WO2022009698A1 - Peptide et complexe le comprenant - Google Patents

Peptide et complexe le comprenant Download PDF

Info

Publication number
WO2022009698A1
WO2022009698A1 PCT/JP2021/024125 JP2021024125W WO2022009698A1 WO 2022009698 A1 WO2022009698 A1 WO 2022009698A1 JP 2021024125 W JP2021024125 W JP 2021024125W WO 2022009698 A1 WO2022009698 A1 WO 2022009698A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
residue
amino acid
arg
peptide
Prior art date
Application number
PCT/JP2021/024125
Other languages
English (en)
Japanese (ja)
Inventor
良雄 林
健太郎 高山
敦彦 谷口
洋一 根岸
史子 伊東
Original Assignee
学校法人東京薬科大学
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 学校法人東京薬科大学 filed Critical 学校法人東京薬科大学
Priority to JP2022535021A priority Critical patent/JPWO2022009698A1/ja
Publication of WO2022009698A1 publication Critical patent/WO2022009698A1/fr

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/69Boron compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/10Peptides having 12 to 20 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • 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

Definitions

  • the present invention relates to peptides and complexes containing them.
  • Muscular dystrophy is a hereditary disease in which degeneration and necrosis of skeletal muscle are the main lesions and muscle weakness progresses.
  • the expression of muscle strength requires a mechanism to transmit the tension generated in the intracellular myofibrils to the extracellular basement membrane via multiple proteins. Defects in the genes that encode the proteins involved in this cause the development of muscular dystrophy. For example, in the most severe Duchenne-type muscular dystrophy, the dystrophin gene is mutated, and it is said that the main cause is deletion or dysfunction of the protein.
  • muscle mass is inhibited by inhibiting the function of myostatin (proliferative differentiation factor-8, GDF-8), which is a factor that negatively controls skeletal muscle mass.
  • GDF-8 proliferation differentiation factor-8
  • Myostatin is a secretory protein that belongs to the TGF- ⁇ family and is highly expressed in skeletal muscle, and is synthesized intracellularly as a precursor protein containing a prodomain on the N-terminal side and a mature domain on the C-terminal side.
  • Myostatin secreted from cells is a propeptide derived from a prodomain called latency assisted protein (LAP), which associates with an active dimer derived from a mature domain and does not eliminate the active dimer, which is the main component that negatively controls skeletal muscle mass.
  • LAP latency assisted protein
  • myostatin stocked in vivo in an inactivated state in this way becomes an active substance by decomposing a propeptide by an enzyme when necessary.
  • the activated myostatin functions as a signal molecule that negatively regulates skeletal muscle mass through binding to a receptor typified by the activin type IIB receptor.
  • WO 2018/030432 (corresponding to US Pat. No. 20130177370) describes a myostatin-inhibiting peptide derived from a myostatin propeptide.
  • the myostatin-inhibiting peptide described in International Publication No. 2018/030432 has high myostatin-inhibiting activity.
  • improvement of stability in vivo is desired.
  • an object of the present invention is to provide a peptide having high myostatin inhibitory activity and improved in vivo stability.
  • the present inventors have conducted diligent research in order to solve the above problems. As a result, it was found that the above-mentioned problem can be solved by a peptide composed of all D-form amino acids, and the present invention has been completed.
  • One embodiment of the present invention relates to a peptide, a pharmaceutically acceptable salt thereof, or a prodrug thereof, which comprises an amino acid sequence represented by the following formula (1) and has 15 to 17 amino acid residues. ..
  • X 0 is an amino acid residue or defect selected from the group consisting of D-Ala, D-Gly and D-2-aminoisobutyric acid
  • X 1 is an amino acid residue or defect selected from the group consisting of D-Leu, D-norleucine, D-Val, D-Ile, D-2-aminobutyric acid, D-norvaline and D-isovaline
  • X 2 is an amino acid residue selected from the group consisting of D-Arg, D-ornithine, D-Lys, D-His, D-2,3-diaminopropionic acid and D-2,4-diaminobutanoic acid.
  • X 3 is an amino acid residue selected from the group consisting of D-2-cyclohexylglycine, D-norleucine, D-Leu, D-Val, D-Ile, D-2-aminobutyric acid, D-norvaline and D-isovaline.
  • X 4 is, D-Lys, D-Arg , D- ornithine, D-His, with an amino acid residue selected from the group consisting of D-2,3-diaminopropionic acid and D-2,4-diaminobutane acid can be;
  • X 5 contains D-Ser, D-Arg, D-2-hydroxyglycine, D-homoserine, D-Lys, D-ornithine, D-His, D-2,3-diaminopropionic acid and D-2,4.
  • X 6 is an amino acid residue selected from the group consisting of D-Trp, D-3- (2-naphthyl) alanine, D-Tyr, D-Phe and D-3- (1-naphthyl) alanine
  • X 7 is an amino acid residue selected from the group consisting of D-2-phenylglycine, D-norleucine, D-Ile, D-Leu, D-Val, D-2-aminobutyric acid, D-norvaline and D-isovaline.
  • X 8 is, D-Gln, D-Arg , D-Asn, D-Lys, D-His, D-2,3- diaminopropionic acid, the group consisting of D-2,4-diaminobutanoic acid and D- ornithine Amino acid residues selected from;
  • X 9 is a D-2-cyclohexylglycine residue;
  • X 10 is an amino acid residue selected from the group consisting of D-Lys, D-Arg, D-ornithine, D-His, D-2,3-diaminopropionic acid and D-2,4-diaminobutanoic acid.
  • X 11 is an amino acid residue selected from the group consisting of D-2-phenylglycine, D-Ile, D-Leu, D-norleucine, D-Val, D-2-aminobutyric acid, D-norvaline and D-isovaline. Is the basis;
  • X 12 is an amino acid residue selected from the group consisting of D-Trp, D-homophenylalanine, D-Tyr and D-Phe;
  • X 13 is an amino acid residue selected from the group consisting of D-Arg, D-ornithine, D-Lys, D-His, D-2,3-diaminopropionic acid and D-2,4-diaminobutanoic acid.
  • X 14 is an amino acid residue selected from the group consisting of D-2-phenylglycine, D-Ile, D-Leu, D-norleucine, D-Val, D-2-aminobutyric acid, D-norvaline and D-isovaline. Is the basis;
  • X 15 is an amino acid residue selected from the group consisting of D-Tyr, D-Trp and D-Phe; and
  • X 16 is from D-Trp, D-homophenylalanine, D-Tyr and D-Phe. Amino acid residues or defects selected from the group consisting of.
  • the peptides of Examples and Comparative Examples show myostatin inhibitory activity.
  • the peptide of the example shows the myostatin inhibitory activity.
  • the peptides of Examples and Comparative Examples show myostatin inhibitory activity.
  • the peptide of the example shows the myostatin inhibitory activity.
  • the results of the stability evaluation of the peptide of the example in a bovine pancreas-derived trypsin solution are shown.
  • the results of the stability evaluation of the peptide of the example in a bovine pancreas-derived ⁇ -chymotrypsin solution are shown.
  • the results of the effect (in vivo evaluation) of the peptide of the example on the Duchenne muscular dystrophy model mdx mouse tibialis anterior muscle are shown.
  • the result of myostatin oxygenation evaluation using the peptide of Example is shown.
  • the results of the effect (in vivo evaluation) on the grip strength of the cancer cachexia model mouse by intramuscular administration of the peptide of the example are shown.
  • the results of the effect (in vivo evaluation) on the gastrocnemius muscle mass of the cancer cachexia model mouse by intramuscular administration of the peptide of the example are shown.
  • the results of the effect (in vivo evaluation) on the muscle fiber area of the cancer cachexia model mouse by intramuscular administration of the peptide of the example are shown.
  • XY indicating a range means "X or more and Y or less”. Unless otherwise specified, the operation and physical properties are measured under the conditions of room temperature (20 to 25 ° C.) / relative humidity of 40 to 50% RH.
  • a peptide containing an amino acid sequence represented by the formula (1) and having 15 to 17 amino acid residues, or a pharmaceutically acceptable salt thereof is simply referred to as “the present invention.” Also called “peptide”.
  • amino acid residue in the present invention means a portion corresponding to one unit of the amino acid constituting the peptide or protein on the peptide or protein molecule. More specifically, it means a divalent group derived from an ⁇ -amino acid, which is represented by the following formula (5):
  • R 0 is a side chain of an amino acid, for example, Gly is a hydrogen atom and Ala is a methyl group.
  • amino acid residues are derived from natural or unnatural ⁇ -amino acids.
  • the peptide according to the present invention is composed of D-form amino acids from the viewpoint of enhancing stability in a living body.
  • amino acid residue is Arg, Lys, Asp, Asn, Glu, Gln, His, Pro, Tyr, Trp, Ser, Thr, Gly, Ala, Met, Cys, Phe, Leu, Val. , And Ile, and their analogs can be exemplified.
  • the analog may be, for example, a derivative in which the side chain of the 20 kinds of amino acid residues is substituted with an arbitrary substituent, and for example, a halogenated derivative of the above 20 kinds of amino acid residues (for example, , 3-Chloroalanine, 4-Fluorophenylalanine, 4-Chlorophenylalanine), 2-aminobutyric acid, 4-aminobutyric acid, norleucine, norvaline, isovalin, 2-aminoisobutyric acid, homophenylalanine, 2,3-diaminopropionic acid, 2 , 4-diaminobutanoic acid, ornithine, 2-hydroxyglycine, homoserine, hydroxylysine, hydroxyproline, 3,4-didehydroproline, homocysteine, homomethionine, aspartic acid ester (eg, aspartic acid-methyl ester, aspartic acid) -Ethyl ester, aspart
  • Natural diastereomers ie, (2R * , 3R * ) for Ile, -2-amino-3-methylpentanoic acid, (2R * , 3S * ) for Thr, -2-amino-3-hydroxybutane Acid
  • amino acid sequence described in the present specification is described in the direction from the N-terminal (amino-terminal) side to the C-terminal (carboxyl-terminal) side according to the convention.
  • each amino acid residue can be replaced with an amino acid residue having similar properties based on the difference in its side chain (conservative substitution).
  • the aliphatic hydrophobic amino acids Val, Leu, Ile, 2-aminobutyric acid (Abu), norleucine (Nle), norvaline (Nva), and isovaline (Iva) can be substituted with each other.
  • Gly, Ala and 2-aminoisobutyric acid (Aib), whose side chains are hydrogen atoms or methyl groups, can be replaced with each other.
  • Phe and homophenylalanine (Hph) whose side chains are phenylalkyl groups, can be substituted with each other.
  • the neutral polar amino acids Asn and Gln can be replaced with each other.
  • the basic amino acids Arg, Lys, His, 2,3-diaminopropionic acid (Dpr), 2,4-diaminobutanoic acid (Dbu), and ornithine (Orn) can be replaced with each other.
  • the acidic amino acids Asp and Glu can be replaced with each other.
  • Ser, 2-hydroxyglycine (Hyg) and homoserine (Hse), whose side chains are hydroxy groups or short hydroxyalkyl groups, can be substituted with each other.
  • Pro and 3,4-didehydroproline (Dhp) having a side chain in which the pyrrolidyl group of the side chain is dehydrogenated can be mutually substituted.
  • Cys and homocysteine (Hcy) whose side chains are short thiolalkyl groups, can be replaced with each other.
  • Met and homomethionine (Hme), whose side chains are short-chain sulfide structures, can be replaced with each other.
  • Trp, Tyr and Phe whose side chains are aromatic, can be replaced with each other.
  • Gly and Pro which are amino acid residues that affect the orientation of the chain, can be replaced with each other.
  • a "pharmaceutically acceptable salt” is a metal salt, ammonium salt, organic acid salt, inorganic acid salt, or organic salt that does not produce an undesired physiological effect after being administered to a patient or subject. It is a salt with a base or an inorganic base.
  • the peptide prodrug according to the present invention is a peptide derivative that converts the peptide according to the present invention, that is, gastric acid or enzyme. It refers to a peptide derivative that undergoes oxidation, reduction, hydrolysis, etc. to change into the peptide according to the present invention.
  • These peptide derivatives are, for example, Bundgard, H. et al. , Design of Prodrugs, pp. It can be produced from the peptide according to the present invention by a conventionally known method described in 7-9, 21-24, Elsevier, Amsterdam 1985 and the like.
  • an ester derivative obtained by reacting the carboxyl group with an alcohol, or by reacting the carboxyl group with an amine can be exemplified. More specifically, for example, an ester in which the carboxyl group of the peptide side chain is represented by -COOR (R is an alkyl group having 1 to 20 carbon atoms), or -CONHR or -CONRR'(R and R'is Independently, a peptide derivatized into an amide group represented by an alkyl group having 1 to 20 carbon atoms can be mentioned.
  • the side chain of the peptide according to the present invention has a hydroxyl group
  • an acyloxy derivative obtained by reacting the hydroxyl group with an acid anhydride or the like to acylate it can be exemplified. More specifically, for example, a peptide obtained by derivatizing the hydroxyl group of the peptide side chain into an acyloxy group represented by ⁇ OCOR (R is an alkyl group having 1 to 20 carbon atoms) can be mentioned.
  • prodrug examples include derivatives in which the amino group is acylated, N-oxidized, alkylated, or phosphorylated when the side chain of the peptide according to the present invention has an amino group. More specifically, for example, a peptide in which the amino group of the side chain is derivatized into an amide group represented by -NHCOR (R is an alkyl group having 1 to 20 carbon atoms) or -NHCOCH (NH 2 ) CH 3. Can be mentioned.
  • -NHCOR R is an alkyl group having 1 to 20 carbon atoms
  • -NHCOCH NH 2
  • the structure of the N-terminal of the peptide according to the present invention is not particularly limited, and may be, for example, a hydrogen atom (that is, unmodified) or a structure in which a modifying group is introduced by a conventionally known method.
  • the N-terminal modifying group includes an alkyl group having 1 to 20 carbon atoms, an alkenyl group having 1 to 20 carbon atoms, an alkynyl group having 1 to 20 carbon atoms, an aromatic hydrocarbon group having 6 to 20 carbon atoms, and a heterocyclic group.
  • a group represented by the following formula (6) a sulfonyl group, a carboxyl group, a glyoxyl group, a formyl group; a polyethylene glycol group (PEGylated), a polyoxyethylene glycol group, a polypropylene glycol group; a tert-butoxycarbonyl group (Boc group).
  • benzyloxycarbonyl group Z group
  • protective group such as fluorenylmethoxycarbonyl group (Fmoc group); cyclopentyloxycarbonyl group, cyclohexyloxycarbonyl group, adamantyloxycarbonyl group, norbornyloxycarbonyl group, iso Cycloalkyloxycarbonyl groups such as Bornyloxycarbonyl groups; Protective groups derived from amino acids such as pyroglutamic acid and morothanic acid; Carbamate-based protective groups; sulfonic acid such as benzenesulfonic acid and protective groups derived from phosphoric acid, etc. , Can be exemplified.
  • the peptide has a hydrogen atom, an alkyl group, an aromatic hydrocarbon group, a heterocyclic group at the N-terminal, a group represented by the following formula (6), a sulfonyl group, a carboxyl group, and the like. It is preferably a glyoxyl group, a formyl group or a polyethylene glycol group, more preferably a hydrogen atom, an acyl group or a polyethylene glycol group, and even more preferably a hydrogen atom.
  • the number of carbon atoms of the alkyl group that can be present at the N-terminal of the peptide is, for example, 1 to 20, preferably 1 to 10.
  • the alkyl group may have a saturated chain, an unsaturated chain, or a cyclic structure, and may have a branched chain structure. More specifically, the alkyl group includes a methyl group, an ethyl group, a propyl group, an isopropyl group, a cyclopropyl group, a butyl group, an isobutyl group, a sec-butyl group, a tert-butyl group, an amyl group, an isoamyl group, and a tert.
  • the carbon number of the aromatic hydrocarbon group that can exist at the N-terminal of the peptide is, for example, 6 to 20, and more specifically, a phenyl group, a naphthyl group, a tolyl group, a phenanthryl group and the like can be exemplified.
  • Heterocyclic groups that can be present at the N-terminal of the peptide include monocyclic, fused bicyclic or fused tricyclic groups containing 1 to 3 heteroatoms selected from nitrogen, oxygen and sulfur atoms in the ring.
  • Examples of the substituent of the structure can be exemplified, and more specifically, a pyrrolidyl group, a pyrrole group, a piperidyl group, a pyridyl group, an imidazolyl group, a pyrazolyl group, an oxazolyl group, a thiazolyl group, a morphoryl group, an indolyl group, a benzoimidazolyl group, a quinolyl group, Examples thereof include a carbazolyl group, a tetrahydrofuranyl group, a tetrahydrothiophenyl group, a furanyl group, a thiophenyl group, a tetrahydropyranyl group, a tetrahydrothiopyranyl group and the like.
  • aromatic hydrocarbon groups and heterocyclic groups are linear or branched alkyl groups having 1 to 6 carbon atoms, linear or branched alkoxy groups having 1 to 6 carbon atoms, amino groups, carboxyl groups and esters. It may be substituted with a group, a carbamoyl group, an amide group, a nitro group, a sulfo group, a sulfonamide group, and / or a further substituent such as a halogen.
  • the N-terminal modifying group may be, for example, a functional group represented by the following formula (6).
  • X 0A is a single bond, an oxygen atom or a sulfur atom, or may have a substituent selected from the group consisting of an amino group, an acetylamino group and a propionylamino group, and is an alkylene having 1 to 3 carbon atoms.
  • Group eg, methylene group, ethylene group, trimethylene group and propylene group
  • oxyalkylene group having 1 to 3 carbon atoms eg, oxymethylene group, oxyethylene group, oxytrimethylene group and oxypropylene group
  • a divalent linking group selected from the group consisting of ⁇ 3 alkyleneoxy groups (eg, methyleneoxy group, ethyleneoxy group, trimethyleneoxy group and propyleneoxy group);
  • R 1A is an alkyl group having 1 to 20 carbon atoms which may have a substituent, as well as an alkyl group having 1 to 20 carbon atoms.
  • R 11 to R 30 are independently hydrogen atom, halogen atom (for example, fluorine atom, chlorine atom, bromine atom, iodine atom) and alkyl group having 1 to 3 carbon atoms (that is, methyl group, ethyl group, propyl group). ), An alkoxy group having 1 to 3 carbon atoms (that is, a methoxy group, an ethoxy group, a propoxy group), a hydroxyl group, and an amino group.
  • halogen atom for example, fluorine atom, chlorine atom, bromine atom, iodine atom
  • alkyl group having 1 to 3 carbon atoms that is, methyl group, ethyl group, propyl group.
  • An alkoxy group having 1 to 3 carbon atoms that is, a methoxy group, an ethoxy group, a propoxy group
  • hydroxyl group and an amino group.
  • R 1A when R 1A is an alkyl group having 1 to 20 carbon atoms which may have a substituent, the fatty chain may have a saturated chain, an unsaturated chain, or a cyclic structure. , May have a branched chain structure.
  • the alkyl group of R 1A preferably has 2 to 12 carbon atoms.
  • the substituent of R 1A includes a hydroxy group, an alkoxy group having 1 to 5 or less carbon atoms (for example, a methoxy group, an ethoxy group, etc.), an amino group, a carboxyl group, an ester group, a carbamoyl group, an amide group, a nitro group, and a sulfo group.
  • Halogen fluorine, chlorine, bromine, iodine
  • R 1A includes a hydroxy group, an alkoxy group having 1 to 5 or less carbon atoms (for example, a methoxy group, an ethoxy group, etc.), an amino group, a carboxyl group, an ester group, a carbamoyl group, an amide group, a nitro group, and a sulfo group.
  • Halogen fluorine, chlorine, bromine, iodine
  • X 0A is a single bond or may have a substituent selected from the group consisting of an amino group and an acetylamino group, and has 1 to 3 carbon atoms. It is a divalent linking group selected from the group consisting of an alkylene group and an oxyalkylene group having 1 to 3 carbon atoms.
  • the group represented by the above formula (6) is an acyl group.
  • Acyl groups include acyl groups derived from various carboxylic acids. More specifically, it may be an acyl group having a fat chain, an aromatic ring or a heterocycle, or a compound selected from the group consisting of an amino acid, a vitamin having an acyl group, and a nucleic acid base having an acyl group. It may be an acyl group to be derived.
  • the acyl group which is an alkyl group having 1 to 20 carbon atoms in which R 1A in the above formula (6) may have a substituent, is an acetyl group, a propionyl group, a butyryl group, or an isobutyryl group.
  • Valeryl group isobarrel group, pivaloyl group, caproyl group, caprinoyle group, methylhexanoyl group, cyclopropanecarbonyl group, aminocyclopropanecarbonyl group, cyclohexanecarbonyl group, cyclohexylacetyl group, cyclopentylpropionyl group, cyclohexylpropionyl group, cyclopentylbuta Noyl group, cyclohexylbutanoyl group, adamantylacetyl group, lauroyl group, myritoyl group, palmitoyl group, stearoyl group, oxalyl group, malonyl group, succinyl group, glutalyl group, adipoyl group, glycol group, lactoyl group, glyceroyl group, pyruboyl group , Acetacetyl group and the like can be exemp
  • vitamins having an acyl group examples include nicotinic acid, pantothenic acid, biotin, pteroylglutamic acid (folic acid), orotic acid, fluoroorotic acid, ⁇ -lipoic acid, pyridoxic acid, biocithin, pteroic acid, and 10-formylpteroin. Acids, 7,8-dihydrofolic acid, homopteroic acid, pterin-6-carboxylic acid, dihydrolipoic acid, hydroorothic acid and the like can be mentioned.
  • the nucleic acid base derivative having an acyl group refers to a base component constituting a nucleotide and a derivative thereof, and preferably a pyrimidine derivative or the like, for example, 5-carboxymethyl uracil, 5-carboxythiouracil and the like can be exemplified.
  • Examples of the sulfonyl group that can exist at the N-terminal of the peptide include those having a structure obtained by converting the carbonyl structure of the above-mentioned acyl group into a sulfone structure.
  • the polyethylene glycol group that may be present at the N-terminal of the peptide is polyethylene glycol or via an ester bond, an amine (-NH-), an acyl group (for example, an acyl group having 1 to 12 carbon atoms), or a combination thereof. It is a structure in which the analogs are linked.
  • the terminal of the polyethylene glycol group opposite to the side connected to the N-terminal of the peptide is an alkyl group having 1 to 6 carbon atoms (for example, a methyl group, an ethyl group, a propyl group, an isopropyl group, a cyclopropyl group, or butyl). Modified with a protective group or amino group commonly used to protect hydroxyl groups, such as group, isobutyl group, sec-butyl group, tert-butyl group, amyl group, isoamyl group, tert-amyl group, hexyl group). It may have been done.
  • the structure of the C-terminal of the peptide according to the present invention is also not particularly limited, and may be a structure modified with a protecting group generally used for protection of a carboxylic acid. More specifically, the structure of the C-terminus of the peptide according to the present invention, for example, a carboxyl group (-COOH), a carboxylate (-COO -), amide (-CONH 2), alkylamide (-CONHR 31, - It may be substituted with a CONR 31 R 32 ), an ester (-COOR 31 ), an acyloxyalkyl (-R 33- OCOR 31 ) such as a pivaloyloxymethyl group, an alkyl group having 1 to 4 carbon atoms or an alkoxy group.
  • a carboxyl group -COOH
  • a carboxylate a carboxylate
  • amide -CONH 2
  • alkylamide -CONHR 31, - It may be substituted with a CONR 31 R 32 ), an
  • a phthalidyl group eg, a phthalidyl group, a dimethylphthalidyl group, a dimethoxyphthalidyl group
  • the C-terminal of the peptide is preferably an amide.
  • R 31 and R 32 in the above alkylamides, esters, and acyloxyalkyls are independently methyl group, ethyl group, propyl group, isopropyl group, cyclopropyl group, butyl group, isobutyl group, sec-butyl group, tert.
  • -Alkyl group having 1 to 6 carbon atoms such as butyl group, amyl group, isoamyl group, tert-amyl group, hexyl group and cyclohexyl group; aryl group having 6 to 10 carbon atoms such as phenyl group and naphthyl; benzyl group and phenethyl An aralkyl group having 7 to 18 carbon atoms such as a group and a benzhydryl group; a sugar such as glucose; an alkyl group having 1 to 6 carbon atoms (for example, a methyl group, an ethyl group, a propyl group, an isopropyl group, a cyclopropyl group, a butyl group, etc.
  • R 33 in the acyloxyalkyl is an alkylene group having 1 to 4 carbon atoms such as a methylene group, an ethylene group, an n-propylene group, an isopropylene group, an n-butylene group, an isobutylene group, an s-butylene group and a t-butylene group. Is.
  • the peptide according to the present invention also includes peptide derivatives chemically modified by covalent bonds with polymers, lipids and the like, and derivatives having further enhanced ⁇ -helix properties contained in the peptides.
  • the derivative having further enhanced ⁇ -helix property include a derivative having a salt bridge formed at the i, i + 4-position and the like as an amino acid arrangement, and a derivative having a crosslinked structure by a disulfide bond, a carbon-carbon bond, and the like.
  • the number of amino acid residues of the peptide according to the present invention is 15 to 17. When the number of amino acid residues is 17 or less, it is advantageous in terms of both synthesis and utilization in vivo. If the number of amino acid residues is less than 14, the effect of the present invention cannot be exerted.
  • One embodiment of the present invention comprises a peptide, a pharmaceutically acceptable salt thereof, or a prodrug thereof, which comprises an amino acid sequence represented by the following formula (1) and has 15 to 17 amino acid residues.
  • X 0 is an amino acid residue or defect selected from the group consisting of D-Ala, D-Gly and D-2-aminoisobutyric acid
  • X 1 is an amino acid residue or defect selected from the group consisting of D-Leu, D-norleucine, D-Val, D-Ile, D-2-aminobutyric acid, D-norvaline and D-isovaline
  • X 2 is an amino acid residue selected from the group consisting of D-Arg, D-ornithine, D-Lys, D-His, D-2,3-diaminopropionic acid and D-2,4-diaminobutanoic acid.
  • X 3 is an amino acid residue selected from the group consisting of D-2-cyclohexylglycine, D-norleucine, D-Leu, D-Val, D-Ile, D-2-aminobutyric acid, D-norvaline and D-isovaline.
  • X 4 is, D-Lys, D-Arg , D- ornithine, D-His, with an amino acid residue selected from the group consisting of D-2,3-diaminopropionic acid and D-2,4-diaminobutane acid can be;
  • X 5 contains D-Ser, D-Arg, D-2-hydroxyglycine, D-homoserine, D-Lys, D-ornithine, D-His, D-2,3-diaminopropionic acid and D-2,4.
  • X 6 is an amino acid residue selected from the group consisting of D-Trp, D-3- (2-naphthyl) alanine, D-Tyr, D-Phe and D-3- (1-naphthyl) alanine
  • X 7 is an amino acid residue selected from the group consisting of D-2-phenylglycine, D-norleucine, D-Ile, D-Leu, D-Val, D-2-aminobutyric acid, D-norvaline and D-isovaline.
  • X 8 is, D-Gln, D-Arg , D-Asn, D-Lys, D-His, D-2,3- diaminopropionic acid, the group consisting of D-2,4-diaminobutanoic acid and D- ornithine Amino acid residues selected from;
  • X 9 is a D-2-cyclohexylglycine residue;
  • X 10 is an amino acid residue selected from the group consisting of D-Lys, D-Arg, D-ornithine, D-His, D-2,3-diaminopropionic acid and D-2,4-diaminobutanoic acid.
  • X 11 is an amino acid residue selected from the group consisting of D-2-phenylglycine, D-Ile, D-Leu, D-norleucine, D-Val, D-2-aminobutyric acid, D-norvaline and D-isovaline. Is the basis;
  • X 12 is an amino acid residue selected from the group consisting of D-Trp, D-homophenylalanine, D-Tyr and D-Phe;
  • X 13 is an amino acid residue selected from the group consisting of D-Arg, D-ornithine, D-Lys, D-His, D-2,3-diaminopropionic acid and D-2,4-diaminobutanoic acid.
  • X 14 is an amino acid residue selected from the group consisting of D-2-phenylglycine, D-Ile, D-Leu, D-norleucine, D-Val, D-2-aminobutyric acid, D-norvaline and D-isovaline. Is the basis;
  • X 15 is an amino acid residue selected from the group consisting of D-Tyr, D-Trp and D-Phe; and
  • X 16 is from D-Trp, D-homophenylalanine, D-Tyr and D-Phe. Amino acid residues or defects selected from the group consisting of.
  • the peptide according to the present invention has high myostatin inhibitory activity, and in addition, since it is composed of all D-form amino acids, it has stability against in vivo enzymes and is effective for living organisms. It can show a long-lasting high effect at the time of administration.
  • X 0 is an amino acid residue or defect selected from the group consisting of D-Ala, D-Gly and D-2-aminoisobutyric acid
  • X 1 is an amino acid residue or defect selected from the group consisting of D-Leu, D-norleucine, D-Val, D-Ile, D-2-aminobutyric acid, D-norvaline and D-isovaline
  • X 2 is an amino acid residue selected from the group consisting of D-Arg, D-ornithine, D-Lys, D-His, D-2,3-diaminopropionic acid and D-2,4-diaminobutanoic acid.
  • X 3 is an amino acid residue selected from the group consisting of D-2-cyclohexylglycine, D-norleucine, D-Leu, D-Val, D-Ile, D-2-aminobutyric acid, D-norvaline and D-isovaline.
  • X 4 is, D-Lys, D-Arg , D- ornithine, D-His, with an amino acid residue selected from the group consisting of D-2,3-diaminopropionic acid and D-2,4-diaminobutane acid can be;
  • X 5 contains D-Ser, D-Arg, D-2-hydroxyglycine, D-homoserine, D-Lys, D-ornithine, D-His, D-2,3-diaminopropionic acid and D-2,4.
  • -Amino acid residues selected from the group consisting of diaminobutanoic acid
  • X 6 is an amino acid residue selected from the group consisting of D-Trp, D-3- (2-naphthyl) alanine, D-Tyr, D-Phe and D-3- (1-naphthyl) alanine.
  • X 7 is an amino acid residue selected from the group consisting of D-2-phenylglycine, D-norleucine, D-Ile, D-Leu, D-Val, D-2-aminobutyric acid, D-norvaline and D-isovaline.
  • X 8 is, D-Gln, D-Arg , D-Asn, D-Lys, D-His, D-2,3- diaminopropionic acid, the group consisting of D-2,4-diaminobutanoic acid and D- ornithine Amino acid residues selected from;
  • X 9 is a D-2-cyclohexylglycine residue;
  • X 10 is an amino acid residue selected from the group consisting of D-Lys, D-Arg, D-ornithine, D-His, D-2,3-diaminopropionic acid and D-2,4-diaminobutanoic acid.
  • X 11 is a D-2-phenylglycine residue
  • X 12 is an amino acid residue selected from the group consisting of D-Trp, D-homophenylalanine, D-Tyr and D-Phe
  • X 13 is an amino acid residue selected from the group consisting of D-Arg, D-ornithine, D-Lys, D-His, D-2,3-diaminopropionic acid and D-2,4-diaminobutanoic acid.
  • X 14 is a D-2-phenylglycine residue
  • X 15 is an amino acid residue selected from the group consisting of D-Tyr, D-Trp and D-Phe
  • X 16 is from D-Trp, D-homophenylalanine, D-Tyr and D-Phe.
  • X 0 is an amino acid residue or defect selected from the group consisting of D-Ala, D-Gly and D-2-aminoisobutyric acid
  • X 1 is an amino acid residue or defect selected from the group consisting of D-Leu, D-norleucine, D-Val, D-Ile, D-2-aminobutyric acid, D-norvaline and D-isovaline
  • X 2 is an amino acid residue selected from the group consisting of D-Arg, D-ornithine, D-Lys, D-His, D-2,3-diaminopropionic acid and D-2,4-diaminobutanoic acid.
  • X 3 is a D-2-cyclohexylglycine residue
  • X 4 is, D-Lys, D-Arg , D- ornithine, D-His, with an amino acid residue selected from the group consisting of D-2,3-diaminopropionic acid and D-2,4-diaminobutane acid can be
  • X 5 contains D-Ser, D-Arg, D-2-hydroxyglycine, D-homoserine, D-Lys, D-ornithine, D-His, D-2,3-diaminopropionic acid and D-2,4.
  • X 6 is an amino acid residue selected from the group consisting of D-Trp, D-3- (2-naphthyl) alanine, D-Tyr, D-Phe and D-3- (1-naphthyl) alanine
  • X 7 is an amino acid residue selected from the group consisting of D-Ile, D-Leu, D-norleucine, D-Val, D-2-aminobutyric acid, D-norvaline and D-isovaline
  • X 8 is, D-Gln, D-Arg , D-Asn, D-Lys, D-His, D-2,3- diaminopropionic acid, the group consisting of D-2,4-diaminobutanoic acid and D- ornithine Amino acid residues selected from;
  • X 9 is a D-2-cyclohexylgly
  • X 11 is an amino acid residue selected from the group consisting of D-Ile, D-Leu, D-norleucine, D-Val, D-2-aminobutyric acid, D-norvaline and D-isovaline
  • X 12 is an amino acid residue selected from the group consisting of D-Trp, D-homophenylalanine, D-Tyr and D-Phe
  • X 13 is an amino acid residue selected from the group consisting of D-Arg, D-ornithine, D-Lys, D-His, D-2,3-diaminopropionic acid and D-2,4-diaminobutanoic acid.
  • X 14 is an amino acid residue selected from the group consisting of D-Ile, D-Leu, D-norleucine, D-Val, D-2-aminobutyric acid, D-norvaline and D-isovaline
  • X 15 is an amino acid residue selected from the group consisting of D-Tyr, D-Trp and D-Phe
  • X 16 is from D-Trp, D-homophenylalanine, D-Tyr and D-Phe.
  • Amino acid residues or defects selected from the group consisting of, more preferably amino acid residues selected from the group consisting of D-Trp, D-homophenylalanine, D-Tyr and D-Phe.
  • X 0 is a D-Ala residue or defect
  • X 1 is a D-Leu residue, a D-norleucine residue or a defect
  • X 2 is a D-Arg residue or a D-ornithine residue
  • X 3 is a D-2-cyclohexylglycine residue or a D-norleucine residue
  • X 4 is, D-Lys residues, be a D-Arg residue, or D- ornithine residues
  • X 5 is a D-Ser residue or a D-Arg residue
  • X 6 is a D-Trp residue or a D-3- (2-naphthyl) alanine residue
  • X 7 is a D-2-phenylglycine residue or a D-Ile residue
  • X 8 is an D-Gln residue or D-Arg residues
  • X 9 is a D-Gln residue or D-Arg residues
  • the peptide represented by the above formula (1) preferably contains any one of the amino acid sequences represented by SEQ ID NOs: 1 to 25.
  • the peptide represented by the above formula (1) is more preferably the amino acid sequence represented by SEQ ID NOs: 1 to 4, 6, 8 to 23 and 25. It comprises any one, and particularly preferably any one of the amino acid sequences represented by SEQ ID NOs: 19-23 and 25.
  • the peptide according to the present invention can be produced by a conventionally known method including a chemical synthesis method and a recombination technique.
  • a chemical synthesis method and a recombination technique.
  • each amino acid is commonly used in peptide chemistry, eg, "The Peptides,” Volume 1 [Schroder and Luhke, Academic Press, New York, U.S.A. S. A. (1966)], "Basics and Experiments of Peptide Synthesis” (Nobuo Izumiya et al., Maruzen Co., Ltd., 1985), etc.
  • the peptide according to the present invention may also be produced by a recombination technique using animal cells, insect cells, microorganisms or the like by a method as described in, for example, Current Protocols in Molecular Biology, Chapter 16 below.
  • the peptide can be purified by a conventionally known method after being produced by cultured cells or microorganisms. Methods for purifying and isolating peptides are known to engineers in the art, and can be carried out by the methods described in, for example, Current Protocols in Molecular Biology, Chapter 16 (Ausube et al., John Wiley and Sons, 2006).
  • Condensation methods for forming peptide bonds include azide method, acid halide method, acid anhydride method, carbodiimide method, carbodiimide-additive method, active ester method, carbonyl imidazole method, oxidation-reduction method, enzyme method, Woodward reagent K. , HATU reagent, method using Bop reagent and the like can be exemplified.
  • the acid anhydride method, the carbodiimide method, and the active ester method are mainly mentioned as the condensation reaction in the solid phase method.
  • the C-terminal amino acid is bound to a support such as a resin that is insoluble in the organic solvent used.
  • a resin a resin having a functional group introduced for the purpose of binding an amino acid to the resin, a resin having a spacer inserted between the resin and the functional group, and the like can also be used depending on the purpose. More specifically, for example, halomethyl resin such as chloromethyl resin, oxymethyl resin, 4- (oxymethyl) -phenylacetamidemethyl resin, 4- (oxymethyl) -phenoxymethyl resin, Rinkamide resin and the like can be mentioned. Can be done.
  • protective means such as a carboxyl group, an amino group, a hydroxyl group, and an amidino group that are not involved in the condensation reaction can be applied by a commonly known means. On the contrary, it is also possible to activate the carboxyl group or amino group directly involved in the condensation reaction.
  • Protecting groups commonly used in organic chemistry as protecting groups for functional groups that are not involved in the condensation reaction of each unit include, for example, "Protective Groups in Organic Synthesis (by Greene, John Wiley & Sons, Inc.). It can be protected by the protecting group described in 1981)) and the like. More specifically, as the protecting group for the carboxyl group, for example, various methyl esters, ethyl esters, benzyl esters, p- Examples of commonly known protecting groups such as nitrobenzyl ester, t-butyl ester and cyclohexyl ester can be mentioned.
  • Examples of the protecting group for the amino group include a benzyloxycarbonyl group, a t-butoxycarbonyl group and an isobornyloxycarbonyl.
  • Groups, 9-fluorenylmethoxycarbonyl group (Fmoc group) and the like can be mentioned.
  • Examples of the activated carboxyl group include acid anhydrides corresponding to the carboxyl group; azide; pentafluorophenol, 2,4-dinitrophenol, cyanomethyl alcohol, p-nitrophenol, and N-hydroxysuccinic acid. Examples thereof include active esters with imide, N-hydroxy-5-norbornen-2,3-dicarboxymid, N-hydroxyphthalimide, 1-hydroxybenzotriazole and the like. Examples of the activated amino group include phosphoric acid amides corresponding to the amino group.
  • the condensation reaction during peptide synthesis is usually carried out in a solvent.
  • the solvent include chloroform, dichloromethane, ethyl acetate, N, N-dimethylformamide, dimethyl sulfoxide, pyridine, dioxane, tetrahydrofuran, N-methylpyrrolidone, water, methanol and the like, or a mixture thereof. ..
  • the reaction temperature of the condensation reaction can be carried out in the range of ⁇ 30 ° C. to 50 ° C., as in the usual case.
  • the type of the protecting group elimination reaction in the peptide production process of the present invention is selected according to the type of protecting group to be used as long as the protecting group can be eliminated without affecting the peptide bond.
  • Examples thereof include alkali treatment with, sodium treatment in liquid ammonia, reduction with palladium carbon, and silylation treatment with trimethylsilyl triflate, trimethylsilyl bromide and the like.
  • the deprotecting group reaction is efficiently carried out by adding a cation scavenger such as anisole, phenol, cresol, thioanisole and ethanedithiol. It is preferable from the viewpoint.
  • the method for cleaving the peptide of the present invention synthesized by the solid phase method from the solid phase also follows a generally known method.
  • the above-mentioned treatment with an acid or a silylating agent can be mentioned as the cutting method.
  • generally known separation and purification means can be fully utilized after the completion of the above series of reactions.
  • the peptide of the present invention can be obtained with higher purity by extraction, partitioning, reprecipitation, recrystallization, solid phase extraction, column chromatography and the like.
  • the obtained peptide can be analyzed by an automatic amino acid analyzer, capillary electrophoresis, reverse phase high performance liquid chromatography, mass spectrometry, etc.
  • various biomolecular interaction analysis methods such as phage display method, two-hybrid method, affinity chromatography, surface plasmon resonance method, co-immunoprecipitation method, protein chip method, three-dimensional structure analysis, far western method, and fluorescence quenching method are used.
  • peptides may be selected using the interaction with myostatin as an index.
  • the peptide according to the present invention may be isolated or purified. "Isolation or purification” means that an operation has been performed to remove components other than the target component.
  • the purity of the isolated or purified peptide according to the present invention is usually 50% or more (for example, 70% or more, 80% or more, 90% or more, 95% or more, 98% or more, 99% or more, 100%). be.
  • One form of the present invention is a complex represented by the following formula (2) or a pharmaceutically acceptable salt thereof.
  • R 1 and R 2 independently represent a halogenoalkyl group or a halogen atom, respectively.
  • R 3 represents a bromine atom, an iodine atom or a selenium atom.
  • R 4 and R 5 each independently represent a hydrogen atom or a substituted or unsubstituted alkyl group.
  • R 6 and R 7 each independently represent a hydrogen atom, a halogen atom, an alkoxy group or a substituted or unsubstituted alkyl group, wherein R 4 and R 6 or R 5 and R 7 are combined.
  • Substituted or unsubstituted alkylene groups or alkenylene groups may be formed.
  • R 8 represents a hydrogen atom or a substituted or unsubstituted alkyl group.
  • R 9 and R 10 each independently represent a hydrogen atom, a halogen atom, an alkoxy group or a substituted or unsubstituted alkyl group, wherein R 8 and R 9 or R 10 are substituted or substituted together. It may form an unsubstituted alkylene group or an alkenylene group.
  • m and n independently represent integers of 1 to 3, respectively. * Is the binding site with L] It is a compound represented by; L represents a linker between Y and Z; Z is the peptide according to the present invention.
  • Y is a compound represented by the above formula (3).
  • the compound represented by the formula (3) is an on / off switch catalyst that enables target-selective photooxygenation.
  • the complex is activated by light having a wavelength of 650 to 800 nm, and myostatin is selectively oxygenated. By making it, it is possible to exert even higher myostatin inhibitory activity.
  • R 1 and R 2 independently represent a halogenoalkyl group or a halogen atom, respectively.
  • the halogenoalkyl group is preferably a linear or branched halogenoalkyl group having 1 to 6 carbon atoms, more preferably a linear or branched halogenoalkyl group having 1 to 4 carbon atoms, and even more preferably a tri. It is a fluoromethyl group or a pentafluoroethyl group, and particularly preferably a trifluoromethyl group.
  • halogen atom examples include a fluorine atom, a chlorine atom, a bromine atom or an iodine atom, and a fluorine atom is preferable.
  • R 1 is preferably a halogen atom and R 2 is a halogenoalkyl group.
  • R 3 represents a bromine atom, an iodine atom or a selenium atom, and is preferably a bromine atom.
  • R 4 and R 5 each independently represent a hydrogen atom or a substituted or unsubstituted alkyl group.
  • R 6 and R 7 each independently represent a hydrogen atom, a halogen atom, an alkoxy group or a substituted or unsubstituted alkyl group. At this time, R 4 and R 6 or R 5 and R 7 may be combined to form a substituted or unsubstituted alkylene group or alkenylene group.
  • R 8 represents a hydrogen atom or a substituted or unsubstituted alkyl group.
  • R 9 and R 10 each independently represent a hydrogen atom, a halogen atom, an alkoxy group or a substituted or unsubstituted alkyl group. At this time, R 8 and R 9 or R 10 may be combined to form a substituted or unsubstituted alkylene group or alkenylene group.
  • R 4 and R 6, R 5 and R 7 and R 8 and R 10 form a substituted or unsubstituted alkylene or alkenylene group, this time the alkylene group
  • the alkaneylene group has 2 or 3 carbon atoms.
  • the alkyl group in R 4 to R 10 is preferably an alkyl group having 1 to 6 carbon atoms in a straight chain or a branched chain, and more preferably an alkyl group having 1 to 4 carbon atoms in a straight chain or a branched chain.
  • Examples of the alkyl group include a methyl group, an ethyl group, an n-propyl group, an isopropyl group, an n-butyl group, an isobutyl group, a tert-butyl group and the like.
  • the group capable of substituting these alkyl groups is preferably 1 to 3 groups selected from a carboxy group, a sulfonic acid group, a hydroxy group, an amino group, -CO-, -CONH- and a triazole group.
  • the group capable of substituting the alkyl group is more preferably selected from a carboxy group, a sulfonic acid group, a hydroxy group and an amino group from the viewpoint of increasing water solubility.
  • the alkoxy group in R 6 , R 7 , R 9 and R 10 is preferably an alkoxy group having 1 to 6 carbon atoms in a straight chain or a branched chain, and an alkoxy group having 1 to 4 carbon atoms in a more linear or branched chain. It is the basis. Specific examples thereof include a methoxy group, an ethoxy group, and a propyloxy group.
  • Examples of the halogen atom in R 6 , R 7 , R 9 and R 10 include a chlorine atom, a bromine atom, an iodine atom and a fluorine atom.
  • R 4 and R 6 , R 5 and R 7 and R 8 and R 10 together form a substituted or unsubstituted alkylene or alkenylene group, wherein said alkylene or alkenylene group.
  • the alkylene group include an ethylene group and a trimethylene group.
  • the alkenylene group include a vinylene group and a propenylene group.
  • Examples of the ring structure formed by these groups together include the following structures.
  • R 4 ⁇ R 7 and R 9 ⁇ R 10 represents a group that does not form an alkylene group or alkenylene group.
  • m and n each independently represent an integer of 1 to 3.
  • m and n are independently, preferably 1 or 2, and more preferably 1.
  • * is the binding site with L.
  • Linker L represents a linker between compound Y represented by formula (3) and peptide Z according to the present invention.
  • the binding site of the linker L to the peptide Z according to the present invention is not particularly limited and may be the N-terminal or the C-terminal of the peptide Z according to the present invention. It may be a side chain of the group.
  • the linker L is attached to the N-terminus of peptide Z according to the present invention.
  • linker unit is not particularly limited, but for example, a substituted or unsubstituted alkylene group having 1 to 20 carbon atoms, a substituted or unsubstituted alkenylene group having 2 to 20 carbon atoms, a substituted or non-substituted alkenyl group.
  • L has a structure represented by the following formula (4).
  • V represents a -CO-, -CONH- or a triazole ring.
  • O represents an integer of 0 or 1.
  • the triazole ring include 1,2,3-triazole-1,4-diyl group and 1,2,4-triazole-1,3-diyl group.
  • l and p each independently represent an integer of 1 to 6.
  • l preferably represents an integer of 1 to 6, and more preferably represents an integer of 1 to 4.
  • p preferably represents an integer of 1 to 6, and more preferably represents an integer of 1 to 4.
  • * 1 is a linking site with the compound represented by the formula (3)
  • * 2 is a linking site with the peptide according to the present invention.
  • R 1 to R 7 and Z are as defined for the above formula (2).
  • the complex represented by the formula (2-1) can be produced according to the following reaction formula.
  • the compound represented by the formula (2-1) can be obtained by condensing the compound a and the compound b to obtain the compound c, and then reacting the compound c with the compound d.
  • the reaction between compound a and compound b is an aldol condensation reaction.
  • reaction between compound c and compound d is a 1,3-dipole addition reaction between alkyne and azide, and easily proceeds at room temperature in a polar solvent such as dimethylformamide in the presence of a copper catalyst.
  • the compounds a to d are the above-mentioned H. Okamoto et al. , Chem. Commun. , 2019, 55, 9108-9111. , International Publication No. 2017/164172 and the like can be appropriately referred to and synthesized.
  • the obtained complex represented by the formula (2-1) can be purified by a known means such as chromatography.
  • a myostatin inhibitor comprising the peptide according to the invention or a prodrug thereof or a complex according to the present invention (hereinafter, "the peptide according to the present invention or a prodrug thereof or the present invention”.
  • a "myostatin inhibitor” containing the complex according to the invention is also simply referred to as a "myostatin inhibitor").
  • the myostatin inhibitor may be composed of one or more peptides according to the present invention, one or more prodrugs thereof, one or more complexes according to the present invention, or a mixture thereof, but usually.
  • a pharmaceutical composition comprising a peptide according to the present invention and a prodrug thereof, one or more selected from the complex according to the present invention, and a pharmaceutically acceptable carrier.
  • One embodiment of the present invention relates to a method for inhibiting myostatin, which comprises administering to a patient an effective amount of a peptide according to the present invention or a prodrug thereof or a complex according to the present invention.
  • One embodiment of the invention also relates to a peptide according to the invention or a prodrug thereof or a complex according to the invention for use in inhibiting myostatin.
  • a prophylactic and / or therapeutic agent for muscular atrophy disorder which comprises the peptide according to the present invention or a prodrug thereof or a complex according to the present invention (hereinafter, "the peptide according to the present invention”).
  • a prodrug thereof or a prophylactic and / or therapeutic agent for muscular atrophy disorder containing the complex according to the present invention is also simply referred to as a "preventive / therapeutic agent for muscular atrophy disorder").
  • the prophylactic / therapeutic agent for muscle atrophy disorder may be composed of one or more peptides according to the present invention, one or more prodrugs thereof, one or more complexes according to the present invention, or a mixture thereof. However, it is usually a pharmaceutical composition comprising a peptide according to the present invention and a prodrug thereof, one or more selected from the complex according to the present invention, and a pharmaceutically acceptable carrier.
  • One embodiment of the present invention relates to a method for preventing and / or treating a muscular atrophy disorder, which comprises administering to a patient an effective amount of a peptide according to the present invention or a prodrug thereof or a complex according to the present invention.
  • One embodiment of the invention also relates to a peptide according to the invention or a prodrug thereof or a complex according to the invention for use in the prevention and / or treatment of muscular atrophy disorder.
  • Myostatin inhibitors, prophylactic / therapeutic agents for muscular atrophy disorders, and the above prophylactic and / or therapeutic methods are also effective for strengthening the tibialis anterior muscle by local administration for elderly people with normal walking disorders. Just strengthening the tibialis anterior muscle facilitates dorsiflexion of the ankle joint, which helps prevent falls. Further, for example, if continuous local administration during staying in space can be carried out, it can contribute to shortening the rehabilitation period after returning.
  • the above-mentioned muscular atrophy disorder is not particularly limited, and is, for example, muscular dystrophy, distal myopathy, congenital myopathy, inflammatory myopathy such as inclusion body myositis, myopathy such as mitochondrial myopathy; and disused muscular atrophy. ; Sarcopenia etc. can be exemplified.
  • the prophylactic / therapeutic agent for muscular atrophy disorder is preferably effectively used for muscular dystrophy and sarcopenia.
  • the preventive / therapeutic agents for muscular dystrophy are more preferably Duchenne muscular dystrophy, Becker muscular dystrophy, Fukuyama muscular dystrophy, melosine deficient muscular dystrophy, limb muscular dystrophy, facial muscular dystrophy, emery muscular dystrophy, and emery muscular dystrophy. , Miyoshi muscular dystrophy, and muscular dystrophy such as infant muscular dystrophy as well as sarcopenia, and is particularly effective against Duchenne muscular dystrophy.
  • Muscle atrophy disorders can also result from chronic diseases such as amyotrophic lateral sclerosis, chronic obstructive pulmonary disease (COPD), cancer, AIDS, renal failure, and rheumatoid arthritis. Muscle atrophy disorders can also result from metabolic disorders such as diabetes and related disorders. Therefore, the preventive / therapeutic agent for muscular atrophy disorder of the present invention, and the above-mentioned preventive and / or therapeutic method can be used for improving cachexia associated with muscular atrophy. In addition, inhibition of myostatin can increase muscle mass, improve bone strength, and reduce osteoporosis and other degenerative bone diseases.
  • COPD chronic obstructive pulmonary disease
  • One embodiment of the present invention relates to a prophylactic and / or therapeutic agent for a muscular atrophy disorder caused by diabetes, which comprises a peptide according to the present invention or a prodrug thereof or a complex according to the present invention.
  • One embodiment of the invention also prevents and / or prevents diabetes-induced muscular atrophy disorders, comprising administering to a patient an effective amount of a peptide according to the invention or a prodrug thereof or a complex according to the invention.
  • the treatment method Regarding the treatment method.
  • One embodiment of the present invention relates to a prophylactic and / or therapeutic agent for muscular atrophy disorder caused by a vicious cancer solution, which comprises a peptide according to the present invention or a prodrug thereof or a complex according to the present invention.
  • One embodiment of the present invention also comprises administering to a patient an effective amount of a peptide according to the invention or a prodrug thereof or a complex according to the present invention to prevent muscular atrophy disorder caused by a vicious cancer solution. And / or regarding treatment methods.
  • a therapeutically “effective amount” is an amount that is effective in producing some desired therapeutic effect commensurate with a reasonable benefit / risk ratio.
  • the "subject” and “patient” include non-human animals including humans and fish, but preferably humans, dogs, cats, mice, rats, hamsters, guinea pigs, and horses (race horses). Included), mammals such as cows, pigs, rabbits, and sheep, and poultry such as chickens, quails, and turkeys, more preferably humans.
  • the above-mentioned pharmaceutically acceptable carrier is not particularly limited, but is an excipient such as lactose, sucrose, mannitol, starch, corn starch, crystalline cellulose, and light anhydrous silicic acid; silica, talc, calcium stearate, stearic acid, etc.
  • Lubricants such as magnesium; binders such as hydroxypropyl cellulose, hydroxypropyl methyl cellulose, carboxymethyl cellulose, sodium carboxymethyl cellulose, methyl cellulose, polyvinylpyrrolidone, crystalline cellulose, dextrin, gelatin; ascorbic acid, sodium sulfite, sodium hydrogen sulfite, tocopherol, etc.
  • Chelating agents such as ethylenediamine tetraacetic acid (EDTA); Buffers such as borates, bicarbonates, Tris-HCl, citrates, phosphates, other organic acids; water for injection, physiological saline Solponics such as water, ethanol, propanol, ethylene glycol, propylene glycol, macrogol, olive oil, corn oil; Pluronic®, polyethylene glycol, sorbitan fatty acid ester, polysorbate, Triton®, lecithin, cholesterol, benza chloride
  • Surface active agents or wetting agents such as luconium, benzethonium chloride, glycerin monostearate; isotonic agents such as sodium chloride, potassium chloride, glycerin, glucose, sorbitol, mannitol; benzoic acid, salicylic acid, thimerosal, phenethyl alcohol, methylparaben
  • Preservatives such as, propylparaben
  • the content of the peptide according to the present invention or the prodrug thereof or the complex according to the present invention in the drug can be 0.01 to 100% by weight based on the whole drug.
  • the dose of the peptide according to the present invention or the prodrug thereof or the complex according to the present invention varies depending on the age, symptoms, administration method, etc., but in the case of oral administration, it is generally given to humans (assuming a body weight of 60 kg). On the other hand, it is about 0.1 to 100 mg, preferably about 1.0 to 50 mg, and more preferably about 1.0 to 20 mg per day.
  • the single dose varies depending on age, symptoms, administration method, etc., but for example, in the form of an injection, it is usually about about a day for a human (assuming a body weight of 60 kg). It is convenient to administer about 0.01 to 30 mg, preferably about 0.1 to 20 mg, and more preferably about 0.1 to 10 mg. In the case of animals other than humans, the amount converted per 60 kg of body weight can also be administered.
  • Trifluoroacetic acid in the presence of m-cresol (0.10 mL), thioanisole (0.10 mL) and 1,2-ethanedithiol (0.10 mL) for removal of various side chain protecting groups and removal of resin.
  • the reaction was carried out in 4.0 mL for 2 hours. After removing the resin by filtration using a funnel with a filter, TFA was distilled off by nitrogen spraying, and 40 mL of diethyl ether was added to precipitate a crude peptide.
  • Peptide 1 was synthesized and purified by the same method as in Synthesis Example 1 using Rink Amide resin (0.37 mmol / g, Watanabe Chemical Industry Co., Ltd.) 30 mg (0.011 mmol) (8.2 mg, 24%).
  • LRMS (MALDI +) calcd for (M + + H) 2392.03, found 2391.04.
  • the peptide as the test material was suspended in DMSO so as to be 10 mM as a stock solution, and stored at -30 ° C. 1 hour before addition to the medium, the cells were suspended in serum-free DMEM medium and allowed to stand at room temperature (25 ° C.) for 20 minutes. Then, the peptide was added to a medium having a final concentration of 0.3 ⁇ M and 8 ng / mL myostatin (Merck Millipore), and then cultured for 4 hours.
  • the propeptide protein (RSD) used as a positive control was suspended using PBS containing 0.1% (v / v) BSA so as to be 10 ⁇ M as a stock solution, and stored at -30 ° C. 1 hour before addition to the medium, the cells were suspended in serum-free DMEM medium and allowed to stand at room temperature (25 ° C.) for 20 minutes. Then, a final concentration of 10 nM of the propeptide protein and 8 ng / mL of myostatin (Merck Millipore) were added to the medium, and then the cells were cultured for 4 hours.
  • the culture medium was removed with an aspirator, and the cells were washed with 1 ⁇ PBS. Then, 50 ⁇ L of Passive Lysis buffer (Promega) was added per well to lyse the cells. The lysate was centrifuged at 4500 rpm for 6 minutes under 4 ° C. conditions. After transferring 20 ⁇ L of the supernatant after centrifugation to a white 96-well plate (Coster), 50 ⁇ L of Luciferase Assay Reagent (Promega) was added, and the luminescence was detected by Luminoscan Ascent (Thermo Fisher Scientific). Firefly luciferase activity was measured. Further, after adding 50 ⁇ L of Stop & Glo Buffer, luminescence was detected by Luminoscan Ascent and the luciferase activity was measured to obtain internal control.
  • Passive Lysis buffer Promega
  • RiDM-35 was dissolved in a 50 mM Tris-HCl buffer (pH 7.5, 0.15 M NaCl, 10 mM CaCl 2 , 0.05% (w / v) Brij-35 containing) solution to a final concentration of 50 ⁇ M and TPCK.
  • Treated bovine pancreatic trypsin Sigma-Aldrich was added to a final concentration of 1 ⁇ g / mL (total volume 100 ⁇ L) and incubated at 37 ° C. for 400 minutes.
  • the column used was COSMOSIL 5C18-AR-II 4.6 ⁇ 150 mm (Nacalai Tesque, Inc.), and a linear gradient of acetonitrile in a water-acetonitrile system containing 0.1% trifluoroacetic acid (25 to 25 to min) at a flow rate of 1 mL / min. By applying 40%, 30 minutes), the peak of the peptide was detected with a UV detector (220 nm).
  • riDM-35 The decomposition product derived from riDM-35 was not identified by mass spectrometry using LCMS-2020 (Shimadzu Corporation). riDM-35 showed extremely high stability in bovine pancreas-derived trypsin solution.
  • riDM-35 Dissolve riDM-35 in 100 mM Tris-HCl buffer (pH 7.8, containing 10 mM CaCl 2 ) solution to a final concentration of 50 ⁇ M, and add TLCK-treated bovine pancreas-derived ⁇ -chymotrypsin (Sigma-Aldrich) to a final concentration of 2 ⁇ g. It was added to / mL (total volume 100 ⁇ L) and incubated at 37 ° C. for 400 minutes.
  • Tris-HCl buffer pH 7.8, containing 10 mM CaCl 2
  • TLCK-treated bovine pancreas-derived ⁇ -chymotrypsin Sigma-Aldrich
  • the column used was COSMOSIL 5C18-AR-II 4.6 ⁇ 150 mm (Nacalai Tesque, Inc.), and a linear gradient of acetonitrile in a water-acetonitrile system containing 0.1% trifluoroacetic acid (25 to 25 to min) at a flow rate of 1 mL / min. By applying 40%, 30 minutes), the peak of the peptide was detected with a UV detector (220 nm).
  • Test Example 4 Effect of riDM-4 on Duchenne muscular dystrophy model mdx mouse tibialis anterior muscle (in vivo evaluation) In order to verify the in vivo grip strength increasing effect of riDM-4 synthesized in Synthesis Example 1, evaluation was performed by the following method.
  • RiDM-4 was dissolved in physiological saline to a concentration of 0.75 mM. 40 ⁇ L intramuscularly was intramuscularly administered to a total of 8 muscles of both hind limbs of a 5-week-old mdx male mouse (purchased from Claire Japan) under anesthesia (40 ⁇ L of saline was similarly administered to the control group). Two weeks later, the same amount of riDM-4 (physiological saline in the control group) was administered again, and four weeks later, the mouse grip strength was measured (Saito-type mouse grip strength measuring device MK-380M, Muromachi Kikai Co., Ltd.). Was done.
  • riDM-4 significantly increased the grip strength of the hind limbs of mdx mice by about 60% based on its myostatin inhibitory activity. The improvement effect was greater than that of the existing MIPE-1686.
  • 16PC-N (3 ⁇ M) and myostatin (1 ⁇ M) in phosphate buffer (10 mM, pH 7.4) were irradiated with light (730 nm, 14 mW) at 37 ° C. for 30 minutes. After reduction with dithiothreitol, digestion with lysylendopeptide dase, and desalting with ZipTip C18, MALDI-TOF MS analysis was performed.
  • Test Example 5 Verification of the effect of intramuscular administration of riDM-35 on improving muscle wasting in a cancer cachexia model mouse (in vivo evaluation)
  • the ri-DM35 synthesized in Synthesis Example 23 was dissolved in PBS (phosphate buffer; 10 mM, pH 7.4) to 1 mM.
  • PBS phosphate buffer; 10 mM, pH 7.4
  • a 5.0 ⁇ 10 6 Lewis lung adenocarcinoma cell (LLC) was transplanted subcutaneously into the back of an anesthetized C57BL6 / J male mouse (purchased from Oriental yeast) to prepare a cancer cachexia model mouse.
  • LLC Lewis lung adenocarcinoma cell
  • mice were performed on the 22nd day of LLC transplantation, and the gastrocnemius muscles of both feet were excised to measure the weight and muscle fiber area.
  • Figure 10 shows the weight of the gastrocnemius muscle per body weight of the cancer cachexia model mouse.
  • the gastrocnemius muscle weight per body weight of the cancer cachexia model mice increased by 19.6%.
  • Figure 11 shows the frequency distribution table of the muscle fiber area of the cancer cachexia model mouse.
  • the ri-DM35-administered group enlarged the muscle fiber area of the cancer cachexia model mice.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biochemistry (AREA)
  • Immunology (AREA)
  • Molecular Biology (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Neurology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)

Abstract

La présente invention concerne un peptide qui a une grande activité d'inhibition de la myostatine et qui présente une stabilité améliorée in vivo. Elle porte sur un peptide représenté par la formule (1) dans la description.
PCT/JP2021/024125 2020-07-06 2021-06-25 Peptide et complexe le comprenant WO2022009698A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
JP2022535021A JPWO2022009698A1 (fr) 2020-07-06 2021-06-25

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
JP2020-116583 2020-07-06
JP2020116583 2020-07-06

Publications (1)

Publication Number Publication Date
WO2022009698A1 true WO2022009698A1 (fr) 2022-01-13

Family

ID=79553070

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/JP2021/024125 WO2022009698A1 (fr) 2020-07-06 2021-06-25 Peptide et complexe le comprenant

Country Status (2)

Country Link
JP (1) JPWO2022009698A1 (fr)
WO (1) WO2022009698A1 (fr)

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2009539357A (ja) * 2006-06-06 2009-11-19 ヘルムホルツ−ツェントルム フュア インフェクツィオンスフォルシュンク ゲーエムベーハー リウマチ熱関連ペプチド(parf)および診断マーカーとしてのその使用
WO2014119753A1 (fr) * 2013-01-31 2014-08-07 学校法人 東京薬科大学 Peptide inhibiteur de la myostatine
JP2016523274A (ja) * 2013-06-26 2016-08-08 ザイジェン インフラメーション リミテッド 種々の疾患を処置するためのjnkシグナル伝達経路の新規の細胞透過性ペプチド阻害剤の使用
JP2017528417A (ja) * 2014-06-12 2017-09-28 ザ ユニヴァーシティ オブ バース 薬物送達強化剤
WO2018030432A1 (fr) * 2016-08-10 2018-02-15 学校法人東京薬科大学 Peptide, sel pharmacologiquement acceptable de celui-ci, et promédicament correspondant
JP2020059663A (ja) * 2018-10-09 2020-04-16 学校法人東京薬科大学 ペプチドもしくはその薬学的に許容される塩、またはそれらのプロドラッグ

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2009539357A (ja) * 2006-06-06 2009-11-19 ヘルムホルツ−ツェントルム フュア インフェクツィオンスフォルシュンク ゲーエムベーハー リウマチ熱関連ペプチド(parf)および診断マーカーとしてのその使用
WO2014119753A1 (fr) * 2013-01-31 2014-08-07 学校法人 東京薬科大学 Peptide inhibiteur de la myostatine
JP2016523274A (ja) * 2013-06-26 2016-08-08 ザイジェン インフラメーション リミテッド 種々の疾患を処置するためのjnkシグナル伝達経路の新規の細胞透過性ペプチド阻害剤の使用
JP2017528417A (ja) * 2014-06-12 2017-09-28 ザ ユニヴァーシティ オブ バース 薬物送達強化剤
WO2018030432A1 (fr) * 2016-08-10 2018-02-15 学校法人東京薬科大学 Peptide, sel pharmacologiquement acceptable de celui-ci, et promédicament correspondant
JP2020059663A (ja) * 2018-10-09 2020-04-16 学校法人東京薬科大学 ペプチドもしくはその薬学的に許容される塩、またはそれらのプロドラッグ

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
H OKAMOTO, TANIGUCHI; USAMI; TAGUCHI; TAKAYAMA; HAYASHI: "Inactivation of myostatin by photo-oxygenation using catalyst-functionalized peptides", CHEM. COMMUN., 1 January 2019 (2019-01-01), pages 9108 - 9111, XP055622123, DOI: 10.1039/C9CC04368C *
TAKAYAMA KENTARO, ASARI TOMO, SAITOH MARIKO, NIRASAWA KEI, SASAKI ERI, ROPPONGI YOSHIMI, NAKAMURA AKARI, SAGA YUSUKE, SHIMADA TAKA: "Chain-Shortened Myostatin Inhibitory Peptides Improve Grip Strength in Mice", ACS MEDICINAL CHEMISTRY LETTERS, AMERICAN CHEMICAL SOCIETY, US, vol. 10, no. 6, 13 June 2019 (2019-06-13), US , pages 985 - 990, XP055893503, ISSN: 1948-5875, DOI: 10.1021/acsmedchemlett.9b00174 *

Also Published As

Publication number Publication date
JPWO2022009698A1 (fr) 2022-01-13

Similar Documents

Publication Publication Date Title
JP5686825B2 (ja) メタスチン誘導体およびその用途
RU2430107C2 (ru) Производные метастина и их применение
EP1165613B1 (fr) Ligands du recepteur de melanocortine
CZ2003788A3 (cs) Ligandy melanokortinového receptoru
JPWO2009131191A1 (ja) メタスチン誘導体およびその用途
CN104822702A (zh) α-MSH和γ-MSH类似物
CN103842373B (zh) 模板-固定的肽模拟物作为fpr1的抑制物
WO2022009698A1 (fr) Peptide et complexe le comprenant
JP7032808B2 (ja) ペプチドもしくはその薬学的に許容される塩、またはそれらのプロドラッグ
TW200410986A (en) Pharamaceutical comiposition comprising cyclic somatostatin analogs II
CN113727991A (zh) Ras抑制肽
JP2020059663A (ja) ペプチドもしくはその薬学的に許容される塩、またはそれらのプロドラッグ
WO2014119753A4 (fr) Peptide inhibiteur de la myostatine
JPWO2010013762A1 (ja) メタスチン誘導体およびその用途
JP7181550B2 (ja) ペプチド、その薬学的に許容される塩、またはそれらのプロドラッグ、およびそれらの用途
JP7201967B2 (ja) ペプチドもしくはその薬学的に許容される塩、またはそれらのプロドラッグ
RU2454425C2 (ru) Производные метастина и их применение
CN118265719A (en) Peptide complexes with TrkB binding Activity
WO2011088462A2 (fr) Peptides d'alpha-fétoprotéine de type « anneau et queue »

Legal Events

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

Ref document number: 21835949

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2022535021

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21835949

Country of ref document: EP

Kind code of ref document: A1