, three or more, four or more, five or more, six or more, or seven of the following features: x10 is Tyr or Phe analog; x11 is Trp, 2-Nal, 1-Nal, Phe (4-O-allyl), Tyr (3-tBu), Phe (4-guanidino), Phe (4-OBzl) or Phe (4-Me); x12 is Arg, hLeu, (D) Asn, or any alpha methyl amino acid including that of Aib, alpha-MeLys, alpha-MeLeu, or alpha-MeOrn; x13 is Lys, Glu, or Lys (Ac); x14 is Phe or Asn; x15 is β -Ala, Gln, Gly, Ser, Ala; and X16 is absent or is AEA. In particular embodiments, the peptide inhibitor of formula III comprises one or more, two or more, three or more, four or more, five or more, six or more, or seven of the following featuresThe method comprises the following steps: x10 is Tyr or Phe analog; x11 is Trp, 2-Nal, 1-Nal, Phe (4-O-allyl), Tyr (3-tBu), Phe (4-guanidino), Phe (4-OBzl) or Phe (4-Me); x12 is Arg, hLeu, (D) Asn, or any alpha methyl amino acid including Aib, alpha-MeLys, alpha-MeLeu, or alpha-MeOrn; x13 is Lys, Glu or Lys (Ac); x14 is Phe or Asn; x15 is Gly, Ser, Ala; and X16 is absent or is AEA. In certain embodiments, the Phe analog is Phe (4-OBzl), Phe (4-OMe), Phe (4-CONH) 2)、Phe(3,4-Cl2) Phe (4-tBu), Phe (4-NH2), Phe (4-Br), Phe (4-CN), Phe (4-carboxy), Phe [4- (2-aminoethoxy)]Or Phe (4-guanidino). In certain embodiments, X11 is 2-Nal or 1-Nal. In certain embodiments, X1, X2, and X3 are absent. In certain embodiments, X4 is Abu and X9 is Cys.
In specific embodiments, the peptide inhibitor of formula III comprises one or more, two or more, three or more, four or more, five or more, six or more, or seven of the following features: x10 is Tyr or Phe analog; x11 is Trp, 2-Nal, 1-Nal, Phe (4-O-allyl), Tyr (3-tBu), Phe (4-guanidino), Phe (4-OBzl) or Phe (4-Me); x12 is Arg, hLeu, (D) Asn, 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Acvc, Agp, Aib, α -diethylGly, α -MeLys (Ac), α -Me-Leu, α -MeOrn, α -MeSer, α -MeVal; x13 is Lys, Glu, or Lys (Ac); x14 is Phe or Asn; x15 is Gly; and X16 is absent or is AEA. In certain embodiments, the Phe analog is Phe (4-OBzl), Phe (4-OMe), Phe (4-CONH)2)、Phe(3,4-Cl2)、Phe(4-tBu)、Phe(4-NH2) Phe (4-Br), Phe (4-CN), Phe (4-carboxy), Phe (4- (2-aminoethoxy)) or Phe (4-guanidino). In certain embodiments, X11 is 2-Nal or 1-Nal. In certain embodiments, X1, X2, and X3 are absent. In certain embodiments, X4 is Abu and X9 is Cys.
In particular embodiments, the peptide inhibitor of formula III comprises one or more, two or more, three or more, four or more, five or more, six or more, seven or more of the following featuresMore, eight or more, nine or more, ten or more, or eleven: x5 is Arg or Gln; x6 is Thr; x7 is Trp; x8 is Gln; x10 is a Phe analog; x11 is Trp, 2-Nal, 1-Nal, Phe (4-O-allyl), Tyr (3-tBu), Phe (4-guanidino), Phe (Bzl) or Phe (4-Me); x12 is Aib, α -MeLys, α -MeLeu, 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Acvc, Agp, Aib, α 0-diethylGly, α 1-MeLys, α 2-MeLys (Ac), α -Me-Leu, α -MeSer, α -MeVal, α -MeOrn; x13 is Lys, Glu, or Lys (Ac); x14 is Phe or Asn; x15 is beta-Ala, Gly, Ser, Ala; and X16 is absent or is AEA. In specific embodiments, the peptide inhibitor of formula III comprises one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, or eleven of the following features: x5 is Arg or Gln; x6 is Thr; x7 is Trp; x8 is Gln; x10 is a Phe analog; x11 is Trp, 2-Nal, 1-Nal, Phe (4-O-allyl), Tyr (3-tBu), Phe (4-guanidino), Phe (Bzl) or Phe (4-Me); x12 is Aib, α -MeLys, α -MeLeu or α -MeOrn; x13 is Lys, Glu, or Lys (Ac); x14 is Phe or Asn; x15 is Gly, Ser, Ala; and X16 is absent or is AEA. In certain embodiments, the Phe analog is Phe (4-OBzl), Phe (4-OMe), Phe [4- (2 aminoethoxy) ]、Phe(4-CONH2)、Phe(3,4-Cl2)、Phe(4-tBu)、Phe(4-NH2)、Phe(4-Br)、Phe(4-CN)、Phe(4-CO2H) Or Phe (4-guanidino). In certain embodiments, X11 is 2-Nal or 1-Nal. In certain embodiments, X1, X2, and X3 are absent. In certain embodiments, X4 is Abu and X9 is Cys.
In particular embodiments, the peptide inhibitor of formula III comprises one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, or eleven of the following features: x5 is Arg or Gln; x6 is Thr; x7 is Trp; x8 is Gln; x10 is Tyr or Phe analog; x11 is Trp, 2-Nal, 1-Nal, Phe(4-O-allyl), Tyr (3-tBu), Phe (4-guanidino), Phe (Bzl), or Phe (4-Me); x12 is Arg, hLeu, (D) Asn, 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Acvc, Aib, α -diethylGly, α -MeLys (Ac), α -Me-Leu, α -MeSer, α -MeVal; x13 is Lys, Glu, or Lys (Ac); x14 is Phe or Asn; x15 is β -Ala, Asn or Gly; and X16 is absent or is AEA. In particular embodiments, the peptide inhibitor of formula III comprises one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, or eleven of the following features: x5 is Arg or Gln; x6 is Thr; x7 is Trp; x8 is Gln; x10 is Tyr or Phe analog; x11 is Trp, 2-Nal, 1-Nal, Phe (4-O-allyl), Tyr (3-tBu), Phe (4-guanidino), Phe (Bzl) or Phe (4-Me); x12 is Arg, hLeu, (D) Asn, α -MeLys, α -MeLeu or α -MeOrn, Aib; x13 is Lys, Glu or Lys (Ac); x14 is Phe or Asn; x15 is Gly; and X16 is absent or is AEA. In certain embodiments, the Phe analog is Phe (4-OBzl), Phe (4OMe), Phe (4-CONH) 2)、Phe(3,4-Cl2)、Phe(4-tBu)、Phe(4-NH2)、Phe(4-Br)、Phe(4-CN)、Phe(4-CO2H) Phe (4- (2-aminoethoxy)) or Phe (4-guanidino). In certain embodiments, X11 is 2-Nal or 1-Nal. In certain embodiments, X1, X2, and X3 are absent. In certain embodiments, X4 is Abu and X9 is Cys.
In certain embodiments, the invention encompasses an optionally cyclized 8 to 20, 8 to 16, or 8 to 12 amino acid peptide comprising or consisting of a core sequence of formula IIIb:
Xaa4-Xaa5-Xaa6-Trp-Xaa8-Xaa9-Xaa10-Xaa11(IIIb))(SEQ ID NO:1408)
wherein Xaa4 and Xaa9 are each independently selected from Abu and Cys, wherein Xaa4 and Xaa9 are not identical; xaa5, Xaa6 and Xaa8 are any amino acid residue; xaa10 is Tyr, a Phe analog, or 2-Nal, and Xaa11 is 2-Nal or Trp, wherein the peptide inhibits the binding of IL-23 to IL-23R. In specific embodiments, Xaa10 is Phe (4-OMe), 2-Nal, or Phe [4- (2-aminoethoxy) ]. In one embodiment, Xaa10 is Phe (4-OMe). In one embodiment, Xaa7 is Phe [4- (2-aminoethoxy) ]. In one embodiment, Xaa11 is 2-Nal. In certain embodiments, the peptide is cyclized via Xaa4 and Xaa 9. In a particular embodiment, the Phe analog is Phe [4- (2 aminoethoxy) ] or Phe (4-OMe). In certain embodiments, Xaa4 is Abu and Xaa9 is Cys, and the peptide is cyclized via Xaa4 and Xaa 9. In particular embodiments, the peptide is a peptide inhibitor of formula III, and wherein in certain embodiments, X1, X2, and X3 are absent. In particular embodiments, the peptide inhibits the binding of IL-23 to IL-23R. In certain embodiments, the peptide of formula IIIb comprises Glu, (D) Glu, Arg, (D) Arg, Phe, (D) Phe, 2-Nal, Thr, Leu, or (D) Gln bound to Xaa 4. In certain embodiments, it is (D) Arg or (D) Phe.
In certain embodiments, the invention encompasses an optionally cyclized 8 to 20, 8 to 16, or 8 to 12 amino acid peptide comprising or consisting of a core sequence of formula IIIc:
Abu-Xaa5-Xaa6-Trp-Xaa8-Cys-[Phe(4-OMe)]-(2-Nal)(IIIc)(SEQ ID NO:1409)
wherein Xaa5, Xaa6 and Xaa8 are any amino acid residue; and wherein the peptide inhibits the binding of IL-23 to IL-23R. In certain embodiments, the peptide is cyclized via Abu of Xaa4 and Cys of Xaa 9. In certain embodiments, the peptide is a peptide inhibitor of formula III, and wherein in certain embodiments, X1, X2, and X3 are absent. In particular embodiments, the peptide inhibits the binding of IL-23 to IL-23R. In certain embodiments, the peptide of formula IIIc comprises Glu, (D) Glu, Arg, (D) Arg, Phe, (D) Phe, 2-Nal, Thr, Leu, or (D) Gln bound to Abu. In certain embodiments, it is (D) Arg or (D) Phe.
In certain embodiments, the invention encompasses an optionally cyclized 8 to 20, 8 to 16, or 8 to 12 amino acid peptide comprising or consisting of a core sequence of formula IIId:
Abu-Xaa5-Xaa6-Trp-Xaa8-Cys-Xaa10-Trp(IIId)(SEQ ID NO:1410)
wherein Xaa5, Xaa6 and Xaa8 are any amino acid residue; xaa10 is a modified Phe; and wherein the peptide inhibits the binding of IL-23 to IL-23R. In a specific embodiment, the modified Phe is Phe (4-tBu), Phe (4-guanidino), Phe [4- (2-aminoethoxy) ]、Phe(4-CO2H)、Phe(4-CN)、Phe(4-Br)、Phe(4-NH2)、PHe(CONH2) Or Phe (4-Me). In a specific embodiment, the modified Phe is Phe (4-tBu), Phe (4-guanidino), Phe [4- (2-aminoethoxy)]、Phe(4-CO2H)、Phe(4-CN)、Phe(4-Br)、Phe(4-NH2) Or Phe (4-Me). In one embodiment, Xaa10 is Phe [4- (2-aminoethoxy)]Or Phe (4-OMe). In one embodiment, Xaa10 is Phe [4- (2-aminoethoxy)]. In certain embodiments, the peptide is cyclized via Abu of Xaa4 and Cys of Xaa 9. In certain embodiments, the peptide is a peptide inhibitor of formula III, and wherein in certain embodiments, X1, X2, and X3 are absent. In particular embodiments, the peptide inhibits the binding of IL-23 to IL-23R. In certain embodiments, the peptide of formula IIId comprises Glu, (D) Glu, Arg, (D) Arg, Phe, (D) Phe, 2-Nal, Thr, Leu, or (D) Gln bound to Abu. In certain embodiments, it is (D) Arg or (D) Phe.
In certain embodiments, the invention encompasses optionally cyclized, optionally 8 to 20, 8 to 16, or 8 to 12 amino acid peptides comprising or consisting of the core sequence of formula IIIe:
Abu-Xaa5-Xaa6-Trp-Xaa8-Cys-Phe [4- (2-aminoethoxy) ] - [2-Nal ] (IIIe) (SEQ ID NO:1411)
Wherein Xaa5, Xaa6 and Xaa8 are any amino acid residue. In certain embodiments, the peptide is cyclized via Abu of Xaa4 and Cys of Xaa 9. In certain embodiments, the peptide is a peptide inhibitor of formula III, and wherein in certain embodiments, X1, X2, and X3 are absent. In particular embodiments, the peptide inhibits the binding of IL-23 to IL-23R. In certain embodiments, the peptide of formula IIIb comprises Glu, (D) Glu, Arg, (D) Arg, Phe, (D) Phe, 2-Nal, Thr, Leu, or (D) Gln bound to Abu. In certain embodiments, it is (D) Arg or (D) Phe.
In one embodiment, Xaa5 and Xaa8 are Gln. In one embodiment, Xaa6 is Thr. In certain embodiments, the peptide is cyclized via Abu of Xaa4 and Cys of Xaa 9.
In particular embodiments of the peptide inhibitor of formula III, the peptide inhibitor has a structure shown in any of table 5 a-5C or comprises an amino acid sequence shown in table 5 a-5C.
In certain aspects, the present invention provides a peptide inhibitor of the interleukin-23 receptor, or a pharmaceutically acceptable salt or solvate thereof, wherein the peptide inhibitor comprises an amino acid sequence of formula (Vf):
X1-X2-X3-Abu-X5-X6-X7-X8-Cys-X10-X11-X12-X13-X14-X15-X16-X17-X18-X19-X20(Vf)(SEQ ID NO:1412),
wherein:
x1 is absent;
x2 is absent or X2 is D-Asp, E, R, D-Arg, F, D-Phe, 2-Nal, T, L, D-Gln or D-Asn;
x3 is D-Arg;
x5 is N, Q, Cit, Lys, or a Lys conjugate (e.g., Lys (IVA), Lys (biotin), Lys (octyl), Lys (Palm), Lys (PEG), Lys (PEG8), Lys (PEG11-Palm), Lys (Ac);
x6 is T, S or V;
x7 is W, 1-Nal or 2-Nal;
x8 is Q, Cit, N, Aib or Lys (Ac);
x10 is Phe [4- (2-aminoethoxy)]Phe [4- (2-acetylaminoethoxy)]Or Phe (4-CONH)2)。
X11 is 2-Nal;
x12 is 4-amino-4-carboxy-tetrahydropyran, Aib, α melleu, α mellys or α mellys conjugate (e.g., α mellys (ac)), α mellys (PEG4-Palm), α mellys (PEG 4-lauryl), α mellys (PEG4 isoglupalm), α mellys (PEG4 isoglulauryl), α mellys (iva), α mellys (biotin) or α mellys (octyl));
X13 is Q, E, Cit or a Lys conjugate (e.g., Lys (Ac), Lys (PEG 4-isoGlu-Palm), Lys (PEG 4-octyl), Lys (PEG4-Palm), Lys (biotin), Lys (octyl), Lys (Palm), Pys (PEG8) or Lys (PEG 11-Palm));
x14 is a N, Cit, Q, L, G, S, Aib, F, 2-Nap, N-Me-Ala, R, W, nLeu, Tic, or Lys conjugate (e.g., Lys (Ac));
x15 is N, Cit, Q, β Ala, Lys (Ac), or Aib; and
x16, X17, X18, X19 and X20 are absent.
In a specific embodiment, X2 is D-Asp, E, R, D-Arg, F, D-Phe, 2-Nal, T, L, D-Gln, or D-Asn.
In certain aspects, the present invention provides a peptide inhibitor of the interleukin-23 receptor, or a pharmaceutically acceptable salt or solvate thereof, wherein the peptide inhibitor comprises an amino acid sequence of formula (Vh):
X1-X2-X3-Abu-X5-X6-X7-X8-Cys-X10-X11-X12-X13-X14-X15-X16-X17-X18-X19-X20(Vh)(SEQ ID NO:1413),
wherein:
x1 is any amino acid or absent;
x2 is any amino acid or absent;
x3 is any D-amino acid or is absent;
x4 is Cys, hCys, Pen, hPen, Abu, Ser, hSer or a chemical moiety capable of forming a bond with X9;
x5 is Ala, α -MeOrn, α -MeSer, Cit, Dap, Dab, Dap (Ac), Gly, Lys, Asn, N-MeGln, N-MeArg, Orn, Gln, Arg, Ser, Glu, or Thr;
x6 is Thr, Ser, Asp, Ile or any amino acid;
X7 is Trp, 6-chloro-Trp, 1-Nap or 2-Nap;
x8 is Glu, Gln, Asn, Lys (Ac), Cit, Cav, Lys (N-e- (N-a-palmitoyl-L-y-glutamyl)) or Lys (N-e-palmitoyl);
x9 is Cys, hCys, Pen, hPen, Abu, or any amino acid or chemical moiety capable of forming a bond with X4;
x10 is 2-Nal, a Phe analog, a Tyr or Tyr analog;
x11 is 1-Nal, 2-Nal, Phe (3, 4-dimethoxy), 5-hydroxy Trp, Phe (3,4-Cl2), Trp or Tyr (3-tBu).
X12 is Aib, 4-amino-4-carboxy-tetrahydropyran, any alpha-methyl amino acid, alpha-ethyl-amino acid, Achc, Acvc, Acbc Acpc, 4-amino-4-carboxy-piperidine, 3-Pal, Agp, alpha-diethylGly, alpha-MeLys (Ac), alpha-MeLeu, alpha-MeOrn, alpha-MeSer, alpha-MeVal, Cav, Cha, Cit, Cpa, D-Asn, Glu, His, hLeu, hArg, Leu, Orn, piperidine, Arg, Ser, Thr, or THP;
x13 is Lys (Ac), Gln, Cit, Glu, or any amino acid;
x14 is Asn, Gln, Lys (Ac), Cit, Cav, Lys (N- ε - (N- α -palmitoyl-L- γ -glutamyl)), Lys (N- ε -palmitoyl), Asp, or any amino acid;
x15 is β -Ala, Asn, Gly, Gln, Ala, Ser, Aib, Asp, or Cit;
X16 is any amino acid or absent;
x17 is any amino acid or absent;
x18 is any amino acid or absent;
x19 is any amino acid or absent; and
x20 is any amino acid or absent.
In certain embodiments of any of the peptide inhibitors described herein, including but not limited to those of formulae (If) and (Ih), the peptide inhibitor is cyclized via a bond (e.g., a thioether bond) between X4 and X9. In certain embodiments, the peptide inhibitor inhibits the binding of interleukin-23 (IL-23) to an IL-23 receptor.
In certain embodiments, X1, X2, and X3 are absent. In certain embodiments, X1 and X2 are absent. In certain embodiments, X1 is a D-amino acid or is absent. In certain embodiments, X2 is a D-amino acid or is absent.
In certain embodiments, X5 is Ala, α -MeOrn, α -MeSer, Cit, Dap, Dab, Dap (Ac), Gly, Lys, Asn, N-MeGln, N-MeArg, Orn, Gln, Arg, Ser, or Thr;
in certain embodiments, X5 is N, X6 is T, X7 is W, or X8 is Q. In certain embodiments, X5 is N, X6 is T, X7 is W, and X8 is Q.
In certain embodiments, X5 is Q, X6 is T, X7 is W, or X8 is Q. In certain embodiments, X5 is Q, X6 is T, X7 is W, and X8 is Q.
In certain embodiments, X5 is N, X6 is T, X7 is W, and X8 is Cit.
In certain embodiments, X10 is Phe [4- (2-aminoethoxy) ].
In certain embodiments, X12 is 4-amino-4-carboxy-tetrahydropyran, Aib, α melleu, or α MeLys. In certain embodiments, X12 is 4-amino-4-carboxy-tetrahydropyran.
In certain embodiments, X13 is E or lys (ac). In certain embodiments, X13 is lys (ac).
In certain embodiments, X14 is Asn, Gln, Lys (Ac), Cit, Cav, Lys (N-. epsilon. - (N-. alpha. -palmitoyl-L-. gamma. -glutamyl)), Lys (N-. epsilon. -palmitoyl), or any amino acid;
in certain embodiments, X15 is β -Ala, Asn, Gly, Gln, Ala, Ser, Aib, or Cit.
In certain embodiments, X14 is N.
In certain embodiments, X15 is N.
In certain embodiments, X16 is a D-amino acid or is absent. In certain embodiments, X17 is a D-amino acid or is absent. In certain embodiments, X18 is a D-amino acid or is absent. In certain embodiments, X19 is a D-amino acid or is absent. In certain embodiments, X20 is a D-amino acid or is absent.
In certain embodiments, X2 is absent; x3 is absent; x5 is Q, X6 is T, X7 is W, and X8 is Q; x10 is Phe [4- (2-aminoethoxy) ]; x12 is 4-amino-4-carboxy-tetrahydropyran, Aib, α melleu, or α mellys; x13 is E or Lys (Ac); x14 is N; and X15 is N. In certain embodiments, X12 is 4-amino-4-carboxy-tetrahydropyran and X13 is lys (ac).
In certain embodiments, any amino acid of the peptide inhibitor is attached via a linker moiety (e.g., PEG).
In certain embodiments, the N-terminus of the peptide inhibitor comprises an Ac group.
In certain embodiments, the C-terminus of the peptide inhibitor comprises NH2A group.
In certain embodiments, the invention encompasses a peptide comprising or consisting of an amino acid sequence set forth in any one of table 4 or table 5A-5B, or a peptide inhibitor (or a pharmaceutically acceptable salt thereof) comprising or consisting of a structure set forth in any one of table 4 or table 5A-5B. In particular embodiments, the peptide does not comprise a conjugated moiety, but comprises an Abu residue. In particular embodiments, the peptide or inhibitor comprises a thioether bond between the two Abu and Cys residues or between the two outermost amino acids in parentheses after the term "ring", indicating the presence of a cyclic structure. In a specific embodiment, the inhibitor is an acetate salt. Peptide sequences of exemplary inhibitors are shown in table 4 and tables 5A-5B from N-terminus to C-terminus, with conjugated moieties, and with the N-terminus Ac and/or C-terminus NH indicated2A group. As shown in table 5A, the cyclic structure is represented by "ring", indicating the presence of a thioether bond between Abu bracketed at X4 and Cys at X9.
TABLE 4 exemplary thioether peptide inhibitors
Figure GDA0003395945150001411
Figure GDA0003395945150001421
Table 5a. exemplary thioether peptide inhibitors
Figure GDA0003395945150001431
Ac-Ring- [ [ Abu]-XXWXC]-[Phe(4-OMe)]-[2-Nal]-XXXX-NH2(SEQ ID NO:1414)
Figure GDA0003395945150001432
Figure GDA0003395945150001441
Figure GDA0003395945150001451
TABLE 5B exemplary thioether peptides
Figure GDA0003395945150001461
Exemplary peptide inhibitors containing cyclic amides
In certain embodiments, the present invention includes a peptide inhibitor of the interleukin-23 receptor, or a pharmaceutically acceptable salt or solvate thereof, wherein the peptide inhibitor has the structure of formula IV:
R1-X-R2(IV)
wherein R is1Is a bond, hydrogen, C1-C6 hydrocarbyl, C6-C12 aryl, C6-C12 aryl, C1-C6 hydrocarbyl, C1-C20 hydrocarboxyl, and includes pegylated forms of any of the foregoing either alone or as a spacer;
R2is a bond, OH or NH2(ii) a And
x is an amino acid sequence of 8 to 20 amino acids comprising or consisting of the sequence of formula IVa:
X1-X2-X3-X4-X5-X6-W-X8-X9-X10-X11-X12-X13-X14-X15-X16-X17-X18-X19-X20(IVa)(SEQ ID NO:1428)
wherein
X1 is absent or is any amino acid;
x2 is absent or is any amino acid;
x3 is absent or is any amino acid;
x4 is Dap, Dab, Glu, Asp, (D) -Asp, or (D) -Dab;
x5 is Gln, Ala, Cys, Cit, Asp, Dab, Dap, Glu, Phe, Gly, His, hCys, Lys, Leu, Met, Asn, N-Me-Ala, N-M-Asn, N-Me-Lys, N-Me-Gln, N-Me-Arg, Orn, Pro, Pen, Gln, Arg, Ser, Thr, or Val;
x6 is Thr, Asp, Glu, Phe, Asn, Pro, Arg, Ser or Thr;
X7 is Trp, Glu, Gly, Ile, Asn, Pro, Arg, Thr, or OctGly;
x8 is Gln, Glu, Phe, Lys, Asn, Pro, Arg, Thr, or Trp;
x9 is Dap, Dab, Glu, Asp, (D) -Asp, or (D) -Dab;
x10 is Tyr (OMe) Phe (4-OMe), 1-Nal, 2-Nal, Aic, a-MePhe, Bip, (D) Cys, Cha, DMT, (D) Tyr), Glu, Phe, His, hPhe (3, 4-dimethoxy), hTyr, N-Me-Tyr, Trp, Phe (4-CONH)2) Phe (4-phenoxy), Thr, Tic, Tyr (3-tBu), Phe (4-CN), Phe (4-Br), Phe (4-NH)2)、Phe(4-F)、Phe(3,5-F2)、Phe(5-F)、Phe(3,4-Cl2)、Phe(4-CF3) Bip, Cha, 4-pyridylalanine, beta hTyr, OctGly, Phe (4-N)3) Phe (4-Br) or Phe [4- (2-aminoethoxy)];
X11 is 2-Nal, 1-Nal, 2, 4-dimethyl Phe, Bip, Phe (3, 4-Cl)2)、Phe(3,5-F2)、Phe(4-CONH2) Phe (4-F), 4-phenylcyclohexylalanine, Phe (4-CF)3) α -MePhe, β hPhe, β hTyr, β hUrp, BIP, Nva (5-phenyl), Phe, His, hPhe, Tic, Tqa, Trp, Tyr, Phe (4-OMe), Phe (4-Me), Trp (2,5, 7-tri-tert-butyl), Phe (4-O allyl), Tyr (3-tBu), Phe (4-guanidino), Tyr (Bzl), or OctGly;
x12 is α -MeLys, α -MeOrn, α -MeLeu, Aib, (D) Ala, (D) Asn, (D) Leu, (D) Asp, (D) Phe, (D) Thr, 3-Pal, Aib, β -Ala, β -Glu, β hAla, β hLeu, β hVal, β -spiro-pip, Cha, Chg, Asp, Dab, Dap, α -diethylGly, Glu, Phe, hLeu, hArg, hLeu, Ile, Lys, Leu, Asn, N-MeLeu, N-MeArg, og, Orn, Pro, Gln, Arg, Ser, Thr, Tle, 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Acvc, α -diethylGly, α -MeLys (Ac), α -MeMe-Leu, α -MeVal;
X13 is Lys (Ac), (D) Asn, (D) Leu, (D) Thr, (D) Phe, Ala, Aib, alpha-MeLeu, Aib, beta-Ala, beta-Glu, beta hAla, beta hLeu, beta hVal, beta-spiro-pip, Cha, Chg, Asp, Dab, Dap, alpha-diethylGly, Glu, Phe, hLeu, Lys (Ac), Leu, Asn, Ogl, Pro, Gln, Arg, Ser, beta-spiro-pip, Thr, Tba, Tlc, Val, or Tyr;
x14 is Asn, Glu, Phe, Gly, His, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Tic, or Tyr;
x15 is β -Ala, Asn, Gly, (D) Ala, (D) Asn, (D) Asp, (D) Leu, (D) Phe, (D) Thr, Ala, AEA, Asp, Glu, Phe, Gly, Lys, Leu, Pro, Gln, Arg, or Ser;
x16 is absent and is Gly, Ala, Asp, Ser, Pro, Asn or Thr;
x17 is absent, Glu, Ser, Gly or Gln;
x18 is absent or is any amino acid;
x19 is absent or is any amino acid; and
x20 is absent or is any amino acid.
In certain embodiments of Iva: x12 is α -MeLys, α -MeOrn, α -MeLeu, Aib, (D) Ala, (D) Asn, (D) Leu, (D) Asp, (D) Phe, (D) Thr, 3-Pal, Aib, β -Ala, β -Glu, β hAla, β hLeu, β hVal, β -spiro-pip, Cha, Chg, Asp, Dab, Dap, α -diethylGly, Glu, Phe, hLeu, hArg, hLeu, Ile, Lys, Leu, Asn, N-MeLeu, N-MeArg, Ogl, Orn, Pro, Gln, Arg, Ser, Thr, or Tle; x13 is Lys (Ac), (D) Asn, (D) Leu, (D) Thr, (D) Phe, Ala, Aib, alpha-MeLeu, Aib, beta-Ala, beta-Glu, beta hAla, beta hLeu, beta hVal, beta-spiro-pip, Cha, Chg, Asp, Dab, Dap, alpha-diethylGly, Glu, Phe, hLeu, Asn, Ogl, Pro, Gln, Arg, Ser, beta-spiro-pip, Thr, Tba, Tlc, Val, or Tyr; x14 is Asn, Glu, Phe, Gly, His, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Tic, or Tyr; and X15 is Gly, (D) Ala, (D) Asn, (D) Asp, (D) Leu, (D) Phe, (D) Thr, Ala, AEA, Asp, Glu, Phe, Gly, Lys, Leu, Pro, Gln, Arg, or Ser.
In particular embodiments of the peptide inhibitor of formula (IV): x5 is Cys, Cit, Asp, Dab, Dap, Gly, His, hCys, Lys, Met, Asn, N-Me-Ala, N-Me-Asn, N-Me-Lys, N-Me-Gln, N-Me-Arg, Orn, Pro, Pen, Gln, Val; x6 is Glu, Arg, Ser; x7 is Trp, Glu, Gly, Ile, Asn, Pro, Arg, Thr, or OctGly; x8 is Phe, Asn, Pro, Arg, Thr, Trp; x10 is Phe (4-OMe), 1-Nal, 2-Nal, Aic, alpha-MePhe, Bip, (D) Cys, Cha, DMT, (D) Tyr, Glu, His, hPhe (3, 4-dimethoxy), hTyr, N-Me-Tyr, Trp, Phe (4-CONH)2) Phe- (4-phenoxy), Thr, Tic, Tyr (3-tBu), Phe (4-CN), Phe (4-Br), Phe (4-NH)2)、Phe(4-F)、Phe(3,5-F2)、PheCH2CO2H、Phe(5-F)、Phe(3,4-Cl2)、Phe(4-CF3) Bip, Cha, 4-pyridylalanine, beta hTyr, OctgGly, Tyr (4-N)3) Phe (4-Br), Phe [4- (2-aminoethoxy)](ii) a X11 is 2-Nal, 1-Nal, 2, 4-dimethyl Phe, Bip, Phe (3, 4-Cl)2)、Phe(3,5-F2)、Phe(4-CONH2) Phe (4-F), 4-phenylcyclohexyl, Phe (4-CF)3) α -MePhe, Nal, β hPhe, β 0hTyr, β 1hTrp, BIP, Nva (5-phenyl), Phe, His, hPhe, Tic, Tqa, Tyr, Phe (4-OMe), Phe (4-Me), Tyr (2,5, 7-tri-t-butyl), Phe (4-O allyl), Phe (3-tBu), Phe (4-guanidino), Tyr (Bzl), OctGly; x12 is β 3-Me-Lys, D-Ala, (D) Asn, (D) Asp, (D) Leu, (D) Phe, (D) Tyr, Aib, β 4-MeLeu, β 5-MeOrn, Aib, β 2-Ala, β 8hAla, β 9hArg, β hLeu, β 0hVal, β 1-spiro-pip, Glu, hArg, Ile, Lys, N-MeLeu, N-MeArg, og, Orn, Pro, Gln, Ser, Thr, Tle, 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Acvc, β 6-diethylGly, β 7-MeLys (Ac), β 3-MeSer, α -MeVal; x13 is Lys, Lys (Ac), (D) Asn, (D) Leu, (D) Phe, (D) Thr, Ala, α -MeLeu, Aib, β 2-Ala, β 4-Glu, β hLeu, β hVal, β -spiro-pip, Cha, Chg, Asp, Dab, Dap, α -diethylGly, hLeu, Asn, Ogl, Pro, Gln, Ser, Thr, Tba, Tle; x14 is Glu, Gly, His, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Tic; x15 is (D) Ala, (D) Asn, (D) Asp, (D) Leu, (D) Phe, (D) Thr, Aea, Asp, Glu, Phe, Gly, Lys, Leu, Pro, Asn, Arg, or β -Ala; x16 is Gly, Ser, Pro, Asn, Thr; or X17 is Glu, Ser, Gly, Gln.
In particular embodiments of the peptide inhibitor of formula (IV): x5 is Cys, Cit, Asp, Dab, Dap, Gly, His, hCys, Lys, Met, Asn, N-Me-Ala, N-Me-Asn, N-Me-Lys, N-Me-Gln, N-Me-Arg, Orn, Pro, Pen, Gln, Val; x6 is Glu, Arg, Ser; x7 is Trp, Glu, Gly, Ile, Asn, Pro, Arg, Thr, or OctGly; x8 is Phe, Asn, Pro, Arg, Thr, Trp; x10 is Phe (4-OMe), 1-Nal, 2-Nal, Aic, α -MePhe, Bip, (D) Cys, Cha, DMT, (D) Tyr, Glu, His, hPhe (3, 4-dimethoxy), hTyr, N-Me-Tyr, Trp, Phe (4-CONH)2) Phe- (4-phenoxy), Thr, Tic, Tyr (3-tBu), Phe (4-CN), Phe (4-Br), Phe (4-NH)2)、Phe(4-F)、Phe(3,5-F2)、PheCH2CO2H、Phe(5-F)、Phe(3,4-Cl2)、Phe(4-CF3) Bip, Cha, 4-pyridylalanine, beta hTyr, OctgGly, Tyr (4-N)3) Phe (4-Br), Phe [4- (2-aminoethoxy)](ii) a X11 is 2-Nal, 1-Nal, 2, 4-dimethyl Phe, Bip, Phe (3, 4-Cl)2)、Phe(3,5-F2)、Phe(4-CONH2) Phe (4-F), 4-phenylcyclohexyl, Phe (4-CF)3) α -MePhe, Nal, β hPhe, β 0hTyr, β 1hTRp, BIP, Nva (5-phenyl), Phe, His, hPhe, Tic, Tqa, Tyr, Phe (4-OMe), Phe (4-Me), Tyr (2,5, 7-tri-t-butyl), Phe (4-O allyl), Phe (3-tBu), Phe (4-guanidino), Tyr (Bzl), OctGly; x12 is β 3-Me-Lys, D-Ala, (D) Asn, (D) Asp, (D) Leu, (D) Phe, (D) Tyr, Aib, β 9-MeLeu, α -MeOrn, Aib, β 2-Ala, β 4hAla, β 5hArg, β 6hLeu, β 7hVal, β 8-spiro-pip, Glu, hArg, Ile, Lys, N-MeLeu, N-MeArg, Ogl, Orn, Pro, Gln, Ser, Thr, Tle; x13 is Lys (Ac), (D) Asn, (D) Leu, (D) Phe, (D) Thr, Ala, α -MeLeu, Aib, β -Ala, β -Glu, β hLeu, β hVal, β -spiro-pip, Cha, Chg, Asp, Dab, Dap, α -diethylGly, hLeu, Asn, og, Pro, Gln, Ser, Thr, Tba, Tle; x14 is Glu, Gly, His, Lys, Leu, Met, Asn, Pro, Gln, Arg, Ser, Thr, Tic; x15 represents (D) Ala, (D) Asn, (D) Asp, (D) Leu, (D) Phe, (D) Thr, Aea, Asp, Glu, Phe, Gly, Lys, Leu, Pro, Arg; x16 is Gly, Ser, Pro, Asn, Thr; or X17 is Glu, Ser, Gly、Gln。
In certain embodiments, the peptide inhibitor is cyclized. In a specific embodiment, the peptide is cyclized via an intramolecular bond between X4 and X9. In a specific embodiment, the intramolecular bond is an amide bond.
In certain embodiments, the peptide inhibitor is linear or not cyclized.
In particular embodiments of the peptide inhibitor of formula IV, one or more, two or more, or all three of X1, X2, and X3 are absent.
In certain embodiments, X3 is Glu, (D) Glu, Arg, (D) Arg, Phe, (D) Phe, 2-Nal, Thr, Leu, or (D) Gln. In certain embodiments, X3 is (D) Arg or (D) Phe.
In particular embodiments of the peptide inhibitor of formula IV, one or more, two or more, or all three of X17, X19, and X20 are absent.
In particular embodiments of the peptide inhibitor of formula IV, X4 is Dap, Dab, or (D) Dab, and X9 is Glu, (D) Asp, or Asp. In particular embodiments of the peptide inhibitor of formula I, X4 is Glu, (D) Asp or Asp, and X9 is Dab, Dap or (D) Dab.
In a specific embodiment of the peptide inhibitor of formula IV, X18 is (D) -Lys. In certain embodiments, X17 is absent, and X18 is (D) -Lys.
In particular embodiments of the peptide inhibitor of formula IV, the peptide inhibitor comprises one or more, two or more, three or more, or all four of the following characteristics: x5 is Gln; x6 is Thr; x7 is Trp; and X8 is Gln.
In particular embodiments of the peptide inhibitor of formula IV, the peptide inhibitor comprises one or more, two or more, three or more, four or more, five or more, six or more, or seven of the following characteristics: x10 is Tyr, Phe [4- (2-aminoethoxy)]、Phe(4-CONH2) Or Phe (4-OMe); x11 is 2-Nal or Trp; x12 is 4-amino-4-carboxy-tetrahydropyran, Achc, Acpc, Acbc, Acvc, Aib, α -diethylGly, α -MeLys (Ac), α -Me-Leu, α -MeOrnalpha-MeSer, alpha-MeVal or Arg; x13 is Glu or Lys (Ac); x14 is Asn; x15 is Gly, Asn or β -Ala; and X16 is AEA. In particular embodiments of the peptide inhibitor of formula IV, the peptide inhibitor comprises one or more, two or more, three or more, four or more, five or more, six or more, or seven of the following characteristics: x10 is Tyr; x11 is Trp; x12 is Arg; x13 is Glu; x14 is Asn; x15 is Gly; and X16 is AEA.
In particular embodiments of the peptide inhibitor of formula IV, the peptide inhibitor comprises one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, or all of the following features: x5 is Gln; x6 is Thr; x7 is Trp; x8 is Gln; x10 is Tyr; x11 is Trp; x12 is Arg; x13 is Glu or Lys (Ac); x14 is Asn; x15 is Gly; and X16 is AEA. In particular embodiments of the peptide inhibitor of formula IV, the peptide inhibitor comprises one or more, two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, or all of the following features: x5 is Gln; x6 is Thr; x7 is Trp; x8 is Gln; x10 is Tyr; x11 is Trp; x12 is Arg; x13 is Glu; x14 is Asn; x15 is Gly; and X16 is AEA.
In certain embodiments of the peptide inhibitor of formula IV, the peptide is cyclized via X4 and X9; x5, X6, X7 and X8 are Gln, Thr, Trp and Gln, respectively; and X10, X11, X12, X13, X14, X15 and X16 are Tyr, Trp, Arg, Glu, Asn, Gly and AEA, respectively.
In certain embodiments, the invention encompasses an optionally cyclized 8 to 20 amino acid peptide comprising or consisting of a core sequence comprising:
xaa4-Xaa5-Xaa6-Trp-Xaa8-Xaa9- [ Phe (4-OMe) ] - [2-Nal ] (formula IVb) (SEQ ID NO:1429)
Wherein Xaa4 and Xaa9 are each independently selected from Dap, Dab, Glu, Asp, (D) -Asp, and (D) -Dab, wherein Xaa4 and Xaa9 are capable of forming an intramolecular bond, e.g., a cyclic amide; and Xaa5, Xaa6, and Xaa8 are any amino acid residue, wherein the peptide inhibits binding of IL-23 to IL-23R. In a specific embodiment, the peptide inhibitor is a peptide inhibitor of formula IV. In particular embodiments, the peptide inhibits the binding of IL-23 to IL-23R.
In certain embodiments of the peptide inhibitor of formula IV, the peptide inhibitor has a structure shown in table E7 or comprises an amino acid sequence shown in table E7.
Certain exemplary peptide inhibitors of the invention are also shown in any of formulas (Va), (Vb), (Vc), (Vd), (Ve), (Vf), (Vg), and (Vh) and tables 2-5, which provide the amino acid sequences of the selected peptide inhibitors. These peptide inhibitors are acetates.
Optional features of peptide inhibitors
Any peptide inhibitor of the present invention can, for example, be further defined as described below. It will be appreciated that each of the further defining features described herein may be applied to any peptide inhibitor in which the specified amino acid at a particular position allows for the presence of the further defining feature.
In certain embodiments of any of the peptide inhibitors described herein, the peptide inhibitor is cyclized.
In certain embodiments of any of the peptide inhibitors described herein, the peptide inhibitor or monomeric subunit thereof is linear or not cyclized. In certain embodiments, wherein the peptide is linear or not cyclized, X4 and X9 can be any amino acid.
In certain embodiments, the peptide inhibitor is cyclized, e.g., by X4 and X9.
In various embodiments, R1Is a bond, hydrogen, a C1-C6 hydrocarbyl group, a C6-C12 aryl group, a C6-C12 aryl group, a C1-C6 hydrocarbyl group, or a C1-C20 hydrocarboxyl group, and includes a PEGylated form of any of the foregoing (e.g., acetyl) alone or as a spacer. It will be appreciated that in addition to the typical amine group located at the amino terminus of the peptide, R is1May be substituted or present. It is also understood that R1May not be present. In thatIn certain embodiments, the peptide inhibitor comprises an N-terminus selected from the group consisting of: hydrogen, C1-C6 hydrocarbyl, C6-C12 aryl, C6-C12 aryl, C1-C6 hydrocarbyl, or C1-C20 hydrocarboxyl, and including pegylated forms (e.g., acetyl) of any of the foregoing, alone or as a spacer. In particular embodiments of any of the peptide inhibitors described herein, R 1Or the N-terminal moiety is hydrogen. In certain embodiments, R1Is a bond, e.g., a covalent bond.
In certain embodiments of any peptide inhibitor having any one of the formulae shown herein, R is1Or the N-terminal moiety is selected from methyl, acetyl, formyl, benzoyl, trifluoroacetyl, isovaleryl, isobutyryl, octyl (octanyl), and conjugated amides of dodecanoic acid, hexadecanoic acid, and γ -Glu-hexadecanoic acid. In one embodiment, R1Or the N-terminal moiety pGlu. In certain embodiments, R1Is hydrogen. In a particular embodiment, R1Is acetyl, whereby the peptide inhibitor is acylated at its N-terminus, e.g., to cap or protect the N-terminal amino acid residue, e.g., the N-terminal Pen or Abu residue.
In certain embodiments of any of the peptide inhibitors described herein, R is1Or the N-terminal moiety is an acid. In certain embodiments, R1Or the N-terminal moiety is an acid selected from: acetic acid, formic acid, benzoic acid, trifluoroacetic acid, isovaleric acid, isobutyric acid, octanoic acid, dodecanoic acid, hexadecanoic acid, 4-biphenylacetic acid, 4-fluorophenylacetic acid, gallic acid, pyroglutamic acid, cyclopentylpropionic acid, glycolic acid, oxalic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, palmitic acid, benzoic acid, 3- (4-hydroxybenzoyl) benzoic acid, cinnamic acid, mandelic acid, 4-methylbicyclo (2.2.2) -oct-2-ene-1-carboxylic acid, glucoheptonic acid, 3-phenylpropionic acid, trimethylacetic acid, tert-butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynaphthoic acid, salicylic acid, stearic acid, muconic acid, hydrocarbyl sulfonic acid, and aryl sulfonic acid.
In a particular embodiment, R1Or the N-terminal moiety is selected from the group consisting of methanesulfonic acid, ethanesulfonic acid, 1, 2-ethane-disulfonic acid andhydrocarbyl sulfonic acids of 2-hydroxyethanesulfonic acid.
In a particular embodiment, R1Or an aryl sulfonic acid having an N-terminal moiety selected from benzenesulfonic acid, 4-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid, 4-toluenesulfonic acid and camphorsulfonic acid.
In some embodiments, wherein the peptide of the invention comprises conjugation to an acidic compound such as, for example, isovaleric acid, isobutyric acid, valeric acid, and the like, the presence of such conjugation is referred to in the acid form. Thus, for example, but not by way of limitation, in some embodiments, the present application refers to such conjugation as isovaleric acid- [ Pen ] -QTWQ [ Pen ] - [ Phe (4-OMe) ] - [2-Nal ] - [ α -MeLys ] - [ Lys (Ac) ] -NG-
NH2(SEQ ID NO:262) instead of indicating the conjugation of isovaleric acid to the peptide by the mentioned isovaleryl group, e.g., isovaleryl- [ Pen]-QTWQ[Pen]-[Phe(4-OMe)]-[2-Nal]-[α-MeLys]-[Lys(Ac)]-NG-
NH2(SEQ ID NO: 262). Reference to conjugation in its acid form is intended to encompass the form present in the peptide inhibitor.
In certain embodiments, the peptide inhibitor comprises a bond selected from OH, NH, or OH2C-terminal of (e.g., R)2Or the C-terminal portion). In certain embodiments, R2Is a bond. In various embodiments of any peptide inhibitor having any of the formulae shown herein, R 2Or C is OH or NH2. It will be appreciated that R is in addition to the carboxyl group normally located at the carboxyl terminus of the peptide2Or the C-terminal moiety may be substituted or present. It is also understood that R2May not be present.
In particular embodiments of any peptide inhibitor having any one of the formulae shown herein, X comprises or consists of: 7 to 35 amino acid residues, 8 to 35 amino acid residues, 9 to 35 amino acid residues, 10 to 35 amino acid residues, 7 to 25 amino acid residues, 8 to 25 amino acid residues, 9 to 25 amino acid residues, 10 to 25 amino acid residues, 7 to 20 amino acid residues, 8 to 20 amino acid residues, 9 to 20 amino acid residues, 7 to 18 amino acid residues, 8 to 18 amino acid residues, 9 to 18 amino acid residues, or 10 to 18 amino acid residues.
In certain embodiments of any formula shown herein, neither or both xs comprises or consists of the amino acid sequence shown in U.S. patent application publication No. US 2013/0029907. In certain embodiments of any formula shown herein, neither or both xs comprises or consists of the amino acid sequence shown in U.S. patent application publication No. US 2013/0172272.
In certain embodiments of any of the peptide inhibitors described herein, the peptide inhibitor, or each monomeric subunit thereof, comprises or consists of: at least 3, at least 4, at least 5, at least 6, or at least 7 amino acid residues from the carboxy terminus of the X9 amino acid residue. In particular embodiments of any of the peptide inhibitors described herein, the peptide inhibitor comprises 3 to 11, 3 to 10, 3 to 9, 3 to 8, 3 to 7, 3 to 6, 3 to 5, 3 to 4, 3, 4, 5, 6, 7, 8, 9, 10, or 11 amino acid residues that are carboxy-terminal to the X9 amino acid residue.
In certain embodiments of any of the peptide inhibitors described herein, the peptide inhibitor, or each monomeric subunit thereof, comprises or consists of 4 amino acid residues between X4 and X9. In one embodiment, both X4 and X9 are cysteine.
In certain embodiments, the peptide inhibitor of any of the formulae described herein comprises an amino acid residue or moiety represented as X4-X15. In particular embodiments, the peptide inhibitor does not include X1-X3 or X16-X20. In certain embodiments, the peptide inhibitor comprises an N-terminal extension of 1 to 3 amino acid residues corresponding to any one of X1-X3. In a particular embodiment, when any one or more of X1, X2, and X3 is present, it is a D-amino acid. In certain embodiments, the peptide inhibitor comprises a C-terminal extension of 1 to 5 amino acid residues corresponding to any one of X16-X20. In particular embodiments, when any one or more of X16, X17, X18, X19, and X20 is present, it is a D-amino acid. Exemplary amino acid residues that may be present in the N-terminal and/or C-terminal extensions are shown in tables 3 and 5. These tables each show a first peptide inhibitor and derivatives thereof comprising an N-terminal extension, a C-terminal extension and/or a conjugate moiety. The present invention includes derivatives of any of the peptide inhibitors described herein comprising one or more such N-terminal extensions, C-terminal extensions and/or conjugate moieties. In certain embodiments, any of the amino acid residues shown in the extended positions in tables 3 and 5 may be present in any combination in the peptide inhibitors of the invention. In particular embodiments, the N-terminal and/or C-terminal extension is associated with an increased half-life, e.g., upon administration to a subject.
In certain embodiments of any of the peptide inhibitors described herein, the peptide inhibitor, or each monomeric subunit thereof, comprises the amino acid sequence motif W-X-X-Y-W (SEQ ID NO:1430) at, for example, X7-X11. In certain embodiments, the peptide inhibitor or each monomeric subunit thereof comprises the amino acid sequence motif C-X-X-W-X-C-Y-W (SEQ ID NO:1431), e.g., at positions X4-X11. In certain embodiments, the peptide inhibitor or each monomeric subunit thereof comprises the amino acid sequence motif Pen-X-X-W-X-Pen-Y-W (SEQ ID NO:1432), e.g., at position X4-X11. In certain embodiments of any of the peptide inhibitors described herein, the peptide inhibitor, or both monomeric subunits thereof, does not comprise the amino acid sequence motif W-X-X-Y-W (SEQ ID NO:1430) at, for example, X7-X11, wherein X is any amino acid.
In certain embodiments of any of the formulae or peptide inhibitors described herein, the peptide inhibitor comprises one or more amino acid residues N-terminal to X4. In particular embodiments, X3 is present. In certain embodiments, X3 is Glu, (D) Glu, Arg, (D) Arg, Phe, (D) Phe, 2-Nal, Thr, Leu, or (D) Gln. In certain embodiments, X3 is (D) Arg or (D) Phe.
In a specific embodiment of any of the formulae or peptide inhibitors described herein, the peptide inhibitor comprises the amino acid at X2. In particular embodiments, X2 is Glu, (D) Asp, Arg, (D) Arg, Phe, (D) Phe, 2-Nal, Thr, Leu, (D) Gln, or (D) Asn. In certain embodiments, X2 and X3 are present. In particular embodiments, X2 is Glu, (D) Asp, Arg, (D) Arg, Phe, (D) Phe, 2-Nal, Thr, Leu, (D) Gln, or (D) As, and X3 is (D) Arg.
In certain embodiments, the peptide inhibitor of the invention, or one or both of its monomeric subunits, optionally comprises at its C-terminus one of the following amino acid sequences:
ENG;
ENN;
[ 4-amino-4-carboxy-tetrahydropyran ] -ENN (SEQ ID NO: 1433);
[Lys(Ac)]-NN;
[α-MeLys]-ENG(SEQ ID NO:1434);
[α-MeLys]-[Lys(Ac)]-NN(SEQ ID NO:1435);
[α-MeLeu]-[Lys(Ac)]-NN(SEQ ID NO:1436);
[α-MeLeu]-ENG(SEQ ID NO:1437);
[α-MeOrn]-[Lys(Ac)]-NG(SEQ ID NO:1438);
[α-MeLeu]-ENG(SEQ ID NO:1439);
Aib-[Lys(Ac)]-NG(SEQ ID NO:1440);
Aib-[Lys(Ac)]-NN(SEQ ID NO:1441);
NG-[AEA]-[(D)-Lys](SEQ ID NO:1442);
[Dapa]-NG-[AEA]-[(D)-Lys](SEQ ID NO:1443);
[Orn]-NG-[AEA]-[(D)-Lys](SEQ ID NO:1444);
[α-MeLys]-ENN(SEQ ID NO:1445);
[ 4-amino-4-carboxy-tetrahydropyran ] - [ Lys (Ac) ] -NN (SEQ ID NO: 1446);
[ Achc ] - [ Lys (Ac) ] -NN (SEQ ID NO: 1447); or
[Acpc]-[Lys(Ac)]-NN(SEQ ID NO:1448)。
In a particular embodiment, one of these amino acid sequences constitutes the terminal C-terminal amino acid of the peptide. In particular embodiments, these amino acid sequences correspond to X13-X15 or X12-X15 or X14-X16 or X13-X17.
In certain embodiments, the peptide inhibitor of the invention, or one or both of its monomeric subunits, optionally comprises at its C-terminus one of the following amino acid sequences:
WQCY-[2-Nal]-[α-MeLys](SEQ ID NO:1449);
WQC-[Phe(4-OMe)]-[2-Nal]-[α-MeLys](SEQ ID NO:1450);
WQC-[Phe(4-OMe)]-[2-Nal]-[Aib](SEQ ID NO:1451);
WQ-[Pen]-[Phe(4-OMe)]-[2-Nal]-[α-MeLys](SEQ ID NO:1452);
W-Xaa8-C-Phe [4- (2-aminoethoxy) ] - [2-Nal ] (SEQ ID NO: 1453);
W-Xaa8-C-Phe [4- (2-aminoethoxy) ] - [1-Nal ] (SEQ ID NO: 1454);
W-Xaa8-C-Phe [4- (2-aminoethoxy) ] (SEQ ID NO: 1455); or
W-Xaa8-C-[Phe(4-OCH3)](SEQ ID NO: 1456). In a particular embodiment, one of these amino acid sequences constitutes the terminal C-terminal amino acid of the peptide. In particular embodiments, these amino acid sequences correspond to X7 to X12 or X7 to X11 or X7 to X10.
In certain embodiments of any of the peptide inhibitors described herein (including the monomeric subunits of the peptide monomer inhibitors and the peptide dimer inhibitors), the peptide monomer inhibitors or the monomeric subunits are cyclized via a peptide bond between their N-terminal amino acid residues and their C-terminal amino acid residues. In particular embodiments, the peptide inhibitor (or monomeric subunit thereof) comprises an intramolecular bond between X4 and X9 and a peptide bond between its N-terminal amino acid residue and its C-terminal amino acid residue. In certain embodiments, the intramolecular linkage is any of those described herein, e.g., a disulfide linkage or a thioether linkage.
In certain embodiments, the invention encompasses peptide inhibitors comprising a core consensus sequence (shown in an N-terminal to C-terminal direction) selected from one of:
X1-X2-X3-Pen-X5-X6-W-X8-Pen-X10-X11-X12-X13-X14-X15(SEQ ID NO:1457);
Pen-X5-X6-W-Q-Pen(SEQ ID NO:1458);
Pen-X5-X6-W-X8-Pen(SEQ ID NO:1459);
Pen-X5-X6-W-X8-Pen- [ Phe (4-CONH2) ] (SEQ ID NO: 1460); and
Pen-X5-X6-W-X8-Pen- [ Phe [4- (2-aminoethoxy) ] ] (SEQ ID NO:1461),
wherein the Pen residues are linked by an intramolecular bond (e.g., a disulfide bond). X1, X2, X3, X5, X6, X8, X10, X11, X12, X13, X14 and X15 can be any amino acid. In some embodiments, X5 is Arg, Asn, Gln, Dap, Orn; x6 is Thr or Ser; and X8 is Gln, Val, Phe, Glu, Lys. In particular embodiments, X1, X2, X3, X5, X6, X8, X10, X11, X12, X13, X14, and X15 are defined as described in any of the formulae and peptide inhibitors described herein.
In certain embodiments, the invention encompasses peptide inhibitors comprising a core consensus sequence (shown in an N-terminal to C-terminal direction) selected from one of:
X1-X2-X3-Abu-X5-X6-W-X8-C-X9-X10-X11-X12-X13-X14-X15(SEQ ID NO:1462);
Abu-X5-X6-W-Q-C(SEQ ID NO:1463);
Abu-X5-X6-W-X8-C(SEQ ID NO:1464);
Abu-X5-X6-W-X8-C- [ Phe (4-CONH2) ] (SEQ ID NO: 1465); and
Abu-X5-X6-W-X8-C- [ Phe [4- (2-aminoethoxy) ] ] (SEQ ID NO:1466),
wherein Abu and C are linked by an intramolecular thioether linkage. X1, X2, X3, X5, X6, X8, X10, X11, X12, X13, X14 and X15 can be any amino acid. In some embodiments, X5 is Arg, Asn, Gln, Dap, Orn; x6 is Thr or Ser; and X8 is Gln, Val, Phe, Glu, Lys. In particular embodiments, X1, X2, X3, X5, X6, X8, X10, X11, X12, X13, X14, and X15 are defined as described in any of the formulae and peptide inhibitors described herein.
In certain embodiments, any of the peptide inhibitors described herein may be further cyclized via a peptide bond between its N-terminal amino acid residue and its C-terminal amino acid residue. In particular embodiments, the peptide inhibitor comprises a peptide bond between X3 or X4 and any one of X9, X10, X11, X12, X13, X14, X15, X16, X17, X18, X19, or X20. In a particular embodiment, the peptide inhibitors of the invention comprise a peptide bond between their N-terminal and C-terminal amino acid residues, and they further comprise an intramolecular bond between X4 and X9. In certain embodiments, the intramolecular bond is a disulfide bond, a thioether bond, a lactam bond, or any other bond described herein.
Peptide dimers
In certain embodiments, the invention includes dimers of the monomeric peptide inhibitors described herein, including dimers of any of the monomeric peptide inhibitors described herein or in the accompanying tables, figures or sequence listings. These dimers fall within the scope of the general term "peptide inhibitors" as used herein. Exemplary dimers of the invention are also shown in the attached table, which in parentheses represent dimerized monomer subunits followed by a linker. Unless otherwise indicated, the subunits are linked via their C-termini. The term "dimer" as in a peptide dimer refers to a compound in which two peptide monomer subunits are linked. The peptide dimer inhibitors of the present invention may comprise two identical monomer subunits, which produce a homodimer, or may comprise two different monomer subunits, which produce a heterodimer. The cysteine dimer comprises two peptide monomer subunits linked by a disulfide bond between a cysteine residue in one monomer subunit and a cysteine residue in the other monomer subunit.
In some embodiments, the peptide inhibitors of the invention are active when in a dimeric conformation, particularly when free cysteine residues are present in the peptide. In certain embodiments, this occurs as a synthetic dimer, or particularly when free cysteine monomer peptides are present and dimerize under oxidative conditions. In some embodiments, the dimer is a homodimer. In other embodiments, the dimer is a heterodimer.
In certain embodiments, a peptide dimer inhibitor of the present invention is a peptide dimer comprising two peptide inhibitors of the present invention, including, but not limited to, a homodimer or heterodimer comprising any of the peptide sequences shown herein, e.g., in table E3 Α -E3H, table E4A, table E4B, table E5 Α -E5C, table E6, table E7, table E8, table E9, table E10, table E11, table E12, table E13, or table 14.
Certain amino acid sequences listed in table E3 a-E3H, table E4A, table E4B, table E5 a-E5C, table E6, table E7, table E8, table E9, table E10, table E11, table E12, table E13, or table 14 are shown using the one letter codes for amino acids. Wherein only the sequence of the monomeric peptide inhibitor is shown; it is to be understood, however, that in certain embodiments, these monomeric peptide inhibitors (i.e., monomeric subunits) are dimerized to form a peptide dimer inhibitor according to the teachings of the present invention and as generally shown, for example, in table E3 a-E3H, table E4A, table E4B, table E5 a-E5C, table E6, table E7, table E8, table E9, table E10, table E11, table E12, table E13, or table 14.
In certain embodiments, the monomer subunits of the invention may dimerize via a suitable linking moiety (e.g., a disulfide bridge between two cysteines (cysteines in each peptide monomer subunit)), or via another suitable linker moiety, including but not limited to those defined herein. Some of the monomeric subunits are shown to have a C-terminus and an N-terminus both comprising free amines. Thus, to produce a peptide dimer inhibitor, the monomer subunits may be modified to eliminate the free amine at the C-or N-terminus, thereby allowing dimerization on the remaining free amine. Additionally, in some examples, the termini of one or more monomeric subunits are acylated with an acylated organic compound selected from: compounds containing trifluoropentyl, acetyl, octonyl, butyl, pentyl, hexyl, palmityl groups; trifluoromethyl butyric acid, cyclopentanecarboxylic acid, cyclopropaneacetic acid, 4-fluorobenzoic acid, 4-fluorophenylacetic acid, 3-phenylpropionic acid, tetrahydro-2H-pyran-4-carboxylic acid, succinic acid, and glutaric acid. In some examples, the monomeric subunits comprise a free carboxy terminus and a free amino terminus, whereby the subunits can be selectively modified by a user to achieve dimerization at a desired terminus. Thus, one skilled in the art understands that the monomeric subunits of the present invention can be selectively modified to obtain a single specific amine for the desired dimerization.
It is also understood that the C-terminal residue of the monomeric subunits disclosed herein are optionally an amide. Additionally, it is understood that in certain embodiments, dimerization of the C-terminus is facilitated by the use of suitable amino acid side chains with amine functionality, as is generally understood in the art. With respect to the N-terminal residue, it is generally understood that dimerization may be achieved by the free amine of the terminal residue, or may be achieved by using an appropriate amino acid side chain with a free amine, as is generally understood in the art.
The linker moiety linking the monomer subunits may comprise any structure, length, and/or size compatible with the teachings herein. In at least one embodiment, the linker moiety is selected from the non-limiting group consisting of: cysteine, lysine, DIG, PEG4, PEG 4-biotin, PEG13, PEG25, PEG1K, PEG2K, PEG3.4K, PEG4K, PEG5K, IDA, ADA, Boc-IDA, glutaric acid, isophthalic acid, 1, 3-phenylenediacetic acid, 1, 4-phenylenediacetic acid, 1, 2-phenylenediacetic acid, triazine, Boc-triazine, ID Α -biotin, PEG 4-biotin, AADA, suitable aliphatic compounds, aromatic compounds, heteroaromatic compounds, and polyethylene glycol-based linkers having a molecular weight of about 400Da to about 40,000 Da. Non-limiting examples of suitable linker moieties are provided in table 2A.
TABLE 2A. exemplary Joint Components
Figure GDA0003395945150001611
Figure GDA0003395945150001621
Figure GDA0003395945150001631
Figure GDA0003395945150001641
In some embodiments, the peptide dimer inhibitor dimerizes via a linker moiety. In some embodiments, the peptide dimer inhibitor dimerizes via an intermolecular disulfide bond formed between two cysteine residues (one cysteine in each monomer subunit). In some embodiments, the peptide dimer inhibitor dimerizes via an intermolecular disulfide bond formed between the linker moiety and the two cysteine residues. In some embodiments, the intramolecular linkage is a thioether, lactam, triazole, selenoether, diselenoether, or alkene linkage, rather than a disulfide linkage.
An exemplary scheme of one embodiment of the dimer is shown below:
Figure GDA0003395945150001651
those skilled in the art will appreciate that the linker (e.g., C-terminal and N-terminal linker) moieties disclosed herein are suitable non-limiting examples, and that the present invention may include any suitable linker moieties. Accordingly, some embodiments of the present invention comprise homodimeric or heterodimeric peptide inhibitors consisting of two monomer subunits selected from the peptides shown in any of the tables herein or comprising or consisting of the sequences presented in any of the tables herein, wherein the C-terminus or N-terminus (or internal amino acid residues) of each monomer subunit is linked by any suitable linker moiety to provide a dimeric peptide inhibitor having IL-23R inhibitory activity. In certain embodiments, the linker binds to the N-terminus or C-terminus of one monomeric subunit and internal amino acid residues of other monomeric subunits that make up the dimer. In certain embodiments, the linker binds to internal amino acid residues of one monomeric subunit and internal amino acid residues of other monomeric subunits that make up the dimer. In other embodiments, the linker is attached to the N-terminus or C-terminus of both subunits.
In particular embodiments, the peptide inhibitors of the invention comprise two or more polypeptide sequences of the monomeric peptide inhibitors described herein.
In one embodiment, the peptide dimer inhibitor of the present invention comprises two peptide monomer subunits linked via one or more linker moieties, wherein each peptide monomer subunit comprises or consists of: 7 to 35 amino acid residues, 8 to 35 amino acid residues, 9 to 35 amino acid residues, 10 to 35 amino acid residues, 7 to 25 amino acid residues, 8 to 25 amino acid residues, 9 to 25 amino acid residues, 10 to 25 amino acid residues, 7 to 20 amino acid residues, 8 to 20 amino acid residues, 9 to 20 amino acid residues, 7 to 18 amino acid residues, 8 to 18 amino acid residues, 9 to 18 amino acid residues or 10 to 18 amino acid residues, and comprising a sequence of formula Ia as described herein.
In particular embodiments, one or both of the monomeric subunits comprises a sequence of any one of formula X, formula I, formula II, formula III, formula IV, or formula V as described herein.
In certain embodiments, the peptide dimer inhibitor comprises two peptide monomer subunits linked via one or more linker moieties, wherein each peptide monomer subunit is 8-20 amino acids in length and comprises a sequence of any one of the formulae described herein, e.g., formula X, formula I, formula II, formula III, formula IV, or formula V. In certain embodiments, the peptide dimer inhibitor comprises two peptide monomer subunits linked via one or more linker moieties, wherein each peptide monomer subunit is 8-20 amino acids in length and comprises a sequence of any one of formula X, formula I, formula II, formula III, formula IV, or formula V.
In certain embodiments, the peptide dimer inhibitor, or a pharmaceutically acceptable salt or solvate thereof, has the structure of formula VI:
(R1-X-R2)2-L(VI)
wherein each R1Independently absent, is a bond (e.g., a covalent bond) or R1 is selected from hydrogen, C1-C6 hydrocarbyl, C6-C12 aryl, C6-C12 aryl C1-C6 hydrocarbyl, C1-C20 hydrocarbonyl, and includes pegylated forms of any of the foregoing alone or as a spacer;
each R is2Independently absent, is a bond (e.g., covalent) or is selected from OH or NH2
L is a linker moiety; and
each X is an independently selected peptide monomer subunit comprising or consisting of an amino acid residue of length: 7 to 35 amino acid residues, 8 to 35 amino acid residues, 9 to 35 amino acid residues, 10 to 35 amino acid residues, 7 to 25 amino acid residues, 8 to 25 amino acid residues, 9 to 25 amino acid residues, 10 to 25 amino acid residues, 7 to 20 amino acid residues, 8 to 20 amino acid residues, 9 to 20 amino acid residues, 7 to 18 amino acid residues, 8 to 18 amino acid residues, 9 to 18 amino acid residues or 10 to 18 amino acid residues, each of which comprises or consists of a sequence of formula Ia as described herein. In particular embodiments, each peptide monomer subunit comprises or consists of: a sequence of formula Ix, formula Ia, formula Ib, formula Ic, formula Id, formula Ie, formula If, formula Ig, formula Ih, formula Ii, formula Ij, formula Ik, formula Il, formula Im, formula In, formula Io, formula Ip, formula Iq', formula Is, formula It, formula IIa, formula IIb, formula IIc, formula IId, formula IIIa, formula IIIb, formula IIIc, formula IIId, formula IIIe, formula IVa, formula IVb, or formula Va-Vh as described herein.
In certain embodiments, one or both peptide monomer subunits of the peptide dimer inhibitor are cyclized, e.g., via an intramolecular bond between X4 and X9. In certain embodiments in which two peptide monomer subunits are cyclized, the intramolecular bonds between the two peptide monomer subunits may be the same or different. In certain embodiments, one or both intramolecular bonds are a disulfide bond, a thioether bond, a lactam bond, a selenoether bond, a diselenide bond, or an alkene bond.
In one embodiment, X4 and X9 in one or both cyclized peptide monomer subunits are independently selected from Cys, Pen, hCys, D-Pen, D-Cys, and D-hCys, and the intramolecular bond is a disulfide bond.
In one embodiment, X4 and X9 in one or both cyclized peptide monomer subunits are independently selected from Glu, Asp, Lys, Orn, Dap, Dab, D-Dap, D-Dab, D-Asp, D-Glu, and D-Lys, and the intramolecular bond is a lactam bond.
In one embodiment, X4 and X9 in one or both cyclized peptide monomer subunits are each independently selected from β -azido-Ala-OH, propargylglycine, and the peptide dimer inhibitor is cyclized through the triazole ring. In one embodiment, X4 and X9 in one or both cyclized peptide monomer subunits are each independently selected from 2-allylglycine, 2- (3 ' -butenyl) glycine, 2- (4 ' -pentenyl) glycine, 2- (5 ' -hexenyl) glycine, and the peptide dimer inhibitor is cyclized via a ring closing metathesis reaction to produce the corresponding alkene/' stapled peptide '.
In one embodiment, X4 of one or both cyclized peptide monomer subunits is 2-chloromethylbenzoic acid, mercaptopropionic acid, mercaptobutyric acid, 2-chloro-acetic acid, 3-chloro-propionic acid, 4-chloro-butyric acid, 3-chloro-isobutyric acid, or hser (cl), X9 of one or both cyclized peptide monomer subunits is hser (cl), Cys, Pen, hCys, D-Pen, D-Cys, or D-hCys, and the intramolecular bond is a thioether bond.
In one embodiment, X4 of one or both cyclized peptide monomer subunits is 2-chloromethylbenzoic acid, 2-chloro-acetic acid, 3-chloro-propionic acid, 4-chloro-butyric acid, 3-chloro-isobutyric acid, hser (cl), or Sec, X9 of one or both cyclized peptide monomer subunits is hser (cl) or Sec, and the intramolecular bond is a selenoether bond.
In certain embodiments, one or both intramolecular bonds are diselenide bonds.
In certain embodiments, one or both peptide monomer subunits are linear or not cyclized.
In a specific embodiment of the peptide dimer inhibitor, each X7 and each X11 is W. In certain embodiments, each X7 and each X11 is W, each X10 is Y, and each X4 and X9 is C. In certain embodiments, each X7 and each X11 is W, each X10 is Y, and each X4 and X9 is an amino acid capable of forming an intramolecular bond, the intramolecular bond being a thioether, lactam, triazole, selenoether, diselenide, or alkene bond.
In certain embodiments of the peptide dimer inhibitor, one or both peptide monomer subunits have the structure shown herein, e.g., in table E3 a-E3H, or comprise an amino acid sequence shown herein, e.g., as shown in table E3 a-E3H, or wherein said peptide dimer inhibitor has the structure shown herein, e.g., in table E3F, or comprise an amino acid sequence shown herein, e.g., as shown in table E3F.
In particular embodiments, each R is1Independently a bond (e.g., covalent bond) or selected from hydrogen, C1-C6 hydrocarbyl, C6-C12 aryl, C6-C12 aryl C1-C6 hydrocarbyl, C1-C20 hydrocarbonyl, and including pre-CA pegylated form of any of the foregoing alone or as a spacer. In particular embodiments, the N-terminus of each subunit comprises a moiety selected from hydrogen, C1-C6 hydrocarbyl, C6-C12 aryl, C6-C12 aryl C1-C6 hydrocarbyl, C1-C20 hydrocarbonyl and comprising the pegylated form of any of the foregoing alone or as a spacer.
In certain embodiments of any peptide inhibitor having any one of the formulae shown herein, each R is 1(or the N-terminal moiety) is selected from the group consisting of methyl, acetyl, formyl, benzoyl, trifluoroacetyl, isovaleryl, isobutyryl, octyl, and conjugated amides of dodecanoic acid, hexadecanoic acid, and γ -Glu-hexadecanoic acid.
In particular embodiments, each R is2(or C-terminal moiety) is independently a bond (e.g., covalent bond) or is selected from OH or NH2
In particular embodiments of any peptide inhibitor having any one of the formulae shown herein, each X comprises or consists of: 7 to 35 amino acid residues, 8 to 35 amino acid residues, 9 to 35 amino acid residues, 10 to 35 amino acid residues, 7 to 25 amino acid residues, 8 to 25 amino acid residues, 9 to 25 amino acid residues, 10 to 25 amino acid residues, 7 to 18 amino acid residues, 8 to 18 amino acid residues, 9 to 18 amino acid residues, or 10 to 18 amino acid residues.
In particular embodiments, one or both X comprise or consist of: a sequence of any one of the formulae as described herein. In certain embodiments of any of the peptide inhibitors (including dimers) or formulae shown herein, X does not comprise or consist of the amino acid sequence shown in U.S. patent application publication No. US 2013/0029907. In certain embodiments of any of the peptide inhibitors (including dimers) or formulae shown herein, X does not comprise or consist of the amino acid sequence shown in U.S. patent application publication No. US 2013/0172272.
In particular embodiments of the peptide inhibitors (monomers and dimers) of the invention comprising a Cys at X4 and a Cys at X9, the Cys at X4 and the Cys at X9 are linked by a disulfide bridge.
In a particular embodiment of the peptide inhibitor of the present invention, each X7 and each X11 is not both W.
In a specific embodiment of the peptide inhibitor of the present invention, each X7 and each X11 is W.
In particular embodiments of the peptide inhibitors of the invention, each X7 and each X11 is W, X10 is Y, and X4 and X9 are both C.
In certain embodiments, at least two cysteines in the peptide dimer inhibitor are linked by an intramolecular or intermolecular disulfide bridge.
In particular embodiments of either or both monomeric subunits (e.g., Ix, Ia-It, where permissible) present in the peptide dimer inhibitor, X4 and X9 are both Cys.
In particular embodiments of either or both monomeric subunits (e.g., Ix, Ia-It, where permissible) present in the peptide dimer inhibitor, X7 and X11 are both W.
In particular embodiments of either or both monomeric subunits (e.g., Ia-It, where permissible) present in the peptide dimer inhibitor, X7 and X11 are both W, X10 is Y, and X4 and X9 are both Cys.
In particular embodiments of either or both monomeric subunits (e.g., Ia-It, where permissible) present in the peptide dimer inhibitor, X15 is Gly or Ser.
In particular embodiments of either or both monomeric subunits (e.g., Ia-It, where permissible) present in the peptide dimer inhibitor, X16 is AEA or AEP.
In particular embodiments of either or both monomeric subunits (e.g., Ia-It, where permissible) present in a peptide dimer inhibitor, X10 is Tyr or Phe or an analog of Tyr or Phe.
In particular embodiments of either or both monomeric subunits (e.g., Ia-It, where permitted) present in the peptide dimer inhibitor, X11 is Trp.
Inhibition of any of the peptide dimers described hereinIn particular embodiments of the agent, either or both R1Is hydrogen.
In particular embodiments of the peptide dimer inhibitors of the present invention, linker moiety (L) is any linker described herein or shown in table 2A. In certain embodiments, L is a lysine linker, a diethylene glycol linker, an iminodiacetic acid (IDA) linker, a β -Ala-iminodiacetic acid (β -Ala-IDA) linker, or a PEG linker.
In various embodiments of any of the peptide dimer inhibitors, the peptide monomer subunits are each linked to a linker moiety via its N-terminal, C-terminal, or internal amino acid residue.
In certain embodiments of any of the peptide dimer inhibitors, the N-termini of the individual peptide monomer subunits are linked by a linker moiety.
In certain embodiments of any peptide dimer inhibitor, the C-termini of the individual peptide monomer subunits are linked by a linker moiety.
In certain embodiments of any peptide dimer inhibitor, the individual peptide monomer subunits are linked by a linker moiety linked to an internal amino acid.
In certain embodiments of the peptide dimer inhibitor, the linker moiety is a diethylene glycol linker, an iminodiacetic acid (IDA) linker, a β -Ala-iminodiacetic acid (β -Ala-IDA) linker, or a PEG linker.
In certain embodiments of the peptide dimer inhibitor, one or both peptide monomer subunits have a structure, or comprise an amino acid sequence, shown in any one of the tables in the examples.
In certain embodiments of any of the peptide inhibitors (including dimers) or formulae shown herein, X does not comprise or consist of the amino acid sequence shown in U.S. patent application publication No. US 2013/0029907. In certain embodiments of any of the peptide inhibitors (including dimers) or formulae shown herein, X does not comprise or consist of the amino acid sequence shown in U.S. patent application publication No. US 2013/0172272.
In a specific embodiment of the peptide inhibitors of the invention, each X7 and each X11 is W, X10 is Y, and X4 and X9 are Pen.
In certain embodiments, at least two cysteine residues in the peptide dimer inhibitor are linked by an intramolecular or intermolecular disulfide bridge.
Peptide inhibitor conjugates and biopolymers
In certain embodiments, the peptide inhibitors (including monomers and dimers) of the present invention comprise one or more conjugated chemical substituents, such as lipophilic substituents and polymeric moieties, which may be referred to herein as half-life extending moieties. Without wishing to be bound by any particular theory, it is believed that the lipophilic substituent binds to albumin in the blood stream, thereby shielding the peptide inhibitor from enzymatic degradation and thus increasing its half-life. In addition, it is believed that the polymeric moiety increases half-life and decreases clearance in the blood stream.
In further embodiments, any peptide inhibitor, such as peptides of formulas (Va) - (Vh), further comprises a linker moiety attached to an amino acid residue present in the inhibitor, e.g., the linker moiety may be conjugated to a side chain of any amino acid of the peptide inhibitor, an N-terminal amino acid of the peptide inhibitor, or a C-terminal amino acid of the peptide inhibitor.
In further embodiments, any peptide inhibitor, such as peptides of formulas (Va) - (Vh), further comprises a half-life extending moiety linked to an amino acid residue present in the inhibitor, e.g., the half-life extending moiety can be conjugated to a side chain of any amino acid of the peptide inhibitor, an N-terminal amino acid of the peptide inhibitor, or a C-terminal amino acid of the peptide inhibitor.
In further embodiments, any peptide inhibitor, such as peptides of formulas (Va) - (Vh), further comprises a half-life extending moiety linked to a linker moiety linked to an amino acid residue present in the inhibitor, e.g., the half-life extending moiety can be linked to a linker moiety that binds to a side chain of any amino acid of the peptide inhibitor, the N-terminal amino acid of the peptide inhibitor, or the C-terminal amino acid of the peptide inhibitor.
In particular embodiments, the IL23R analog comprises a half-life extending moiety having the structure shown below, wherein n-0 to 24 or n-14 to 24:
Figure GDA0003395945150001721
in certain embodiments, the IL23R analogs of the invention comprise a half-life extending moiety as shown in table 7.
TABLE 7 exemplary half-life extending moieties
Figure GDA0003395945150001722
Figure GDA0003395945150001731
In certain embodiments, the half-life extending moiety is directly conjugated to the peptide inhibitor, while in other embodiments, the half-life extending moiety is conjugated to the peptide inhibitor via a linker moiety, such as any of the linker moieties described in table 6 or table 8.
TABLE 8 exemplary Joint segment
Figure GDA0003395945150001732
Figure GDA0003395945150001741
Figure GDA0003395945150001751
In particular embodiments, the peptide inhibitors of the present invention comprise any linker moiety shown in table 8 and any half-life extending moiety shown in table 7, including any of the following combinations shown in table 9a.
Table 9a. exemplary combinations of linker and half-life extending moieties in peptide inhibitors
Figure GDA0003395945150001752
Figure GDA0003395945150001761
Figure GDA0003395945150001762
Figure GDA0003395945150001771
Figure GDA0003395945150001772
In some embodiments, there may be multiple linkers between the peptide and the conjugated moiety (e.g., half-life extending moiety), e.g., as described in table 9b.
TABLE 9b exemplary combinations of linker and half-life extending moieties in peptide inhibitors
Figure GDA0003395945150001781
Illustrative examples of peptide inhibitors of the invention are shown below, including peptide inhibitors having a conjugate linker and/or half-life extending moiety. Unless otherwise indicated, all amino acids are L amino acids. The invention also includes salt forms of any of these peptide inhibitors, including but not limited to acetate salts thereof.
Figure GDA0003395945150001782
Example 1: ring [ [ Abu ]]-QTWQC]- [ Phe [4- (2-amino)Radical ethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN-NH2(SEQ ID NO:1290)
Figure GDA0003395945150001791
Example 1 a: ac- [ (D) -Arg]-cyclo [ [ Abu ]]-QTWQC]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN-NH2(SEQ ID NO:1415)
Figure GDA0003395945150001792
Example 2: ac- [ Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran ]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1185)
Figure GDA0003395945150001801
Example 3: ring [ [ Abu ]]-QTWQC]- [ Phe [4- (2-aminoethoxy) - (linker-half-life extending moiety)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN-NH2(SEQ ID NO:1467)
Figure GDA0003395945150001802
Example 3 a: ac- [ (D) -Arg]Ring [ [ Abu ]]-QTWQC]- [ Phe [4- (2-aminoethoxy) - (linker-half-life extending moiety)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN-NH2(SEQ ID NO:1468)
Figure GDA0003395945150001811
Example 4: ring [ [ Abu ]]-QTWQC]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]- [ Lys (linker-half-life extending moiety)]-NN-NH2(SEQ ID NO:1469)
Figure GDA0003395945150001812
Example 4 a: ac- [ (D) -Arg]-cyclo [ [ Abu ]]-QTWQC]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]- [ Lys (linker-half-life extending moiety)]-NN-NH2(SEQ ID NO:1470)
Figure GDA0003395945150001821
Example 5: ring [ [ Abu ]]-QTWQC]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN- [ Lys (linker-half-life extending moiety)]-NH2(SEQ ID NO:1471)
Figure GDA0003395945150001822
Example 5a Ac [ (D) -Arg]-cyclo [ [ Abu ]]-QTWQC]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN- [ Lys (linker-half-life extending moiety)]-NH2(SEQ ID NO:1472)
Figure GDA0003395945150001831
Example 6 [ half-life extending moiety-linker]- [ Ring [ [ Abu ]]-QTWQC]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN-NH2(SEQ ID NO:1473)
Figure GDA0003395945150001832
Example 6a [ half-life extending moiety-linker]-[(D)-Arg]- [ Ring [ [ Abu ]]-QTWQC]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN-NH2(SEQ ID NO:1474)
Figure GDA0003395945150001841
Example 7 half-life extending moiety-linker]-[Pen]-NTWQ-[Pen]- [ Phe [4- (aminoethoxy) ]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1475)
Figure GDA0003395945150001842
Example 8 Ac- [ Pen]-NTWQ-[Pen]- [ Phe [4- (aminoethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-[Lys(Ac)]-NN- [ Lys (linker-half-life extending moiety)]-NH2(SEQ ID NO:1476)
Figure GDA0003395945150001851
Example 9 Ac- [ Pen]-NTWQ-[Pen]- [ Phe [4- (aminoethoxy) - (linker-half-life extending moiety)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1477)
Figure GDA0003395945150001852
Example 10: ac- [ Pen]-NTWQ-[Pen]- [ Phe [4- (aminoethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]- [ Lys (linker-half-life extending moiety)]-NN-NH2(SEQ ID NO:1478).
In certain embodiments, a peptide inhibitor of the invention comprising a conjugated chemical substituent (i.e., half-life extending moiety) has a half-life that is at least 100%, at least 120%, at least 150%, at least 200%, at least 250%, at least 300%, at least 400%, or at least 500% of the half-life of the same peptide inhibitor (but without the conjugated chemical substituent). In certain embodiments, the lipophilic substituent and/or the polymeric moiety increases the permeability of the peptide inhibitor through the epithelium and/or the retention of the peptide inhibitor in the lamina propria. In certain embodiments, the permeability through the epithelium and/or retention in the lamina propria of a peptide inhibitor of the invention comprising a conjugated chemical substituent is 100%, at least 120%, at least 150%, at least 200%, at least 250%, at least 300%, at least 400%, or at least 500% of the half-life of the same peptide inhibitor (but without the conjugated chemical substituent).
In one embodiment, the side chain of one or more amino acid residues (e.g., Lys residues) in the peptide inhibitors of the invention is conjugated (e.g., covalently linked) to a lipophilic substituent. The lipophilic substituent may be covalently bonded to an atom in the amino acid side chain, or alternatively may be conjugated to the amino acid side chain via one or more spacers. The spacer, when present, may provide a separation between the peptide analog and the lipophilic substituent. In particular embodiments, the peptide inhibitor comprises any of the conjugated moieties shown in tables 2-4 or tables 5A-5B.
In certain embodiments, the lipophilic substituent may comprise a hydrocarbon chain having from 4 to 30C atoms, for example at least 8 or 12C atoms, and preferably 24C atoms or less or 20C atoms or less. The hydrocarbon chain may be straight or branched and may be saturated or unsaturated. In certain embodiments, the hydrocarbon chain is substituted with a moiety that forms a linking moiety with an amino acid side chain or spacer, such as an acyl group, a sulfonyl group, an N atom, an O atom, or an S atom. In some embodiments, the hydrocarbon chain is substituted with an acyl group, and thus the hydrocarbon chain may form a hydrocarbon acyl moiety, such as a palmitoyl, hexanoyl, lauroyl, myristoyl, or stearoyl group.
The lipophilic substituent may be conjugated to any amino acid side chain in the peptide inhibitors of the invention. In certain embodiments, the amino acid side chain comprises a carboxyl, hydroxyl, thiol, amide, or amine group for forming an ester, sulfonyl ester, thioester, amide, or sulfonamide with a spacer or lipophilic substituent. For example, the lipophilic substituent may be conjugated to Asn, Asp, Glu, gin, His, Lys, Arg, Ser, Thr, Tyr, Trp, Cys, or Dbu, Dpr, or Orn. In certain embodiments, the lipophilic substituent is conjugated to Lys. The amino acid shown as Lys in any of the formulae provided herein may be substituted, for example, with Dbu, Dpr or Orn (where lipophilic substituents are added).
In certain embodiments, the peptide inhibitors of the invention may be modified by conjugating a chemical moiety to one or more amino acid side chains within the peptide, for example, to enhance stability, increase permeability, or enhance drug-like properties. For example, the N (. epsilon.) of lysine N (. epsilon.), the β -carboxyl group of aspartic acid or the γ -carboxyl group of glutamic acid may be suitably functionalized. Thus, to produce a modified peptide, amino acids within the peptide may be modified as appropriate. Additionally, in some examples, the side chain is acylated with an acylated organic compound selected from: trifluoropentyl, acetyl, Octonyl, butyl, pentyl, hexyl, palmityl, trifluoromethylbutyric acid, cyclopentanecarboxylic acid, cyclopropylacetic acid, 4-fluorobenzoic acid, 4-fluorophenylacetic acid, 3-phenylpropionic acid, tetrahydro-2H-pyran-4-carboxylic acid, succinic acid, glutaric acid or bile acid. One skilled in the art understands that a range of conjugates can be attached, for example, PEG4, isoglu, and combinations thereof. Those skilled in the art understand that amino acids within a peptide can be substituted isosterically, e.g., Lys can be substituted with Dap, Dab, α -MeLys, or Orn. Examples of modified residues within peptides are shown in table 1B.
Examples of modified lysine, Asp and Asn within peptides
Figure GDA0003395945150001871
Figure GDA0003395945150001881
In other embodiments of the invention, alternatively or additionally, the side chain of one or more amino acid residues in the peptide inhibitor of the invention is conjugated to a polymer moiety, e.g. in order to increase solubility and/or half-life in vivo (e.g. in plasma) and/or bioavailability. Such modifications are also known to reduce the clearance of therapeutic proteins and peptides (e.g., renal clearance).
As used herein, "polyethylene glycol" or "PEG" is a polyether compound of the general formula H- (O-CH2-CH2) n-OH. PEG is also known as polyethylene oxide (PEO) or Polyoxyethylene (POE), depending on their molecular weight, PEO, PEE or POG, as used herein, refers to oligomers or polymers of ethylene oxide. The three names are chemically synonymous, but PEG tends to refer to oligomers and polymers having a molecular mass of less than 20,000Da, PEO tends to refer to polymers having a molecular mass of greater than 20,000Da, and POE tends to refer to polymers of any molecular mass. PEG and PEO are liquids or low melting solids, depending on their molecular weight. Throughout this disclosure, these 3 names are used indiscriminately. PEG is prepared by polymerization of ethylene oxide and is commercially available in a wide range of molecular weights from 300Da to 10,000,000 Da. Although different molecular weight PEGs and PEO are used in different applications and have different physical properties (e.g., viscosity) due to chain length effects, their chemical properties are nearly identical. The polymer portion is preferably water-soluble (amphiphilic or hydrophilic), non-toxic, and pharmaceutically inert. Suitable polymer moieties include polyethylene glycol (PEG), homopolymers or copolymers of PEG, monomethyl-substituted polymers of PEG (mPEG) or polyoxyethylene glycerol (POG). See, e.g., int.j. hematology 68:1 (1998); bioconjugate chem.6:150 (1995); and crit. rev. therap. drug Carrier sys.9:249 (1992). Also contemplated are PEGs prepared for the purpose of extending half-life, e.g., mono-activated alkoxy-terminated polyalkylene oxides (POA's), such as mono-methoxy-terminated polyethylene glycols (mPEG's); also included are dual activated polyethylene oxide (ethylene glycol) or other PEG derivatives. The weight of suitable polymers varies widely, and a weight of about 200Da to about 40,000Da, or about 200Da to about 60,000Da, is generally selected for purposes of the present invention. In certain embodiments, PEG with a molecular weight of 200 to 2,000 or 200 to 500 is used. Different forms of PEG may also be used depending on the initiator used in the polymerization process-a common initiator is monofunctional methyl ether PEG or methoxy poly (ethylene glycol) (abbreviated mPEG).
Low molecular weight PEG can also be obtained in the form of pure oligomers, which are referred to as monodisperse, homogeneous or discrete. These are used in certain embodiments of the invention.
PEGs with different geometries are also available: branched PEG has three to ten PEG chains emanating from a central core group; star-shaped PEG has 10 to 100 PEG chains emanating from a central core group; and comb-shaped PEG has multiple PEG chains typically grafted onto a polymer backbone. PEG may also be linear. The numbers typically included in the names of PEGs represent their average molecular weight (e.g., a PEG with n-9 will have an average molecular weight of about 400 daltons and will be labeled PEG 400).
As used herein, "pegylation" is the act of covalently coupling a PEG structure to a peptide inhibitor of the invention, which is then referred to as a "pegylated peptide inhibitor". In certain embodiments, the PEG of the pegylated side chain is a PEG having a molecular weight of about 200 to about 40,000. In some embodiments, the spacer of the peptide of formula I, formula I', or formula I "is pegylated. In certain embodiments, the PEG of the pegylation spacer is PEG3, PEG4, PEG5, PEG6, PEG7, PEG8, PEG9, PEG10, or PEG 11. In certain embodiments, the PEG of the pegylation spacer is PEG3 or PEG 8.
Other suitable polymer moieties include polyamino acids such as polylysine, polyaspartic acid and polyglutamic acid (see, e.g., Gombotz, et al (1995), Bioconjugate chem., vol.6:332, 351; Hudecz, et al (1992), Bioconjugate chem., vol.3,49-57 and Tsukada, et al (1984), J.Natl.cancer Inst. vol.73, 721-.
In some embodiments, the peptide inhibitors of the invention may comprise two or more such polymeric moieties, in which case the overall molecular weight of all such moieties will generally fall within the ranges provided above.
In some embodiments, the polymer moiety is coupled (by covalent attachment) to an amino, carboxyl, or thiol group of an amino acid side chain. Certain examples are the thiol group of a Cys residue and the epsilon amino group of a Lys residue, and may also involve the carboxyl group of Asp and Glu residues.
The skilled person will be familiar with suitable techniques which may be used to carry out the coupling reaction. For example, a PEG moiety bearing a methoxy group may be coupled to the thiol group of Cys via a maleimide linkage using reagents commercially available from Nektar Therapeutics AL. For details on suitable chemistry, see also WO 2008/101017 and the references cited above. Maleimide functionalized PEG may also be conjugated to the thiol group of the side chain Cys residue.
As used herein, oxidation of disulfide bonds can occur in one step or in a two-step process. As used herein, for single step oxidation, trityl protecting groups are typically used during assembly, which allow deprotection during cleavage, followed by solution oxidation. When a second disulfide bond is desired, native oxidation or selective oxidation is selected. For selective oxidations that require orthogonal protecting groups, Acm and trityl groups are used as protecting groups for cysteine. Cleavage results in the removal of a protected pair of cysteines, allowing oxidation of the pair. A second oxidative deprotection step of the cysteine protected by the Acm group is then carried out. For native oxidation, trityl protecting groups were used for all cysteines, allowing native folding of the peptide. The skilled person will be familiar with suitable techniques which may be used to carry out the oxidation step.
Several chemical moieties, including poly (ethylene) glycol, are reacted with functional groups present in the twenty naturally occurring amino acids, such as, for example, the epsilon amino group in a lysine amino acid residue, a thiol or other nucleophilic amino acid side chain present in a cysteine amino acid residue. When multiple naturally occurring amino acids are reacted in a peptide inhibitor, these non-specific chemical reactions produce a final peptide inhibitor that contains a number of peptide isomers conjugated to one or more poly (ethylene) glycol chains at various positions within the peptide inhibitor.
One advantage of certain embodiments of the present invention includes the ability to add one or more chemical moieties (e.g., PEG) by incorporating one or more unnatural amino acids with unique functional groups that chemically react with activated PEG that is not reactive with the natural amino acids present in the peptide inhibitor. For example, azide groups and alkyne groups do not react with all naturally occurring functional groups in proteins. Thus, unnatural amino acids can be incorporated at one or more specific sites in a peptide inhibitor where PEG or another modification is desired, without undesirable non-specific reactions. In certain embodiments, the particular chemistry involved in the reaction results in a stable covalent linkage between the PEG chain and the peptide inhibitor. Furthermore, such reactions can be carried out under mild aqueous conditions without destroying most peptides. In certain embodiments, the unnatural amino acid residue is AHA.
Chemical moieties attached to the natural amino acids are limited in number and range. In contrast, a chemical moiety attached to an unnatural amino acid can employ a significantly larger range of useful chemicals, thereby attaching the chemical moiety to the target molecule. Essentially, any target molecule, including any protein (or portion thereof) that comprises an unnatural amino acid (e.g., an unnatural amino acid, such as an aldehyde-or ketone-derived amino acid, that contains a reactive site or side chain to which a chemical moiety can be attached) can serve as a substrate to which a chemical moiety is attached.
Many chemical moieties can be bound or linked to a particular molecule by a variety of methods known in the art. A number of such methods are described in U.S. patent No. 8,568,706. As an illustrative example, an azide moiety may be useful in conjugating a chemical moiety such as PEG or other chemical moieties described herein. The azide moiety acts as a reactive functional group and is not present in most naturally occurring compounds (and thus is not reactive with the natural amino acids in the naturally occurring compounds). Azides also selectively attach to a limited number of reaction partners, and azides are small and can be introduced into biological samples without significantly changing molecular size. One reaction that allows for the incorporation or introduction of azides into molecules is copper-mediated Huisgen [3+2] cycloaddition of azides. This reaction can be used for selective pegylation of peptide inhibitors (Tornoe et al, J.org.chem.67:3057,2002; Rostovtsev et al, Angew.chem., int.Ed.41:596, 2002; and Wang et al, J.Am.chem.Soc.125:3192,2003; Speers et al, J.Am.chem.Soc.,2003,125,4686).
Exemplary peptide inhibitors and peptide dimer inhibitors, and methods of making peptide inhibitors and peptide dimer inhibitors Method of
The present invention thus provides a variety of peptide inhibitors that bind to or associate with IL-23 to disrupt or block the binding between IL-23 and IL-23R.
Exemplary peptide inhibitors and peptide dimer inhibitors of the invention shown in table E3a-E3H, table E4A, table E4B, table E5a-E5C, table E6, table E7, table E8, table E9, table E10, table E11, table E12, table E13, or table 14 provide the amino acid sequences of selected monomeric peptide inhibitors and peptide dimer inhibitors, and indicate the linker moiety present in the peptide dimer inhibitor. According to the protocol discussed herein, a number of peptide inhibitors and peptide dimer inhibitors shown in the attached table were synthesized and cyclized. Tables E3A-E3H, E4A, E4B, E5A-E5C, E6, E7, E8, E9, E10, E11, E12, E13, or 14 provide IC50 values for selected monomeric peptide inhibitors and peptide dimer inhibitors to inhibit binding of IL-23 to IL-23R or to inhibit IL-23 signaling, as determined by measuring changes in phosphorylation-STAT 3 levels, as described in the appended examples. Exemplary peptide inhibitors of the invention are shown in formula (V) and tables 2-4 and 5A, which provide the amino acid sequence of the selected peptide inhibitor. These peptide inhibitors are acetates.
The peptide inhibitors of the present invention can be synthesized by a number of techniques known to those skilled in the art. In certain embodiments, the monomeric subunits are synthesized, purified, and dimerized using the techniques described in the accompanying examples. In certain embodiments, the invention provides methods of producing a peptide inhibitor of the invention (or a monomeric subunit thereof) comprising chemically synthesizing a peptide comprising, consisting of, or consisting essentially of a peptide having an amino acid sequence described herein, including but not limited to formula I, formula II, formula III, formula IV, formula V, or formula VI, or any amino acid sequence shown in any of the tables described herein. In other embodiments, the peptides are recombinantly synthesized, rather than chemically synthesized. In certain embodiments, the peptide inhibitor is a dimer, and the method comprises synthesizing two monomeric subunits of the peptide dimer inhibitor, and then dimerizing the two monomeric subunits to produce the peptide dimer inhibitor. In various embodiments, dimerization is achieved via any of the various methods described herein. In particular embodiments, the method of producing a peptide inhibitor (or monomeric subunit thereof) further comprises cyclizing the peptide inhibitor (or monomeric subunit thereof) after its synthesis. In particular embodiments, cyclization is achieved via any of the various methods described herein. In certain embodiments, the present invention provides methods of producing a peptide inhibitor of the present invention (or a monomeric subunit thereof) comprising introducing an intramolecular bond, e.g., a disulfide bond, an amide bond, or a thioether bond, between two amino acid residues within a peptide comprising, consisting of, or consisting essentially of a peptide having an amino acid sequence described herein, including but not limited to any amino acid sequence shown in formula I, formula II, formula III, formula IV, formula V, or formula VI, or any of the accompanying examples, tables, or sequence listings.
In related embodiments, the invention includes polynucleotides encoding polypeptides having a sequence as set forth in formula I, formula II, formula III, formula IV, formula V, or formula VI, or any of the accompanying examples, tables, or sequence listings.
In addition, the invention includes vectors, e.g., expression vectors, comprising a polynucleotide of the invention.
Method of treatment
In certain embodiments, the invention includes a method of inhibiting the binding of IL-23 to IL-23R on a cell, comprising contacting IL-23 with a peptide inhibitor of the invention. In certain embodiments, the cell is a mammalian cell. In particular embodiments, the method is performed in vitro or in vivo. Inhibition of binding can be determined by various routine experimental methods and assays known in the art.
In certain embodiments, the invention includes a method of inhibiting IL-23 signaling in a cell comprising contacting IL-23 with a peptide inhibitor of the invention. In certain embodiments, the cell is a mammalian cell. In particular embodiments, the method is performed in vitro or in vivo. In particular embodiments, inhibition of IL-23 signaling can be determined by measuring changes in the level of phospho-STAT 3 in the cell.
In some embodiments, the invention provides methods for treating a subject having a condition or indication associated with IL-21 or IL-23R (e.g., activation of the IL-23/IL-23R signaling pathway), wherein the method comprises administering to the subject a peptide inhibitor of the invention. In one embodiment, a method is provided for treating a subject having a condition or indication characterized by inappropriate, deregulated or increased IL-23 or IL-23R activity or signaling comprising administering to the subject a peptide inhibitor of the invention in an amount sufficient to (partially or completely) inhibit the binding of IL-23 to IL-23R in the subject. In particular embodiments, inhibition of IL-23 binding to IL-23R occurs in a specific organ or tissue of the subject, e.g., the stomach, small intestine, large intestine/colon, intestinal mucosa, lamina propria, peyer's patches, mesenteric lymph nodes, or lymphatic vessels.
In some embodiments, the methods of the invention comprise providing a subject in need thereof with a peptide inhibitor of the invention. In particular embodiments, a subject in need thereof has been diagnosed with a disease or disorder associated with IL-23/IL-23R or has been determined to be at risk of developing a disease or disorder associated with IL-23/IL-23R. In a specific embodiment, the subject is a mammal.
In certain embodiments, the disease or disorder is autoimmune inflammation and related diseases and disorders, such as multiple sclerosis, asthma, rheumatoid arthritis, Inflammatory Bowel Disease (IBD), juvenile IBD, crohn's disease, sarcoidosis, systemic lupus erythematosus, ankylosing spondylitis (axial spine arthritis), psoriatic arthritis, or psoriasis. In particular embodiments, the disease or disorder is psoriasis (e.g., plaque psoriasis, guttate psoriasis, reverse psoriasis, pustular psoriasis, palmoplantar pustulosis, psoriasis vulgaris or erythrodermopathy), dermatitis odorifera, ace ectoica, ulcerative colitis, crohn's disease, celiac disease (non-tropical sprue), enteropathy associated with seronegative arthropathy, microscopic colitis, collagenous colitis, eosinophilic gastroenteritis/esophagitis, colitis associated with radiotherapy or chemotherapy, colitis associated with an innate immune disorder such as leukocytic adhesion deficiency type 1, chronic granulomatosis, glycogen storage disease type 1b, herskmann-Pudlak syndrome, Chediak-Higashi syndrome, Wiskott-Aldrich syndrome, proctorey and ileoconvasitis resulting from anastomosis, Gastrointestinal cancer, pancreatitis, insulin-dependent diabetes mellitus, mastitis, cholecystitis, cholangitis, primary biliary cirrhosis, virus-associated bowel disease, peribiliary inflammation, chronic bronchitis, chronic sinusitis, asthma, uveitis, or graft-versus-host disease.
In certain related embodiments, the invention provides a method of selectively inhibiting IL-23 or IL-23R signaling (or IL-23 binding to IL-23R) in a subject in need thereof, comprising providing to the subject a peptide inhibitor of the invention. In particular embodiments, the invention includes a method of selectively inhibiting IL-23 or IL-23R signaling (or binding of IL-23 to IL-23R) in the GI tract of a subject in need thereof, comprising providing to the subject by oral administration a peptide inhibitor of the invention. In specific embodiments, the administered peptide inhibitor has an exposure in GI tissue (e.g., small intestine or colon) that is at least 10-fold, at least 20-fold, at least 50-fold, or at least 100-fold greater than the exposure in blood. In particular embodiments, the invention includes a method of selectively inhibiting IL23 or IL23R signaling (or binding of IL23 to IL 23R) in the GI tract of a subject in need thereof, comprising providing to the subject a peptide inhibitor, wherein the peptide inhibitor does not block the interaction between IL-6 and IL-6R or does not antagonize the IL-12 signaling pathway. In further related embodiments, the invention includes a method of inhibiting GI inflammation and/or neutrophil infiltration into the GI comprising providing a subject in need thereof with a peptide inhibitor of the invention. In some embodiments, the methods of the invention comprise providing a peptide inhibitor of the invention (i.e., a first therapeutic agent) in combination with a second therapeutic agent to a subject in need thereof. In certain embodiments, the second therapeutic agent is provided to the subject prior to and/or concurrently with and/or after administration of the peptide inhibitor to the subject. In particular embodiments, the second therapeutic agent is an anti-inflammatory agent. In certain embodiments, the second therapeutic agent is a nonsteroidal anti-inflammatory drug, a steroid, or an immunomodulator. In another embodiment, the method comprises administering to the subject a third therapeutic agent. In certain embodiments, the second therapeutic agent is an antibody that binds IL-23 or IL-23R.
In a specific embodiment, the peptide inhibitor or the pharmaceutical composition comprising the peptide inhibitor is suspended in a slow release matrix. As used herein, a sustained release matrix is a matrix made of a material (typically a polymer) that can be degraded by enzymatic or acid-base hydrolysis or by dissolution. Once embedded in the body, the matrix is acted upon by enzymes and body fluids. The sustained release matrix is desirably selected from biocompatible materials such as liposomes, polylactides (polylactic acid), polyglycolides (polymers of glycolic acid), polylactide-co-glycolide (copolymers of lactic and glycolic acid) polyanhydrides, poly (ortho) esters, polypeptides, hyaluronic acid, collagen, chondroitin sulfate, carboxylic acids, fatty acids, phospholipids, polysaccharides, nucleic acids, polyamino acids, amino acids (such as phenylalanine, tyrosine, isoleucine), polynucleotides, polyethylene propylene, polyvinylpyrrolidone, and silicone. One embodiment of the biodegradable matrix is a matrix of one of polylactide, polyglycolide, or polylactide-co-glycolide (a copolymer of lactic acid and glycolic acid).
In certain embodiments, the invention encompasses pharmaceutical compositions comprising one or more peptide inhibitors of the invention and a pharmaceutically acceptable carrier, diluent, or excipient. A pharmaceutically acceptable carrier, diluent or excipient refers to a non-toxic solid, semi-solid or liquid filler, diluent, encapsulating material or formulation aid of any type. Prevention of the action of microorganisms can be ensured by including various antibacterial and antifungal agents (e.g., parabens, chlorobutanol, phenol sorbic acid, and the like). It may also be desirable to include isotonic agents, for example, sugars, sodium chloride, and the like.
In certain embodiments, the composition is administered orally, parenterally, intracisternally, intravaginally, intraperitoneally, intrarectally, topically (e.g., by powder, ointment, drops, suppositories, or transdermal patches), by inhalation (e.g., intranasal spray), ocularly (e.g., intraocularly), or buccally. The term "parenteral" as used herein refers to modes of administration which include intravenous, intramuscular, intraperitoneal, intrasternal, subcutaneous, intradermal and intraarticular injection and infusion. Thus, in certain embodiments, the compositions are formulated for delivery by any of these routes of administration.
In certain embodiments, the pharmaceutical compositions for parenteral injection comprise pharmaceutically acceptable sterile aqueous or non-aqueous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions just prior to use. Examples of suitable aqueous and nonaqueous carriers, diluents, solvents or vehicles include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), carboxymethylcellulose and suitable mixtures thereof, beta-cyclodextrin, vegetable oils (such as olive oil), and injectable organic esters (such as ethyl oleate). Suitable fluidity can be maintained by: for example, by the use of coating materials such as lecithin, by maintaining the desired particle size in the case of dispersions, and by the use of surfactants. These compositions may also contain adjuvants such as preserving, wetting, emulsifying, and dispersing agents. Prolonged absorption of the injectable pharmaceutical form is brought about by the inclusion of agents which delay absorption such as aluminum monostearate and gelatin.
Injectable depot forms include those made by forming a microcapsule matrix of the peptide inhibitor in one or more biodegradable polymers such as polylactide-polyglycolide, poly (orthoester), poly (anhydride), and (poly) glycols such as PEG. Depending on the ratio of peptide to polymer and the nature of the particular polymer employed, the rate of release of the peptide inhibitor can be controlled. Depot injectable formulations are also prepared by entrapping the peptide inhibitor in liposomes or microemulsions which are compatible with body tissues.
The injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
Topical administration includes application to the skin or mucous membranes, including surfaces of the lungs and eyes. Compositions for topical pulmonary administration (including those for inhalation and intranasal) may comprise solutions and suspensions in aqueous and non-aqueous formulations, and may be prepared as dry powders, which may be pressurized or atmospheric. In an atmospheric pressure powder composition, the active ingredient in finely divided form may be employed in admixture with a pharmaceutically acceptable inert carrier of larger size containing, for example, particles of up to 100 microns in diameter size. Suitable inert carriers include sugars such as lactose.
Alternatively, the composition may be pressurised and contain a compressed gas, such as nitrogen or a liquefied gas propellant. The liquefied propellant medium, and indeed the total composition, may be such that the active ingredient is not dissolved therein to any significant extent. The pressurized composition may also contain a surfactant, such as a liquid or solid nonionic surfactant, or may be a solid anionic surfactant. Preferably, a solid anionic surfactant in the form of a sodium salt is used.
Other forms of topical administration are to the eye. The peptide inhibitors of the invention may be delivered in the form of a pharmaceutically acceptable ophthalmic vehicle such that the peptide inhibitor remains in contact with the ocular surface for a sufficient period of time to allow the peptide inhibitor to penetrate into the corneal and internal regions of the eye, such as, for example, the anterior chamber, the posterior chamber, the vitreous body, the aqueous humor, the vitreous humor, the cornea, the iris/eyelashes, the lens, the choroid/retina, and the sclera. The pharmaceutically acceptable ophthalmic vehicle may be, for example, an ointment, a vegetable oil, or an encapsulating material. Alternatively, the peptide inhibitors of the invention may be injected directly into the vitreous and aqueous humor.
Compositions for rectal or vaginal administration include suppositories which can be prepared by mixing the peptide inhibitors of the invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax, which are solid at room temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
The peptide inhibitors of the invention may also be administered in the form of liposomes or other lipid-based carriers. Liposomes are generally derived from phospholipids or other lipid substances, as is known in the art. Liposomes are formed by single or multiple layers of hydrated liquid crystals dispersed in an aqueous medium. Any non-toxic, physiologically acceptable and metabolizable lipid capable of forming liposomes can be used. The present composition in liposome form may contain a stabilizer, a preservative, an excipient, and the like, in addition to the peptide inhibitor of the present invention. In certain embodiments, the lipids include natural and synthetic phospholipids, including phosphatidylcholine (lecithin) and phosphatidylserine. Methods of forming liposomes are known in the art.
Pharmaceutical compositions suitable for parenteral administration to be used in the present invention may comprise sterile aqueous solutions and/or suspensions of the peptide inhibitors, which are typically made isotonic with the blood of the recipient using sodium chloride, glycerol, glucose, mannitol, sorbitol and the like.
In some aspects, the present invention provides pharmaceutical compositions for oral delivery. The compositions and peptide inhibitors of the invention may be prepared for oral administration according to any of the methods, techniques, and/or delivery vehicles described herein. In addition, those skilled in the art understand that the peptide inhibitors of the present invention may be modified or may be incorporated into systems or delivery vehicles not disclosed herein, but well known and compatible in the art for oral delivery of peptides.
In certain embodiments, formulations for oral administration may include adjuvants (e.g., resorcinol and/or non-ionic surfactants such as polyoxyethylene oleyl ether and n-hexadecyl polyethylene ether) to artificially increase the permeability of the intestinal wall, and/or enzyme inhibitors (e.g., pancreatic trypsin inhibitor, diisopropyl fluorophosphate (DFF) or aprotinin) to inhibit enzymatic degradation. In certain embodiments, the peptide inhibitor in a solid dosage form for oral administration may be mixed with at least one additive such as sucrose, lactose, cellulose, mannitol, trehalose, raffinose, maltitol, dextran, starch, agar, alginate, chitin, chitosan, pectin, gum tragacanth, gum acacia, gelatin, collagen, casein, albumin, synthetic or semi-synthetic polymers, or glycerides. These dosage forms may also contain other types of additives, for example, inactive diluents, lubricants (e.g., magnesium stearate, parabens), preservatives (e.g., sorbic acid, ascorbic acid, alpha-tocopherol), antioxidants such as cysteine, disintegrants, binders, thickeners, buffering agents, pH adjusters, sweeteners, flavoring agents, or flavoring agents.
In particular embodiments, an oral dosage form or unit dose compatible with the use of the peptide inhibitors of the present invention may comprise a mixture of the peptide inhibitor and a non-pharmaceutical component or excipient, as well as other single use materials, which may be considered ingredients or packaging. Oral compositions can include at least one of liquid, solid, and semi-solid dosage forms. In some embodiments, an oral dosage form is provided comprising an effective amount of a peptide inhibitor, wherein the dosage form comprises at least one of a pill, a tablet, a capsule, a gel, a paste, a drink, a syrup, an ointment, and a suppository. In some examples, oral dosage forms are provided that are designed and configured to achieve delayed release of a peptide inhibitor in the small intestine and/or colon of a subject.
In one embodiment, an oral pharmaceutical composition comprising a peptide inhibitor of the present invention comprises an enteric coating designed to delay the release of the peptide inhibitor in the small intestine. In at least some embodiments, a pharmaceutical composition comprising a peptide inhibitor of the invention and a protease inhibitor (e.g., aprotinin) is provided in a delayed-release pharmaceutical formulation. In some examples, the pharmaceutical compositions of the present invention comprise an enteric coating that is soluble in gastric fluid at a pH of about 5.0 or higher. In at least one embodiment, a pharmaceutical composition is provided comprising an enteric coating comprising a polymer having separable carboxyl groups, such as cellulose derivatives, including hydroxypropylmethylcellulose phthalate, cellulose acetate phthalate, and cellulose acetate trimellitate, and similar cellulose derivatives and other carbohydrate polymers.
In one embodiment, the pharmaceutical composition comprising the peptide inhibitor of the present invention is provided in an enteric coating designed to protect the pharmaceutical composition and release it in a controlled manner within the lower gastrointestinal system of the subject and designed to avoid systemic side effects. In addition to enteric coatings, the peptide inhibitors of the present invention may be encapsulated, coated, conjugated or otherwise associated within any compatible oral drug delivery system or component. For example, in some embodiments, the peptide inhibitors of the present invention are provided in a lipid carrier system comprising at least one of a polymeric hydrogel, a nanoparticle, a microsphere, a micelle, and other lipid systems.
To overcome degradation of peptides in the small intestine, some embodiments of the invention include a hydrogel polymer carrier system comprising a peptide inhibitor of the invention, whereby the hydrogel polymer protects the peptide inhibitor from proteolysis in the small intestine and/or colon. The peptide inhibitors of the invention may also be formulated for use with carrier systems designed to increase the dissolution kinetics of the peptide and enhance intestinal absorption. These methods include the use of liposomes, micelles, and nanoparticles to increase GI tract penetration of peptides.
Various bioresponse systems may also be combined with one or more peptide inhibitors of the invention to provide a medicament for oral delivery. In some embodiments, the peptide inhibitors of the invention are combined with a polymer such as hydrogels and mucoadhesive polymers with hydrogen bonding groups (e.g., PEG, poly (methacrylic) acid [ PMAA)]Cellulose, cellulose,
Figure GDA0003395945150002001
Chitosan and alginate) to provide a therapeutic agent for oral administration. Other embodiments include methods of optimizing or extending the drug residence time of the peptide inhibitors disclosed herein, wherein the surface of the peptide inhibitor surface is modifiedThe mucoadhesive properties are decorated with polymers containing mucin attached by hydrogen bonding or/and hydrophobic interactions. These modified peptide molecules may exhibit increased drug residence time in a subject, consistent with the desirable features of the present invention. Furthermore, the target mucoadhesion system can specifically bind to receptors on the surface of intestinal and M cells, thereby further increasing the uptake of particles containing peptide inhibitors.
Other embodiments include methods of oral delivery of the peptide inhibitors of the present invention, wherein the peptide inhibitor is provided to a subject in combination with a penetration enhancer that facilitates transport of the peptide across the intestinal mucosa by increasing paracellular or transcellular penetration. Various penetration enhancers and methods for Oral delivery of therapeutic agents are described in Brayden, D.J., Mrsny, R.J.,2011, Oral peptide delivery technologies, the. delivery 2(12), 1567-1573.
In certain embodiments, the pharmaceutical compositions and formulations of the present invention comprise a peptide inhibitor of the present invention and one or more penetration enhancers. Examples of absorption enhancers may include, for example, bile salts, fatty acids, surfactants (anionic, cationic and non-anionic), chelating agents, Zonular (Zonular) OT, esters, cyclodextrins, dextran sulfate, azone, crown ethers, EDTA, sucrose esters, and phosphatidylcholine. Although absorption enhancers are not generally carriers per se, they are also widely combined with other carriers to improve oral bioavailability through transport of peptides and proteins across the intestinal mucosa. Such materials may be added to the formulation as excipients or incorporated to form non-specific interactions with the intended peptide inhibitor.
Dietary components and/or other naturally occurring substances that are determined to enhance tight junction penetration and are Generally Regarded As Safe (GRAS) include, for example, asglycerides, acylcarnitines, bile salts, and medium chain fatty acids. Sodium salts of Medium Chain Fatty Acids (MCFAS) are also considered penetration enhancers. The most widely studied MCFAS is sodium caprate, which is the decanoate salt, with decanoic acid accounting for 2-3% of the fatty acids in the milk fat fraction. To date, sodium caprate has been used primarily as a suppository formulation (Doktacillin) TM) To improve rectal ampicillinAbsorption of penicillin. The osmotic properties of another diet MCFAS, sodium caprylate (8 carbon), showed lower in vitro than sodium caprate. Sodium caprylate and peptide drugs were formulated to produce an Oily Suspension (OS) of enhanced permeability mixed with other excipients in oil (Tuvia, s. et al, Pharmaceutical Research, vol.31, No.8, pp.2010-2021 (2014)).
For example, in one embodiment, a permeation enhancer is combined with the peptide inhibitor, wherein the permeation enhancer comprises at least one of a medium chain fatty acid, a long chain fatty acid, a bile salt, an amphiphilic surfactant, and a chelating agent. In certain embodiments, the medium chain fatty acid salt facilitates absorption by increasing the paracellular permeability of the intestinal epithelium. In one embodiment, a permeation enhancer comprising sodium N- [ hydroxybenzoyl) amino ] caprylate is used to form a weak non-covalent association with the peptide inhibitor of the present invention, wherein the permeation enhancer facilitates membrane transport and further disassociates once it reaches the blood circulation. In another embodiment, the peptide inhibitors of the present invention are conjugated to oligoarginine, thereby increasing cellular penetration of the peptide into various cell types. Additionally, in at least one embodiment, a non-covalent bond is provided between the peptide inhibitor of the present invention and a permeation enhancer selected from the group consisting of Cyclodextrins (CDs) and dendrimers, wherein the permeation enhancer reduces peptide aggregation and increases the stability and solubility of the peptide inhibitor molecule.
In certain embodiments, a pharmaceutical composition or formulation comprises a peptide inhibitor of the present invention and a Transient Permeability Enhancer (TPE). Penetration enhancers and TPEs can be used to increase oral bioavailability or peptide inhibitors. One example of a TPE that can be used is an oily suspension formulation with a powder containing sodium caprylate and a therapeutic agent dispersed (Tuvia, s. et al, Pharmaceutical Research, vol.31, No.8, pp.2010-2021 (2014)).
In certain embodiments, pharmaceutical compositions and formulations may comprise a peptide inhibitor of the invention and one or more absorption enhancers, enzyme inhibitors, or mucoadhesive polymers.
In particular embodiments, the peptide inhibitors of the present invention are formulated in a formulation vehicle, such as an emulsion, liposome, microsphere, or nanoparticle.
Other embodiments of the invention provide methods of treating a subject with a peptide inhibitor of the invention having an increased half-life. In one aspect, the invention provides peptide inhibitors that: the peptide inhibitor has a half-life in vitro or in vivo (e.g., when administered to a human subject) of at least several hours to one day, sufficient to administer a therapeutically effective amount once daily (q.d.) or twice daily (b.i.d.). In another embodiment, the peptide inhibitor has a half-life of three days or more, sufficient to be administered in a therapeutically effective amount once a week (q.w.). In addition, in another embodiment, the peptide inhibitor has a half-life of eight days or more, sufficient to administer a therapeutically effective amount once a week (b.i.w.) or once a month. In another embodiment, the peptide inhibitor is derivatized or modified such that it has a longer half-life compared to the underivatized or unmodified peptide inhibitor. In another embodiment, the peptide inhibitor contains one or more chemical modifications to increase serum half-life.
When used in at least one of the therapeutic or delivery systems described herein, the peptide inhibitors of the invention may be used in pure form or, when such form is present, in the form of a pharmaceutically acceptable salt.
The total daily amount of the peptide inhibitors and compositions of the present invention to be used may be determined by the attending physician within the scope of sound medical judgment. The specific therapeutically effective dose level for any particular subject will depend upon a variety of factors including: a) the condition being treated and the severity of the condition; b) the activity of the particular compound used; c) the particular composition used, the age, weight, general health, sex, and diet of the patient; d) the time of administration, route of administration, and rate of excretion of the particular peptide inhibitor used; e) the duration of the treatment; f) drugs used in combination or concomitantly with the particular peptide inhibitor employed and like factors well known in the medical arts.
In particular embodiments, the total daily dose of the peptide inhibitors of the invention administered to a human or other mammalian host in single or divided doses is in an amount of, for example, from 0.0001 to 300mg/kg body weight per day or from 1 to 300mg/kg body weight per day.
Non-invasive detection of enteritis
The peptide inhibitors of the invention can be used for the detection, assessment and diagnosis of enteritis by microPET imaging as part of a non-invasive diagnostic procedure, wherein the peptide inhibitors are labeled with a chelating group or a detectable label. In one embodiment, the peptide inhibitor is conjugated to a bifunctional chelating agent. In another embodiment, the peptide inhibitor is radiolabeled. The labeled peptide inhibitor is then administered to the subject orally or rectally. In one embodiment, the labeled peptide inhibitor is contained in drinking water. Following ingestion of the peptide inhibitor, inflammation throughout the subject's intestinal and digestive tract may be visualized using microPET imaging.
Identification of peptide inhibitors that inhibit IL-23 signaling
As described herein, in certain embodiments, the peptide inhibitors of the invention bind preferentially to human IL-23R and/or rat IL-23R as compared to mouse IL-23R. In contrast to human IL-23R or rat IL-23R, mouse IL-23R contains additional amino acids in the region corresponding to about amino acid residue 315 to about amino acid residue 340 of the mouse IL23R protein (e.g., amino acid region NWQPWSSPFVHQTSQETGKR) (see, e.g., FIG. 4). In a specific embodiment, the peptide inhibitor binds to the region of about amino acid 230 to about amino acid residue 370 of human IL-23R.
The present invention provides novel methods of identifying inhibitors (e.g., peptide inhibitors) of IL-23R based on identifying agents (e.g., peptides) that preferentially bind human IL-23R or rat IL-23R as compared to mouse IL-23R. In certain embodiments, the method comprises: (a) determining the amount of binding of the candidate substance to the human IL-23R polypeptide or the rat IL-23R polypeptide; (b) determining the amount of binding of the candidate substance to the mouse IL-23R polypeptide; and (c) comparing the determined amount of bound human IL-23R polypeptide or rat IL-23R polypeptide with the determined amount of bound mouse IL-23R polypeptide, wherein the candidate compound is an inhibitor of IL-23R if the determined amount of bound human IL-23R polypeptide or rat IL-23R polypeptide is greater than the amount of bound mouse IL-23R polypeptide. In particular embodiments, the candidate compound is identified as an inhibitor of IL-23R if the amount of bound human IL-23R polypeptide or rat IL-23R polypeptide determined is at least 1.5 fold, at least 2 fold, at least 3 fold, at least 4 fold, at least 5 fold, at least 10 fold, at least 20 fold, at least 30 fold, at least 40 fold, at least 50 fold, at least 100 fold, at least 200 fold, at least 500 fold, or at least 100 fold greater than the amount of bound mouse IL-23R polypeptide determined. In a specific embodiment, the candidate compound is a peptide. In a specific embodiment, the peptide is a peptide of one of the formulae described herein. In particular embodiments, the human IL-23 polypeptide or rat IL-23R polypeptide comprises or consists of a full-length human IL-23R or rat IL-23R protein, respectively. In other embodiments, the human IL-23R polypeptide is a fragment of a full length human IL-23R protein comprising 8 or more amino acid residues within the region of about amino acid residue 230 to about amino acid residue 370 of human IL-23R. In other embodiments, the rat IL-23R polypeptide is a fragment of a full length rat IL-23R protein comprising 8 or more amino acid residues within the region of about amino acid residue 245 to about amino acid residue 385 of rat IL-23R.
In another embodiment, the invention provides a new method of identifying inhibitors (e.g., peptide inhibitors) of IL-23R based on identifying agents that bind to human IL-23R or a region of rat IL-23 that results in disruption of mouse IL-23R due to the presence of additional amino acids from about amino acid residue 315 to about amino acid residue 340 of the mouse IL23R protein (e.g., amino acid region NWQPWSSPFVHQTSQETGKR) (see, e.g., FIG. 4). In certain embodiments, the method comprises: (a) determining the amount of binding of the candidate substance to a fragment of a human IL-23R polypeptide falling within about amino acid residue 230 to about amino acid residue 370 or to a fragment of a rat IL-23R polypeptide falling within about amino acid residue 245 to about amino acid residue 385; (b) determining the amount of candidate substance bound to a negative control (e.g., a negative control peptide not related to human IL-23R or rat-IL-23R); and (c) comparing the amount of the fragment that binds to the human IL-23R polypeptide or the fragment that binds to the rat IL-23R polypeptide to the amount of the binding negative control, wherein if the amount of the fragment that binds to the human IL-23R polypeptide or the fragment that binds to the rat IL-23R polypeptide is greater than the amount of the binding negative control, the candidate compound is an inhibitor of IL-23R. In particular embodiments, a candidate compound is identified as an inhibitor of IL-23R if the amount of bound human IL-23R polypeptide fragment or rat IL-23R polypeptide fragment determined is at least 1.5-fold, at least 2-fold, at least 3-fold, at least 4-fold, at least 5-fold, at least 10-fold, at least 20-fold, at least 30-fold, at least 40-fold, at least 50-fold, at least 100-fold, at least 200-fold, at least 500-fold, or at least 100-fold the amount of bound negative control determined. In a specific embodiment, the candidate compound is a peptide. In a specific embodiment, the peptide is a peptide of one of the formulae described herein. In specific embodiments, fragments of human IL-23R include at least 8, at least 12, at least 20, at least 50, or at least 100 amino acid residues or all amino acid residues within the region of about amino acid residue 230 to about amino acid residue 370 of human IL-23R. In other embodiments, a fragment of a rat IL-23R polypeptide includes at least 8, at least 12, at least 20, at least 50, or at least 100 amino acid residues, or all amino acid residues, within a region of about amino acid residue 245 to about amino acid residue 385 of rat IL-23R.
Methods of determining binding of a candidate compound to an IL-23 polypeptide are known in the art and include, but are not limited to, in vitro and cell-based binding assays, including those described herein. For example, labeled candidate compounds can be incubated with a solid support-bound recombinantly produced IL-23R polypeptide or negative control under conditions and for a time sufficient to allow binding, and binding then determined by measuring the amount of label associated with the bound IL-23R polypeptide.
Non-invasive detection of enteritis
The peptide inhibitors of the invention can be used for the detection, assessment and diagnosis of enteritis by microPET imaging as part of a non-invasive diagnostic procedure, wherein the peptide inhibitors are labeled with a chelating group or a detectable label. In one embodiment, the peptide inhibitor is conjugated to a bifunctional chelating agent. In another embodiment, the peptide inhibitor is radiolabeled. The labeled peptide inhibitor is then administered to the subject orally or rectally. In one embodiment, the labeled peptide inhibitor is contained in drinking water. Following ingestion of the peptide inhibitor, inflammation throughout the subject's intestinal and digestive tract may be visualized using microPET imaging.
Animal model of IBD
The invention includes models of animal diseases, including inflammatory diseases and disorders, such as inflammatory bowel disease, e.g., crohn's disease and colitis. As described in the accompanying examples, several animal models of the development of inflammatory diseases and disorders.
In one embodiment, the invention includes a method of assessing the ability of a candidate compound to inhibit or reduce an inflammatory disease condition, comprising:
(a) providing to a rat Dextran Sodium Sulfate (DSS) in an amount sufficient to induce IBD;
(b) providing an amount of a candidate compound to a rat; and
(c) measuring the amount of IBD symptoms present in the rat following provision of the DSS and the candidate compound;
wherein the candidate compound inhibits or reduces the inflammatory disease or disorder if the amount of IBD symptoms measured in (c) is significantly lower than the amount measured in a control rat provided with the amount of DSS and an amount of the control compound or no peptide (e.g., vehicle control).
In certain embodiments, the rats are provided DSS for about 5 to 12 days, e.g., about 9 days. In particular embodiments, the DSS is provided to the rat by providing the rat with potable water that is freely exposed to a DSS (e.g., about 1% to about 10% DSS, about 2% to about 5% DSS, or about 3% DSS). In specific embodiments, the rat is provided with from about 5mg/kg to about 100mg/kg, or from about 10mg/kg to about 50mg/kg, or about 20mg/kg or about 30mg/kg of the test compound. In particular embodiments, the test compound is provided to the rat orally (e.g., in drinking water). In certain embodiments, the DSS analysis is performed as described in the accompanying examples.
In another embodiment, the invention includes a method of evaluating the ability of a candidate compound to inhibit or reduce an inflammatory disease condition, comprising:
(a) providing 2,4, 6-trinitrobenzenesulfonic acid (TNBS) to rats in an amount sufficient to induce IBD;
(b) providing an amount of a candidate compound to a rat; and
(c) measuring the amount of IBD symptoms present in the rat following the provision of TNBS and the candidate compound;
wherein the candidate compound inhibits or reduces the inflammatory disease or disorder if the amount of IBD symptoms measured in (c) is significantly lower than the amount measured in a control rat that provided the amount of TNBS and an amount of a control compound or no peptide (e.g., vehicle control).
In certain embodiments, about 10mg/kg to about 200mg/kg TNBS is provided to the animal, for example, about 10mg/kg, about 20mg/kg, about 30mg/kg, about 40mg/kg, about 50mg/kg, about 60mg/kg, about 70mg/kg, about 80mg/kg, about 90mg/kg, about 100mg/kg, about 120mg/kg, about 150mg/kg, or about 200mg/kg TNBS. In certain embodiments, the TNBS is in an alcohol, e.g., in 45% -50% ethanol. In a specific embodiment, the TNBS is administered intrarectally. In specific embodiments, the rat is provided with from about 5mg/kg to about 100mg/kg, or from about 10mg/kg to about 50mg/kg, or about 20mg/kg or about 30mg/kg of the test compound. In particular embodiments, the test compound is provided to the rat orally (e.g., in drinking water). In certain embodiments, the TNBS analysis is performed as described in the accompanying examples.
In particular embodiments, the symptoms of IBD are measured immediately after the DSS or TNBS and the candidate compound (or test compound or no compound) are administered, or subsequently, for example, about 3 days, 5 days or 9 days after the initial administration of the DSS or TNBS and the candidate compound (or test compound or no compound). In particular embodiments, the measured IBD symptoms include one or more of percent weight loss, stool consistency, quantitative hemoccult score, and colon weight to colon length ratio. In certain embodiments, IBD symptoms are measured using a Disease Activity Index (DAI) score and/or a colon weight to colon length ratio, wherein the DAI score consists of a ranking from three parameters, including percent weight loss, stool consistency, and a quantitative hemoccult score, and a maximum of three units can be achieved.
In certain embodiments, a neutralizing anti-IL-23 p19 antibody is used as a comparator or positive control.
In certain embodiments, to assess the extent of inflammatory response, the animal is observed daily for clinical signs, including percentage weight loss and signs of loose or diarrhea, for example. After a period of incubation with DSS or TNBS (e.g., 5, 6 or 7 days), the rats are sacrificed and the entire colon length and colon weight from the cecum to the rectum are recorded. The severity of colitis can be assessed by a pathologist blinded to the nature of the treatment. In addition to colon wall thickness, gross colon lesions can be assessed on a scale of 0-4 according to table E19 below, and histopathological scores determined based on the parameters below (table E20 and table E21).
In certain embodiments, the symptoms of IBD are measured in three groups of rats, each group having at least 3 animals, e.g., six animals per group, wherein three groups include: vehicle, DSS or TNBS, and DSS or TNBS with positive control (e.g., sulfasalazine, QD administered at 100mg/kg PO).
Examples
Example 1
Synthesis of peptide monomers
Peptide monomers of the invention were synthesized on a Protein Technology's Symphony multichannel synthesizer using Merrifield solid phase synthesis Technology. The peptides were assembled using coupling conditions of HBTU (O-benzotriazole-N, N' -tetramethyl-uronium-hexafluoro-phosphate), Diisopropylethylamine (DIEA). For some amino acids, PyAOP (7-azabenzotriazol-1-yloxy) tripyrrolidinylphosphonium hexafluorophosphate) and DIEA coupling conditions were used. Rink Amide MBHA resin (100-200 mesh, 0.57mmol/g) was used for peptides with C-terminal amides and preloaded Wang resin with N-. alpha. -Fmoc protected amino acids was used for peptides with C-terminal acids. Coupling reagents were prepared at a concentration of 100mmol (pre-mixed HBTU and DIEA). Similarly, a 100mmol concentration solution of the amino acid was prepared. Peptide inhibitors of the invention are identified based on medical chemistry optimization and/or phage display, and screening is performed to identify those peptide inhibitors that have superior binding and/or inhibitory properties.
Assembly
The peptides were assembled using the standard Symphony protocol. The peptide sequences were assembled as follows: the resin (250mg, 0.14mmol) in each reaction vial was washed twice with 4ml DMF and subsequently treated with 2.5ml 20% 4-methylpiperidine (Fmoc deprotection) for 10 minutes. The resin was then filtered, washed twice with DMF (4ml) and treated again with N-methylpiperidine for a further 30 minutes. The resin was washed three more times with DMF (4ml) followed by the addition of 2.5ml of amino acid and 2.5ml of HBTU-DIEA mixture. After stirring frequently for 45 minutes, the resin was filtered and washed three times with DMF (4ml each). For a typical peptide of the invention, double coupling is performed. After completion of the coupling reaction, the resin was washed three times with DMF (4ml each) before proceeding to the next round of amino acid coupling.
Ring closing metathesis reaction to form olefins
The resin (100. mu. mol) was washed with 2ml DCM (3X 1 min) and then with 2ml DCE before treatment with 2ml of a solution of 6mM Grubbs first generation catalyst solution in DCE (4.94mg ml-1; 20 mol% for resin substitution). The solution was refluxed overnight under nitrogen (12 hours) before being discharged. The resin was washed three times with DMF (4ml each time); washed with DCM (4ml) before being dried and cleaved.
Cutting of
After peptide assembly is complete, the peptide is cleaved from the resin by treatment with a cleavage reagent such as reagent K (82.5% trifluoroacetic acid, 5% water, 5% thiophenylmethane, 5% phenol, 2.5% 1, 2-ethanedithiol). The cleavage reagent was able to successfully cleave the peptide from the resin and cleave all remaining side chain protecting groups.
The cleaved peptide was precipitated in cold ether, followed by two washes with ether. The filtrate was decanted, a second aliquot of cold ether was added, and the procedure repeated. The crude peptide was dissolved in acetonitrile: water (7:3, 1% TFA) and filtered. The mass of the linear peptide was then verified using electrospray ionization mass spectrometry (ESI-MS) (Micromass/Waters ZQ) prior to purification.
Formation of disulfide bonds via oxidation
Peptides containing free thiols (e.g., diPen) were assembled on Rink Amide-MBHA resin following the general Fmoc-SPPS procedure. The peptide was cleaved from the resin by treatment with a cleavage reagent (90% trifluoroacetic acid, 5% water, 2.5% 1, 2-ethanedithiol, 2.5% triisopropylsilane). The cleaved peptide was precipitated in cold ether, followed by two washes with ether. The filtrate was decanted, a second aliquot of cold ether was added, and the procedure was repeated. The crude peptide was dissolved in acetonitrile: aqueous (7:3, 1% TFA) and filtered to give the desired crude peptide as unoxidized peptide.
Dissolving crude cleaved peptides wherein X4 and X9 are Cys, Pen, hCys, (D) Pen, (D) Cys or (D) hCys in 20ml of water: in acetonitrile. Acetic acid containing saturated iodine was then added dropwise with stirring until the yellow color persisted. The solution was stirred for 15 minutes and the reaction was monitored by analytical HPLC and LCMS. When the reaction was complete, solid ascorbic acid was added until the solution was clear. The solvent mixture was then purified by: it was first diluted with water and then loaded onto a reverse phase HPLC machine (Luna C18 support, 10u, 100A, mobile phase a: water with 0.1% TFA, mobile phase B: Acetonitrile (ACN) with 0.1% TFA, gradient starting from 5% B and changing over 60 minutes to 50% B, flow rate 15 ml/min). The fractions containing the pure product were then freeze-dried on a freeze-dryer.
Formation of lactam bonds
100mg of the crude cleaved peptide (about 0.12mmol) was dissolved in 100ml of anhydrous dichloromethane. HOBt (1-hydroxybenzotriazole hydrate) (0.24mmol, 2 equiv.) was added followed by DIEA (N, N-diisopropylethylamine) (1.2mmol, 10 equiv.) and TBTU (O- (benzotriazol-1-yl) -N, N, N ', N' -tetramethyluronium tetrafluoroborate) (0.24mmol, 2 equiv.). The mixture was stirred overnight and the reaction was followed by HPLC. When the reaction was complete, dichloromethane was evaporated, diluted with water and acetonitrile and then loaded onto a reverse phase HPLC machine (Luna C18 support, 10u, 100A, mobile phase a: water with 0.1% TFA, mobile phase B: Acetonitrile (ACN) with 0.1% TFA, gradient starting from 5% B and changing to 50% B over 60 minutes, flow rate 15 ml/min). The fractions containing the pure product were then freeze-dried on a freeze-dryer.
Formation of triazole linkage
The purified peptide containing the relevant amino acids alkyne and azide was stirred (1mg/2ml) in phosphate/MeOH (2: 1) at pH 7.4 at room temperature. Adding CuSO4 & 5H2O (10 equivalents) and sodium ascorbate (10 equivalents), and the mixture was stirred at room temperature for 36 hours. MeOH was removed and the solution was acidified to pH 3 with 1% TFA water mixture. The solution was then filtered before being loaded into HPLC for purification of the peptide.
Formation of thioether linkages
Peptides containing free thiols (e.g., Cys) and hSer (OTBDMS) were assembled on Rink Amide-MBHA resin following the general Fmoc-SPPS procedure. Chlorination by PPh in DCM3(10 equivalents) and Cl3CCN (10 equivalents) was carried out for 2 hours. The peptide was cleaved from the resin by treatment with a cleavage reagent (90% trifluoroacetic acid, 5% water, 2.5% 1, 2-ethanedithiol, 2.5% triisopropylsilane). The cleaved peptide was precipitated in cold ether, followed by two washes with ether. The filtrate was decanted, a second aliquot of cold ether was added, and the procedure was repeated. The crude peptide was dissolved in acetonitrile: aqueous (7:3, 1% TFA) solution and filtration gave the desired crude acyclic peptide.
The crude peptide with free thiol (e.g., Cys, Pen, hCys, (D) Pen, (D) Cys or (D) hCys) and alkyl halide (hser (cl)) at X4 and X9 or X9 and X4 was dissolved in 0.1M TRIS buffer ph 8.5. Cyclization was allowed to occur overnight at room temperature. The solvent mixture was then purified by: it was first diluted two-fold with water and then loaded onto a reverse phase HPLC machine (Luna C18 support, 10u, 100A, mobile phase a: water with 0.1% TFA, mobile phase B: Acetonitrile (ACN) with 0.1% TFA, gradient starting from 5% B and changing to 50% B over 60 minutes, flow rate 15 ml/min). The fractions containing the pure product were then freeze-dried on a freeze-dryer.
Formation of selenoether linkages
The crude peptide containing the thiol-protected selenoamino acid and alkyl halide at positions X4 and X9 was dissolved in 0.1M sodium phosphate buffer (40 eq) containing DTT at pH 5.5. The cyclization was allowed to occur over 24 hours at room temperature. The solution was then diluted two-fold with water and the final cyclized peptide was purified using RP-HPLC to provide the selenoether.
Formation of diselenide bonds
The diselenide precursor was dissolved in 0.1M phosphate buffer at pH 6.0 and a DTT-containing isopropanol solution (40 equivalents) and the reaction mixture was incubated at 37 ℃. After 20 hours, additional DTT (10 equivalents) was added to the reaction. After a total of 32 hours, the cyclization reaction was then diluted with two-fold water and the final cyclized peptide was purified using RP-HPLC to provide the diselenide.
Purification of
Analytical reverse phase High Performance Liquid Chromatography (HPLC) was performed on a Gemini C18 column (4.6 mm. times.250 mm) (Phenomenex). Semi-preparative reverse phase HPLC was performed on a Gemini 10. mu. m C18 column (22 mm. times.250 mm) (Phenomenex) or Jupiter 10. mu.m, 300A ℃ 18 column (21.2 mm. times.250 mm) (Phenomenex). Separation was achieved using a linear gradient of buffer B in A (mobile phase A: water with 0.15% TFA, mobile phase B: Acetonitrile (ACN) with 0.1% TFA) at a flow rate of 1 mL/min (analytical) and 15 mL/min (preparative). Separation was achieved using a linear gradient of buffer B in A (mobile phase A: water with 0.15% TFA, mobile phase B: Acetonitrile (ACN) with 0.1% TFA) at a flow rate of 1 mL/min (analytical) and 15 mL/min (preparative).
Linker activation and dimerization
The peptide monomer subunits are linked to form a peptide dimer inhibitor as described below.
Small scale DIG linker activation procedure:5mL of NMP was added to a glass vial containing IDA diacid (304.2mg, 1mmol), N-hydroxysuccinimide (NHS, 253.2mg, 2.2 equivalents, 2.2mmol) and a stir bar. The mixture was stirred at room temperature to solidifyThe bulk raw material is completely dissolved. N, N' -dicyclohexylcarbodiimide (DCC, 453.9mg, 2.2 eq, 2.2mmol) was then added to the mixture. Precipitation occurred within 10 minutes and the reaction mixture was further stirred at room temperature overnight. The reaction mixture was then filtered to remove precipitated Dicyclohexylurea (DCU). The activated linker was stored in a sealed vial prior to use for dimerization. The nominal concentration of activated linker was about 0.20M.
For dimerization with PEG linkers, no pre-activation step is involved. Commercially available pre-activated bifunctional PEG linkers were used.
Dimerization procedure:2mL of anhydrous DMF was added to a vial containing peptide monomer (0.1 mmol). The pH of the peptide was adjusted to 8-9 with DIEA. The activated linker (IDA or PEG13, PEG 25) (0.48 equivalents relative to monomer, 0.048mmol) was then added to the monomer solution. The reaction mixture was stirred at room temperature for one hour. The completion of the dimerization reaction was monitored using analytical HPLC. The time for completion of the dimerization reaction varies depending on the linker. After completion of the reaction, the peptide was precipitated in cold ether and centrifuged. The upper ether layer was discarded. The precipitation step was repeated twice. The crude dimer was then purified using reverse phase HPLC (Luna C18 support, 10u, 100A, mobile phase A: water with 0.1% TFA, mobile phase B: Acetonitrile (ACN) with 0.1% TFA, 15% B and a gradient to 45% B over 60 minutes, flow rate 15 ml/min). The fractions containing the pure product were then freeze-dried on a freeze-dryer.
Example 2
Characterization of peptides to inhibit binding of interleukin-23 to interleukin-23 receptor
Optimization of peptides to identify at low concentrations (e.g., IC 50)<10nM) active while exhibiting Gastrointestinal (GI) stability. As described below, certain peptides were tested to identify peptides that inhibit IL-23 binding to human IL-23R and inhibit IL-23/IL-23R functional activity. The peptides tested included peptides containing various cyclization chemistries including, for example, cyclic amides (side chain cyclization), peptides containing disulfide bonds between, for example, two Pen residues, and peptides containing thioether bonds. Peptide inhibitors of the invention include, but are not limited toA peptide having any of the structures described herein. In addition, the peptide inhibitors of the invention include those having the same amino acid sequence as the peptides or structures described herein, without necessarily having the same or any N-or C-terminal "capping" groups, such as, for example, Ac or NH2
Assays performed to determine peptide activity are described below, and the results of these assays are provided in tables E3 a-E3H, tables E4A and E4B, tables E5 a-E5C, tables E6, tables E7 and table E8. Human ELISA represents the competitive binding assay for IL23-IL23R as described below, rat ELISA represents the competitive binding ELISA assay for rat IL-23R as described below, and pStat3HTRF represents the assay for DB cell IL-23R pSTAT3 as described below. The peptides described in tables E3B-E3E are cyclized via a disulfide bridge formed between two cysteines in these peptides. As shown, the peptides described in table E3F were partially dimerized via a linker moiety or through an internal cysteine. The peptides described in tables E4A and E4B are cyclized via the two Pen residues present in each of these peptides. The peptides described in table E5A were cyclized via a thioether bond between the indicated amino acid residues. Table E5B provides exemplary structures describing thioether cyclization, which is indicated in the table by the term "ring", followed immediately by the bracketed ring region. The monomer subunits of the peptide dimers shown in table E5C are cyclized by the term "ring" as shown and are linked to each other via the linker shown. The peptides shown in table E6 were cyclized via ring closing metathesis of the indicated residues. Table E7 provides two exemplary structures describing cyclization via cyclic amide side chains, and the peptides in this table are cyclized as indicated by the term "ring". Table E8 describes peptides cyclized via cysteine and Pen residues.
Peptide inhibitors of the invention include cyclized forms of the peptides set forth herein, as well as non-cyclized forms. For certain peptides, the residue Abu is present at the indicated position, while in other embodiments involving non-cyclized forms, Abu may be referred to as a hSer (Cl) or high Ser residue.
IL23-IL23R competitive binding ELISA
Will be provided with
Figure GDA0003395945150002131
4HBX plates were coated with 50 ng/well IL23R _ huFC and incubated overnight at 4 ℃. Wells were washed four times with PBST, blocked with 3% skim milk in PBS for 1 hour at room temperature, and washed four more times with PBST. To each well was added serial dilutions of test peptide and IL-23 diluted in assay buffer (PBS with 1% skim milk) to a final concentration of 2nM and incubated for 2 hours at room temperature. After washing the wells, the wells were washed by mixing with goat anti-p 40 polyclonal antibody (R) diluted in assay buffer at 50 ng/well&D Systems # AF309) was incubated at room temperature for 1 hour to detect bound IL-23. The wells were washed four more times with PBST. Secondary antibody (HRP-conjugated donkey anti-goat IgG diluted at 1:5000 in assay buffer (Jackson ImmunoResearch Laboratories # 705-. The plates were subjected to final washing as above. The signal was visualized with TMB One Component HRP Membrane Substrate (TMB One Component HRP Membrane Substrate), quenched with 2M sulfuric acid and read spectrophotometrically at 450 nm. The IC50 values determined for the various test peptides from these data are shown in tables E3 a-E3H, tables E4A and 4EB, tables E5 a-E5C, table E6, table E7 and table E8.
Rat IL-23R competitive binding ELISA
Assay plates were coated with 300 ng/well rat IL-23R _ huFC and incubated overnight at 4 ℃. The wells were washed, blocked and rewashed. To each well, serial dilutions of test peptide and IL-23 were added at a final concentration of 7nM and incubated for 2 hours at room temperature. After washing the wells, bound IL-23 was detected with goat anti-p 40 polyclonal antibody followed by HRP conjugated donkey anti-goat IgG. Signal was visualized with TMB single component HRP membrane substrate and quenched with 2M sulfuric acid. The IC50 values determined from these data for the various test peptides are shown in table E3G, table E3H, table E4A, table E4B, table E5B, table E5C, and table E8.
DB cell IL23R pSTAT3 cell analysis
IL-23 plays a major role in supporting and maintaining Th17 differentiation in vivo. This process is thought to be mediated primarily by signal transducer and activator of transcription 3(STAT3), phosphorylation of STAT3 (to produce pSTAT3) leading to RORC and promotionInflammation of the IL-17 upregulation. This cell assay examined pSTAT3 levels in DB cells expressing IL-23R when stimulated with IL-23 in the presence of test compounds. DB cells (ATCC # CRL-2289) cultured in RPMI-1640 medium (ATCC #30-2001) supplemented with 10% FBS and 1% glutamine were seeded at 5X10E5 cells/well on 96-well tissue culture plates. To each well was added serial dilutions of test peptide and IL-23 at a final concentration of 0.5nM, and 5% CO at 37 ℃ 2Was incubated in the humidified incubator for 30 minutes. Changes in the level of phospho-STAT 3 in cell lysates were detected using the Cisbio HTRF pSTAT3 cell assay kit according to the manufacturer's two-plate assay protocol. The IC50 values determined from these data are shown in tables E3E, table E3G, table E3H, table E4A, table E4B, table E5B, table E5C, and table E8 as absolute values or ranges. Where not shown, no data was measured.
Table e3a. exemplary acyclic peptides and activities
Figure GDA0003395945150002141
Figure GDA0003395945150002151
Exemplary peptides containing the CXXXXC motif (SEQ ID NO:1479) of IC50>1uM in IL23-IL23R competitive binding ELISA
Figure GDA0003395945150002152
Figure GDA0003395945150002161
Exemplary peptides containing the CXXXXC motif (SEQ ID NO:1479) with an IC50 of 500nM to 1000nM in an IL23-IL23R competitive binding ELISA
Figure GDA0003395945150002162
Figure GDA0003395945150002171
Exemplary peptides containing the CXXXXC motif (SEQ ID NO:1479) with IC50<500nM in an IL23-IL23R competitive binding ELISA
Figure GDA0003395945150002172
Figure GDA0003395945150002181
TABLE E3E. IC50 for active exemplary peptides containing the CXXXXC motif (SEQ ID NO:1479)
Figure GDA0003395945150002191
*=<10nM;**=10-25nM***=25-100nM,****=100-1000nM,*****=1000-10,000nM。
Table e3f. IC50 of exemplary peptide dimers
Figure GDA0003395945150002192
Figure GDA0003395945150002201
*=<10nM;**=10-25nM***=25-100nM,****=100-1000nM,*****=1000-10,000nM。
TABLE E3G IC50 for an exemplary peptide comprising the CXXWXCXXXXXXX- [ (D) Lys ] motif (SEQ ID NO:1480)
Figure GDA0003395945150002202
Figure GDA0003395945150002211
Figure GDA0003395945150002221
Figure GDA0003395945150002231
Figure GDA0003395945150002241
Figure GDA0003395945150002251
TABLE E3H IC50 for exemplary peptides containing the CXXWXCXXXX motif (SEQ ID NO:1481)
Figure GDA0003395945150002252
Figure GDA0003395945150002261
Figure GDA0003395945150002271
Figure GDA0003395945150002281
Figure GDA0003395945150002291
TABLE E4A IC50 for an illustrative example of dimers of peptides containing Ac- [ Pen ] -XXWX- [ Pen ] -XXXXXXX motif (SEQ ID NO:1482) and analogs
Figure GDA0003395945150002292
Figure GDA0003395945150002301
*=<10nM;**=10-25nM,***=25-100nM,****=100-1000nM,*****=1000-10,000nM
TABLE E4B IC50 for exemplary peptides containing Ac- [ Pen ] -XXWX- [ Pen ] -XXXXXX motifs and analogs
Figure GDA0003395945150002302
Figure GDA0003395945150002311
Figure GDA0003395945150002321
Figure GDA0003395945150002331
Figure GDA0003395945150002341
Figure GDA0003395945150002351
Figure GDA0003395945150002361
*=<10nM;**=10-25nM***=25-100nM,****=100-1000nM,*****=1000-10,000nM。
TABLE E5A. IC50 for exemplary peptide inhibitors (thioethers)
Figure GDA0003395945150002362
Figure GDA0003395945150002371
TABLE E5B IC50 for exemplary peptide inhibitors (thioethers)
Figure GDA0003395945150002372
Ac-Ring- [ [ Abu]XXWXC]-[Phe(4-OMe)]-[2-Nal]-XXX-NH2(SEQ ID NO:1483)
Figure GDA0003395945150002381
Figure GDA0003395945150002391
Figure GDA0003395945150002401
Figure GDA0003395945150002411
Figure GDA0003395945150002421
Figure GDA0003395945150002431
Figure GDA0003395945150002441
Figure GDA0003395945150002451
Figure GDA0003395945150002461
Figure GDA0003395945150002471
Figure GDA0003395945150002481
Figure GDA0003395945150002491
Figure GDA0003395945150002501
Figure GDA0003395945150002511
*=<10nM;**=10-25nM***=25-100nM,****=100-1000nM,*****=>1000nM。
Table e5c. IC50 for synthetic exemplary thioether-peptide dimers
Figure GDA0003395945150002512
Figure GDA0003395945150002521
*=<10nM;**=10-25nM***=25-100nM,****=100-1000nM,*****=>1000nM。
Table e5d
Figure GDA0003395945150002522
Figure GDA0003395945150002531
*=≤1nM;**=1nM-10nM;***=10nM-100nM
TABLE E6. IC50 of peptide inhibitors (Ring closing metathesis)
Figure GDA0003395945150002532
Figure GDA0003395945150002541
*=<10nM;**=10-25nM***=25-100nM,****=100-1000nM,*****=1000-10,000nM。
TABLE E7. IC50 of exemplary peptides containing cyclic amides (side chain cyclization)
Figure GDA0003395945150002542
SEQ ID NO:1484
Figure GDA0003395945150002543
TABLE E8. IC50 of exemplary peptides containing Ac- [ Pen ] -XXWXXXXXXX motif (SEQ ID NO:1485) and Ac-XXXXWX- [ Pen ] -XXXXXXX (SEQ ID NO:1486) analogs
Figure GDA0003395945150002544
Figure GDA0003395945150002551
*=<10nM;**=10-25nM***=25-100nM,****=100-1000nM,*****=1000-10,000nM。
SAR analysis of the activity of the tested peptide inhibitors showed that CXXXXC (SEQ ID NO:1479) disulfide was associated with high activity. Two Trp residues and Phe residues are also associated with high activity, but it will be appreciated that these amino acids can be readily exchanged with analogous homologues (e.g., 1-Nal substituted for Trp and/or Phe substituted for Tyr). Furthermore, the data indicate that the presence of one or more basic residues at the C-terminus is associated with high activity. Furthermore, His-9 may be replaced by Arg or another homologue and maintain or improve activity. The following scheme provides an exemplary consensus sequence showing certain residues associated with high activity.
Figure GDA0003395945150002552
(SEQ ID NO:275)
Example 3
Stability of peptide inhibitors under Simulated Intestinal Fluid (SIF), Simulated Gastric Fluid (SGF) and redox conditions
Studies were performed in Simulated Intestinal Fluid (SIF) and Simulated Gastric Fluid (SGF) to evaluate gastric stability of the peptide inhibitors of the invention. In addition, studies were conducted to evaluate the redox stability of the peptide inhibitors of the present invention.
SIF was prepared by adding 6.8g of monopotassium phosphate and 10.0g of pancreatin to 1.0L of water. After dissolution, the pH was adjusted to 6.8 using NaOH. DMSO stock solutions (2mM) for test compounds were first prepared. Aliquots of the DMSO solution were dispensed into 6 separate tubes, each containing 0.5mL SIF, which were preheated to 37 ℃. The final test compound concentration was 20 μ M. The vial was kept on bench for the duration of the experiment
Figure GDA0003395945150002561
In (1). At each time point (0, 5, 10, 20, 40, 60 or 360 minutes or 24 hours), 1.0mL of acetonitrile containing 1% formic acid was added to one vial to stop the reaction. The samples were stored at 4 ℃ until the end of the experiment. After sampling at the last time point, the tubes were mixed and then centrifuged at 3,000rpm for 10 minutes. An aliquot of the supernatant was removed, diluted 1:1 into distilled water containing an internal standard and analyzed by LCMS/MS. The remaining percentage of each time point was calculated based on the peak area response ratio of the test compound to the internal standard. Time 0 is set to 100% and all subsequent time points are calculated relative to time 0. The half-life was calculated by fitting to a first order exponential decay equation using Graphpad. Stability in SIF analysis is shown in table E9 and table E10.
SGF (final pH 2) was prepared by adding 20mg NaCl, 32mg porcine pepsin (MP Biochemicals, cat: 02102599) and 70. mu.l HCl to 10ml water. Aliquots of SGF (0.5 ml each) were preheated at 37 ℃. To initiate the reaction, 1 μ l peptide stock solution (10mM in DMSO) was added to 0.5ml SGF and mixed well to give a final peptide concentration of 20 μ M. The reaction was incubated at 37 ℃ with gentle shaking. At each time point (0, 15, 30, 60 min), a 50 μ l aliquot was removed and added to 200ul of acetonitrile containing 0.1% formic acid to quench the reaction. The samples were stored at 4 ℃ until the end of the experiment and centrifuged at 10,000rpm for 5 minutes. An aliquot of the supernatant was removed, diluted 1:1 into distilled water containing an internal standard and analyzed by LCMS/MS. The remaining percentage of each time point was calculated based on the peak area response ratio of the test compound to the internal standard. Time 0 is set to 100% and all subsequent time points are calculated relative to time 0. The half-life was calculated by fitting to a first order exponential decay equation using Graphpad. Stability in SGF analysis is shown in tables E9 and E10.
TABLE E9. stability of exemplary peptides containing the Ac- [ Pen ] -XXWX- [ Pen ] -XXXXXXX motif (SEQ ID NO:1482) and analogs in Simulated Intestinal Fluid (SIF) and Simulated Gastric Fluid (SGF)
Figure GDA0003395945150002571
Figure GDA0003395945150002581
Figure GDA0003395945150002591
The substrate used is a 100-fold dilution of standard SIF concentration
>360 minutes; 180 ═ 360 minutes; 120 ═ 120-; (ii) 60-120 minutes; (ii) 60 minutes
TABLE E10 stability of exemplary peptides containing thioether motifs and analogs within Simulated Intestinal Fluid (SIF) and Simulated Gastric Fluid (SGF)
Figure GDA0003395945150002601
Figure GDA0003395945150002611
Figure GDA0003395945150002621
Figure GDA0003395945150002631
Figure GDA0003395945150002641
The matrix is 100 times diluted solution of standard SIF concentration
>360 minutes; 180 ═ 360 minutes; 120 ═ 120-; (ii) 60-120 minutes; (ii) 60 minutes
For each peptide tested, DTT stability analysis was performed by adding 5. mu.l of a 10mM peptide stock solution in DMSO (final peptide concentration 50. mu.M) to 1ml of 100mM Tris-Cl, pH 7.5. At 0 min, 5 μ l of freshly thawed 100mM DTT solution was added to the peptide-containing incubation tube to give a final DTT concentration of 0.5 mM. The reaction was incubated at room temperature. At various time points up to 120 min (20 min, 40 min, 80 min, 120 min), 50 μ l aliquots were removed and the reaction was quenched by the addition of 10 μ l of 5M acetic acid. To measure the disappearance of the parent peptide, the quenched sample (30 μ l) was analyzed by reverse phase HPLC and UV absorbance at 220 nm. The remaining oxidized fractions were plotted against time and the half-life was calculated by fitting to a first-order exponential decay equation using Excel. The results of these studies are shown in table E11. Peptides with half-lives >120 minutes were considered stable.
TABLE E11 stability of exemplary peptides in DTT assays
Figure GDA0003395945150002642
10-120 minutes
Example 4
Cross-reactivity of peptide inhibitors
The amino acids of the extracellular domain of human IL-23R are 95%, 77% and 70% identical to cynomolgus IL-23R, rat IL-23R and mouse IL-23R, respectively. Interestingly, the mouse receptor contains an insertion of 21 residues that are not present in human, mouse, chimpanzee, canine and bovine receptors. These additional amino acids are located in the region: the region is believed to be where human IL-23R binds to IL-23.
To identify peptide inhibitors that cross-react with IL-23R of species other than human, the ability of certain peptide inhibitors to inhibit human IL-23R, cynomolgus IL-23R, rat IL-23R, and mouse IL-23R was analyzed by ELISA. Consistent with the observations regarding sequence differences between human IL-23R and mouse IL-23R, the tested peptide antagonists showed either a lack of inhibitory activity or very weak inhibitory activity in the mouse IL-23R ELISA (see Table E12). In contrast, the antagonists tested to date exhibited comparable potency against the rat receptor, and slightly less activity against the cynomolgus monkey receptor.
Various bioassays performed to determine the potency, cross-reactivity and selectivity of IL-23R antagonists are described below.
Analysis of Selectivity of specific IL-23R antagonists
Human IL-12R beta 1 ELISA
Assay plates were coated with 100 ng/well human IL-12R β 1_ huFC and incubated overnight at 4 ℃. The wells were washed, blocked and washed again. To each well was added serial dilutions of the test peptide and IL-23 at a final concentration of 2.5nM and incubated for 2 hours at room temperature. After washing the wells, bound IL-23 was detected with goat anti-p 40 polyclonal antibody followed by detection with HRP conjugated donkey anti-goat IgG. Signal was visualized with TMB single component HRP membrane substrate and quenched with 2M sulfuric acid.
Mouse IL-23R competitive binding ELISA
Assay plates were coated with 50 ng/well mouse IL-23R _ huFC and incubated overnight at 4 ℃. The wells were washed, blocked and washed again. To each well was added serial dilutions of the test peptide and IL-23 at a final concentration of 4nM and incubated for 2 hours at room temperature. After washing the wells, bound IL-23 was detected with goat anti-p 40 polyclonal antibody followed by detection with HRP conjugated donkey anti-goat IgG. Signal was visualized with TMB single component HRP membrane substrate and quenched with 2M sulfuric acid.
Rat IL-23R competitive binding ELISA
Assay plates were coated with 300 ng/well rat IL-23R _ huFC and incubated overnight at 4 ℃. The wells were washed, blocked and washed again. To each well was added a serial dilution of the test peptide and IL-23 at a final concentration of 7nM and incubated for 2 hours at room temperature. After washing the wells, bound IL-23 was detected with goat anti-p 40 polyclonal antibody followed by detection with HRP conjugated donkey anti-goat IgG. Signal was visualized with TMB single component HRP membrane substrate and quenched with 2M sulfuric acid.
Kiwi IL-23R competitive binding ELISA
Assay plates were coated with 50 ng/well cynomolgus IL-23R _ huFC and incubated overnight at 4 ℃. The wells were washed, blocked and washed again. To each well was added serial dilutions of the test peptide and IL-23 at a final concentration of 2nM and incubated for 2 hours at room temperature. After washing the wells, bound IL-23 was detected with goat anti-p 40 polyclonal antibody followed by detection with HRP conjugated donkey anti-goat IgG. Signal was visualized with TMB single component HRP membrane substrate and quenched with 2M sulfuric acid.
TABLE E12 Cross-reactivity of exemplary peptide inhibitors
Figure GDA0003395945150002661
+ + + + denotes 0-250nM
+ represents 251-1000nM
+ represents 1001-10,000nM
-represents >25,000nM
Example 5
NK cell analysis
Natural Killer (NK) cells purified from human peripheral blood of healthy donors by negative selection (Miltenyi Biotech, Cat #130-092-657) were cultured in complete medium (RPMI 1640 containing 10% FBS, L-glutamine and penicillin-streptomycin) in the presence of 25ng/mL IL-2(RnD, Cat # 202-IL-010/CF). After 7 days, the cells were centrifuged and resuspended at 1E6 cells/mL in complete medium. Recombinant IL-23 and 10ng/mL IL-18(RnD, Cat # B003-5) of pre-determined EC50 to EC75 were mixed with different concentrations of peptide and added to NK cells seeded at 1E5 cells per well. After 20 to 24 hours, IFN γ in the supernatant was quantified using Quantikine ELISA (RnD, Cat # DIF 50).
Table e13. IC50 of exemplary peptide inhibitors in primary cell lines (NK cell analysis)
Figure GDA0003395945150002671
Figure GDA0003395945150002681
*=<25nM
TABLE 14 IC50(NK cell analysis) of exemplary peptides containing the Ac- [ Pen ] -XXWX- [ Pen ] -XXXXXXX motif (SEQ ID NO:1482) and analogs
Figure GDA0003395945150002682
Figure GDA0003395945150002691
*=<10nM;**=10-25nM
Example 6
Bioanalytical characterization of peptide inhibitors
The potency, cross-reactivity and selectivity of certain peptide inhibitors were determined using various bioassays developed for this purpose and described below.
Rat splenocyte analysis
The new assay developed was a rat splenocyte assay. This assay examined the level of IL-17A in activated rat splenocytes after stimulation with IL-23 in the presence of test compounds.
Briefly, splenocytes freshly isolated from rats were seeded in 96-well tissue culture plates containing complete medium of IL-1 β. Serial dilutions of test compounds were mixed with rat IL-23 at EC50 to EFinal concentrations between C80 values were assigned to each well together; the plates were then incubated at 37 ℃ with 5% CO2Was incubated in the humidified incubator of (1) for 3 days. Changes in IL-17A levels in the supernatants were detected using ELISA.
Rat colitis model: 9-day drinking water containing 3% DSS
There is a large body of evidence in the literature that supports the pathogenic role of IL-23/IL-23R signaling in animal models of colitis. For IL-23 ligands, the demand has been shown in a variety of models, including IL-10 -/-Spontaneous colitis model, Helicobacter hepaticus-driven colitis model, anti-CD 40 congenital colitis model, and chronic CD45RBHigh (a)CD4+T-cell transfer model. The need for IL-23 receptors for colitis development has been shown in acute colitis models induced by DSS or by anti-CD 40, and chronic CD45RBHeight ofCD4+T-cell transfer model. Since certain peptide inhibitors of the invention do not cross-react with the mouse-derived IL-23 receptor but recognize the rat-derived IL-23 receptor, a rat model of IBD associated with the IL-23 pathway was developed.
In this model, colitis was induced in SD rats by ad libitum exposure to 3% DSS in drinking water for 9 days. Three study groups (n ═ 6 rats/group) were compared: disease Activity Index (DAI) scores and colon weight to colon length ratios of vehicle, 3% DSS and 3% DSS to positive control (sulfasalazine, QD administered at 100mg/kg PO). The DAI score consists of a rating from three parameters including percent weight loss, stool consistency, and quantitative hemoccult score, and a maximum of three units can be achieved. Animals exposed to DSS exhibited significantly increased DAI scores from the fourth day before (compared to vehicle control), with DAI values reaching a peak of about 2.5 at the end of the study (day 9). Treatment of rats exposed to DSS with the positive control (sulfasalazine) reduced the disease score from day 5 (compared to DSS alone). The differences observed in the final ratio of colon weight to colon length were also significant for DSS-induced disease animals treated with and without sulfasalazine.
In vitro Activity and stability
Two peptides (compound a and compound B) were selected for further biological studies (shown below). One containing thioether bonds and the other containing Pen-Pen disulfide bonds. Provided herein are the activity, selectivity and ex vivo stability characteristics of two compounds.
Assays for the selectivity of peptide inhibitors include the human IL-12Rb1 ELISA and the measurement of IL-12 production in PHA-activated human PBMC, which are briefly described below.
Human IL-12R beta 1 ELISA
Assay plates were coated with 100 ng/well human IL-12Rb1_ huFC and incubated overnight at 4 ℃. The wells were washed, blocked and washed again. To each well was added serial dilutions of the test peptide and IL-23 at a final concentration of 2.5nM and incubated for 2 hours at room temperature. After washing the wells, bound IL-23 was detected with goat anti-p 40 polyclonal antibody followed by detection with HRP conjugated donkey anti-goat IgG. Signal was visualized with TMB single component HRP membrane substrate and quenched with 2M sulfuric acid. Data from these analyses are provided herein.
IFN γ production by IL-12 in PHA-activated human PBMC
This assay examines the ability of IL-23R antagonists to neutralize IFN γ protein production in IL-12 stimulated human PBMCs. IL-23R peptide inhibitors specific for the IL-23/IL-23R pathway are not expected to alter the levels of IFN γ produced. Compound a and compound B were tested in this assay and figure 2 provides a graph showing that they did not alter the level of IFN γ produced at most concentrations tested.
Figure GDA0003395945150002711
In vivo Activity
Acute colitis was induced by feeding female Sprague Dawley rats with 3% (wt/vol) DSS dissolved in drinking water. Starting on the same day as DSS for nine days, compound a or B was administered orally at 20mg/kg or 30mg/kg three times daily. Compound A was also administered intraperitoneally at 30mg/kg three times daily. Neutralizing anti-IL-23 p19 antibody was used as a comparator and was administered intraperitoneally at 4mg/kg the same day as and the fifth day after the start of DSS. To quantify the clinically active colitis, the Disease Activity Index (DAI) of each animal was determined daily as the average of three parameters: body weight change (grade 0-3), fecal consistency (grade 0-3), and hemoccult blood (grade 0-3), as shown in table E15. At necropsy, the entire colon from the cecum to the rectum was removed. The length of the colon was measured, rinsed with PBS to remove fecal matter, weighed and cut longitudinally to determine a macroscopic score. The visible lesions of the colon were scored on a scale of 0-3 as shown in table E16.
Table E17 shows that at day 7, treatment with compounds a and B significantly improved the DAI score compared to the vehicle-treated group. Fig. 1 shows the results of DAI values from day 7. In addition, a significant reduction in the ratio of colon weight to colon length and macroscopic score of the colon was also observed (fig. 3). The reduction in inflammation observed with orally delivered peptides was similar to the effect observed with the neutralizing anti-IL 23p19 monoclonal antibody. Statistical significance analysis was compared to vehicle-treated groups and determined using student T-test (GraphPad Prism). Differences were recorded as significant p <0.05, p <0.01, p <0.001, p < 0.0001.
TABLE E15 Scoring of disease Activity index
Score of Percent change in body weight Consistency of manure Hemoccult score
0 Is composed of Is normal Is normal
1 1 to 7 Semi-solid Guaiazulene
2 8 to 15 Loose stool Blood haemorrhage treating medicine
3 >15 Diarrhea (diarrhea) Blood treatment +
TABLE E16 scoring of gross morphological lesions of the colon
Figure GDA0003395945150002721
Figure GDA0003395945150002731
Table e17 disease activity index score and individual parameter scores at day 7, colon weight to length ratio and colon macroscopic score at day 9.
Figure GDA0003395945150002732
Example 7
In vitro analysis and Surface Plasmon Resonance (SPR) analysis
In vitro analysis and SPR were performed to further characterize the exemplary compound, compound C:
Figure GDA0003395945150002733
Ac-Ring- [ [ Abu]-QTWQC]- [ Phe [4- (2-aminoethoxy)]-W-[α-MeLys]-ENG-NH2(SEQ ID NO:259)
Compound C
The analysis described in the previous examples was performed to demonstrate that compound C is a potent, selective and competitive inhibitor of IL-23R, showing IL-23 dependent up-regulation of phospho-STAT 3(pSTAT3) in human DB cells and effective inhibition of IFN γ production in human peripheral blood natural killer (PB NK) cells. Furthermore, compound C was selective, showing little inhibition in cell-free elisa (cell free elisa) of human IL6R, or in IL-12 dependent production of IFNg in PBMC. The data are shown in table E18A below. Compound C also cross-reacted with cynomolgus IL-23R (IC 507 nM) and rat IL-23R (IC5017nM) and inhibited IL-23-dependent IL-17A production in rat splenocytes (IC 50130 nM) (data not shown).
Table e18A. in vitro characterization of Compound C
Figure GDA0003395945150002741
After oral administration (PO) at 20mg/kg to rats, the exposure of compound C was also limited to GI, with an AUC value of 355ug.h/g for the small intestinal mucosa; for colonic mucosa, the AUC value was 77 ug.h/g; and AUC values for plasma of 0.3ug.h/mL, with 40% recovery in the excreta.
Compound C is also stable in various GI fluids and reducing environments, with a SIF half-life >24 hours; an SGF half-life of >24 hours; human intestinal fluid half-life >24 hours, and half-life >2 hours in DTT assay.
SPR experiments were performed using a Biacore 2000 instrument equipped with Biacore CM4 and Xantec HC1500m sensor chips and a T100 optical biosensor. Recombinant human IL-23R _ huFC (RnD) or recombinant human IL-12R β 1_ huFC (RnD) or a mixture of both receptor subunits are captured on an anti-human IgG surface. Recombinant human IL-23(Humanzyme) or Compound C was used as the analyte. Fitting SPR sensorgrams to a one-to-one interaction model yields the binding rate constant (k) of the complexon) Dissociation rate constant (k)off) And dissociation constant (K)D) As shown in table E18B. The data show that compound C does not bind to IL-12R β 1, and binds to IL-23R and the mixed surface of IL-12R β 1 and IL-23R with similar potency of 2.42nM and 2.56nM, respectively. The affinity for IL-23R is comparable to that from IL-23. In contrast, the affinity of IL-23 to the mixing surface was about 14x stronger than that from compound C.
TABLE E18B. binding characteristics of IL-23 and Compound C to IL-12R β 1, IL-23R, or mixed IL-12R β 1 and IL-23 as determined by SPR.
Figure GDA0003395945150002751
Example 8
Efficacy of IL-23R antagonists in TNBS-induced colitis in rats
To further evaluate the efficacy of IL-23R antagonists in animal disease models, acute colitis was induced by: on day 0, 7-week old female Sprague-Dawley rats were provided with an intrarectal administration of 45% -50% ethanol (TNBS/ethanol) containing 60mg/ kg 2,4, 6-trinitrobenzenesulfonic acid (TNBS). Compound C (described in example 7) was administered orally at 20mg/kg or 6.7mg/kg three times a day and provided in drinking water at 0.6mg/mL or 0.2mg/mL, respectively, starting approximately 24 hours (day-1) before TNBS vaccination for 8 days. Neutralizing anti-IL-23 p19 antibody was used as a comparator and was administered intraperitoneally at 4mg/kg on days-1 and 3. All animals received orally a PBS (pH 7.4) vehicle used to formulate compound C. The study design is shown in figure 5.
To assess the extent of the inflammatory response, animals were observed daily for clinical signs including percent weight loss and signs of loose or diarrhea. Six days after TNBS inoculation, rats were sacrificed and the entire colon length and colon weight from cecum to rectum were recorded for each animal. The severity of colitis was assessed by a pathologist blinded to the nature of treatment. Gross colon lesions were scored according to a scale of 0-4 as described in table E19 below, in addition to colon wall thickness, and histopathological scores were determined based on the parameters below (table E20 and table E21).
Table E19: definition of colon Macro-Scoring
Score of General morphology of the colon
0 Is normal
1 Erythema
2 Erythema, mild edema, minor erosion
3 Bleeding ulcer, inflammation, and moderate adhesion at two or more sites
4 Severe ulcer, dilated stenosis, severe adhesion
Table E20: definition of histopathology
Figure GDA0003395945150002761
Table E21: scoring criteria
Figure GDA0003395945150002762
Figure GDA0003395945150002763
Figure GDA0003395945150002771
Score of Gland loss
0 Lossless, normal crypt epithelium and mucosa
0.5 Very little loss, no more than 1-2 areas of affected mucosa/gland
1 Very small, 1-10% of the mucosal/glandular area affected
2 Mild, 11-25% of the mucosal/glandular area affected
3 Moderate, 26-50% of the mucosal/glandular area affected
4 It is evident that 51-75% of the mucosal/glandular area is affected
5 In the severe case of the disease, the side chain,>75% of the mucosal/glandular area is affected
Score of Thickness of colon
0 Normal ═<350 micron or less
0.5 Very small 400 microns
1 Very small 400-
2 501-
3 Medium 601-700 mu m
4 Obviously 701 and 800 microns
5 Severe ═ severe>801 micron
Rats challenged with TNBS experienced dramatic weight loss, showing increased incidence of loose stools, and increased colon weight to length ratio compared to sham. These data were confirmed by macroscopic examination of the colon, which revealed mild colon lesions characterized by erythema, edema, and small erosions. Treatment with compound C attenuated these changes compared to the TNBS colitis group. At high doses, compound C was significantly effective in reducing colon weight to length ratio, reducing colon wall thickness, and more importantly, improving the gross pathological score of the colon to normal in 70% of the animals. In all of the above indications, except for colon wall thickness (although the trend is significant), statistical significance was observed at low doses. The reduction in inflammation observed with orally delivered compound C was similar to the effect observed from neutralizing the anti-IL-23 p19 monoclonal antibody (figure 6).
Histological examination of the H & E stained distal colon showed that most of the lesions observed from the vehicle group were transmural and characterized by necrosis of inflammatory cells traversing the entire thickness of the colon, the presence of necrotic tissue debris at the luminal surface, and a crypt free of mucosa. Animals treated with compound C often showed localized lesions localized to mucosal and submucosal regions, with colonic tissue showing potential signs of healing at the site of necrosis. Specifically, animals treated with 160mg/kg/d of compound C showed a significant reduction in inflammation, mucosal necrosis and colon wall thickness, resulting in a significant reduction in overall histological score, comparable to that obtained from the anti-IL-23 p19 antibody control (figure 7).
Concentration analysis of samples collected 1 hour after the last PO dose showed that the plasma concentration of compound C detected from all animals was 2X lower than the IC75 of compound, IC75 as determined in rat splenocyte/IL-17A cell based assays or in rat IL-23R ELISA, suggesting that the efficacy observed from oral treatment is most likely due to its local activity in the colon (see figure 8). Collectively, these data highlight the protective role of IL-23R antagonists in the development of TNBS colitis.
These studies indicate that the peptides of the invention are potent, selective, and orally effective IL-23R peptide antagonists, which are promising therapeutic agents for the treatment of IBD and other disorders. As shown herein, the present invention provides peptides that: it is an effective blocker of IL-23/IL-23R signaling in human cell lines and human primary cells; is selective for IL-23R and does not inhibit binding to IL-6R or inhibit signal transduction by IL-12R; cross-reactive against rat and cynomolgus homologs, but not with mouse homologs, enabling in vivo studies in these species; resistance to the proteolytic and reductive environment of the GI, resulting in high drug levels in intestinal tissues and limited drug concentrations in circulation, providing potential safety advantages compared to systemically delivered therapeutics; and was effective in attenuating TNBS-induced colitis in the rat colitis model and was most similar in activity by GI-limitation, as the anti-IL 23p19 monoclonal antibody.
Example 9
In vitro characterization of exemplary IL-23R antagonists
To evaluate the efficacy properties, SEQ ID NO: 980 (peptide 980), 993 (peptide 993) and 1185 (peptide 1185) to determine the efficacy, selectivity and stability of the peptides. Peptides directed against IL in human (Hu), cynomolgus monkey (Cyno) and Rat (Rat) as measured by quantitative ELISA for IL-23/IL-23R competitive binding assays (performed as described in example 2 above) IC of binding of-23 and IL-23R50The values are shown in Table E22. The efficacy of the peptides was also assessed by IL-23R activity assay, as described above. By reduction of phosphorylation-STAT 3(pSTAT3) levels in IL-23 exposed human DB cells (Hu DB cells (pSTAT 3); performed as described in example 2 above); reduction of IFN γ production by human NK cells exposed to IL-23 (Hu NK cells; performed as described in example 5 above); and determining the IC of the peptides by the reduction of IL-17A produced by activated splenocytes exposed to IL-23 (rat (spleen); performed as described in example 6 above)50The value is obtained. By measuring the IC of peptides that inhibit the human IL-23/IL-12 Rbeta 1 interaction (see example 4) or the human IL-6/IL-6R interaction (see example 6)50To evaluate selectivity. Peptide stability was determined by measuring the half-life of peptides exposed to Simulated Intestinal Fluid (SIF), Simulated Gastric Fluid (SGF) or human intestinal fluid (SIF).
TABLE E22 Properties of exemplary IL-23R antagonist peptides
Figure GDA0003395945150002791
The structure of peptide 993 is shown below:
Figure GDA0003395945150002801
Ac-[(D)-Arg]-cyclo [ [ Abu ]]-QTWQC]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN-NH2(SEQ ID NO:993)
The structure of peptide 1185 is shown below:
Figure GDA0003395945150002802
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran ]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1185)
The structure of peptide 980 is shown below:
Figure GDA0003395945150002811
Ac-Ring- [ [ Abu]-QTWQC]- [ Phe [4- (2-aminoethoxy)]]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN-NH2(SEQ ID NO:980)
The results summarized in Table E22 indicate that the peptides effectively and selectively inhibit IL-23/IL-23R binding compared to IL-23/IL-12R β 1 or IL-6/IL-6R interactions. This inhibition was confirmed by cell culture assays measuring IL-23R-dependence, such as STAT3 phosphorylation, IFN γ production, and IL-17A production. The peptides were also determined to have high stability when exposed to simulated intestinal fluid, simulated gastric fluid, or human intestinal fluid.
Data from the human DB cell (pSTAT3) experiment was used for Schild analysis (see figure 17). For the Schild assay, peptide 993 was tested at concentrations of 0.3nM, 1nM, 3nM, 10nM, 30nM, 100 nM. The Schild slope was determined to be 1.068, indicating that peptide 993 behaves as a simple competitive antagonist. Also to a polypeptide having the sequence of SEQ ID NO: the Schild analysis was performed on a peptide of structure 1169, which is highly similar to the structure of peptide 1185. The Schild slope was determined to be 0.91, indicating that peptides with similar structures, including peptide 1185, may behave as simple competitive antagonists. Also for SEQ ID NO: 1211 is subjected to Schild analysis, and has a structure similar to that of peptide 980. The Schild slope was determined to be 0.76. However, when the slope is fixed to 1, R 2The value was 0.975. These data indicate that peptides with similar structures, including peptide 1185, may behave as simple competitive antagonists.
Example 10
In vivo pharmacokinetics of exemplary IL-23R antagonists
The pharmacokinetic properties of exemplary peptides were measured in vivo. Sprague Dawley rats were administered 10mg/kg of peptide 993 (P.O.).
A single oral dose of peptide 993 was administered to normal female Sprague-Dawley rats (3 rats per time point) with or without ad libitum provision of drinking water doses (see figure 11). Exposure of peptide 993 to plasma was determined at 0.25, 0.5, 1, 3, 6, 8, and 24 hours post-dose following oral dosing. Levels of peptide 993 were also determined in the small intestine, colon, small intestinal mucosa, colonic mucosa, small intestinal mucus, peyer's patches and Mesenteric Lymph Nodes (MLNs) at 1, 3, 6, 8 and 24 hours post-administration. Urine and feces were collected at 6 and 24 hours to determine excretion of peptide 993. Plasma, fecal and tissue samples were stored at-80 ± 10 ℃ prior to analysis. For plasma, compounds were extracted by protein precipitation using 50 μ L samples using a quenching solution with internal standards (MeOH: ACN w/0.1% formic acid, 50:50 volumes). For the excrements, the samples were homogenized with a 0.1% aqueous formic acid solution (ratio of water to excrements 4: 1) before extraction. Compounds were extracted by protein precipitation using 50. mu.L of fecal homogenate using a quenching solution with internal standards (MeOH: ACN w/0.1% formic acid, 50:50 vol). For tissues such as the colon or small intestine, the samples were homogenized with 0.1% formic acid in water (3: 1 ratio of water to tissue) prior to extraction. For tissues such as peyer's patches and mesenteric lymph nodes, the samples were homogenized with 0.1% aqueous formic acid (water: tissue ratio 20:1) prior to extraction. Compounds were extracted by protein precipitation using 50. mu.L of tissue homogenate using a quench solution with internal standards (MeOH: ACN w/0.1% formic acid, 50:50 vol.). Precipitated proteins were removed by filtration plate and the collected supernatant was dried and redissolved. The treated samples were analyzed on an AB/MDS Sciex API 4000 mass spectrometer. Positive ions were monitored in Multiple Reaction Monitoring (MRM) mode. Quantification was performed as peak area ratio.
No detectable levels of peptide 993 were observed in rat plasma 0-24 hours after administration (see fig. 11A). In contrast, detectable levels of peptide 993 were present in peyer's patches and small intestine for at least 6 hours (see fig. 11B and 11C), and at least 8 hours after colonic administration (see fig. 11D). At 24 hours post-administration, levels greater than 5% of the total administered dose of peptide 993 were detected in rat feces, further indicating that peptide 993 is highly orally stable. Taken together, these results demonstrate that peptide 993 is an orally stable GI-restricted peptide that exhibits high GI content and limited systemic distribution following oral administration.
Sprague Dawley rats were administered 10mg/kg of peptide 1185 (P.O.). A single oral dose of peptide 1185 was administered to normal female Sprague-Dawley rats (3 rats per time point). Following oral dosing, samples taken up to 8 hours after dosing were assayed for exposure of peptide 1185 in plasma. Several hours of urine and feces were collected to determine excretion of peptide 1185 (fig. 18).
Sprague Dawley rats were administered 10mg/kg of peptide 980 (P.O). A single oral dose of peptide 980 was administered to normal female Sprague-Dawley rats (3 rats per time point). Following oral dosing, exposure of peptide 980 to plasma was determined in samples taken up to 8 hours after dosing. Several hours of urine and feces were collected to determine excretion of peptide 980 (fig. 19).
Example 11
Safety profiles for exemplary IL-23R antagonists
A safety profile of exemplary peptide inhibitors was characterized. Peptides 993 and 1185 were evaluated in the safety group which examined binding to the 44 target groups. Targets include G protein-coupled receptors (GPCRs), transporters (e.g., dopamine transporters (DAT)), and ion channels. These peptides showed no activity for all targets, as defined by a change (inhibition or stimulation) in target activity of greater than 25%. Table E24 lists the targets tested in the safety group. Peptides 993 and 1185 were determined to be inactive at concentrations up to 10 μ M for each target. The safety profile of peptide 980 was evaluated by test compounds selected from table E24. For all compounds tested, peptide 980 showed only moderate activity (33%) in the acetylcholinesterase assay, and did not otherwise show any activity, as defined by a change in target activity of greater than 25%.
TABLE E24 peptide 993 and peptide 1185 are inactive in the safety group
Figure GDA0003395945150002831
Figure GDA0003395945150002841
Example 12
Effect of exemplary peptide inhibitors in rat model of acute colitis
To evaluate the efficacy of the exemplary peptide inhibitor peptide 1185 in an animal disease model, acute colitis was induced by providing 7-week-old female Sprague-Dawley rats with intrarectal administration of 64mg/kg TNBS in 50% ethanol on day 0. Peptide 1185 (in combination with PO and in drinking water), PO BID, was administered orally at 37 mg/kg/day on days-1 to 7. A sham-operated group not exposed to TNBS and a TNBS-treated group receiving vehicle treatment were used as control groups. For the comparison agent, neutralizing anti-IL-23 p19 antibody was administered intraperitoneally at 4mg/kg on day-1 and again intraperitoneally on day 3 and prednisolone at 10 mg/day p.o. All animals received water orally as a vehicle for formulation of peptide 993.
As noted above, the animals were observed daily for clinical signs including percent weight loss and signs of loose stools or diarrhea. Rats were sacrificed 6 days after TNBS inoculation and the total colon length and colon weight were recorded for each animal. The severity of colitis was assessed by a pathologist. In addition to colon wall thickness, gross colon lesions were scored on a scale of 0-5 according to table E29, and histopathological scores were determined based on the parameters listed in table E30.
Treatment with peptide 1185 significantly reduced some of the disease parameters observed in the TNBS rat model of acute colitis. Rats exposed to TNBS and treated with vehicle experienced weight loss as rats in the sham group continued to increase in weight throughout the study. Oral treatment with prednisolone or systemic treatment with anti-IL-23 p19 prevented weight loss in TNBS-exposed rats. Treatment with orally administered peptide 1185 did not significantly prevent weight loss in TNBS challenged rats (see figure 13). A significant decrease in the ratio of colon weight to colon length was also observed after treatment with prednisolone or anti-IL-23 p19 compared to vehicle treatment. Oral administration of peptide 1185 resulted in a similar decrease in the ratio of colon weight to colon length and colon macroscopic score in TNBS-exposed rats. A higher macroscopic colon score indicates a higher degree of colon pathology. Colon macroscopic scores were determined by adding scores for adhesions, stenosis, ulcers and colon wall thickness (all significantly reduced by treatment with prednisolone, anti-IL-23 p19 or peptide 1185) compared to vehicle-treated controls. These data indicate that orally administered peptide 1185 has comparable efficacy to systemically administered anti-IL-23 p19 monoclonal antibody.
The pathological features of the histological sections of the colon of rats taken from the sham-operated group, the vehicle group, the anti-IL-23 p19 and the peptide 1185 group were examined. Mucosal inflammation, transmural inflammation, gland loss and erosion were scored according to the criteria listed in table E29. For all of these features, treatment with anti-IL-23 p19 or prednisolone reduced the histopathological score associated with TNBS exposure. Treatment with peptide 1185 did not significantly reduce histopathological scores.
Example 13
Effect of exemplary peptide inhibitors in rat model of acute colitis
To evaluate the efficacy of the exemplary IL-23R peptide inhibitor peptide 993 in an animal disease model, acute colitis was induced by providing 7 week old female Sprague-Dawley rats on day 0 with intrarectal administration of 64mg/kg TNBS in 50% ethanol. Starting approximately 24 hours before TNBS vaccination (day-1), peptide 993 was administered orally at 10mg/kg 2 times per day for a total of 42 mg/kg/day for 8 days. A sham-operated group not exposed to TNBS and a TNBS-treated group receiving vehicle treatment were used as control groups. All animals received water orally as a vehicle for formulation of peptide 993.
As noted above, the animals were observed daily for clinical signs including percent weight loss and signs of loose stools or diarrhea. Rats were sacrificed 6 days after TNBS inoculation and the total colon length and colon weight were recorded for each animal. The severity of colitis was assessed by a pathologist. In addition to colon wall thickness, gross colon lesions were scored on a scale of 0-5 according to table E29, and histopathological scores were determined based on the parameters listed in table E30.
Treatment with peptide 993 significantly reduced all disease parameters observed in the TNBS rat model of acute colitis. Rats exposed to TNBS and treated with vehicle experienced weight loss as rats in the sham group continued to increase in weight throughout the study. Treatment with orally administered peptide 993 also prevented weight loss in TNBS-challenged rats (see figure 12). In addition, a significant decrease in the ratio of colon weight to colon length was also observed after oral administration of 993 peptide for treatment. A higher colon macroscopic score indicates a higher degree of colon pathology and the colon macroscopic score is significantly reduced by treatment with peptide 993 compared to vehicle-treated controls. Orally administered peptide 993 had comparable efficacy to systemically administered anti-IL-23 p19 monoclonal antibody (used as a positive control). Histopathological scores were significantly reduced in the colon from peptide 993 treated rats compared to the vehicle group.
Example 14
Effect of exemplary peptide inhibitors in rat model of acute colitis
To evaluate the efficacy of the exemplary IL-23R peptide inhibitor peptide 980 in animal disease models, acute colitis was induced by providing 7-week-old female Sprague-Dawley rats on day 0 with intrarectal administration of 64mg/kg TNBS in 50% ethanol. Peptide 980 (in combination with PO and in drinking water), PO BID, was administered orally at 37 mg/kg/day from day-1 to day 7. A sham-operated group not exposed to TNBS and a TNBS-treated group receiving vehicle treatment were used as control groups. All animals received PBS orally as a vehicle for formulation of peptide 980.
As noted above, the animals were observed daily for clinical signs including percent weight loss and signs of loose stools or diarrhea. Rats were sacrificed 6 days after TNBS inoculation and the entire colon length and colon weight were recorded for each animal. The severity of colitis was assessed by a pathologist. In addition to colon wall thickness, gross colon lesions were scored on a scale of 0-5 according to table E29, and histopathological scores were determined based on the parameters listed in table E30. Treatment with peptide 980 significantly reduced all disease parameters observed in the TNBS rat model of acute colitis.
Treatment with orally administered peptide 980 prevented weight loss in TNBS challenged rats (see figure 14). In addition, a significant decrease in the ratio of colon weight to colon length was also observed after oral administration of peptide 980 for treatment. A higher colon macroscopic score indicates a higher degree of colon pathology and the colon macroscopic score is significantly reduced by treatment with peptide 980 compared to vehicle-treated controls (see fig. 14).
The pathological features of the histological sections of the colon of rats taken from the sham-operated group, vehicle group and peptide 980-treated group were examined. Mucosal inflammation, transmural inflammation, gland loss and erosion were scored according to the criteria listed in table E30 (see fig. 14D). Treatment with peptide 980 significantly reduced the sum of histopathological scores compared to vehicle.
Example 15
Levels of biomarkers following treatment with peptide inhibitors in a rat model of acute colitis
Levels of inflammatory markers were detected in the colon. Distal colon tissue samples designated for protein expression analysis were snap-frozen after collection. For protein extraction, the samples were thawed, weighed and homogenized in extraction buffer (PBS supplemented with protease inhibitors, pH 7.2, 3 x volume: weight). The homogenate was centrifuged at 13krpm for 15 minutes at 4 ℃ in total twice to remove debris. The supernatant was stored in multiple aliquots at-80 ℃ and subsequently used for protein expression analysis on ELISA. Total protein in each sample was quantified using BCA assay. The expression of MPO, IL-1. beta., IL-6, IL-17A and IL-22 proteins in distal colon samples was analyzed using a commercially available rat ELISA kit.
Treatment with peptide 993 reduced the levels of inflammatory markers present in the colon. Treatment with peptide 993 reduced the disease defined by (MPO, IL-6 and IL-1. beta.) and biomarkers for IL-23 (IL-22 and IL-17A) compared to vehicle-treated controls (see FIG. 15). These data indicate that administration of peptide 993 in an amount that reduces pathology in vivo also reduces the levels of biomarkers associated with IL-23R activity present in the colon. Treatment with peptide 980 reduced the levels of MPO and IL-22 compared to vehicle-treated controls (see fig. 16). Treatment with peptide 1185 did not significantly reduce the levels of MPO, IL-22 or IL-17A at the doses tested.
TABLE E29 Colon Macro-Scoring
Figure GDA0003395945150002871
Figure GDA0003395945150002881
Table e30a. histopathology-mucosal/submucosal inflammation score
Figure GDA0003395945150002882
TABLE E30b
Figure GDA0003395945150002883
Figure GDA0003395945150002891
TABLE E30c
Figure GDA0003395945150002892
TABLE E30d
Figure GDA0003395945150002893
TABLE E30E
Figure GDA0003395945150002894
TABLE E30f
Figure GDA0003395945150002895
Figure GDA0003395945150002901
Example 16
Characterization of additional peptides to inhibit binding of interleukin-23 to interleukin-23 receptor
Optimization of peptides to identify at low concentrations (e.g., IC 50)<10nM) while exhibiting Gastrointestinal (GI) stability. As described below, certain peptides were tested to identify peptides that inhibit IL-23 binding to human IL-23R and inhibit IL-23/IL-23R functional activity. The peptides tested include peptides containing various cyclization chemistries, including, for example, peptides containing disulfide bonds between two Pen residues and peptides containing thioether bonds. Peptide inhibitors of the invention include, but are not limited to, peptides having any of the structures described herein. In addition, the peptide inhibitors of the invention include those having the same amino acid sequence as the peptides or structures described herein, without the need to have the same or any N-or C-terminal "capping" groups, such as Ac or NH2
The assays performed to determine peptide activity were performed as described in example 2 above. Human ELISA means IL23-IL23R competitive binding assay, rat ELISA means rat IL-23R competitive binding ELISA assay, and pStat3HTRF means DB cell IL-23R pSTAT3 cell assay. The peptides described in table E31 were cyclized via a disulfide bridge formed between two residues in these peptides. The peptides described in table E32 were cyclized via a thioether bond between the indicated amino acid residues. Table E32 provides exemplary structures describing thioether cyclization, which may also be represented in the table by the term "ring", followed immediately by a bracketed ring region.
TABLE E31 exemplary peptides containing Ac- [ Pen ] -XXWX- [ Pen ] -XXXXXXX motif (SEQ ID NO:1482) and analogs
Figure GDA0003395945150002902
Figure GDA0003395945150002911
Figure GDA0003395945150002921
Figure GDA0003395945150002931
Figure GDA0003395945150002941
TABLE E32 IC50 for exemplary peptide inhibitors (thioethers)
Figure GDA0003395945150002942
Ac-Ring- [ [ Abu]-XXWXC]-[Phe(4-OMe)]-[2-Nal]-XXX-NH2(SEQ ID NO:1483)
Figure GDA0003395945150002943
Figure GDA0003395945150002951
10 nM; at ≥ 10 and <100 nM; not less than 100 and not more than 1,000nM
Example 17
Stability of additional peptide inhibitors mimicking intestinal fluid (SIF), mimicking gastric fluid (SGF) and redox conditions
Studies were performed in Simulated Intestinal Fluid (SIF) and Simulated Gastric Fluid (SGF) to evaluate gastric stability of additional peptide inhibitors of the invention. In addition, studies were conducted to evaluate the redox stability of additional peptide inhibitors of the present invention.
TABLE E33 thioether and Dipens
Figure GDA0003395945150002961
Greater than 90 minutes less than or equal to 180 minutes and greater than 90 minutes; 90 minutes or less and greater than 45 minutes >45 min; >10 minutes less than or equal to 45 minutes and greater than 10 minutes; less than 10 minutes.
Example 18
NK cell analysis
Natural Killer (NK) cells purified from human peripheral blood of healthy donors by negative selection (Miltenyi Biotech, catalog #130-092-657) were cultured in complete medium (RPMI 1640 containing 10% FBS, L-glutamine and penicillin-streptomycin) in the presence of 25ng/mL of IL-2(RnD, catalog # 202-IL-010/CF). After 7 days, the cells were centrifuged and resuspended at 1E6 cells/mL in complete medium. Predetermined recombinant IL-23 of EC50 to EC75 and 10ng/mL IL-18(RnD, catalog # B003-5) were mixed with different concentrations of peptide and added to NK cells seeded at 1E5 cells/well. After 20-24 hours, IFN γ in the supernatant was quantified using Quantikine ELISA (RnD, catalog # DIF 50). The results are shown in Table E34. Multiple results shown for a single peptide are from separate analyses.
TABLE E34 Primary cell analysis (thioether and Dipens)
Figure GDA0003395945150002971
Figure GDA0003395945150002981
All of the above U.S. patents, U.S. patent application publications, U.S. patent applications, foreign patents, foreign patent applications and non-patent publications referred to in this specification and/or listed in the application data sheet, are incorporated herein by reference, in their entirety.
From the foregoing it will be appreciated that, although specific embodiments of the invention have been described herein for purposes of illustration, various modifications may be made without deviating from the spirit and scope of the invention. Accordingly, the invention is not limited except as by the appended claims.

Claims (20)

1. A peptide inhibitor of an interleukin-23 receptor, or a pharmaceutically acceptable salt thereof, comprising an amino acid sequence selected from the group consisting of:
Ac-Ring- [ [ Abu]-QTWQC]- [ Phe [4- (2-aminoethoxy)]]-[2-Nal]-[Achc]-ENN-NH2(SEQ ID NO:977);
Ac-Ring- [ [ Abu]-QTWQC]- [ Phe [4- (2-aminoethoxy)]]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN-NH2(SEQ ID NO:980);
Ac-Ring- [ [ Abu]-QTWQC]- [ Phe [4- (2-aminoethoxy)]]-[2-Nal]-[α-MeLeu]-QN-[βAla]-NH2(SEQ ID NO:984);
Ac- (D) Phe-cyclo- [ [ Abu ]]-QTWQC]- [ Phe [4- (2-aminoethoxy)]]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN-NH2(SEQ ID NO:992);
Ac-[(D)Arg]-Ring- [ [ Abu [ ]]-QTWQC]- [ Phe [4- (2-aminoethoxy)]]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN-NH2(SEQ ID NO:993);
Ac-Ring- [ [ Abu]-QTWQC]- [ Phe [4- (2-aminoethoxy)]]-[2-Nal]-[Acpc]-ENN-NH2(SEQ ID NO:1043);
Ac-[(D)Arg]-cyclo [ [ Abu ]]-QTWQC]- [ Phe [4- (2-aminoethoxy) ]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1272);
Ac-R-[(D)Arg]-cyclo [ [ Abu ]]-QTWQC]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN-NH2(SEQ ID NO:1275);
[Alexa488]-[PEG4]-cyclo [ [ Abu ]]-QTWQC]- [ Phe [4- (2-aminoethoxy)]-[2-Na l]- [ 4-amino-4-carboxylic acidTetrahydro-pyrans]-ENN-NH2(SEQ ID NO:1285);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:632)
Ac- [ (D) Phe ] - [ Pen ] -NTWQ [ Pen ] - [ Phe (4-OMe) ] - [2-Nal ] - [ 4-amino-4-carboxy-tetrahydropyran ] - [ Cit ] -NN-NH2(SEQ ID NO: 679);
Ac-[(D)Phe]-[Pen]-NTWQ[Pen]-[Phe(4-OMe)]-[2-Nal]-[Achc]-ENN-NH2(SEQ ID NO:681);
Ac-[Pen]-NTWQ[Pen]-[Phe(CONH2)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:682);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[α-MeLeu]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1183);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[α-MeLeu]-[L ys(Ac)]-N-[βAla]-NH2(SEQ ID NO:1186);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[α-MeLys]-[Ly s(Ac)]-N-[βAla]-NH2(SEQ ID NO:1187);
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-[Lys(Ac)]-N-[βAla]-NH2(SEQ ID NO: 1188); and
Ac-[Pen]-NTW-[Cit]-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1194);
Wherein the peptide inhibitor is cyclized via a thioether bond between Abu and C or a disulfide bond between two Pen, and
wherein 2-Nal is L-2-naphthylalanine, Abu is 2-aminobutyric acid, α -Me-Lys is α -methyl-L-lysine, α -Me-Leu is α -methyl-L-leucine, α -Me-Ser is α -methyl-L-serine, α -Me-Val is α -methyl-L-valine, Achc is 1-aminocyclohexanecarboxylic acid, Acvc is 1-aminocyclopentanecarboxylic acid, Acpc is 1-aminocyclopropylcarboxylic acid, Acbc is 1-aminocyclobutanecarboxylic acid, and Aib is 2-aminoisobutyric acid.
2. The peptide inhibitor or pharmaceutically acceptable salt thereof according to claim 1, wherein the peptide inhibitor or pharmaceutically acceptable salt thereof is:
Ac-Ring- [ [ Abu]-QTWQC]- [ Phe [4- (2-aminoethoxy)]]-[2-Nal]-[acpc]-ENN-NH2(SEQ ID NO:1043);
Ac-Ring- [ [ Abu]-QTWQC]- [ Phe [4- (2-aminoethoxy)]]-[2-Nal]-[achc]-ENN-NH2(SEQ ID NO: 977); or
Ac-Ring- [ [ Abu]-QTWQC]- [ Phe [4- (2-aminoethoxy)]]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN-NH2(SEQ ID NO:980),
Or a pharmaceutically acceptable salt thereof, wherein the peptide inhibitor is cyclized via a thioether bond between Abu and C.
3. The peptide inhibitor or pharmaceutically acceptable salt thereof according to claim 1, wherein the peptide inhibitor or pharmaceutically acceptable salt thereof is:
Ac-Ring- [ [ Abu]-QTWQC]- [ Phe [4- (2-aminoethoxy)]]-[2-Nal]- [ alpha-methyl-L-leucine]-QN-[βAla]-NH2(SEQ ID NO:984) or a pharmaceutically acceptable salt thereof, wherein the peptide inhibitor is cyclized via a thioether bond between Abu and C.
4. The peptide inhibitor or pharmaceutically acceptable salt thereof according to claim 1, wherein the peptide inhibitor or pharmaceutically acceptable salt thereof is:
ac- (D) Phe-cyclo- [ [ Abu ]]-QTWQC]- [ Phe [4- (2-aminoethoxy)]]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN-NH2(SEQ ID NO:992) or a pharmaceutically acceptable salt thereof, wherein the peptide inhibitor is cyclized via a thioether bond between Abu and C.
5. The peptide inhibitor or pharmaceutically acceptable salt thereof according to claim 1, wherein the peptide inhibitor or pharmaceutically acceptable salt thereof is:
Ac-[(D)Arg]-Ring- [ [ Abu [ ]]-QTWQC]- [ Phe [4- (2-aminoethoxy)]]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN-NH2(SEQ ID NO:993) or a pharmaceutically acceptable salt thereof, wherein the peptide inhibitor is via AbCyclization of the thioether bond between u and C.
6. The peptide inhibitor or pharmaceutically acceptable salt thereof according to claim 1, wherein the peptide inhibitor or pharmaceutically acceptable salt thereof is:
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:632);
Or a pharmaceutically acceptable salt thereof, wherein the peptide inhibitor is cyclized via a disulfide bond between Pen.
7. The peptide inhibitor or pharmaceutically acceptable salt thereof according to claim 1, wherein the peptide inhibitor or pharmaceutically acceptable salt thereof is:
Ac-[(D)Phe]-[Pen]-NTWQ[Pen]-[Phe(4-OMe)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-[Cit]-NN-NH2(SEQ ID NO:679);
Ac-[(D)Phe]-[Pen]-NTWQ[Pen]-[Phe(4-OMe)]-[2-Nal]-[Achc]-ENN-NH2(SEQ ID NO: 681); or
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[α-MeLeu]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1183), or a pharmaceutically acceptable salt thereof, wherein the peptide inhibitor is cyclized via a disulfide bond between Pen.
8. The peptide inhibitor or pharmaceutically acceptable salt thereof according to claim 1, wherein the peptide inhibitor or pharmaceutically acceptable salt thereof is:
Ac-[Pen]-NTWQ[Pen]-[Phe(4-CONH2)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:682), or a pharmaceutically acceptable salt thereof, wherein the peptide inhibitor is cyclized via a disulfide bond between Pen.
9. A pharmaceutical composition comprising the peptide inhibitor of claim 1, or a pharmaceutically acceptable salt thereof.
10. Use of an effective amount of the peptide inhibitor of claim 1, or a pharmaceutically acceptable salt thereof, or the pharmaceutical composition of claim 9, in the manufacture of a medicament for treating Inflammatory Bowel Disease (IBD) in a subject.
11. The use of claim 10, wherein the medicament is formulated for oral delivery.
12. The use of claim 10, wherein the IBD is ulcerative colitis.
13. The use of claim 10, wherein the IBD is crohn's disease.
14. The use of claim 10, wherein the IBD is pouchitis resulting from proctocotomy and ileoanal anastomosis.
15. The use of claim 10, wherein the peptide inhibitor or pharmaceutically acceptable salt thereof is:
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:632);
Ac-[(D)Phe]-[Pen]-NTWQ[Pen]-[Phe(4-OMe)]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-[Cit]-NN-NH2(SEQ ID NO:679);
Ac-[(D)Phe]-[Pen]-NTWQ[Pen]-[Phe(4-OMe)]-[2-Nal]-[Achc]-ENN-NH2(SEQ ID NO:681);
Ac-[Pen]-NTWQ[Pen]-[Phe(4-CONH2)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO: 682); or
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[α-MeLeu]-[Lys(Ac)]-NN-NH2(SEQ ID NO:1183),
Or a pharmaceutically acceptable salt thereof, wherein the peptide inhibitor is cyclized through a disulfide bond between two Pen.
16. The use of claim 10, wherein the peptide inhibitor or pharmaceutically acceptable salt thereof is:
Ac-Ring- [ [ Abu]-QTWQC]- [ Phe [4- (2-aminoethoxy)]]-[2-Nal]-[Achc]-ENN-NH2(SEQ ID NO:977);
Ac-Ring- [ [ Abu ]-QTWQC]- [ Phe [4- (2-aminoethoxy)]]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN-NH2(SEQ ID NO:980);
Ac-Ring- [ [ Abu]-QTWQC]- [ Phe [4- (2-aminoethoxy)]]-[2-Nal]-[α-MeLeu]-QN-[βAla]-NH2(SEQ ID NO:984);
Ac- (D) Phe-cyclo- [ [ Abu ] -QTWQC ] - [ Phe [4- (2-aminoethoxy) ] ] - [2-Nal ] - [ 4-amino-4-carboxy-tetrahydropyran ] -ENN-NH2(SEQ ID NO: 992);
Ac-[(D)Arg]-cyclo- [ [ Abu]-QTWQC]- [ Phe [4- (2-aminoethoxy)]]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN-NH2(SEQ ID NO: 993); or
Ac-Ring- [ [ Abu]-QTWQC]- [ Phe [4- (2-aminoethoxy)]]-[2-Nal]-[acpc]-ENN-NH2(SEQ ID NO:1043),
Or a pharmaceutically acceptable salt thereof, wherein the peptide inhibitor is cyclized via a thioether bond between Abu and Cys.
17. The use of claim 10, wherein the peptide inhibitor or pharmaceutically acceptable salt thereof is:
Ac-[Pen]-NTWQ-[Pen]- [ Phe [4- (2-aminoethoxy)]-[2-Nal]-[Aib]-[Lys(Ac)]-NN-NH2(SEQ ID NO:632), or a pharmaceutically acceptable salt thereof, wherein the peptide inhibitor is cyclized through a disulfide bond between two Pen.
18. The use of claim 10, wherein the peptide inhibitor or pharmaceutically acceptable salt thereof is:
Ac-Ring- [ [ Abu]-QTWQC]- [ Phe [4- (2-aminoethoxy)]]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN-NH2(SEQ ID NO:980), or a pharmaceutically acceptable salt thereof, wherein the peptide inhibitor is cyclized via a thioether bond between Abu and Cys.
19. The use of claim 10, wherein the peptide inhibitor or pharmaceutically acceptable salt thereof is:
Ac- (D) Phe-cyclo- [ [ Abu ] -QTWQC ] - [ Phe [4- (2-aminoethoxy) ] ] - [2-Nal ] - [ 4-amino-4-carboxy-tetrahydropyran ] -ENN-NH2(SEQ ID NO:992), or a pharmaceutically acceptable salt thereof, wherein the peptide inhibitor is cyclized via a thioether bond between Abu and Cys.
20. The use of claim 10, wherein the peptide inhibitor or pharmaceutically acceptable salt thereof is:
Ac-[(D)Arg]-Ring- [ [ Abu [ ]]-QTWQC]- [ Phe [4- (2-aminoethoxy)]]-[2-Nal]- [ 4-amino-4-carboxy-tetrahydropyran]-ENN-NH2(SEQ ID NO:993), or a pharmaceutically acceptable salt thereof, wherein the peptide inhibitor is cyclized via a thioether bond between Abu and Cys.
CN201680049562.2A 2015-07-15 2016-07-15 Peptide inhibitors of interleukin-23 receptor and their use to treat inflammatory diseases Active CN108348580B (en)

Applications Claiming Priority (9)

Application Number Priority Date Filing Date Title
US14/800,627 US9624268B2 (en) 2014-07-17 2015-07-15 Oral peptide inhibitors of interleukin-23 receptor and their use to treat inflammatory bowel diseases
US14/800,627 2015-07-15
PCT/US2015/040658 WO2016011208A1 (en) 2014-07-17 2015-07-15 Oral peptide inhibitors of interleukin-23 receptor and their use to treat inflammatory bowel diseases
USPCT/US15/40658 2015-07-15
US201562264820P 2015-12-08 2015-12-08
US62/264,820 2015-12-08
US201662281123P 2016-01-20 2016-01-20
US62/281,123 2016-01-20
PCT/US2016/042680 WO2017011820A2 (en) 2015-07-15 2016-07-15 Peptide inhibitors of interleukin-23 receptor and their use to treat inflammatory diseases

Publications (2)

Publication Number Publication Date
CN108348580A CN108348580A (en) 2018-07-31
CN108348580B true CN108348580B (en) 2022-05-10

Family

ID=57757647

Family Applications (1)

Application Number Title Priority Date Filing Date
CN201680049562.2A Active CN108348580B (en) 2015-07-15 2016-07-15 Peptide inhibitors of interleukin-23 receptor and their use to treat inflammatory diseases

Country Status (23)

Country Link
EP (1) EP3341011A4 (en)
JP (1) JP6858174B2 (en)
KR (1) KR102513978B1 (en)
CN (1) CN108348580B (en)
AU (1) AU2016293619B2 (en)
BR (1) BR112018000691A2 (en)
CA (1) CA2991984A1 (en)
CL (3) CL2018000128A1 (en)
CO (1) CO2018000349A2 (en)
CR (1) CR20180029A (en)
DO (1) DOP2018000010A (en)
EA (1) EA035733B9 (en)
EC (1) ECSP18002929A (en)
HK (2) HK1257747A1 (en)
IL (1) IL256827A (en)
MX (1) MX2018000542A (en)
NI (1) NI201800008A (en)
PE (1) PE20180571A1 (en)
PH (1) PH12018500086A1 (en)
SG (1) SG10201912066SA (en)
SV (1) SV2018005614A (en)
UA (1) UA123772C2 (en)
WO (1) WO2017011820A2 (en)

Families Citing this family (39)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PT3176170T (en) 2012-06-13 2019-02-05 Incyte Holdings Corp Substituted tricyclic compounds as fgfr inhibitors
EP4091624A1 (en) 2013-03-15 2022-11-23 Protagonist Therapeutics, Inc. Hepcidin analogues and uses thereof
PE20152033A1 (en) 2013-04-19 2016-01-21 Incyte Holdings Corp BICYCLE HETEROCYCLES AS FGFR INHIBITORS
CA2949215C (en) 2014-05-16 2023-03-14 Protagonist Therapeutics, Inc. .alpha.4.beta.7 integrin thioether peptide antagonists
KR102482790B1 (en) 2014-07-17 2022-12-29 프로타고니스트 테라퓨틱스, 인코포레이티드 Oral peptide inhibitors of interleukin-23 receptor and their use to treat inflammatory bowel diseases
MX2017004300A (en) 2014-10-01 2017-12-18 Protagonist Therapeutics Inc NOVEL A4ß7 PEPTIDE MONOMER AND DIMER ANTAGONISTS.
US10301371B2 (en) 2014-10-01 2019-05-28 Protagonist Therapeutics, Inc. Cyclic monomer and dimer peptides having integrin antagonist activity
US10851105B2 (en) 2014-10-22 2020-12-01 Incyte Corporation Bicyclic heterocycles as FGFR4 inhibitors
MA41551A (en) 2015-02-20 2017-12-26 Incyte Corp BICYCLIC HETEROCYCLES USED AS FGFR4 INHIBITORS
EP3617205B1 (en) 2015-02-20 2021-08-04 Incyte Corporation Bicyclic heterocycles as fgfr inhibitors
US10787490B2 (en) 2015-07-15 2020-09-29 Protaganist Therapeutics, Inc. Peptide inhibitors of interleukin-23 receptor and their use to treat inflammatory diseases
GB2545898B (en) 2015-12-21 2019-10-09 Sutura Therapeutics Ltd Improved drug delivery by conjugating oligonucleotides to stitched/stapled peptides
CA3009834A1 (en) 2015-12-30 2017-07-06 Protagonist Therapeutics, Inc. Analogues of hepcidin mimetics with improved in vivo half lives
US10407468B2 (en) 2016-03-23 2019-09-10 Protagonist Therapeutics, Inc. Methods for synthesizing α4β7 peptide antagonists
EP3570864A4 (en) * 2017-01-18 2020-12-30 Protagonist Therapeutics, Inc. Peptide inhibitors of interleukin-23 receptor and their use to treat inflammatory diseases
AR111960A1 (en) 2017-05-26 2019-09-04 Incyte Corp CRYSTALLINE FORMS OF A FGFR INHIBITOR AND PROCESSES FOR ITS PREPARATION
GB2563875B (en) * 2017-06-28 2020-08-19 Sutura Therapeutics Ltd Improvements in drug delivery
US10278957B2 (en) 2017-09-11 2019-05-07 Protagonist Therapeutics, Inc. Opioid agonist peptides and uses thereof
WO2019157268A1 (en) 2018-02-08 2019-08-15 Protagonist Therapeutics, Inc. Conjugated hepcidin mimetics
CA3099116A1 (en) 2018-05-04 2019-11-07 Incyte Corporation Salts of an fgfr inhibitor
CN112867716A (en) 2018-05-04 2021-05-28 因赛特公司 Solid forms of FGFR inhibitors and methods for their preparation
TW202019948A (en) * 2018-07-12 2020-06-01 美商領導醫療有限公司 Peptide inhibitors of interleukin-23 receptor and their use to treat inflammatory diseases
WO2020185532A1 (en) 2019-03-08 2020-09-17 Incyte Corporation Methods of treating cancer with an fgfr inhibitor
CN110015978B (en) * 2019-04-29 2021-03-19 康化(上海)新药研发有限公司 Synthesis method of O- [2- [ [ (tert-butyloxycarbonyl) amino ] ethyl ] -N- [ fluorenylmethoxycarbonyl ] -L-tyrosine
WO2021007269A1 (en) 2019-07-09 2021-01-14 Incyte Corporation Bicyclic heterocycles as fgfr inhibitors
CN114341161A (en) * 2019-07-10 2022-04-12 领导医疗有限公司 Peptide inhibitors of interleukin-23 receptor and their use for the treatment of inflammatory diseases
JOP20220083A1 (en) 2019-10-14 2023-01-30 Incyte Corp Bicyclic heterocycles as fgfr inhibitors
US11566028B2 (en) 2019-10-16 2023-01-31 Incyte Corporation Bicyclic heterocycles as FGFR inhibitors
EP3819307A1 (en) 2019-11-07 2021-05-12 CytoKi Pharma ApS Therapeutic derivatives of interleukin-22
CA3162010A1 (en) 2019-12-04 2021-06-10 Incyte Corporation Derivatives of an fgfr inhibitor
CA3163875A1 (en) 2019-12-04 2021-06-10 Incyte Corporation Tricyclic heterocycles as fgfr inhibitors
EP4090670A1 (en) 2020-01-15 2022-11-23 Janssen Biotech, Inc. Peptide inhibitors of interleukin-23 receptor and their use to treat inflammatory diseases
JP7397239B2 (en) * 2020-11-20 2023-12-12 ヤンセン ファーマシューティカ エヌ.ベー. Compositions of peptide inhibitors of interleukin-23 receptors
US11939331B2 (en) 2021-06-09 2024-03-26 Incyte Corporation Tricyclic heterocycles as FGFR inhibitors
AU2022310347A1 (en) * 2021-07-14 2024-02-29 Janssen Biotech, Inc. Bicyclic peptide inhibitors of interleukin-23 receptor
TW202332682A (en) 2021-12-01 2023-08-16 丹麥商西蘭製藥公司 Peptide inhibitors of interleukin-23 receptor
WO2023172648A2 (en) * 2022-03-09 2023-09-14 Glympse Bio, Inc. Fluorogenic substrates for aminopeptidase detection in biofluids
CN114751962B (en) * 2022-03-17 2023-11-07 北京大学 Staple peptide, preparation method and pharmaceutical application thereof
WO2024003313A1 (en) 2022-06-30 2024-01-04 Sanofi New peptides as selective il-23 receptor antagonists

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101990547A (en) * 2007-07-06 2011-03-23 瓦勒瑞萨欣Hsj有限合伙公司 Il-23 receptor antagonists and uses thereof

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5942492A (en) * 1996-11-12 1999-08-24 Angstrom Pharmaceuticals, Inc. Cyclic peptides that bind to urokinase-type plasminogen activator receptor
KR20090119876A (en) 2007-02-15 2009-11-20 인디애나 유니버시티 리서치 앤드 테크놀로지 코퍼레이션 Glucagon/glp-1 receptor co-agonists
CA2707840A1 (en) 2007-08-20 2009-02-26 Allozyne, Inc. Amino acid substituted molecules
US9523073B2 (en) 2010-03-31 2016-12-20 Medical Diagnostic Laboratories, Llc Elisa for a naturally-occurring soluble truncated form of IL-23 receptor
US9169292B2 (en) * 2011-06-14 2015-10-27 Medical Diagnostic Laboratories, Llc Polypeptides that bound to IL-23 receptor and inhibit binding of IL-23 and cell signaling thereof
US8946150B2 (en) 2011-06-14 2015-02-03 Medical Diagnostic Laboratories, LLC. Polypeptides that bound to IL-23 receptor and inhibit binding of IL-23 and cell signaling thereof
CA2926685A1 (en) * 2013-10-09 2015-04-16 Synergy Pharmaceuticals, Inc. Agonists of guanylate cyclase useful for downregulation of pro-inflammatory cytokines
KR102482790B1 (en) 2014-07-17 2022-12-29 프로타고니스트 테라퓨틱스, 인코포레이티드 Oral peptide inhibitors of interleukin-23 receptor and their use to treat inflammatory bowel diseases

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101990547A (en) * 2007-07-06 2011-03-23 瓦勒瑞萨欣Hsj有限合伙公司 Il-23 receptor antagonists and uses thereof

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Specific targeting of the IL-23 receptor, using a novel small peptide noncompetitive antagonist, decreases the inflammatory response;C. Quiniou等;《Am J Physiol Regul Integr Comp Physiol》;20140820;第307卷(第10期);第R1216-R1230页 *

Also Published As

Publication number Publication date
KR102513978B1 (en) 2023-03-27
HK1257747A1 (en) 2019-10-25
AU2016293619A1 (en) 2018-02-01
EA201890325A1 (en) 2018-06-29
JP2018522008A (en) 2018-08-09
CN108348580A (en) 2018-07-31
DOP2018000010A (en) 2018-03-30
ECSP18002929A (en) 2018-03-31
EP3341011A4 (en) 2019-02-20
WO2017011820A2 (en) 2017-01-19
JP6858174B2 (en) 2021-04-14
WO2017011820A3 (en) 2017-02-23
PE20180571A1 (en) 2018-04-04
BR112018000691A2 (en) 2018-09-18
SG10201912066SA (en) 2020-02-27
EA035733B9 (en) 2021-01-14
CA2991984A1 (en) 2017-01-19
UA123772C2 (en) 2021-06-02
NI201800008A (en) 2018-06-29
PH12018500086A1 (en) 2018-07-30
CL2018003322A1 (en) 2019-01-18
KR20180030663A (en) 2018-03-23
CL2021000343A1 (en) 2021-08-06
HK1259149A1 (en) 2019-11-29
AU2016293619B2 (en) 2021-02-25
EA035733B1 (en) 2020-07-31
IL256827A (en) 2018-03-29
CO2018000349A2 (en) 2018-07-10
CR20180029A (en) 2018-06-05
EP3341011A2 (en) 2018-07-04
CL2018000128A1 (en) 2018-06-15
MX2018000542A (en) 2018-06-27
SV2018005614A (en) 2018-05-22

Similar Documents

Publication Publication Date Title
CN108348580B (en) Peptide inhibitors of interleukin-23 receptor and their use to treat inflammatory diseases
US11884748B2 (en) Oral peptide inhibitors of interleukin-23 receptor and their use to treat inflammatory bowel diseases
US10787490B2 (en) Peptide inhibitors of interleukin-23 receptor and their use to treat inflammatory diseases
US20200207822A1 (en) Peptide inhibitors of interleukin-23 receptor and their use to treat inflammatory diseases
US20190270786A1 (en) Peptide inhibitors of interleukin-23 and their use to treat inflammatory diseases
AU2021209086A1 (en) Peptide inhibitors of interleukin-23 receptor and their use to treat inflammatory diseases
CN114341161A (en) Peptide inhibitors of interleukin-23 receptor and their use for the treatment of inflammatory diseases
US20240132558A1 (en) Peptide inhibitors of interleukin-23 receptor and their use to treat inflammatory diseases

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 1259149

Country of ref document: HK

GR01 Patent grant
GR01 Patent grant