CN117320710A - Combination therapy for fumarate-related diseases - Google Patents

Combination therapy for fumarate-related diseases Download PDF

Info

Publication number
CN117320710A
CN117320710A CN202180047680.0A CN202180047680A CN117320710A CN 117320710 A CN117320710 A CN 117320710A CN 202180047680 A CN202180047680 A CN 202180047680A CN 117320710 A CN117320710 A CN 117320710A
Authority
CN
China
Prior art keywords
seq
amino acid
peptide
fumarate
group
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.)
Pending
Application number
CN202180047680.0A
Other languages
Chinese (zh)
Inventor
让-马里·圣-雷米
吕克·范德埃尔斯特
文森特·卡利尔
米洛斯·埃拉克
让·万兰佩尔博格
马塞勒·万梅赫伦
大卫·瓦尔格拉夫
杰弗瑞·格卢瓦尔
让·斯马尔
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.)
Imcyse SA
Original Assignee
Imcyse SA
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 Imcyse SA filed Critical Imcyse SA
Priority claimed from PCT/EP2021/061972 external-priority patent/WO2021144478A2/en
Publication of CN117320710A publication Critical patent/CN117320710A/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/21Esters, e.g. nitroglycerine, selenocyanates
    • A61K31/215Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids
    • A61K31/22Esters, e.g. nitroglycerine, selenocyanates of carboxylic acids of acyclic acids, e.g. pravastatin
    • A61K31/225Polycarboxylic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/4015Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil having oxo groups directly attached to the heterocyclic ring, e.g. piracetam, ethosuximide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0008Antigens related to auto-immune diseases; Preparations to induce self-tolerance
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia

Abstract

The present invention relates to pharmaceutical formulations (combinations or pharmaceutical compositions or kits) comprising one or more dosage units of a fumarate compound and an immunogenic or tolerogenic peptide comprising an oxidoreductase motif and a NKT cell epitope or MHC class II T cell epitope of a (self) antigen involved in a fumarate-related disease or disorder. The invention also relates to the medical use of such a pharmaceutical formulation.

Description

Combination therapy for fumarate-related diseases
Technical Field
The present invention relates to a pharmaceutical formulation (combination or pharmaceutical composition or kit of parts) comprising a fumarate-based component and an immunogenic peptide comprising an oxidoreductase motif and a NKT cell epitope or an MHC class II T cell epitope of an autoantigen involved in a disease treatable with said fumarate-based component or a tolerogenic peptide comprising a NKT cell epitope or an MHC class II T cell epitope. The invention also relates to the medical use of such a pharmaceutical formulation.
Background
Fumarate compounds such as monomethyl fumarate or its prodrug dimethyl fumarate have been implemented in several diseases ranging from demyelinating disorders to cancer to transplant rejection. Fumaric acid ester (fumarate) or fumaric acid (fumaric acid) is a hydrolysate of monomethyl fumarate (MMF), which is in turn a hydrolysate of dimethyl fumarate (DMF). Fumarates have also been reported to be involved in the succination (secretion) of cysteine residues in certain proteins such as KEAP1, CTSZ, GAPDH, MGST, NUBP1, PRDX1 and PRDX3, TXN, and UCHL1, thereby impeding their function and leading to defects. DMF is converted to its active metabolite MMF, which binds Nrf 2. Subsequently, nrf2 translocates to the nucleus and binds to the antioxidant response element (antioxidant response element, ARE). This induces the expression of a number of cytoprotective genes including NAD (P) H quinone oxidoreductase 1 (NAD (P) H quinone oxidoreductase 1, NQO 1), thioredoxin 1 (thiofidoxin 1, srxn 1), heme oxygenase-1 (HO 1, HMOX 1), superoxide dismutase 1 (superoxide dismutase 1, SOD 1), gamma-glutamylcysteine synthetase (gamma-glutamylcysteine synthetase, gamma-GCS), thioredoxin reductase-1 (thioredoxin reductase-1, TXNRD 1), glutathione S-transferase (GST), glutamate-cysteine ligase catalytic subunit (glutamate-cysteine ligase catalytic subunit, gclc) and glutamate-cysteine ligase regulatory subunit (glutamate-cysteine ligase regulatory subunit, gclm); this also increases the synthesis of the antioxidant Glutathione (GSH). The intra-neuronal synthesis of GSH may protect neuronal cells from damage due to oxidative stress. DMF has also been shown to inhibit nuclear factor- κB (NF-kB) mediated pathways, regulate the production of certain cytokines, and induce apoptosis of certain T cell subsets. Its radiosensitizing activity is due to the ability of such agents to bind to and sequester (sequester) intracellular GSH, thereby depleting it and preventing its antioxidant effect. This enhances the cytotoxicity of ionizing radiation in hypoxic cancer cells. Leucine zipper transcription factor Nrf2 plays a key role in redox homeostasis and cytoprotection against oxidative stress.
DMF is currently being studied in a number of diseases, such as: multiple sclerosis (Multiple Sclerosis, MS), neuromyelitis optica (Neuromyelitis Optica, NMO), psoriasis, rheumatoid arthritis (Rheumatoid Arthritis, RA), asthma, atopic dermatitis, scleroderma, ulcerative colitis, cancer and transplant rejection.
WO2008017517 describes such a technique: allow for CD4 + T cells are polarized to a cytolytic phenotype, allowing them to be on peptide-MHC IClass I homologies recognize to induce APC apoptosis and thus suppress immune responses against specific antigens. This can be achieved by increasing the intensity of synapses produced with peptide-MHC complexes due to the addition of an oxidoreductase motif within the flanking residues of the class II restriction epitope. This technique also prevents or inhibits immune responses to multiple antigens, as APC apoptosis also prevents CD4 + T cells are activated to either a surrogate epitope for the autoantigen from which the peptide is derived, or to an epitope for the relevant autoantigen. In addition, CD4 polarized to a cytolytic phenotype + T cells eliminate bystander CD4 through apoptosis + T cells, provided that they are activated at the surface of the same APC. Finally, WO2008017517 discloses cytolytic CD4 produced using the above technique + T cells have a memory phenotype, allowing long term function.
WO2008/017517 shows this concept for allergies and autoimmune diseases (e.g. type 1 diabetes), wherein insulin may be used as an autoantigen. These immunogenic peptides are obtained by direct vaccination or for the administration of CD4 + In vitro transformation of T cells into cytolytic CD4 + Cells are used.
WO2012069568 patent application describes the same concept, but with cytolytic transformation of CD1 d-restricted NKT cells due to the use of CD1 d-restricted peptide epitopes fused to the oxidoreductase motif. These immunogenic peptides are used by direct vaccination or for in vitro transformation of CD1 d-restricted NKT cells.
WO2017182528 describes the use of immunogenic peptides comprising myelin oligodendrocyte glycoprotein (Myelin Oligodendrocyte Glycoprotein, MOG) epitopes for the treatment of multiple sclerosis.
In addition, tolerogenic peptides comprising T cell epitopes have been used to induce tolerance to certain autoantigens or autoantigens. For example, patent application WO0216410 describes antigen processing independent epitopes of suitable size to be presented by immature APCs that are not antigen processed, which would be advantageous for immune tolerance. WO2018182495 also discloses tolerogenic peptides comprising T cell epitopes to treat multiple sclerosis.
In searching for improved methods of treatment of the above diseases and conditions, the present invention provides synergistic combination therapies of fumarate compounds with immunogenic or tolerogenic peptides comprising an oxidoreductase motif.
Disclosure of Invention
Accordingly, the present invention provides the following aspects:
aspect 1. A kit comprising:
a) One or more dosage forms of a fumarate compound of formula (I):
wherein R is 1 And R is 2 Each independently selected from: OH, O - And optionally substituted (C) 1-10 ) Alkoxy, preferably optionally substituted (C 1-6 ) Alkoxy, or optionally substituted (C 1-3 ) An alkoxy group, an amino group,
wherein R is 3 And R is 4 Each independently selected from: h or deuterium is used as a carrier for the electron beam,
wherein each group independently may be optionally substituted as described elsewhere herein; and
b) An immunogenic or tolerogenic peptide of one or more dosage forms comprising, consisting of, or consisting essentially of: t cell epitopes of antigen proteins involved in fumarate-related diseases or disorders, more preferably T cell epitopes or NKT cell epitopes of antigen proteins involved in fumarate-related diseases or disorders, which T cell epitopes are capable of binding to MHC class I or class II molecules.
Aspect 2 the kit of aspect 1, wherein the peptide is an immunogenic peptide having an oxidoreductase motif linked to the T cell epitope, the oxidoreductase motif having a sequence of the general formula:
Z m -[CST]-X n -C- (SEQ ID NOS: 1 to 25) or Z m -C-X n -[CST]- (SEQ ID NO:26 to 50), wherein n is an integer selected from 0 to 6, preferably wherein n is 2, 1, 3 or 0, whereinm is an integer selected from 0 to 3, wherein X is any amino acid, wherein Z is any amino acid, wherein [ CST]Represents any one of cysteine (C), serine (S) or threonine (T);
wherein the oxidoreductase motif and the T cell epitope are separated by a linker having 0 to 7 amino acids,
wherein the hyphen (-) in the oxidoreductase motif represents the point of attachment of the oxidoreductase motif to the N-terminal end of a linker or epitope or to the C-terminal end of a linker or T-cell epitope.
In some embodiments, the fumarate compound is a deuterated form of any of the foregoing fumarate compounds, or a clathrate, solvate, tautomer, stereoisomer, or non-toxic pharmaceutically acceptable salt thereof, such as an acid addition salt, or a combination of any of the foregoing.
In some embodiments, the pharmaceutically acceptable salt is a salt of a metal (M) cation, where M may be an alkali metal, alkaline earth metal, or transition metal, such as Li, na, K, ca, zn, sr, mg, fe or Mn.
In a preferred embodiment, the oxidoreductase motif is not part of the following: a repeat of a standard C-XX- [ CST ] or [ CST ] -XX-C oxidoreductase motif, for example a repeat of said motif which may be separated from each other by one or more amino acids (e.g. CXXC X CXXC X CXXC (SEQ ID NO: 196)), such as a repeat adjacent to each other (CXXCXXCXXCXXC (SEQ ID NO: 197)) or such as a repeat overlapping each other (CXXXXCXXC (SEQ ID NO: 198) or CXCCXCXCC (SEQ ID NO: 199)), especially when n is 0 or 1 and m is 0 in the general formula as defined in aspect 2.
Aspect 3 the kit of aspects 1 or 2, wherein the fumarate compound is selected from the group consisting of: dialkyl fumarate, monoalkyl fumarate, a combination of dialkyl fumarate and monoalkyl fumarate, such as a combination of dimethyl fumarate and monomethyl fumarate, or a combination of any of the foregoing.
Aspect 4. The kit according to aspects 1, 2 or 3, wherein the fumarate compound is dimethyl fumarate-DMF (R1 is OCH3 and R2 is OCH 3-formula (II) below), or monomethyl fumarate-MMF (R1 is OCH3 and R2 is O-or OH-formula (III) below), or a combination thereof, or deuterated forms, clathrates, solvates, tautomers, stereoisomers or pharmaceutically acceptable salts thereof.
In some preferred embodiments of the fumarate compound of formula (I), R 1 Or R is 2 In (C) 1-10 ) The alkoxy groups may be selected from: ethoxy, methoxy, (C) 1-5 ) Alkoxy, (C) 1-4 ) Alkoxy, (C) 1-3 ) Alkoxy, (C) 2-3 ) Alkoxy, (C) 2-4 ) Alkoxy, (C) 2-5 ) Alkoxy and (C) 1-6 ) An alkoxy group.
Some preferred examples of such fumarate compounds are mono alkyl fumarates or more particularly prodrugs of mono methyl fumarates, i.e. compounds which are metabolizable in vivo to mono methyl fumarates, such as those of formula (I) wherein R 1 Is C 1 -C 3 Alkoxy, e.g. methoxy, ethoxy or propoxy, and wherein R 2 Is optionally substituted C 1 -C 3 Alkoxy, such as methoxy, ethoxy or propoxy.
Further preferred examples are where R 1 Is methoxy and R 2 Are those of methoxy or optionally substituted ethoxy.
In some embodiments, the fumarate compound is a prodrug of a monoalkyl fumarate, such as duloxetine fumarate (diroximel fumarate) (formula (IV)):
further examples of fumarate compounds that can be used in combination with an immunogenic or tolerogenic peptide as disclosed herein are discussed below.
In other illustrative embodiments, the fumarate compound is a calcium salt of MMF (Ca-MMF) or DMF (Ca-DMF), optionally in deuterated form, wherein one or more alkyl groups are deuterated alkyl groups comprising at least one deuterium atom, such as deuterated methyl groups. Some examples of deuterated methyl groups include: -CDH2, -CD2H and-CD 3. Some examples of deuterated ethyl groups include: -CHDCH3, -CD2CH3, -CHDCDH2, -CHDCD2H, CHDCD3, -CD2CDH2, CD2H and CD2CD3.
Aspect 5 the kit of any one of aspects 1 to 4, wherein the antigenic protein is a self antigen, a soluble allofactor (soluble allofactor), an alloantigen shed by a graft, an antigen of an intracellular pathogen, an antigen of a viral vector for gene therapy or gene vaccination, a tumor-associated antigen or an allergen.
Aspect 6 the kit of any one of aspects 1 to 5, wherein the fumarate-related disease or disorder is an autoimmune disorder, a demyelinating disorder, a graft rejection, or a cancer, preferably a demyelinating disorder.
Some preferred examples of autoimmune fumarate-related diseases and disorders are: multiple Sclerosis (MS), neuromyelitis optica (NMO), NMO induced preferably by MOG (i.e. MO caused by anti-MOG antibodies or MOG autoantigens), psoriasis, rheumatoid Arthritis (RA), polyarthritis, asthma, atopic dermatitis, scleroderma, ulcerative colitis, juvenile diabetes (juveline diabetes), thyroiditis (tyroiditis), grave's disease, systemic lupus erythematosus (Systemic Lupus Erythromatosis, SLE), sjogren's syndrome @syndrome), pernicious anemia (anemia perniciosa), chronic active hepatitis, transplant rejection, and cancer.
In some embodiments, the demyelinating disorder is selected from: multiple Sclerosis (MS), neuromyelitis optica (NMO), optic neuritis, acute disseminated encephalomyelitis, baluo's Disease, HTLV-I related myelopathy, hilder's Disease, transverse myelitis, idiopathic inflammatory demyelinating diseases, vitamin B12-induced central nervous system neuropathy, central myelination of the bridge, myelopathy including tuberculosis, leukodystrophies such as adrenoleukodystrophy, leukoencephalopathy such as progressive multifocal leukoencephalopathy (Progressive multifocal leukoencephalopathy, PML), vanishing leukopathy and rubella-induced mental retardation.
In some preferred embodiments, the demyelinating disorder is caused or exacerbated by MOG autoantigens and/or anti-MOG antibodies, and is therefore selected from the group consisting of: multiple Sclerosis (MS), neuromyelitis optica (NMO), optic neuritis, acute disseminated encephalomyelitis, transverse myelitis, adrenoleukodystrophy, vanishing leukopathy and rubella-induced mental retardation. In some more preferred embodiments, the demyelinating disorder is Multiple Sclerosis (MS) or neuromyelitis optica (NMO). In certain embodiments, the MS is selected from clinically isolated syndrome (Clinically Isolated Syndrome, CIS), relapsing-remitting MS (RRMS), secondary progressive MS (secondary progressive MS, SPMS), primary progressive MS (primary progressive MS, PPMS), acute fulminant multiple sclerosis, and radiological isolated syndrome (radiology isolated syndrome, RIS) of suspected MS.
Aspect 7 the kit of any one of aspects 1 to 6, wherein the fumarate-related disease or disorder is MS, and wherein the autoantigen is selected from the group consisting of: myelin Oligodendrocyte Glycoprotein (MOG), myelin basic protein (Myelin basic protein, MBP), proteolipid protein (Proteolipid protein, PLP), myelin-associated antigen (myelin-associated antigen, MAG), oligodendrocyte-specific protein (OSP), myelin-associated Oligodendrocyte basic protein (myelin-associated Oligodendrocyte basic protein, MOBP), 2',3' -cyclic nucleotide 3' -phosphodiesterase (CNPase), S100 β protein and transaldolase H, preferably MOG; or wherein the fumarate-related disease or disorder is a MOG autoantigen-induced disease or disorder, preferably MS or NMO, wherein the antigenic protein is MOG.
Aspect 8 the kit of any one of aspects 1 to 7, wherein the fumarate-related disease or disorder is Rheumatoid Arthritis (RA), and wherein the antigenic protein is selected from the group comprising: GRP78, HSP60, 60kDa chaperonin 2, gelsolin, chitinase-3-like protein 1, cathepsin S, serum albumin, focal adhesion proteins and cathepsin D.
Aspect 9 the kit of any one of aspects 1 to 8, wherein the fumarate-related disease or disorder is psoriasis, and wherein the antigenic protein is selected from the group consisting of: ADAMTSL5, PLA2G4D, keratins such as keratin 14 or 17, antigens from wheat (Triticum aestivum), pso p27, antimicrobial peptides, cetrorofil defensin 1 and LL37, preferably LL37.
Aspect 10 the kit of any one of aspects 1 to 9, wherein the (self) antigen involved in a fumarate-related disease or disorder does not naturally comprise an oxidoreductase motif within the 11 amino acids of the N-or C-terminus adjacent to the epitope.
Aspect 11. The kit of aspect 10, wherein in the immunogenic peptide, the epitope does not naturally comprise an oxidoreductase motif in its sequence.
Aspect 12 the kit according to any one of aspects 1 to 11, wherein in the immunogenic or tolerogenic peptide the T-cell epitope is an MHC class I or class II T-cell epitope or a NKT-cell epitope.
MHC class II epitopes are typically 7 to 20 amino acids in length, more typically 8 to 20 or 9 to 20 amino acids in length, even more preferably 7 to 17, 8 to 17, 9 to 17, 10 to 17, 11 to 17, 12 to 17, 13 to 17 amino acids, e.g. 14 to 16 amino acids in length. Peptides that bind to MHC class II molecules may also be longer, as these peptides are located in an extended conformation along an MHC class II peptide binding groove (groove) that is open at both ends (unlike an MHC class I peptide binding groove). Peptides are held in place primarily by contacting the backbone atoms with conserved residues arranged in the peptide binding groove.
MHC class I T cell epitopes are typically 7 to 13 amino acids in length, more preferably 8 to 10 amino acids in length. Peptide binding is stabilized at both ends by contact between atoms in the peptide backbone and invariant sites in the peptide binding groove of all MHC class I molecules. At both ends of the groove are invariant sites for the amino-and carboxy-termini of the binding peptide. The change in peptide length is accommodated by a kink in the peptide backbone, typically at a proline or glycine residue, which allows flexibility of the chain;
An NKT cell epitope may be recognized and bound by a receptor at the cell surface of NKT cells, in particular by a CD1d molecule. Such epitopes are typically 7 to 20 amino acids in length, more typically 7 to 17 amino acids in length, even more preferably 8 to 17, 9 to 17, 10 to 17, 11 to 17, 12 to 17, 13 to 17 amino acids, e.g. 14 to 16 amino acids in length. Such epitopes typically have the motif [ FWHY ] -XX- [ ILMV ] -XX- [ FVVTHY ] [ SEQ ID NO:51] or [ FW ] -XX- [ ILMV ] -XX- [ FW ] [ SEQ ID NO:52].
Aspect 13 the kit of any one of aspects 1 to 12, wherein the T cell epitope is an immunodominant epitope, a subdominant epitope, a cryptic epitope or a secondary epitope, preferably an immunodominant or subdominant epitope, more preferably an immunodominant epitope.
Aspect 14 the kit according to any one of aspects 2 to 13, wherein in the immunogenic peptide the oxidoreductase motif is located at the N-terminus of the linker or epitope, or at the C-terminus of the linker or epitope, preferably at the N-terminus of the linker or epitope, and/or wherein the oxidoreductase motif is located at the N-terminus or the C-terminus of the immunogenic peptide, preferably wherein Z corresponds to the N-terminus or the C-terminus of the immunogenic peptide.
Aspect 15: the kit according to any one of aspects 1 to 14, wherein in the immunogenic or tolerogenic peptide the T-cell epitope of an antigen protein is a NKT-cell epitope or an MHC class II T-cell epitope, preferably wherein when the T-cell epitope of an antigen protein is a NKT-cell epitope it is 7 to 25 amino acids in length; or wherein when said T cell epitope of the antigen protein is an MHC class II T cell epitope, it is 9 to 25 amino acids in length.
Aspect 16: the kit of any one of aspects 1 to 15, wherein the immunogenic or tolerogenic peptide is 7 to 50 amino acids in length, and/or wherein the immunogenic or tolerogenic peptide comprising an MHC class II T cell epitope is 9 to 50 amino acids in length.
Aspect 17 the kit of any one of aspects 2 to 16, wherein in the immunogenic peptide the linker between the oxidoreductase motif and the T cell epitope has 0 to 4 amino acids.
Aspect 18 the kit according to any one of aspects 2 to 17, wherein in the immunogenic peptide there is a sequence Z as defined in aspect 2 m -[CST]-X n -C-or Z m -C-X n -[CST]-said oxidoreductase motif is selected from the following amino acid motifs:
(a) Z as defined in aspect 2 m -[CST]-X n -C-or Z m -C-X n -[CST]-, wherein n is 0; and:
wherein m is an integer selected from 0 to 3,
wherein Z is any amino acid, preferably a basic amino acid selected from the group consisting of: H. k, R and a non-natural basic amino acid as defined herein, such as L-ornithine, preferably K or H.
In some preferred embodiments of motif (a), m is 1 or 2, and Z is a basic amino acid selected from the group consisting of: H. k, R and a non-natural basic amino acid as defined herein, such as L-ornithine, preferably K or H.
Some particularly preferred but non-limiting examples of such motifs are:
CC, KCC, KKCC (SEQ ID NO: 53), RCC, RRCC (SEQ ID NO: 54), RKCC (SEQ ID NO: 55), or KRCC (SEQ ID NO: 56);
(b) Z as defined in aspect 2 m -[CST]-X n -C-or Z m -C-X n -[CST]-, wherein n is 1,
wherein X is any amino acid, preferably a basic amino acid selected from the group consisting of: H. k, R and unnatural basic amino acids such as L-ornithine, preferably K or R, most preferably R,
wherein m is an integer selected from 0 to 3,
wherein Z is any amino acid, preferably a basic amino acid selected from the group consisting of: H. k, R and a non-natural basic amino acid as defined herein, such as L-ornithine, preferably K or H.
In some preferred embodiments of motif (b), m is 1 or 2, and Z is a basic amino acid selected from the group consisting of: H. k, R and a non-natural basic amino acid as defined herein, such as L-ornithine, preferably K or H.
Some particularly preferred but non-limiting examples of such motifs are:
CRC, CKC, KCXC (SEQ ID NO: 57), KKKCXC (SEQ ID NO: 58), RCXC (SEQ ID NO: 59), RRCXC (SEQ ID NO: 60), RKCXC (SEQ ID NO: 61), KCXC (SEQ ID NO: 52), KCKCKC (SEQ ID NO: 63), KKKCKC (SEQ ID NO: 64), KCRC (SEQ ID NO: 65), KKCCRC (SEQ ID NO: 66), RCRC (SEQ ID NO: 67), RRCRC (SEQ ID NO: 68), RKCKC (SEQ ID NO: 69), or KCKC (SEQ ID NO: 70);
(c) Z as defined in aspect 2 m -[CST]-X n -C-or Z m -C-X n -[CST]-, where n is 2, thereby producing an internal X within the oxidoreductase motif 1 X 2 Amino acid conjugate, wherein m is an integer selected from 0 to 3, wherein Z is any amino acid, preferably a basic amino acid selected from the group consisting of: H. k, R and a non-natural basic amino acid as defined herein, such as L-ornithine, preferably K or H. Preferred are motifs wherein m is 1 or 2.
In some preferred embodiments, m is 1 and Z is a basic amino acid selected from the group consisting of: H. k or R or a non-natural basic amino acid as defined herein, such as L-ornithine, preferably K or H.
In some preferred embodiments, X 1 And X 2 Each independently may be any amino acid selected from the group consisting of: g, A, V, L, I, M, F, W, P, S, T, C, Y, N, Q, D, E, K, R, and H, or an unnatural amino acid. Preferably, X in the motif 1 And X 2 Is any amino acid other than C, S or T. In a specific embodiment, X in the motif 1 Or X 2 At least one of which is a basic amino acid selected from the group consisting of: H. k or R or a non-natural basic amino acid as defined herein, such as L-ornithine. In another embodiment, X in the motif 1 Or X 2 At least one of which is P or Y. Internal X within the oxidoreductase motif 1 X 2 Some specific examples of amino acid conjugates are: PY, HY, KY, RY, PH, PK, PR, HG, KG, RG, HH, HK, HR, GP, HP, KP, RP, GH, GK, GR, GH, KH, and RH.
Some particularly preferred motifs of this type are:
HCPYC (SEQ ID NO: 71), KCPYC (SEQ ID NO: 72), RCPYC (SEQ ID NO: 73), HCGHC (SEQ ID NO: 74), KCGHC (SEQ ID NO: 75), RCGHC (SEQ ID NO: 76), KHCPYC (SEQ ID NO: 77), KCPYC (SEQ ID NO: 78), KCPYC (SEQ ID NO: 79), KHCGHC (SEQ ID NO: 80), KKCCGHC (SEQ ID NO: 81), and KRCGGC (SEQ ID NO: 82);
(d) Z as defined in aspect 2 m -[CST]-X n -C-or Z m -C-X n -[CST]-, where n is 3, thereby producing an internal X within the oxidoreductase motif 1 X 2 X 3 Amino acid segment, wherein m is an integer selected from 0 to 3, wherein Z is any amino acid, preferably a basic amino acid selected from the group consisting of: H. k, R and a non-natural basic amino acid as defined herein, such as L-ornithine, preferably K or H. Preferred are motifs wherein m is 1 or 2.
In some embodiments, X 1 、X 2 And X 3 Each independently may be any amino acid selected from the group consisting of: g, A, V, L, I, M, F, W, P, S, T, C, Y, N, Q, D, E, K, R, and H, or an unnatural amino acid. Preferably, X in the motif 1 、X 2 And X 3 Is any amino acid other than C, S or T. In a specific embodiment, X in the motif 1 、X 2 Or X 3 At least one of which is a basic amino acid selected from the group consisting of: H. k or R or a non-natural basic amino acid as defined herein, such as L-ornithine.
Internal X within the oxidoreductase motif 1 X 2 X 3 Some specific examples of amino acid segments are: XPY, PXY and PYX, wherein X may be any ammoniaBasic acids, preferably basic amino acids such as K, R or H or unnatural basic amino acids such as L-ornithine. Some non-limiting examples are:
KPY, RPY, HPY, GPY, APY, VPY, LPY, IPY, MPY, FPY, WPY, PPY, SPY, TPY, CPY, YPY, NPY, QPY, DPY, EPY, and KPY; or alternatively
PKY, PRY, PHY, PGY, PAY, PVY, PLY, PIY, PMY, PFY, PWY, PPY, PSY, PTY, PCY, PYY, PNY, PQY, PDY, PEY, and PLY; or alternatively
PYK, PYR, PYH, PYG, PYA, PYV, PYL, PYI, PYM, PYF, PYW, PYP, PYS, PYT, PYC, PYY, PYN, PYQ, PYD, PYE, and PYL;
XHG, HXG and HGX, wherein X may be any amino acid, for example in the following:
KHG, RHG, HHG, GHG, AHG, VHG, LHG, IHG, MHG, FHG, WHG, PHG, SHG, THG, CHG, YHG, NHG, QHG, DHG, EHG, and KHG; or alternatively
HKG, HRG, HHG, HGG, HAG, HVG, HLG, HIG, HMG, HFG, HWG, HPG, HSG, HTG, HCG, HYG, HNG, HQG, HDG, HEG, and HLG; or alternatively
HGK, HGR, HGH, HGG, HGA, HGV, HGL, HGI, HGM, HGF, HGW, HGP, HGS, HGT, HGC, HGY, HGN, HGQ, HGD, HGE, and HGL;
XGP, GXP, and GPX, where X can be any amino acid, for example in the following:
KGP, RGP, HGP, GGP, AGP, VGP, LGP, IGP, MGP, FGP, WGP, PGP, SGP, TGP, CGP, YGP, NGP, QGP, DGP, EGP, and KGP; or alternatively
GKP, GRP, GHP, GGP, GAP, GVP, GLP, GIP, GMP, GFP, GWP, GPP, GSP, GTP, GCP, GYP, GNP, GQP, GDP, GEP, and GLP; or alternatively
GPK, GPR, GPH, GPG, GPA, GPV, GPL, GPI, GPM, GPF, GPW, GPP, GPS, GPT, GPC, GPY, GPN, GPQ, GPD, GPE, and GPL;
XGH, GXH and GHX, wherein X may be any amino acid, for example in the following:
KGH, RGH, HGH, GGH, AGH, VGH, LGH, IGH, MGH, FGH, WGH, PGH, SGH, TGH, CGH, YGH, NGH, QGH, DGH, EGH, and KGH; or alternatively
GKH, GRH, GHH, GGH, GAH, GVH, GLH, GIH, GMH, GFH, GWH, GPH, GSH, GTH, GCH, GYH, GNH, GQH, GDH, GEH, and GLH; or alternatively
GHK, GHR, GHH, GHG, GHA, GHV, GHL, GHI, GHM, GHF, GHW, GHP, GHS, GHT, GHC, GHY, GHN, GHQ, GHD, GHE, and GHL;
XGF, GXF, and GFX, where X may be any amino acid, for example, in the following:
KGF, RGF, HGF, GGF, AGF, VGF, LGF, IGF, MGF, FGF, VVGF, PGF, SGF, TGF, CGF, YGF, NGF, QGF, DGF, EGF, and KGF; or alternatively
GKF, GRF, GHF, GGF, GAF, GVF, GLF, GIF, GMF, GFF, GVVF, GPF, GSF, GTF, GCF, GYF, GNF, GQF, GDF, GEF, and GLF; or alternatively
GFK, GFR, GFH, GFG, GFA, GFV, GFL, GFI, GFM, GFF, GFW, GFP, GFS, GFT, GFC, GFY, GFN, GFQ, GFD, GFE, and GFL;
XRL, RXL and RLX, where X can be any amino acid, for example in the following:
KRL, RRL, HRL, GRL, ARL, VRL, LRL, IRL, MRL, FRL, WRL, PRL, SRL, TRL, CRL, YRL, NRL, QRLRL, DRL, ERL, and KRL; or alternatively
GKF, GRF, GHF, GGF, GAF, GVF, GLF, GIF, GMF, GFF, GVVF, GPF, GSF, GTF, GCF, GYF, GNF, GQF, GDF, GEF, and GLF; or alternatively
RLK, RLR, RLH, RLG, RLA, RLV, RLL, R L, RLM, RLF, RLW, RLP, RLS, RLT, RLC, RLY, RLN, RLQ, RLD, RLE, and RLL;
XHP, HXP and HPX, where X can be any amino acid, for example in the following:
KHP, RHP, HHP, GHP, AHP, VHP, LHP, IHP, MHP, FHP, WHP, PHP, SHP, THP, CHP, YHP, NHP, QHP, DHP, EHP, and KHP; or alternatively
HKP, HRP, HHP, HGP, HAF, HVF, HLF, HIF, HMF, HFF, HWF, HPF, HSF, HTF, HCF, HYP, HNF, HQF, HDF, HEF, and HLP; or alternatively
HPK, HPR, HPH, HPG, HPA, HPV, HPL, HPI, HPM, HPF, HPW, HPP, HPS, HPT, HPC, HPY, HPN, HPQ, HPD, HPE, and HPL;
some particularly preferred examples are:
CRPP YC (SEQ ID NO: 83), KCRPYC (SEQ ID NO: 84), KHCRPC (SEQ ID NO: 85), RCRPYC (SEQ ID NO: 86), HCRPYC (SEQ ID NO: 87), CPRYC (SEQ ID NO: 88), KCPRYC (SEQ ID NO: 89), RCPRYC (SEQ ID NO: 90), HCPRYC (SEQ ID NO: 91), CPYRC (SEQ ID NO: 92), KCPYRC (SEQ ID NO: 93), RCPYRC (SEQ ID NO: 94), HCPYRC (SEQ ID NO: 95), CKPYC (SEQ ID NO: 96), KCKPYC (SEQ ID NO: 97), RCKPYC (SEQ ID NO: 98), HCKPYC (SEQ ID NO: 99), CPKYC (SEQ ID NO: 100), KCYC (SEQ ID NO: 101), RCYC (SEQ ID NO: 102), HCPKYC (SEQ ID NO: 103), YKC (SEQ ID NO: 104), PKKC (SEQ ID NO: 106) and HCPYC (SEQ ID NO: 106);
(e) Z as defined in aspect 2 m -[CST]-X n -C-or Z m -C-X n -[CST]-, where n is 4, thereby producing an internal X within the oxidoreductase motif 1 X 2 X 3 X 4 (SEQ ID NO: 111) amino acid segment, wherein m is an integer selected from 0 to 3, wherein Z is any amino acid, preferably a basic amino acid selected from the group consisting of: H. k, R and a non-natural basic amino acid as defined herein, such as L-ornithine, preferably K or H. Preferred are motifs wherein m is 1 or 2. X is X 1 、X 2 、X 3 And X 4 Each independently may be any amino acid selected from the group consisting of: g, A, V, L, I, M, F, W, P, S, T, C, Y, N, Q, D, E, K, R, and H, or an unnatural amino acid as defined herein. Preferably, X in the motif 1 、X 2 、X 3 And X 4 Is any amino acid other than C, S or T. In a specific embodiment, X in the motif 1 、X 2 、X 3 Or X 4 At least one of them is alkaline ammonia selected from the followingBase acid: H. k or R or a non-natural basic amino acid as defined herein.
Some specific examples are: LAVL (SEQ ID NO: 108), TVQA (SEQ ID NO: 109) or GAVH (SEQ ID NO: 110), and variants thereof, for example: x is X 1 AVL,LX 2 VL,LAX 3 L, or LAVX 4 ;X 1 VQA,TX 2 QA,TVX 3 A, or TVQX 4 ;X 1 AVH,GX 2 VH,GAX 3 H, or GAVX 4 (corresponding to SEQ ID NOS: 112 to 122); wherein X is 1 、X 2 、X 3 And X 4 Each independently may be any amino acid selected from the group consisting of: g, A, V, L, I, M, F, W, P, S, T, C, Y, N, Q, D, E, K, R, and H, or a non-natural basic amino acid as defined herein;
(f) Z as defined in aspect 2 m -[CST]-X n -C-or Z m -C-X n -[CST]-, where n is 5, thereby producing an internal X within the oxidoreductase motif 1 X 2 X 3 X 4 X 5 (SEQ ID NO: 125) amino acid segment, wherein m is an integer selected from 0 to 3, wherein Z is any amino acid, preferably a basic amino acid selected from the group consisting of: H. k, R and a non-natural basic amino acid as defined herein, such as L-ornithine, preferably K or H. Preferred are motifs wherein m is 1 or 2. X is X 1 、X 2 、X 3 、X 4 And X 5 Each independently may be any amino acid selected from the group consisting of: g, A, V, L, I, M, F, W, P, S, T, C, Y, N, Q, D, E, K, R, and H, or an unnatural amino acid. Preferably, X in the motif 1 、X 2 、X 3 、X 4 And X 5 Is any amino acid other than C, S or T. In a specific embodiment, X in the motif 1 、X 2 、X 3 、X 4 Or X 5 At least one of which is a basic amino acid selected from the group consisting of: H. k or R or a non-natural basic amino acid as defined herein.
Some specific examples are: PAFPL (SEQ ID NO: 123) or DQGGE (SEQ ID NO: 124), and variants thereof such as :X 1 AFPL,PX 2 FPL,PAX 3 PL,PAFX 4 L, or PAFPX 5 ;X 1 QGGE,DX 2 GGE,DQX 3 GE,DQGX 4 E, or DQGGX 5 (corresponding to SEQ ID NOS: 126 to 135), wherein X 1 、X 2 、X 3 、X 4 And X 5 Each independently may be any amino acid selected from the group consisting of: g, A, V, L, I, M, F, W, P, S, T, C, Y, N, Q, D, E, K, R, and H, or an unnatural amino acid as defined herein;
(g) Z as defined in aspect 2 m -[CST]-X n -C-or Z m -C-X n -[CST]-, where n is 6, thereby producing an internal X within the oxidoreductase motif 1 X 2 X 3 X 4 X 5 X 6 (SEQ ID NO: 137) amino acid segment, wherein m is an integer selected from 0 to 3, wherein Z is any amino acid, preferably a basic amino acid selected from the group consisting of: H. k, R and a non-natural basic amino acid as defined herein, such as L-ornithine, preferably K or H, and wherein B is any amino acid. Preferred are motifs wherein m is 1 or 2. X is X 1 、X 2 、X 3 、X 4 、X 5 And X 6 Each independently may be any amino acid selected from the group consisting of: g, A, V, L, I, M, F, W, P, S, T, C, Y, N, Q, D, E, K, R, and H, or an unnatural amino acid. Preferably, X in the motif 1 、X 2 、X 3 、X 4 、X 5 And X 6 Is any amino acid other than C, S or T.
In a specific embodiment, X in the motif 1 、X 2 、X 3 、X 4 、X 5 Or X 6 At least one of which is a basic amino acid selected from the group consisting of: H. k or R or a non-natural basic amino acid as defined herein.
Some specific examples are: DIADKY (SEQ ID NO: 136), or variants thereof, for example: x is X 1 IADKY,DX 2 ADKY,DIX 3 DKY,DIAX 4 KY,DIADX 5 Y, or DIADKX 6 (corresponding to)In SEQ ID NO:138 to 143), wherein X 1 、X 2 、X 3 、X 4 、X 5 And X 6 Each independently may be any amino acid selected from the group consisting of: g, A, V, L, I, M, F, W, P, S, T, C, Y, N, Q, D, E, K, R, and H, or a non-natural basic amino acid as defined herein; or alternatively
(h) Z as defined in aspect 2 m -[CST]-X n -C-or Z m -C-X n -[CST]-, where n is 0 to 6 and where m is 0, and where C or [ CST ]]One of the residues has been modified to carry an acetyl, methyl, ethyl or propionyl group on the N-terminal amide or on the C-terminal carboxyl group of the amino acid residue of the motif (SEQ ID NOS: 144 to 163).
In some preferred embodiments of such motifs, n is 2 and m is 1 or 2, wherein internal X 1 X 2 Each independently may be any amino acid selected from the group consisting of: g, A, V, L, I, M, F, W, P, S, T, C, Y, N, Q, D, E, K, R, and H, or an unnatural amino acid. Preferably, X in the motif 1 And X 2 Is any amino acid other than C, S or T. In another example, X in the motif 1 Or X 2 At least one of which is a basic amino acid selected from the group consisting of: H. k or R or a non-natural basic amino acid as defined herein, such as L-ornithine. In another example of a motif, X in the motif 1 Or X 2 At least one of which is P or Y. Internal X within the oxidoreductase motif 1 X 2 Some specific non-limiting examples of amino acid conjugates are: PY, HY, KY, RY, PH, PK, PR, HG, KG, RG, HH, HK, HR, GP, HP, KP, RP, GH, GK, GR, GH, KH, and RH. Preferably, the modification results in N-terminal acetylation of the first cysteine (N-acetyl-cysteine) in the motif.
Aspect 19 the kit of any one of aspects 1 to 18, wherein the epitope is derived from a Myelin Oligodendrocyte Glycoprotein (MOG) antigen amino acid sequence. More preferably, the epitope is selected from the group comprising amino acid residues: consists of the following SEQ ID NOs: 208 from amino acid residues 40 to 60, 41 to 55, 43 to 57, 44 to 58, 45 to 59, and 35 to 55 of the mature MOG amino acid sequence defined by seq id no:
such as those selected from the group comprising:
or a combination thereof.
Aspect 20 the kit according to any one of aspects 1 to 18, wherein the epitope in the immunogenic or tolerogenic peptide is derived from the myelin proteolipid protein (also known as proteolipid protein (PLP) or lipophilic (1 ipoline)) antigenic amino acid sequence. More preferably, with reference to patent application WO2014111841, the epitope is selected from the group comprising amino acid residues: consists of the following SEQ ID NOs: 207 Amino acid residues 36 to 61, 179 to 206, 207 to 234, 39 to 57, 180 to 198, 208 to 222, 39 to 53, 42 to 56, 43 to 57, 180 to 194, 181 to 195, 182 to 196, 183 to 197, 184 to 198, 208 to 222, 36 to 61, 179 to 206, and 207 to 234 of the PLP amino acid sequence defined by (UniProtKB-P60201 (mypr_human)).
Such as those selected from the group comprising:
PLP bits 36 to 61: HEALTGTEKLIETYFSKNYQDYEYLI (SEQ ID NO: 209);
PLP bits 179 to 206: TWTTCQSIAFPSKTSASIGSLCADARMY (SEQ ID NO: 210);
PLP bits 207 to 234: GVLPWNAFPGKVCGSNLLSICKTAEFQM (SEQ ID NO: 211)
PLP positions 39 to 57: LTGTEKLIETYFSKNYQDY (SEQ ID NO: 212)
PLP bits 180 to 198: WTTCQSIAFPSKTSASIGS (SEQ ID NO: 213)
PLP bits 208 to 222: VLPWNAFPGKVCGSN (SEQ ID NO: 214)
PLP bits 39 to 53: LTGTEKLIETYFSKN (SEQ ID NO: 215)
PLP bits 42 to 56: TEKLIETYFSKNYQD (SEQ ID NO: 216)
PLP bits 43 to 57: EKLIETYFSKNYQDY (SEQ ID NO: 217)
PLP bits 180 to 194: WTTCQSIAFPSKTSA (SEQ ID NO: 218)
PLP bits 181 to 195: TTCQSIAFPSKTSAS (SEQ ID NO: 219)
PLP positions 182 to 196: TCQSIAFPSKTSASI (SEQ ID NO: 220)
PLP positions 183 to 197: CQSIAFPSKTSASIG (SEQ ID NO: 221)
PLP bits 184 to 198: QSIAFPSKTSASIGS (SEQ ID NO: 222)
PLP bits 208 to 222: VLPWNAFPGKVCGSN (SEQ ID NO: 223)
PLP bits 36 to 61: HEALTGTEKLIETYFSKNYQDYEYLI (SEQ ID NO: 224)
PLP bits 179 to 206: TWTTCQSIAFPSKTSASIGSLCADARMY (SEQ ID NO: 225) and
PLP bits 207 to 234: GVLPWNAFPGKVCGSNLLSICKTAEFQM (SEQ ID NO: 226) or combinations thereof.
Aspect 21 the kit of any one of aspects 1 to 18, wherein the epitope in the immunogenic peptide or tolerogenic peptide is derived from Myelin Basic Protein (MBP) antigen amino acid sequence. More preferably, the MBP epitope is selected from the group comprising:
LSRFSWGAEGQRPG(SEQ ID NO:254),
or a combination thereof,
or any one or more of the fragments defined by amino acid residues 30 to 44, 80 to 94, 83 to 99, 81 to 95, 82 to 96, 83 to 97, 84 to 98, 110 to 124, 130 to 144, 131 to 158, 131 to 145, 140 to 148, 142 to 152, 132 to 146, 134 to 148, 135 to 149, 136 to 150, 137 to 151, 138 to 152, 139 to 153, 140 to 154, and 133 to 147 of an MBP amino acid sequence defined by the following SEQ ID NOs: 255 (UniProtKB-P02686-5 (mbp_command)), defined by:
in a preferred embodiment, the MBP epitope is selected from the group consisting of SEQ ID NO:227 to 230, or any 2, 3 or 4 combinations thereof. Particularly preferred is reference to streeteter et al, 2015,Neurol Neuroimmunol Neuroinflamm.2015Jun;2 (3): e93 reports the use of SEQ ID NO: clinical trials of a specific mixture of all 4 peptides defined in 227 to 230 (termed ATX-MS-1467).
Aspect 22 the kit of any one of aspects 2 to 21, wherein the immunogenic peptide has an oxidoreductase motif comprising the sequence: the CC is used to determine the specific parameters of the cell,
KCC, RCC, CRC, CKC, KCRC (SEQ ID NO: 65), KCKCKC (SEQ ID NO: 63), RCKC (SEQ ID NO: 171), RCRC (SEQ ID NO: 67), CPYC (SEQ ID NO: 172), HCPYC (SEQ ID NO: 71), KCPYC (SEQ ID NO: 72), RCPYC (SEQ ID NO: 73), CRPYC (SEQ ID NO: 83), CPRYC (SEQ ID NO: 88), CPYRC (SEQ ID NO: 92), CKPYC (SEQ ID NO: 96), CPKYC (SEQ ID NO: 100), CPYKC (SEQ ID NO: 104), RCRPYC (SEQ ID NO: 86), RCPRYC (SEQ ID NO: 90), RCPYC (SEQ ID NO: 94), RCKPYC (SEQ ID NO: 98), RCYC (SEQ ID NO: 102), RCPYC (SEQ ID NO: 106), RPYC (SEQ ID NO: 84), KCYC (SEQ ID NO: 89), PKKC (SEQ ID NO: 101) or PKKC (SEQ ID NO: 97)
Aspect 23 the kit of any one of aspects 2 to 22, wherein the immunogenic peptide has a linker between the oxidoreductase motif and the T cell epitope with the sequence VRY.
Aspect 24 the kit of any one of aspects 2 to 23, wherein the immunogenic peptide comprises or consists essentially of the amino acid sequence:
Aspect 25 the kit according to any one of aspects 1 to 24 for use in the treatment of a fumarate-related disease or disorder, ameliorating symptoms of a fumarate-related disease or disorder and/or preventing a fumarate-related disease or disorder, preferably selected from the group consisting of: autoimmune disorders, demyelinating disorders, graft rejection, or cancer. Some preferred examples of such diseases and conditions are: multiple Sclerosis (MS), neuromyelitis optica (NMO), psoriasis, rheumatoid Arthritis (RA), asthma, atopic dermatitis, scleroderma, ulcerative colitis, cancer and transplant rejection.
Aspect 26. The kit for use according to aspect 25, wherein the fumarate compound and the immunogenic or tolerogenic peptide are administered simultaneously, sequentially and/or separately.
Aspect 26 the kit for use according to aspect 25 or 26, wherein the immunogenic peptide or tolerogenic peptide is administered prior to the fumarate composition, preferably at least 12 hours prior to the start of treatment with the fumarate compound, for example at least 24 hours prior, more preferably at least 1 to 20 days or at least 1 to 10 days prior. In certain embodiments, administration (injection) of the immunogenic peptide or tolerogenic peptide is repeated once, twice, three times, four times, five times, or six times, each time 1 to 20 days apart, or 1 to 10 days apart.
Aspect 27. The kit for use according to aspects 25 or 26, wherein the following chronologically sequential treatment regimen is applied:
1) Fumarate compound treatment comprising administering the fumarate compound once or twice daily for a period of at least 4 weeks, for example for a period of at least 1, 2 or 3 months to as long as 4, 5 or 6 months;
2) Initiating immunogenic peptide or tolerogenic peptide treatment (injection) for at least 1, 2, 3 or 4 times, each time at intervals of 1 to 10 days, for example 5 to 9 days, for example about 7 days, optionally while maintaining fumarate treatment as in 1) during peptide treatment;
optional step 3) wherein the fumarate compound treatment as in 1) is maintained when needed after steps 1) and 2) are completed;
the optional step 4) may be performed with one or more boost administrations of the immunogenic peptide or tolerogenic peptide, which are performed 1, 2 or 3 months after the last administration of the immunogenic peptide, each boost re-administration being at intervals of 1 to 20 days or 1 to 10 days, for example 5 to 9 days, for example about 7 days.
Some preferred dosage regimens for fumarate compounds and immunogenic or tolerogenic peptides are defined in more detail elsewhere in this application.
A particularly preferred treatment regimen for DMF fumarate compounds is 120mg twice daily for the first 7 days, after which it is raised to 240mg twice daily.
One specific but non-limiting dosage regimen of an immunogenic or tolerogenic peptide as defined herein is 50 to 1500 μg, preferably 450 to 1500 μg. The dosage regimen may comprise simultaneous or sequential administration in a single dose or in 2, 3, 4, 5, 6 or more doses.
Aspect 28 the kit for use according to any one of aspects 25 to 27, wherein the treatment with fumarate is performed daily or twice daily, and/or wherein the treatment with the immunogenic peptide or tolerogenic peptide is performed 1 to 6 times, such as 1 to 4 times, preferably every 5 to 9 days, such as about every 7 days.
Aspect 29 the kit for use according to any one of aspects 25 to 28, wherein the fumarate composition is administered before, during and optionally after administration of the immunogenic or tolerogenic peptide.
Aspect 30 the kit for use according to any one of aspects 25 to 29, wherein the fumarate compound is administered orally once or twice a day, and/or wherein the immunogenic peptide or tolerogenic peptide is administered by subcutaneous injection.
Aspect 30. A method of treating, ameliorating the symptoms of, and/or preventing a fumarate-related disease or disorder in a patient in need thereof, comprising the step of administering an effective amount of a dosage unit of the kit of any one of aspects 1 to 24.
Aspect 31 the method of aspect 30, wherein the fumarate-related disease or disorder is an autoimmune disorder, a demyelinating disorder, a graft rejection, or a cancer. Some preferred examples of such diseases and conditions are: multiple Sclerosis (MS), psoriasis, neuromyelitis optica (NMO), rheumatoid Arthritis (RA), polyarthritis, asthma, atopic dermatitis, scleroderma, ulcerative colitis, juvenile diabetes, thyroiditis, grave's disease, systemic Lupus Erythematosus (SLE), sjogren's syndrome, pernicious anemia, chronic active hepatitis, transplant rejection and cancer.
Aspect 32 the method of aspects 30 or 31, wherein the fumarate compound and the immunogenic or tolerogenic peptide are administered simultaneously, sequentially and/or separately.
Aspect 33 the method of any one of aspects 30 to 32, wherein the immunogenic peptide or tolerogenic peptide is administered prior to the fumarate composition, preferably at least 12 hours, such as at least 24 hours, more preferably at least 1 to 20 days, such as at least 1 to 10 days, prior to the initiation of the treatment with the fumarate compound, such as 5 to 9 days, such as about 7 days, prior to the initiation of the treatment with the fumarate compound. In certain embodiments, administration (injection) of the immunogenic or tolerogenic peptide is repeated once, twice, three times, four times, five times or six times, each time at intervals of 1 to 20 days, such as 1 to 10 days, such as 5 to 9 days, such as about 7 days.
Alternatively, the following chronological treatment regimen is applied:
1) Fumarate compound treatment comprising administering the fumarate compound once or twice daily for a period of at least 4 weeks, for example for a period of at least 1, 2 or 3 months to as long as 4, 5 or 6 months;
2) Initiating immunogenic peptide or tolerogenic peptide treatment (injection) for at least 1, 2, 3 or 4 times, each time at intervals of 1 to 20 days, for example 1 to 10 days, for example 5 to 9 days, for example about 7 days, optionally while maintaining fumarate treatment as in 1) during peptide treatment;
optional step 3) wherein the fumarate compound treatment as in 1) is maintained when needed after steps 1) and 2) are completed;
the optional step 4) may be performed with one or more administration of the immunogenic or tolerogenic peptide 1, 2 or 3 months after the last administration of the immunogenic or tolerogenic peptide, each boost being given at an interval of 1 to 20 days, for example 1 to 10 days, for example 5 to 9 days, for example about 7 days.
Some preferred dosage regimens for fumarate compounds and immunogenic or tolerogenic peptides are defined in more detail elsewhere in this application. A particularly preferred treatment regimen for DMF fumarate compounds is 120mg twice daily for the first 7 days, after which it is raised to 240mg twice daily.
Aspect 34 the method according to any one of aspects 30 to 33, wherein the treatment with fumarate is performed daily or twice daily, and/or wherein the treatment with the immunogenic peptide or tolerogenic peptide is performed 1 to 6 times, such as 1 to 4 times, preferably every 5 to 9 days, such as about every 7 days.
The method of any one of aspects 30 to 34, wherein the fumarate composition is administered before, during, and optionally after administration of the immunogenic or tolerogenic peptide.
Aspect 40 the method of any one of aspects 30 to 35, wherein the fumarate compound is administered orally once or twice a day, and/or wherein the immunogenic peptide or tolerogenic peptide is administered by subcutaneous injection.
Aspect 41 preferably, the tolerogenic peptide is administered by: delivery via mucous membranes, for example, by nasal, oral, buccal, pulmonary, ocular, vaginal or rectal delivery; or by intradermal, transdermal or subcutaneous injection. Preferably, the tolerogenic peptide should be administered in soluble form in the absence of an adjuvant.
Aspect 42 preferably, the immunogenic peptide is administered by intradermal, transdermal or subcutaneous injection. Preferably, the immunogenic peptide should be administered in soluble form in the presence of an adjuvant.
Aspect 43. A nucleic acid encoding an immunogenic or tolerogenic peptide according to any of the aspects or examples disclosed herein, preferably selected from isolated deoxyribonucleic acid (desoxyribonucleic acid, DNA), plasmid DNA (pDNA), coding DNA (cDNA), ribonucleic acid (RNA), messenger RNA (mRNA), or modified forms thereof. In some embodiments, the nucleic acid may be part of an expression cassette, optionally incorporated into a (viral) vector or plasmid useful in gene therapy or may be present in the form of encapsulated or naked DNA or RNA for administration according to techniques known in the pharmaceutical and gene therapy arts.
Aspect 44. In any of the aspects disclosed herein that relate to methods of treatment or medical use of tolerogenic or immunogenic peptides, the peptides may also be administered as nucleic acids encoding the corresponding peptides according to aspect 43.
Aspect 45 in any one of the aspects described herein,
-when the fumarate-related disease or disorder is MS, the antigen is preferably recognized in the context of HLA-DRB1 x 15:01, HLA-DRB1 x 03:01, HLA-DRB1 x 04:01, HLA-DRB1 x 07:01, HLA DRB5 x 0101 or DQ6 types. More preferred are patients with HLA-DRB1 type 15:01;
-when the fumarate-related disease or disorder is NMO, the antigen is preferably recognized in the context of HLA-DRB1 x 03:01 or HLA-DPB1 x 05:01 (for asia); or alternatively
-when the fumarate-related disease or disorder is RA, the antigen is preferably recognized in the context of HLA-DRB1 x 01:01, 04:01 or 04:04.
Drawings
Fig. 1: blind evaluation of clinical EAE scores (0 to 5) performed daily from day 7 to day 28 is shown. Mice were prophylactically immunized with or without IMCY-0189, then MOG on day 0 35 to 55 Injection to induce EAE and treatment with or without BG-12 (see table 1 for details). The average clinical score for each group of mice was determined daily.
Fig. 2: AUC calculated from EAE scores shown in fig. 1 for each group of mice is shown. The significance differences were referenced as follows: * p <0.05, < p <0.01, < p <0.001, < p <0.0001.
Fig. 3: MMS calculated from EAE scores shown in fig. 1 for each group of mice are shown. The significance differences were referenced as follows: * p <0.05, < p <0.01, < p <0.001, < p <0.0001.
Fig. 4: the inflammatory levels of the groups of mice shown in table 1 are shown. Inflammatory lesions were counted for about 20 cells in each H & E stained section. When inflammatory infiltrates consisted of more than 20 cells, an estimate was made of how many 20-cell lesions were present. The significance differences were referenced as follows: * p <0.05, < p <0.01, < p <0.001, < p <0.0001.
Fig. 5: the demyelination levels of the groups of mice shown in table 1 are shown. Demyelination was scored in each anti-MBP (using immunohistochemistry) staining section. The demyelination score represents an estimate of the demyelination area of each slice. The significance differences were referenced as follows: * p <0.05, < p <0.01, < p <0.001, < p <0.0001.
Fig. 6: plasma neurofilament levels for each group of mice shown in table 1 are shown. In Quantix TM Pass through NF-lightThe assay dominance kit quantifies the level of neurofilament light (NF-L) protein. The significance differences were referenced as follows: * P is p<0.05,**p<0.01,***p<0.001,****p<0.0001。
Fig. 7: blind evaluation of clinical EAE scores (0 to 5) performed daily from day 7 to day 28 is shown. Mice were injected with MOG on day 0 35 to 55 To induce EAE and without IMCY-0189 or MOG in combination with BG-12 or without BG-12 35 to 55 By treatment or with IMCY-0189 or MOG 35 to 55 Therapeutic treatments were performed (see table 3 for details). The average clinical score for each group of mice was determined daily.
Fig. 8: AUC calculated from EAE scores shown in fig. 7 for each group of mice is shown. The significance differences were referenced as follows: * p <0.05, < p <0.01, < p <0.001, < p <0.0001.
Fig. 9: MMS calculated from EAE scores shown in fig. 7 for each group of mice are shown. The significance differences were referenced as follows: * p <0.05, < p <0.01, < p <0.001, < p <0.0001.
Fig. 10: blind evaluation of clinical EAE scores (0 to 5) performed daily from day 7 to day 28 is shown. Mice were injected with MOG on day 0 35 to 55 To induce EAE and without treatment with IMCY-0189, IMCY-0453 or IMCY-0455 or therapeutic treatment with IMCY-0189, IMCY-0453 or IMCY-0455 in combination with BG-12 or without BG-12 (see Table 4 for details). The average clinical score for each group of mice was determined daily.
Fig. 11: AUC calculated from EAE scores shown in fig. 10 for each group of mice is shown. The significance differences were referenced as follows: * p <0.05, < p <0.01, < p <0.001, < p <0.0001.
Fig. 12: MMS calculated from EAE scores shown in fig. 10 for each group of mice are shown. The significance differences were referenced as follows: * p <0.05, < p <0.01, < p <0.001, < p <0.0001.
Fig. 13: serum neurofilament levels for each group of mice shown in table 4 are shown. In Quantix TM Pass through NF-lightThe assay dominance kit quantifies the level of neurofilament light (NF-L) protein. The significance differences were referenced as follows: * P is p<0.05,**p<0.01,***p<0.001,****p<0.0001。
Fig. 14: blind evaluation of clinical EAE scores (0 to 5) performed daily from day 7 to day 28 is shown. Mice were injected with MOG on day 0 35 to 55 To induce EAE and, in combination with BG-12 or without BG-12, without treatment with IMCY-0189 or P4 or with IMCY-0189 or P4 (see Table 5 for details). The average clinical score for each group of mice was determined daily.
Fig. 15: AUC calculated from EAE scores shown in fig. 14 for each group of mice is shown. The significance differences were referenced as follows: * p <0.05, < p <0.01, < p <0.001, < p <0.0001.
Fig. 16: MMS calculated from EAE scores shown in fig. 14 for each group of mice are shown. The significance differences were referenced as follows: * p <0.05, < p <0.01, < p <0.001, < p <0.0001.
Fig. 17: serum neurofilament levels for each group of mice shown in table 5 are shown. In Quantix TM Pass through NF-lightThe assay dominance kit quantifies the level of neurofilament light (NF-L) protein. The significance differences were referenced as follows: * P is p<0.05,**p<0.01,***p<0.001,****p<0.0001。
Detailed Description
Unless the context clearly indicates otherwise, nouns without quantitative word modifications as used herein mean one and more. By way of example, an immunogenic peptide refers to one or more than one immunogenic peptide.
The terms "comprising" and "consisting of … …" are used herein synonymously with "including" or "comprising" and are inclusive or open-ended, and do not exclude additional, non-recited members, elements, or method steps. The term also encompasses embodiments that "consist essentially of … …" and "consist of … …".
As used herein, the term "for" as used in "formulation for treating a disease" shall also disclose a corresponding method of treatment and use of a corresponding formulation for the preparation of a medicament for treating a disease.
Recitation of numerical ranges by endpoints includes all numbers subsumed within that range and fractions subsumed therein, and the recited endpoints. The term "about" as used herein in reference to measurable values such as parameters, amounts, time intervals (temporal duration), etc., is intended to encompass variations of the specified value or +/-10% or less, preferably +/-5% or less, more preferably +/-1% or less, and still more preferably +/-0.1% or less relative to the specified value, within which such variations are suitable for implementation in the disclosed invention. It is to be understood that the value itself referred to by the modifier "about/approximately" is also specifically and preferably disclosed.
The term "any/any" when used in relation to an aspect, claim or embodiment as used herein refers to any single (i.e., any) and all combinations of the aspect, claim or embodiment concerned.
All references cited in this specification are incorporated herein by reference in their entirety. In particular, the teachings of all references specifically mentioned herein are incorporated by reference.
Unless otherwise defined, all terms (including technical and scientific terms) used to describe the invention have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. By way of further guidance, term definitions are included to better understand the teachings of the present invention.
As used herein, the terms "kit", "pharmaceutical combination", "pharmaceutical composition" or "kit of parts" are used interchangeably and are specifically defined in the following sense: the different active ingredients, i.e. the fumarate compound and the immunogenic or tolerogenic peptide as defined herein, may be administered independently, i.e. in different unit doses or dosage forms, in the kit. The separate dosage forms may be administered simultaneously and/or at different time points, e.g. chronologically staggered, i.e. at different time points and at the same or different time intervals for any part of the kit of parts. The proportion of the total amount of combination partners to be administered in the combined preparation may vary. The combination partners may be administered by the same route or by different routes.
The term "fumarate composition" or "fumarate agent" as used herein refers to a composition according to the general formula (I):
Wherein R is 1 And R is 2 Each independently selected from: OH, O - And optionally substituted (C) 1-10 ) Alkoxy, preferably optionally substituted (C 1-6 ) Alkoxy, or optionally substituted (C 1-3 ) An alkoxy group, an amino group,
wherein R is 3 And R is 4 Each independently selected from: h or deuterium is used as a carrier for the electron beam,
wherein each group may independently be optionally substituted to form a prodrug of MMF as described elsewhere herein. Note that in some embodiments DMF is actually a prodrug of the active ingredient MMF.
In some preferred embodiments, the (C 1-10 ) The alkoxy groups may be selected from: (C) 1-5 ) Alkoxy, (C) 1-4 ) Alkoxy, (C) 1-3 ) Alkoxy, ethoxy, (C) 2-3 ) Alkoxy, (C) 2-4 ) Alkoxy, (C) 2-5 ) Alkoxy and (C) 1-6 ) An alkoxy group.
In some preferred embodiments, the fumarate compound is a mono-alkyl fumarate, a di-alkyl fumarate, or a combination thereof.
In some non-limiting illustrative embodiments, the fumarate compound of formula (I) is: dimethyl fumarate-DMF (R) 1 Is OCH 3 And R is 2 Is OCH 3 -formula (II)) or monomethyl fumarate-MMF (R) 1 Is OCH 3 And R is 2 Is O - Or OH-formula (III)), or in the form of a prodrug of monomethyl fumarate.
The term "substituted" or "optionally" as used herein Substituted "refers to groups in which one or more hydrogen atoms may each independently be replaced by the same or different substituents. In certain embodiments, each substituent is independently halogen, -OH, -CN, -CF 3 、=O、-NO 2 Benzyl, -C (O) NH 2 、-R”、-OR”、-C(O)R”、-COOR”、-S(O) 2 R' or-NR 2 ", wherein each R" is independently hydrogen or (C) 1-6 ) Alkyl, cycloalkyl, cycloalkylalkyl, alkenyl, alkynyl, arylalkyl, aryl, alkanediyl, heteroalkyl, heterocycloalkyl, heteroaryl or heteroarylalkyl, heterocycloalkylalkyl. In certain embodiments, each substituent is independently halogen, -OH, -CN, -CF 3 、-NO 2 Benzyl, -R ', -OR' OR-NR 2 ", wherein each R" is independently hydrogen or (C) 1-4 ) An alkyl group. In certain embodiments, each substituent is independently halogen, -OH, -CN, -CF 3 、=O、-NO 2 Benzyl, -C (O) NR 2 ", -R", -OR ", -C (O) R", -COOR "OR-NR" 2 ", wherein each R" is independently hydrogen or (C) 1-4 ) An alkyl group. In certain embodiments, each substituent is independently-OH, (C) 1-4 ) Alkyl and NH 2
A "prodrug of monomethyl fumarate" is a compound of formula (I) wherein R1, R2, R3 or R4 are each independently optionally substituted with a chemical group that is capable of being removed in vivo, i.e., after administration to a patient. Thus, a prodrug is a compound that can be metabolized in vivo to monomethyl fumarate and produce an active form, monomethyl fumarate.
Some preferred examples of such fumarate compounds are mono alkyl fumarates or more particularly prodrugs of mono methyl fumarates, i.e. compounds which are metabolizable in vivo to mono methyl fumarates, such as those of formula (I) wherein R 1 Is C 1 -C 3 Alkoxy (e.g. methoxy, ethoxy or propoxy) and wherein R 2 Is optionally substituted C 1 -C 3 Alkoxy (e.g., methoxy, ethoxy, or propoxy).
Further preferred examples areWherein R is 1 Is methoxy, and R 2 Are those of optionally substituted methoxy or optionally substituted ethoxy.
Some non-limiting examples of such prodrugs are disclosed in any of the following patent applications or patents: WO2016081355, WO2015105757A1, W02014/096425, WO2014031901, WO2014152494, WO2013/119677, U.S. patent No.8,669,281B1 and US2014/0179779. Some preferred examples of prodrugs of MMF are DMF (formula (II)), duloxetyl fumarate (formula (IV)), or tepilamide fumarate (formula (V)).
In other preferred embodiments, the fumarate compound of formula (I) is in the form of a pharmaceutically acceptable salt of monomethyl fumarate or dimethyl fumarate, such as an acid addition salt. The acid addition salts are formed by mixing a solution of the fumarate with a solution of a pharmaceutically acceptable non-toxic acid, such as hydrochloride, hydrobromide, hydroiodide, nitrate, sulfate, bisulfate, phosphate, acid phosphate, isonicotinate, acetate, lactate, salicylate, citrate, tartrate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisate, gluconate, glucuronate, sucrate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate and pamoate. Acceptable base salts include aluminum, calcium, lithium, magnesium, potassium, sodium, zinc and diethanolamine salts. Base addition salts of fumaric acid esters provided herein include, but are not limited to, metal salts made of aluminum, calcium, lithium, magnesium, potassium, sodium and zinc or organic salts made of lysine, N' -dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine) and procaine. Suitable non-toxic acids include, but are not limited to, inorganic and organic acids such as acetic acid, alginic acid, anthranilic acid, benzenesulfonic acid, benzoic acid, camphorsulfonic acid, citric acid, ethanesulfonic acid, formic acid, fumaric acid, furanoic acid (furoic), galacturonic acid, gluconic acid, glucuronic acid, glutamic acid, glycolic acid, hydrobromic acid, hydrochloric acid, isethionic acid, lactic acid, maleic acid, malic acid, mandelic acid, methanesulfonic acid, mucic acid, nitric acid, pamoic acid, pantothenic acid, phenylacetic acid, phosphoric acid, propionic acid, salicylic acid, stearic acid, succinic acid, sulfanilic acid (sulfanilic acid), sulfuric acid, tartaric acid, and p-toluenesulfonic acid. Some specific non-toxic acids include hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid, and methanesulfonic acid. Others are well known in the art, see, for example, remington's Pharmaceutical Sciences, 18 th edition, mack publication, easton PA (1990), or Remington: the Science and Practice of Pharmacy, 19 th edition, mack publication, easton PA (1995). In some preferred embodiments, the pharmaceutically acceptable salt is a salt of a metal (M) cation, where M may be an alkali metal, alkaline earth metal, or transition metal, such as Li, na, K, ca, zn, sr, mg, fe or Mn. In a preferred embodiment, the salt is Ca-MMF or Ca-DMF.
The fumarate compounds as defined herein may be formulated into compositions.
The term "stereoisomer" as used herein refers to one stereoisomer of the fumarate compound of formula (I) that is substantially free of other stereoisomers of the fumarate. For example, a "stereoisomerically pure" fumarate having one chiral center will be substantially free of the opposite enantiomer of the fumarate. A "stereoisomerically pure" fumarate having two chiral centers will be substantially free of the other diastereomers of the fumarate. Typical stereoisomerically pure fumarate compounds comprise: more than about 80% by weight of one stereoisomer of the fumarate and less than about 20% by weight of the other stereoisomers of the fumarate, more than about 90% by weight of one stereoisomer of the fumarate and less than about 10% by weight of the other stereoisomers of the fumarate, more than about 95% by weight of one stereoisomer of the fumarate and less than about 5% by weight of the other stereoisomers of the fumarate, or more than about 97% by weight of one stereoisomer of the fumarate and less than about 3% by weight of the other stereoisomers of the fumarate. The fumarate compounds can have chiral centers and can exist as racemates, individual enantiomers or diastereomers, and mixtures thereof. All such isomeric forms are included in some of the embodiments disclosed herein, including mixtures thereof. The use of stereoisomerically pure forms of such fumarates, as well as the use of mixtures of those forms, are encompassed by some embodiments disclosed herein. For example, mixtures comprising equal or unequal amounts of enantiomers of a particular fumarate can be used in the methods and compositions disclosed herein. These isomers may be asymmetrically synthesized or resolved using standard techniques such as chiral columns or chiral resolving agents. See, e.g., jacques, j., et al, enantiomers, racemates and Resolutions (Wiley Interscience, new York, 1981); wilen, S.H., et al, tetrahedron33:2725 (1977); eliel, e.l., stereochemistry of Carbon Compounds (McGraw Hill, NY, 1962); and Wilen, s.h., tables of Resolving Agents and Optical Resolutions p.268 (e.l. eliel, ed., univ. of note Dame Press, note Dame, IN, 1972).
In other embodiments, the fumarate compound of formula (I) is deuterated, i.e., wherein one or more hydrogen atoms in formula (I) are deuterated, e.g., in the form of a deuterated alkyl group, e.g., a deuterated methyl group comprising at least one deuterium atom. Some examples of deuterated methyl groups include: -CDH 2 、-CD 2 H and-CD 3 . Some examples of deuterated ethyl groups include: -CHDCH 3 ,-CD 2 CH 3 、-CHDCDH 2 、-CHDCD 2 H、CHDCD 3 、-CD 2 CDH 2 、-CD 2 CD 2 H and-CD 2 CD 3
The term "alkoxy" as used herein is an alkyl (carbon and hydrogen chain) group bonded to oxygen in particular.
The term "alkyl" as used herein refers to a fully saturated branched or unbranched hydrocarbon moiety. In one embodiment, the alkyl group comprises 1 to 10 carbon atoms, 1 to 16 carbon atoms, 1 to 10 carbon atoms, 1 to 6 carbon atoms, or 1 to 3 carbon atoms. Some representative examples of alkyl groups include, but are not limited to: methyl, ethyl, n-propyl, isopropyl, n-butyl, sec-butyl, isobutyl, tert-butyl, n-pentyl, isopentyl, neopentyl, n-hexyl, 3-methylhexyl, 2-dimethylpentyl, 2, 3-dimethylpentyl, n-heptyl, n-octyl, n-nonyl or n-decyl.
The term "alkanediyl" as used herein refers to a linear or branched alkyl chain having for example from 1 to 6 carbon atoms. Some representative examples of alkanediyl groups include, but are not limited to:
-CH 2 -,-(CH 2 ) 2 ,-CH(CH 3 )-,-(CH 2 ) 3 -,-CH 2 CH(CH 3 )-,-CH(CH 3 )CH 2 -,-CH(C 2 H 5 )-,-C(CH 3 ) 2 -,-(CH 2 ) 4 -,-(CH 2 ) 2 CH(CH 3 )-,-CH 2 CH(CH 3 )CH 2 -,-CH(CH 3 )(CH 2 ) 2 -,-CH(C 2 H 5 )CH 2 -,-CH 2 CH(C 2 H 5 )-,-C(CH 3 ) 2 CH 2 -,-CH 2 C(CH 3 ) 2 -,-CH(CH 3 )CH(CH 3 ),-CH(C 3 H 7 )-,-(CH 2 ) 5 ,-(CH 2 ) 3 CH(CH 3 ),-(CH 2 ) 2 CH(CH 3 )CH 2 -,-CH 2 CHCH 3 (CH 2 ) 2 -,-CH 2 C(CH 3 ) 2 CH 2 -,-(CH 2 ) 2 C(CH 3 ) 2 -,-(CH 2 ) 6 -,-(CH 2 ) 4 CH(CH 3 )-,-(CH 2 ) 3 CH(CH 3 )CH 2 -,-CH 2 CHCH 3 (CH 2 ) 3 -,-(CH 2 ) 3 C(CH 3 ) 2 - (CH) 2 ) 2 C(CH 3 ) 2 CH 2 -。
The term "alkenyl" as used herein refers to monovalent straight or branched chain hydrocarbons having two to six carbons and at least one carbon-carbon double bond. Some representative examples of alkenyl groups include, but are not limited to: -ch=ch 2 、-CH=CH-CH 3 、-CH 2 -CH=CH-CH 3 or-CH (CH) 3 )-CH=CH-CH 3
The term "alkynyl" as used herein refers to monovalent straight or branched hydrocarbons having two to six carbons and at least one carbon-carbon triple bond. Some representative examples of alkynyl groups include, but are not limited to: 2-propynyl, 3-butynyl, 2-butynyl, 4-pentynyl, 3-pentynyl.
The term "aryl" as used herein refers to a monocyclic, bicyclic or tricyclic aromatic hydrocarbon group having, for example, 5 to 14 carbon atoms in the ring portion. In one embodiment, aryl refers to monocyclic and bicyclic aromatic hydrocarbon groups having 6 to 10 carbon atoms. Some representative examples of aryl groups include, but are not limited to, phenyl, naphthyl, fluorenyl, and anthracenyl.
The term "arylalkyl" as used herein refers to a radical wherein the radical is bonded to a carbon atom (typically terminal or sp 3 Carbon atom) one of the hydrogen atoms bonded is an acyclic alkyl group substituted by an aryl group. Some representative examples of arylalkyl groups include, but are not limited to, benzyl, 2-phenylethan-1-yl, naphthylmethyl, 2-naphthylethan-1-yl, naphthobenzyl or 2-naphthophenylethan-1-yl. In certain embodiments, arylalkyl is C 7-30 Arylalkyl groups, e.g. the alkyl portion of arylalkyl groups is C 1-10 And the aryl moiety is C 6-20 . In certain embodiments, arylalkyl is C 6-18 Arylalkyl groups, e.g. the alkyl portion of arylalkyl groups is C 1-8 And the aryl moiety is C 6-10 . In certain embodiments, arylalkyl is C 7-12 An arylalkyl group.
The term "cycloalkyl" as used herein refers to a saturated or partially unsaturated cyclic alkyl group. Some representative examples of cycloalkyl groups include, but are not limited to, cyclopropane, cyclobutane, cyclopentane, or cyclohexane. In one embodiment, cycloalkyl is C 3-15 Cycloalkyl, C 3-12 Cycloalkyl or C 3-8 Cycloalkyl groups.
The term "cycloalkylalkyl" as used herein refers to a radical wherein the radical is bonded to a carbon atom (typically terminal or sp 3 Carbon atom) one of the hydrogen atoms bonded is an acyclic alkyl group substituted by a cycloalkyl group. In certain embodiments, cycloalkylalkyl is C 4-30 Cycloalkyl alkylAnd the alkyl portion of the cycloalkylalkyl group is, for example, C 1-10 And cycloalkyl moiety is C 3-20 . In another embodiment, cycloalkylalkyl is C 3-20 Cycloalkylalkyl groups, and the alkyl portion of the cycloalkylalkyl group is, for example, C 1-8 And cycloalkyl moiety is C 3-12 . In a specific embodiment, cycloalkylalkyl is C 4-12 Cycloalkyl alkyl.
The term "halogen" as used herein refers to fluorine, chlorine, bromine or iodine.
The term "heteroalkyl," as used herein, by itself or as part of another substituent, refers to an alkyl group in which one or more of the carbon atoms (and some related hydrogen atoms) are independently replaced with a heteroatom group. Some examples of heteroatom groups include, but are not limited to: -O-, -S-, -O-, -S-, -O-S-, -NR ',n-n=, -n=n-NR', -PR '-, -P (O) 2-, -POR' -, -O-P (O) 2-, -SO2-, and-Sn (R ') 2-, wherein each R' is independently hydrogen, C 1-6 Alkyl, substituted C 1-6 Alkyl, C 6-12 Aryl, substituted C 6-12 Aryl, C 7-18 Arylalkyl, substituted C 7-18 Arylalkyl, C 3-7 Cycloalkyl, substituted C 3-7 Cycloalkyl, C 3-7 Heterocycloalkyl, substituted C 3-7 Heterocycloalkyl, C 1-6 Heteroalkyl, substituted C 1-6 Heteroalkyl, C 6-12 Heteroaryl, substituted C 6-12 Heteroaryl, C 7-18 Heteroarylalkyl or substituted C 7-18 Heteroaryl alkyl. In one embodiment, C 1-6 Heteroalkyl means, for example, C in which at least one carbon atom (and some related hydrogen atoms) is replaced by a heteroatom 1-6 An alkyl group. In one embodiment, for example, C 1-6 Heteroalkyl includes groups having five carbon atoms and one heteroatom, groups having four carbon atoms and two heteroatoms, and the like. In one embodiment, each R' is independently hydrogen or C 1-3 An alkyl group. In a further embodiment of the present invention, heteroatom group is-O-, a-S-, -NH-, -N (CH) 3 ) -or-SO 2 -. In one placeIn a specific embodiment, the heteroatom group is-O-.
The term "heteroaryl" as used herein refers to, for example, a 5 to 14 membered monocyclic, bicyclic, or tricyclic ring system having 1 to 10 heteroatoms independently selected from N, O or S, wherein N and S can optionally be oxidized to various oxidation states, and wherein at least one ring in the ring system is aromatic. In one embodiment, the heteroaryl group is a single ring and has 5 or 6 ring members. Some representative examples of monocyclic heteroaryl groups include, but are not limited to: pyridyl, thienyl, furyl, pyrrolyl, pyrazolyl, imidazolyl,Azolyl, iso->Oxazolyl, thiazolyl, isothiazolyl, triazolyl,/-yl>Diazolyl, thiadiazolyl, and tetrazolyl. In another embodiment, the heteroaryl is bicyclic and has 8 to 10 ring members. Some representative examples of bicyclic heteroaryl groups include indolyl, benzofuranyl, quinolinyl (quinolyl), isoquinolinyl indazolyl, indolinyl, isoindolyl, indolizinyl, benzimidazolyl (benzamidazolyl), quinolinyl (quinolyl), 5,6,7, 8-tetrahydroquinoline, and 6, 7-dihydro-5H-pyrrolo [3,2-d ] ]Pyrimidine. In another embodiment, the heteroaryl is bicyclic and has 8 to 10 ring members.
The term "heteroarylalkyl" as used herein refers to an acyclic alkyl group in which one of the hydrogen atoms bonded to a carbon atom (typically a terminal or sp3 carbon atom) is replaced by a heteroaryl group. In certain embodiments, the heteroarylalkyl is C 7-12 Heteroarylalkyl, and the alkyl portion of the heteroarylalkyl is, for example, C 1-2 And the heteroaryl moiety is C 6-10
The term "heterocycle" as used herein refers to any ring structure (saturated or partially unsaturated) that contains at least one ring heteroatom (e.g., N, O or S). Some examples of heterocycles include, but are not limited to, morpholine, pyrrolidine, tetrahydrothiophene, piperidine, piperazine, and tetrahydrofuran.
The term "heterocycloalkyl" as used herein refers to a saturated or unsaturated cyclic alkyl group in which one or more carbon atoms (and some related hydrogen atoms) are independently replaced by one or more heteroatoms; or a parent aromatic ring system in which one or more carbon atoms (and some related hydrogen atoms) are independently replaced by one or more heteroatoms such that the ring system no longer comprises at least one aromatic ring. Some representative examples of heteroatoms replacing carbon atoms include, but are not limited to: n, P, O, S and Si. Some representative examples of heterocycloalkyl groups include, but are not limited to: epoxide, aziridine, thiurane, imidazolidine, morpholine, piperazine, piperidine, pyrazolidine, pyrrolidine, and quinuclidine. In one embodiment, the heterocycloalkyl is C 5-10 Heterocycloalkyl, C 5-8 A heterocycloalkyl group. In a specific embodiment, the heterocycloalkyl is C 5-6 A heterocycloalkyl group.
The term "heterocycloalkyl alkyl" as used herein refers to a radical wherein the radical is bonded to a carbon atom (typically terminal or sp 3 Carbon atom) one of the hydrogen atoms bonded is an acyclic alkyl group replaced by a heterocycloalkyl group. In certain embodiments, the heterocycloalkyl alkyl is C 7-12 Heterocycloalkyl, and the alkyl portion of the heterocycloalkyl alkyl is, for example, C 1-2 And the heterocycloalkyl moiety is C 6-10
In one embodiment, the fumarate compound as defined herein for use in the kits and methods of the invention is in the form of a fumarate pharmaceutical composition or dosage form comprising a therapeutically effective amount of the fumarate compound as defined herein and a pharmaceutically acceptable carrier or pharmaceutically acceptable excipient.
In a specific embodiment, the fumarate pharmaceutical composition or dosage form comprises a fumarate compound selected from the group comprising: dialkyl fumarate, monoalkyl fumarate, a combination of dialkyl fumarate and monoalkyl fumarate, a prodrug of monoalkyl fumarate, deuterated forms of any of the foregoing, or inclusion compounds, solvates, tautomers or stereoisomers of any of the foregoing, or a combination of any of the foregoing.
In a specific embodiment, the fumarate pharmaceutical composition or dosage form consists essentially of DMF and/or MMF.
The fumarate pharmaceutical composition or dosage form can be administered in a number of ways. For example, U.S. patent nos. 6,509,376 and 6,436,992 disclose some possible formulations comprising DMF and/or MMF. With respect to the route of administration, the compositions may be administered orally, intranasally, transdermally, subcutaneously, intradermally, vaginally, intraoral, intraocular, intramuscular, buccal, rectal, transmucosally, or by inhalation or intravenous administration. In some embodiments, DMF or MMF is administered orally.
In a particular embodiment, the fumarate pharmaceutical composition or dosage form may be an oral dosage form, such as a solid oral dosage form, e.g., a micro-pellet (micro-pellet), a micro-tablet, a capsule (e.g., a soft gelatin capsule or a hard gelatin capsule), a granule, or a tablet. In a specific embodiment, the fumarate pharmaceutical composition or dosage form is in the form of a micro-pill or micro-tablet, a capsule, or a capsule comprising a micro-tablet or micro-pill.
Optionally, the microtablets or micropellets or capsules are enteric coated. In a specific embodiment, the fumarate pharmaceutical composition or dosage form is in the form of an enterically coated tablet or minitablet (optionally contained in a capsule) wherein once the enteric coating is dissolved in the gastrointestinal tract it acts as an immediate release dosage form.
In another specific embodiment, the fumarate pharmaceutical composition or dosage form is a controlled release or sustained release composition, optionally enterically coated. Such formulations may be prepared by a variety of techniques by those skilled in the art. For example, the formulation may comprise a therapeutic compound, a rate controlling polymer (i.e., a substance that controls the rate of release of the therapeutic compound from the dosage form), and optionally other excipients. Some examples of rate controlling polymers are hydroxyalkyl celluloses, hydroxypropyl alkyl celluloses (e.g., hydroxypropyl methyl cellulose, hydroxypropyl ethyl cellulose, hydroxypropyl isopropyl cellulose, hydroxypropyl butyl cellulose, and hydroxypropyl hexyl cellulose), polyethylene oxides, alkyl celluloses (e.g., ethyl cellulose and methyl cellulose), carboxymethyl cellulose, hydrophilic cellulose derivatives, and polyethylene glycols, the compositions being described in WO 2006/037342.
The fumarate compounds or formulations defined herein can be combined with pharmaceutically acceptable excipients or carriers and optionally a sustained release matrix (e.g., a biodegradable polymer) to form a pharmaceutical formulation. In such pharmaceutical formulations, the active ingredient, alone or in combination with additional active ingredients, may be administered to animals and humans in unit administration form as a mixture with conventional pharmaceutical supports. Suitable unit administration forms include: oral route forms (e.g., tablets, gel capsules, powders, granules, and oral suspensions or solutions), sublingual and buccal administration forms, aerosols, implants, subcutaneous, transdermal, topical, intraperitoneal, intramuscular, intravenous, subcutaneous, transdermal, intrathecal, and intranasal administration forms, and rectal administration forms.
Preferably, the pharmaceutical formulation or dosage form comprises a pharmaceutically acceptable carrier of the injectable formulation. These may in particular be isotonic sterile saline solutions (sodium or disodium phosphate, sodium chloride, potassium chloride, calcium or magnesium chloride, etc. or mixtures of such salts), or as the case may be, after addition of sterile water or physiological saline, allowing the construction of dry, in particular lyophilized, compositions of the injectable solution.
The fumarate pharmaceutical compositions or formulations described herein are prepared in a manner known per se, for example by conventional mixing, granulating, dragee-making, dissolving, or lyophilizing processes. Thus, a fumarate pharmaceutical formulation for oral use can be obtained by: the fumarate is combined with solid excipients, optionally after adding suitable excipients, if desired or necessary, the resulting mixture is ground and the mixture of granules is processed to obtain tablets or dragee cores.
Suitable excipients are in particular fillers, for example: sugars (e.g., lactose or sucrose, mannitol or sorbitol), cellulose preparations and/or calcium phosphates (e.g., tricalcium phosphate or calcium hydrogen phosphate), and binders (e.g., starch pastes), using, for example, corn starch, wheat starch, rice starch, potato starch, gelatin, tragacanth, methyl-cellulose, hydroxypropyl-methyl-cellulose, sodium carboxymethyl-cellulose, and/or polyvinylpyrrolidone. If desired, disintegrating agents can be added, such as the starches mentioned above as well as carboxymethyl starch, crosslinked polyvinylpyrrolidone, agar, or alginic acid or a salt thereof (e.g., sodium alginate). Adjuvants are first flow regulators and lubricants, for example, silicon dioxide, talc, stearic acid or salts thereof (for example, magnesium stearate or calcium stearate) and/or polyethylene glycols. Dragee cores are provided with suitable coatings that are resistant to gastric juice, if desired. For this purpose, concentrated sugar solutions may be used, which may optionally comprise gum arabic, talc, polyvinyl pyrrolidone, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. In order to produce a coating that is resistant to gastric juice, a solution of a suitable cellulose formulation (e.g., acetyl cellulose phthalate or hydroxypropyl methylcellulose phthalate) is used. Dyes or pigments may be added to the tablets or dragee coatings, for example for identification or for characterization of the active compound dose combinations.
In one embodiment, the fumarate pharmaceutical formulation or dosage form described herein comprises a capsule containing the pharmaceutical composition described herein in the form of an enterically coated microtablet. The coating of the microtablets may consist of different layers. The first layer may be a methacrylic acid-methyl methacrylate copolymer/isopropyl solution that isolates the tablet core from potential hydrolysis from the next applied aqueous suspension.
Enteric coating of the tablets may then be imparted by aqueous methacrylic acid-ethyl acrylate copolymer suspension.
The number of excipients that can be included in the composition is not limited.
Some examples of fillers or binders include, but are not limited to, ammonium alginate, calcium carbonate,Calcium phosphate, calcium sulfate, cellulose acetate, compressible sugar (compressible sugar), sugar of confectionary (dextrose), dextrates (dextrates), dextrin, dextrose, erythritol, ethylcellulose, fructose, glyceryl palmitostearate (glyceryl palmitostearate), hydrogenated vegetable oil type I, isomalt, kaolin, lactitol, lactose, mannitol, magnesium carbonate, magnesium oxide, maltodextrin, maltose, mannitol, medium chain triglycerides, microcrystalline cellulose, polydextrose, polymethacrylates, dimethicone, sodium alginate, sodium chloride, sorbitol, starch, sucrose, sugar spheres, sulfobutyl ether β -cyclodextrin, talc, tragacanth, trehalose, polysorbate 80, and xylitol. In one embodiment, the filler is microcrystalline cellulose. The microcrystalline cellulose may be, for example, PROSOLV 50、PROSOLV />90、PROSOLVHD90、PROSOLV/>90 LM, and any combination thereof.
Some examples of disintegrants include, but are not limited to: hydroxypropyl starch, alginic acid, calcium alginate, carboxymethylcellulose calcium, carboxymethylcellulose sodium, powdered cellulose, chitosan, colloidal silicon dioxide, croscarmellose sodium, crospovidone, docusate sodium, guar gum, hydroxypropyl cellulose, low substituted hydroxypropyl cellulose, magnesium aluminum silicate, methylcellulose, microcrystalline cellulose, polacrilin potassium (polacrilin potassium), povidone, sodium alginate, sodium starch glycolate, starch and pregelatinized starch. In one embodiment, the disintegrant is croscarmellose sodium.
Some examples of glidants include, but are not limited to, calcium phosphate, calcium silicate, powdered cellulose, magnesium silicate, magnesium triplicate, silica, talc and colloidal silicon dioxide, and anhydrous colloidal silicon dioxide. In one embodiment, the glidant is anhydrous colloidal silicon dioxide, talc, or a combination thereof.
Some examples of lubricants include, but are not limited to: canola oil, hydroxyethyl cellulose, lauric acid, leucine, mineral oil, poloxamer (poloxamer), polyvinyl alcohol, talc, octyldodecanol, sodium hyaluronate, sterilizable corn starch, triethanolamine, calcium stearate, magnesium stearate, glyceryl monostearate, glyceryl behenate, glyceryl palmitostearate, hydrogenated castor oil, hydrogenated vegetable oil type I, light mineral oil, magnesium lauryl sulfate, medium chain triglycerides, mineral oil, myristic acid, palmitic acid, poloxamer, polyethylene glycol, potassium benzoate, sodium chloride, sodium lauryl sulfate, stearic acid, talc and zinc stearate. In one embodiment, the lubricant is magnesium stearate.
Fumarate pharmaceutical compositions or dosage forms suitable for the above methods include, but are not limited to, those formulated for once-daily (QD) administration or multiple times-daily (e.g., twice-daily (BID) administration or three times-daily (TTD) administration). In some embodiments, the pharmaceutical composition is formulated for QD administration, wherein a therapeutically effective amount of a fumarate compound as defined herein (e.g., DMF or MMF) is contained in one unit dosage form or provided in a kit containing multiple unit dosage forms. In some embodiments, the pharmaceutical composition is formulated for BID or TID administration, wherein a therapeutically effective amount of a fumarate compound as defined herein (e.g., DMF or MMF) is divided, for example, equally into two or three daily administrations.
The therapeutically effective amount of the fumarate compound (e.g., DMF or MMF) as defined herein can be any therapeutically effective dose. In some embodiments, the neurological disorder is multiple sclerosis, wherein a therapeutically effective amount of a fumarate compound (e.g., DMF or MMF) as defined herein is an amount effective to treat or prevent multiple sclerosis, e.g., an amount effective to treat or prevent multiple sclerosis in a subject characterized as not responsive to interferon beta treatment.
In some embodiments, the fumarate agent is DMF, and the appropriate (i.e., therapeutically effective) dose of DMF may be any dose of DMF from 20mg to 1 g. In some embodiments, the DMF in the pharmaceutical composition is about 60mg, about 80mg, about 100mg, about 120mg, about 160mg, about 200mg, about 240mg, about 320mg, about 360mg, about 400mg, about 480mg, about 600mg, about 720mg, about 800mg, about 900mg, about 1000mg of DMF, or any range thereof. In some embodiments, the therapeutically effective amount of DMF is about 480mg or about 720mg per day. In some embodiments, about 480mg of DMF is provided in two unit dosage forms, each comprising about 240mg of DMF, and is administered to the subject about 6 to about 14 hours apart during the day. In some embodiments, about 720mg of DMF is provided in three unit dosage forms, each comprising about 240mg of DMF, and is administered to the subject at intervals of about 4 hours to about 8 hours in a day.
In one embodiment, the administration of dimethyl fumarate is 240mg twice daily.
In one embodiment, 120mg of dimethyl fumarate is administered twice daily for 7 days, followed by 240mg of dimethyl fumarate twice daily as a maintenance dose.
In one embodiment, no more than 720mg of total fumarate is administered per day.
In one embodiment, no more than 480mg of total fumarate is administered per day.
In one embodiment, the pharmaceutical composition consists essentially of dimethyl fumarate, and no more than 720mg of dimethyl fumarate per day is administered.
In one embodiment, the pharmaceutical composition consists essentially of dimethyl fumarate, and no more than 480mg of dimethyl fumarate is administered per day.
In a preferred embodiment, the fumarate compound is dimethyl fumarate or a derivative thereof, e.g. under the trade name TECFIDERA TM Lower commercial drugs. (formula II):
in another embodiment, the fumarate compound is under the trade name FUMADERM TM The following commercial forms of pharmaceutical compositions comprising as active ingredients: dimethyl fumarate, the calcium salt of monoethyl fumarate (ethyl hydrogen fumarate), the magnesium salt of monoethyl fumarate and the zinc salt of monoethyl fumarate.
The term "peptide" as used herein refers to a molecule comprising an amino acid sequence, which in the case of NKT cell epitopes of a minimum of 7 amino acids is 9 to 50 amino acids linked by peptide bonds or in the case of MHC class II T cell epitopes of a minimum length of 7, 8 or 9 amino acids is 9 to 50 amino acids linked by peptide bonds, preferably 11 to 50 amino acids, but may comprise a non-amino acid structure. The peptide according to the invention may comprise any conventional 20 amino acids or modified forms thereof, or may comprise non-naturally occurring amino acids incorporated by chemical peptide synthesis or by chemical or enzymatic modification.
The peptides of the invention can be produced in bacteria, yeast, insect cells, plant cells or mammalian cells using recombinant DNA technology. In view of the limited length of the peptide, it can be prepared by chemical peptide synthesis, in which the peptide is prepared by coupling different amino acids to each other. Chemical synthesis is particularly suitable for inclusion of e.g. D-amino acids, amino acids with non-naturally occurring side chains or natural amino acids with modified side chains, etc.
Chemical peptide synthesis methods have been fully described and peptides can be ordered from companies such as Applied Biosystems and others.
In any aspect of the invention, and unless otherwise indicated, the term "peptide" may mean an immunogenic peptide or tolerogenic peptide as defined herein.
The length of the immunogenic or tolerogenic peptides of the invention may vary significantly. Peptides may be up to 20, 25, 30, 40 or 50 amino acids in length (i.e., consisting of 7, 8 or 9 amino acid epitopes, 2 to about 11 amino acid modified oxidoreductase motifs adjacent thereto) and not equal. For example, the peptide may comprise an endosomal targeting sequence of 40 amino acids, a flanking sequence of about 2 amino acids, an oxidoreductase motif as described herein of 2 to about 11 amino acids, a linker of 4 to 7 amino acids, and a T cell epitope peptide of minimum length of 7, 8, or 9 amino acids.
Thus, in some embodiments, the intact peptide consists of 9 amino acids up to 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 40, 50, 75, or 100 amino acids. More particularly, where the reducing compound is a modified oxidoreductase motif as described herein, the length of the (artificial or natural) sequence comprising the epitope and the modified oxidoreductase motif (referred to herein as the "epitope-modified oxidoreductase motif" sequence) optionally linked by a linker, without an endosomal targeting sequence, is critical. An "epitope-modified oxidoreductase motif" more particularly has a length of 9, 10, 11, 12, 13, 14, 15, 16, 17, 18 or 19 amino acids. Such 9, 10, 11, 12, 13 or 14 to 19 amino acid peptides may optionally be coupled with less critical size endosomal targeting signals.
In a specific embodiment, the peptides of the invention are 9 to 30 or 11 to 30 amino acids in length.
According to one embodiment, the immunogenic or tolerogenic peptide of the invention comprises an NKT cell epitope and is 9 to 30 amino acids in length.
According to another embodiment, the immunogenic or tolerogenic peptide of the invention comprises a MCH class II T cell epitope and is 11 to 30 amino acids in length.
The term "basic amino acid" refers to any amino acid that functions like a Bronsted-Lowry base and a Lewis base, and includes natural basic amino acids such as arginine (R), lysine (K), or histidine (H), or non-natural basic amino acids such as, but not limited to:
■ Lysine variants such as Fmoc-beta-Lys (Boc) -OH (CAS number 219967-68-7); fmoc-Orn (Boc) -OH, also known as L-ornithine or ornithine (CAS number 109425-55-0); fmoc-beta-Homolys (Boc) -OH (CAS number 203854-47-1); fmoc-Dap (Boc) -OH (CAS number 162558-25-0) or Fmoc-Lys (Boc) OH (DiMe) -OH (CAS number 441020-33-3);
■ Tyrosine/phenylalanine variants such as Fmoc-L-3Pal-OH (CAS number 175453-07-3); fmoc-beta-homoPhe (CN) -OH (CAS number 270065-87-7); fmoc-L-beta-homoAla (4-pyridyl) -OH (CAS number 270065-69-5) or Fmoc-L-Phe (4-NHBoc) -OH (CAS number 174132-31-1);
■ Proline variants such as Fmoc-Pro (4-NHBoc) -OH (CAS number 221352-74-5) or Fmoc-Hyp (tBu) -OH (CAS number 122996-47-8);
■ Arginine variants such as Fmoc-beta-Homoarg (Pmc) -OH (CAS number 700377-76-0).
Amino acids are referred to herein by their full names, their three-letter abbreviations or their single-letter abbreviations.
The motifs of the amino acid sequences are written herein according to the format Prosite. Motifs are used to describe a certain sequence variation at a specific part of a sequence. The symbol X is used at the position where any amino acid is accepted. By being placed in brackets ("[ V ]]") acceptable amino acids at a given position are listed to indicate substitution. For example: [ CST ]]Represents an amino acid selected from Cys, ser or Thr. Amino acids that are excluded as alternatives are indicated by listing them between brackets ("{ }"). For example: { AM } represents any amino acid other than Ala and Met. The different elements in the motif are optionally separated from each other by hyphens (-). In the context of the motifs disclosed in this specification, the disclosed universal oxidoreductase motifs are typically accompanied by hyphens and do not form linkages to different elements outside the motif. These "open" hyphens indicate the location of the physical linkage of the motif to another portion of the immunogenic peptide, such as a linker sequence or epitope sequence. For example, form "Z m -C-X n -[CST]Motif indication [ CST ]]Is an amino acid linked to the rest of the immunogenic peptide and Z is the terminal amino acid of the immunogenic peptide. The preferred physical linkage is a peptide bond. Repetition of the same element in a motif can be indicated by the inclusion of a numerical value or range of numerical values between parentheses after the element. For example in the In aspects, "X n "refers to n" X ". X (2) corresponds to X-X or XX; x (2, 5) corresponds to 2, 3, 4 or 5X amino acids and A (3) corresponds to A-A-A or AAA. To distinguish amino acids, those other than the oxidoreductase motif can be referred to as external amino acids and those within the oxidoreductase motif can be referred to as internal amino acids. Unless otherwise indicated, X represents any amino acid, particularly an L-amino acid, more particularly one of the 20 naturally occurring L-amino acids.
The term "antigen" as used herein refers to a structure of a macromolecule, typically a protein (with or without a polysaccharide), or a structure composed of a protein composition comprising one or more haptens and comprising T or NKT cell epitopes. The term "antigenic protein" as used herein refers to a protein comprising one or more T or NKT cell epitopes. As used herein, "autoantigen" or "autoantigen protein" refers to a human or animal protein or fragment thereof that is present in the body that elicits an immune response in the same human or animal body.
The term "epitope" refers to one or several parts of an antigen protein (which may define conformational epitopes) which are specifically recognized and bound by antibodies or parts thereof (Fab ', fab2', etc.) or receptors present on the cell surface of B, T or NKT cells, and which B, T or NKT cells are capable of inducing an immune response by said binding.
In the context of the present invention, the term "T cell epitope" refers to a dominant (domino), sub-dominant (sub-domino) or minor (minor) T cell epitope, i.e. a portion of an antigen protein, which is specifically recognized and bound by a receptor at the cell surface of T lymphocytes. Whether an epitope is dominant, subdominant, or minor depends on the immune response elicited against the epitope. The dominance depends on the frequency with which such epitopes are recognized by T cells and are able to activate them among all possible T cell epitopes of the protein. In the context of the immunogenic peptides defined herein, T cell epitopes may be epitopes recognized by MHC class II molecules, consisting of a sequence of +/-9 amino acids suitable for the groove of MHC class II molecules. In the peptide sequence representing the T cell epitope, the amino acid numbers in the epitope are P1 to P9, the N-terminal amino acid numbers of the epitope are P-1, P-2 and the like, and the C-terminal amino acid numbers of the epitope are P+1, P+2 and the like. Peptides recognized by MHC class II molecules, but not MHC class I molecules, are referred to as MHC class II restricted T cell epitopes. Alternatively, in the context of the immunogenic peptides defined herein, T cell epitopes may be epitopes recognized by CD1d molecules, consisting of a sequence of +/-7 amino acids that bind to CD1d molecules. In the peptide sequence representing the T cell epitope, the amino acid numbers in the epitope are P1 to P7, the N-terminal amino acid numbers of the epitope are P-1, P-2 and the like, and the C-terminal amino acid numbers of the epitope are P+1, P+2 and the like. Peptides recognized by CD1d molecules, but not MHC molecules, are referred to as CD1d or NKT-restricted T cell epitopes.
According to tolerogenic peptides as defined herein, the T cell epitope may be an MHC (e.g. MHC i or MHC ii) or NKT epitope, and may be longer.
The identification and selection of T cell epitopes from antigenic proteins is known to those skilled in the art.
This discovery provides a rule-based method for selecting tolerogenic T-cell epitopes that does not require in vivo examination of peptide tolerogenicity. This is particularly advantageous in developing strategies for treating or preventing diseases for which no animal model is available. Even for diseases with animal models, the selection method should make the development of tolerance-inducing compositions simpler and safer, since the selection method provides the mechanism: the peptides may be tested for tolerance-inducing capacity in vitro in human T cells (recognizing antigens bound to human MHC molecules) prior to their use in vivo.
Typical methods for selecting immunogenic or tolerogenic peptides include the step of selecting peptides capable of binding to MHC class I or MHC class II molecules, as reported for example in WO0216410 A2. In a preferred embodiment, the peptide is capable of binding to an MHC class II molecule.
Many methods for screening for immunogenic tolerogenic peptides capable of acting as T cell epitopes for a given antigen are known in the art. Thus, in general, this approach will be used to select tolerogenic peptides from a variety of peptides each comprising a T cell epitope.
The term "tolerogenic" means capable of inducing tolerance, i.e., substantially no response to an antigen. Tolerance to self or autoantigens is an essential feature of the immune system and disorders therein can lead to autoimmune diseases. Tolerance usually results from the thymus (central tolerance), in which autoreactive immature T lymphocytes undergo apoptosis. However, there are also mechanisms: by this mechanism, mature autoreactive T lymphocytes in peripheral tissues acquire tolerance (peripheral tolerance). In the context of the present application, tolerogenic peptides do not comprise an oxidoreductase motif as defined herein.
Mechanisms of central and peripheral tolerance are reported, for example, in Anderton et al (1999) (Immunological Reviews 169:123-137). Tolerance may be produced or characterized by: induction of anergy in at least a portion of cd4+ T cells. To activate T cells, peptides must be associated with "professional" APCs that are capable of delivering both signals to T cells. The first signal (signal 1) is delivered by MHC-peptide complexes on the surface of APC cells and received by T cells through the TCR. The second signal (signal 2) is delivered by costimulatory molecules on the surface of APC, such as CD80 and CD86, and is received by CD28 on the surface of T cells. It is believed that when a T cell receives signal 1 in the absence of signal 2, it is not activated and becomes virtually anergic. Anergic T cells are refractory to subsequent antigen challenge and may be able to suppress other immune responses. Anergic T cells are thought to be involved in mediating T cell tolerance. It has been shown that antigen-specific cd4+ T cells have a reduced ability to proliferate when tolerance is induced by peptide inhalation. In addition, IL-2, IFN-y and IL-4 production by these cells is down-regulated, but IL-10 production is increased. Neutralization of IL-10 in mice has been shown to completely restore susceptibility to disease in a state of peptide-induced tolerance. It has been proposed to regulate the persistence of cell populations in a tolerogenic state that produces IL-10 and mediates immunomodulation (Burkhart et al (1999) int. Immunol. 11:1625-1634). Thus, induction of tolerance can be monitored by a variety of techniques including: (a) Reducing susceptibility to a disease in which the peptide is a target epitope in vivo; (b) Inducing anergy in cd4+ T cells (which can be detected by subsequent in vitro challenge with antigen); (c) Changes in the CD4+ T cell population, including (i) decreased proliferation, (ii) down-regulated IL-2, IFN-y, and IL-4 production, and (iii) increased IL-10 production.
Tolerogenic peptides as used herein encompass all antigen-derived peptides and T cell epitopes that induce tolerance (anergy) against the antigen from which they are derived.
Epitope selection includes the step of selecting peptides capable of binding to MHC class I or MHC class II proteins. Epitopes can be immunodominant, i.e., hot spots in an antigen that are presented by, typically APCs, as compared to others. The immunodominant determinant region may be a good toleragen and thus in a preferred embodiment the tolerogenic peptide or epitope of the invention is immunodominant epitope-based. However, during the development of autoimmune disease, epitope diffusion into the subdominant determinants can occur (Lehmann et al (1992) Nature 358:155-157). Presentation of the subdominant epitope may thus also be important in triggering autoimmunity, and the tolerogenic peptide or epitope of the invention may thus be based on the subdominant epitope. Last but not least, the tolerogenic peptide or epitope of the invention may be a cryptic epitope, i.e., an epitope that stimulates a T cell response when administered as a peptide but fails to produce a response to an antigen when administered as a whole.
Naturally processed epitopes can be identified by mass spectrophotometric analysis (mass spectrophotometric analysis) of peptides eluted from antigen-loaded APCs, i.e. APCs that have been prompted to ingest antigen or that have been forced to produce proteins intracellularly by transformation with appropriate genes. Typically, APCs are incubated with proteins in solution or proteins that are appropriately targeted to the surface of the APC cells. After incubation at 37 ℃, the cells are lysed in a detergent and the class II proteins are purified by e.g. affinity chromatography. Treatment of the purified MHC with a suitable chemical medium (e.g., acidic conditions) results in elution of the peptide from the MHC. The pool of peptides was isolated and the spectra were compared to peptides from control APCs treated in the same manner. Peaks specific to cells expressing the protein are analyzed (e.g., by mass spectrometry) and the peptide fragment is identified. This procedure typically yields information about the range of peptides (typically present in a "nested set") that are produced from a particular antigen by antigen processing.
Another method for identifying epitopes is to screen synthetic libraries of peptides overlapping and spanning the length of the antigen in an in vitro assay. For example, peptides of 15 amino acids in length and overlapping by 5 or 10 amino acids may be used. Peptides were tested in an antigen presenting system comprising antigen presenting cells and T cells. For example, the antigen presentation system can be a murine spleen cell preparation, a human cell preparation from tonsils, or PBMC T cell activation can be measured by T cell proliferation (e.g., using 3H-thymidine incorporation) or cytokine production. Activation of THI type CD4+ T cells can be detected, for example, by IFNy production, which can be detected by standard techniques such as ELISPOT assay. Such overlapping peptide studies generally indicate the region of the antigen where the epitope is located. The minimal epitope of a particular T cell can then be assessed by measuring the response to the truncated peptide. For example, if a response is obtained to peptides comprising residues 1 to 15 in the overlapping library, the truncated set at both ends (i.e., positions 1 to 14, 1 to 13, 1 to 12, etc., and positions 2 to 15, 3 to 15, 4 to 15, etc.) can be used to identify the smallest epitope.
The identification of immunodominant regions of antigens using in vitro assays (especially those using T cell lines) to exhibit skewed patterns of peptide reactivity was predicted by the present inventors. Kinetic response assays can be used in which proliferation of PBMCs from patients and healthy individuals is measured against overlapping peptide libraries. This determination is based on the finding that: while T cells from normal individuals and patients respond in a similar manner to purified protein antigens, they respond in a different manner to peptides based on antigen sequences. T cells from autoimmune patients respond to peptide autoantigens with greater amplitude and faster kinetics when compared to normal healthy donors. This enables screening and identification of epitopes that respond to a particular patient at a particular time.
To identify epitopes suitable in the context of the present invention, the isolated peptide sequences of the antigen proteins are tested, for example, by T cell biotechnology, to determine whether the peptide sequences elicit a T cell response. Those peptide sequences found to elicit a T cell response are defined as having T cell stimulatory activity.
Human T cell stimulatory activity can also be tested by: t cells obtained from an individual suffering from a fumarate-related disease or disorder are incubated with a peptide/epitope derived from an autoantigen involved in the disease or disorder, and it is determined whether proliferation of T cells occurs in response to the peptide/epitope, as measured, for example, by cellular uptake of tritiated thymidine (tritiated thymidine). The stimulation index of T cell responses to peptides/epitopes can be calculated as the maximum CPM in response to the peptide/epitope divided by the control CPM. T cell stimulation index (stimulation index, s.i.) equal to or greater than twice background levels was considered "positive". Positive results were used to calculate the average stimulation index for each peptide/epitope of the subject peptide/epitope group.
Non-natural (or modified) T cell epitopes can also optionally be tested for their binding affinity to MHC class II molecules. This can be done in different ways. For example, soluble HLA class II molecules are obtained by lysing cells that are homozygous for a given class II molecule. The latter is purified by affinity chromatography. The soluble class II molecule is incubated with a biotin-labeled reference peptide, which is produced based on its strong binding affinity to the MHC class II molecule. The peptides to be evaluated for class II binding were then incubated at different concentrations and their ability to displace the reference peptide from their class II binding was calculated by adding neutravidin.
In order to determine the optimal T cell epitope by, for example, fine mapping techniques, peptides having T cell stimulatory activity and thus comprising at least one T cell epitope (as determined by T cell biotechnology) are modified by adding or deleting amino acid residues at the amino or carboxy terminus of the peptide and tested to determine changes in T cell reactivity against the modified peptide. If two or more peptides sharing overlapping regions in the native protein sequence are found to have human T cell stimulatory activity as determined by T cell biotechnology, additional peptides comprising all or part of such peptides can be produced and these additional peptides can be tested by similar procedures. According to this technique, peptides are selected and recombinantly or synthetically produced. T cell epitopes or peptides are selected based on a variety of factors including the intensity of T cell responses to the peptides/epitopes (e.g., stimulation index) and the frequency of T cell responses to the peptides in a population of individuals.
Additionally and/or alternatively, one or more in vitro algorithms may be used to identify T cell epitope sequences in an antigen protein. Suitable algorithms include, but are not limited to, those described in the following: zhang et al (2005) Nucleic Acids Res, W180-W183 (PREDBALB); salomon & flow (2006) BMC Bioinformatics 7,501 (MHCBN); schulter et al (2007) Methods mol. Biol.409,75-93 (SYFPEITHI); donnes & Kohlbacher (2006) Nucleic Acids Res.34, W194-W197 (SVMHC); kolaskar & Tongaonkar (1990) FEBS Lett.276,172-174, guan et al (2003) appl.Bioinformation 2,63-66 (MHCPred) and Singh and Raghava (2001) Bioinformation 17,1236-1237 (Propred). More particularly, such algorithms allow prediction of one or more octapeptide or nonapeptide sequences in the groove of an MHC II molecule to be adapted to an antigen protein, and this is also true for different HLA types.
In the context of the present invention, the term "immunogenic" means capable of inducing so-called cytolytic CD4 + T cells, i.e. T cells with apoptotic properties against APC, as described in detail in WO2009101207 and Carlier et al (2012) Plos one 7,10e 45366.
The term "immunogenic peptide" refers to a peptide comprising an oxidoreductase motif as defined herein.
The terms "oxidoreductase motif", "thiol-oxidoreductase motif", "thioreductase motif", "thioredox motif" or "redox motif" are used synonymously herein and refer to the general sequence thioreductase sequence motif C-X n -[CST]- (SEQ ID NO:26 to 30) or [ CST ]]-X n -a motif of C- (SEQ ID NO:1 to 5), wherein n is an integer from 0 to 6. Such peptide motifs exert reducing activity on disulfide bonds on proteins (e.g., enzymes) through redox-active cysteines within the conserved active domain consensus sequence: C-X n -[CST]-or [ CST ]]-X n -C-, for example as in C-XX-C, C-XX-S, C-XX-T, S-XX-C, T-XX-C (SEQ ID NO:187 to 191) (Fomenko et al (2003) Biochemistry 42,1 1214-1 1225), wherein "X" represents any amino acid, wherein C represents cysteine, S represents serine, T represents threonine, and X represents any amino acid other than tyrosine, phenylalanine or tryptophan. In another embodiment thereof, the oxidoreductase motif is located N-terminal to a T cell epitope. Alternatively, the immunogenic peptide may comprise an oxidoreductase motif of the general amino acid form: z as defined elsewhere herein m -[CST]-X n -C- (SEQ ID NOS: 1 to 25) or Z m -C-X n -[CST]- (SEQ ID NO:26 to 50), wherein n is an integer selected from 0 to 6, wherein m is an integer selected from 0 to 3, wherein X is any amino acid, wherein Z is any amino acid, wherein C represents cysteine, S represents serine, and T represents threonine.
When used in reference to amino acid residues present in the oxidoreductase motifs disclosed herein, the terms "cysteine" ("C"), "serine" ("S") and "threonine" ("T") refer to naturally occurring cysteine, serine or threonine amino acids, respectively. Unless explicitly stated differently, the term thus excludes chemically modified cysteines, serines and threonines, such as those modified to carry acetyl, methyl, ethyl or propionyl groups on the N-terminal amides of amino acid residues of the motif or on the C-terminal carboxyl groups.
The identification and selection of T cell epitopes from antigenic proteins is known to those skilled in the art.
To identify epitopes suitable in the context of the present invention, the isolated peptide sequences of the antigen proteins are tested, for example, by T cell biotechnology, to determine whether the peptide sequences elicit a T cell response. Those peptide sequences found to elicit a T cell response are defined as having T cell stimulatory activity.
Human T cell stimulatory activity can also be tested by: t cells obtained from an individual suffering from a fumarate-related disease or disorder are incubated with a peptide/epitope derived from an autoantigen involved in the disease or disorder, and it is determined whether proliferation of T cells occurs in response to the peptide/epitope, as measured, for example, by cellular uptake of tritiated thymidine. The stimulation index of T cell responses to peptides/epitopes can be calculated as the maximum CPM in response to the peptide/epitope divided by the control CPM. T cell stimulation index (s.i.) equal to or greater than twice background levels was considered "positive". Positive results were used to calculate the average stimulation index for each peptide/epitope of the subject peptide/epitope group.
Non-natural (or modified) T cell epitopes can also optionally be tested for their binding affinity to MHC class II molecules. This can be done in different ways. For example, soluble HLA class II molecules are obtained by lysing cells that are homozygous for a given class II molecule. The latter is purified by affinity chromatography. The soluble class II molecule is incubated with a biotin-labeled reference peptide, which is produced based on its strong binding affinity to the MHC class II molecule. The peptides to be evaluated for class II binding were then incubated at different concentrations and their ability to displace the reference peptide from their class II binding was calculated by adding neutravidin.
In order to determine the optimal T cell epitope by, for example, fine mapping techniques, peptides having T cell stimulatory activity and thus comprising at least one T cell epitope (as determined by T cell biotechnology) are modified by adding or deleting amino acid residues at the amino or carboxy terminus of the peptide and tested to determine changes in T cell reactivity against the modified peptide. If two or more peptides sharing overlapping regions in the native protein sequence are found to have human T cell stimulatory activity as determined by T cell biotechnology, additional peptides comprising all or part of such peptides can be produced and these additional peptides can be tested by similar procedures. According to this technique, the peptide is selected and produced recombinantly or synthetically. T cell epitopes or peptides are selected based on a variety of factors including the intensity of T cell responses to the peptides/epitopes (e.g., stimulation index) and the frequency of T cell responses to the peptides in a population of individuals.
Additionally and/or alternatively, one or more in vitro algorithms may be used to identify T cell epitope sequences in an antigen protein. Suitable algorithms include, but are not limited to, those described in the following: zhang et al (2005) Nucleic Acids Res, W180-W183 (PREDBALB); salomon & flow (2006) BMC Bioinformatics 7,501 (MHCBN); schulter et al (2007) Methods mol. Biol.409,75-93 (SYFPEITHI); donnes & Kohlbacher (2006) Nucleic Acids Res.34, W194-W197 (SVMHC); kolaskar & Tongaonkar (1990) FEBS Lett.276,172-174, guan et al (2003) appl.Bioinformation 2,63-66 (MHCPred) and Singh and Raghava (2001) Bioinformation 17,1236-1237 (Propred). More particularly, such algorithms allow prediction of one or more octapeptide or nonapeptide sequences in the groove of an MHC II molecule to be adapted to an antigen protein, and this is also true for different HLA types.
The term "MHC" refers to "major histocompatibility antigen (major histocompatibility antigen)". In humans, the MHC gene is called the HLA ("human leukocyte antigen (human leukocyte antigen)") gene. Although not always followed, some documents refer to HLA protein molecules using HLA and to genes encoding HLA proteins using MHC. Thus, as used herein, the terms "MHC" and "HLA" are equivalents. The HLA system in humans has its equivalent, the H2 system, in mice. The most deeply studied HLA genes are nine so-called classical MHC genes: HLA-A, HLA-B, HLA-C, HLA-DPA1, HLA-DPB1, HLA-DQA1, HLA DQB1, HLA-DRA and HLADRB1. In humans, MHC is divided into three regions: I. class II and III. A. The B and C genes belong to MHC class I, while the six D genes belong to class II. MHC class I molecules consist of a single polymorphic chain comprising 3 domains (α1, 2 and 3) that associate with β2 microglobulin on the cell surface. Class II molecules consist of 2 polymorphic chains, each comprising 2 chains (α1 and 2, and β1 and 2). Class I MHC molecules are expressed on almost all nucleated cells. Since the HLA system inherits in a Mendelian manner, HLA series genes or haplotypes can be distinguished in subjects of a given population.
In the global MS patient population, about 50% to 60% have HLA-DRB1 type 15:01. In addition, more than 75% of the MS patient population have HLA-DRB1 x 15:01, HLA-DRB1 x 03:01, HLA-DRB1 x 04:01, or HLA-DRB1 x 07:01 types of HLA. In view of the present invention, the preferred HLA type of the patient is thus selected from: DRB1 x 15:01, HLA-DRB1 x 03:01, HLA-DRB1 x 04:01 and HLA-DRB1 x 07:01. More preferred are patients with HLA-DRB1 x type 15:01. Further preferred are RRMS diagnosed MS patients with HLA types selected from the group consisting of: DRB1 x 15:01, HLA-DRB1 x 03:01, HLA-DRB1 x 04:01 and HLA-DRB1 x 07:01. Further preferred are RRMS diagnosed MS patients with HLA type HLA-DRB 1:15:01.
Preferred HLA haplotypes in NMO are HLA-DRB 1X 03:01 and HLA-DPB 1X 05:01 (for Asia).
Preferred HLA haplotypes in RA are HLA-DRB 1. Times.01:01, 04:01 and 04:04.
In humans, MHC is divided into three regions: I. class II and III. A. The B and C genes belong to MHC class I, while the six D genes belong to class II. MHC class I molecules consist of a single polymorphic chain comprising 3 domains (α1, 2 and 3) that associate with β2 microglobulin on the cell surface. Class II molecules consist of 2 polymorphic chains, each comprising 2 chains (α1 and 2, and β1 and 2). Class I MHC molecules are expressed on almost all nucleated cells. Peptide fragments presented in the context of class I MHC molecules are recognized by cd8+ T lymphocytes (cytotoxic T lymphocytes or CTLs). Cd8+ T lymphocytes often mature into cytotoxic effectors that lyse cells bearing stimulatory antigens. Class II MHC molecules are expressed predominantly on activated lymphocytes and antigen presenting cells. Cd4+ T lymphocytes (helper T lymphocytes or Th) are activated by recognition of unique peptide fragments presented by MHC class II molecules, which fragments are typically present on antigen presenting cells (e.g. macrophages, B cells or dendritic cells). CD4+ T lymphocytes proliferate and secrete cytokines that support antibody-mediated and cell-mediated responses, such as IL-2, IFN-gamma, and IL-4.
Functional HLA is characterized by deep binding grooves to which endogenous as well as exogenous, potential antigenic peptides bind. The groove is also characterized by a well-defined shape and physicochemical properties. The HLAI-like binding site is blocked because the peptide ends are fixed in the ends of the groove. They also participate in hydrogen bonding networks with conserved HLA residues. With these limitations in mind, the length of the bound peptide is limited to 7, 8, 9 or 10 residues. However, peptides of up to 12 amino acid residues have been shown to be able to bind HLAI classes as well. Comparison of different HLA complex structures determines a general binding pattern in which the peptide adopts a relatively linear, extended conformation or may involve the protrusion of a central residue out of the groove (bulge out).
In contrast to the HLAI binding site, the class II site is open at both ends. This allows the peptide to extend from the actual binding region, thus "overhang" at both ends. Thus, HLA class II can bind peptide ligands of variable length having 7 to more than 25 amino acid residues. Similar to HLA class I, the affinity of class II ligands is determined by "constant" and "variable" components. The constant portion is also created by a network of hydrogen bonds formed between the conserved residues in the HLA class II groove and the backbone of the binding peptide. However, this hydrogen bonding pattern is not limited to the N-terminal and C-terminal residues of the peptide, but is distributed throughout the chain. The latter is important because it limits the conformation of the complex peptide to a strictly linear binding mode. This is common to all class II allotypes. The second component determining peptide binding affinity is variable due to certain polymorphic positions within the class II binding site. Different allotypes form different complementary pockets within the groove, explaining subtype-dependent selection or specificity of peptides. Importantly, the restriction of the amino acid residues retained in the class II pocket is generally more "gentle" than for class I. There is much more peptide cross-reactivity between different HLA class II allotypes. Sequences of +/-9 amino acids (i.e., 8, 9 or 10) suitable for MHC class II T cell epitopes of the groove of MHC 11 molecules are generally numbered P1 to P9. The N-terminal amino acids of the other epitopes are numbered P-1, P-2 and the like, and the C-terminal amino acids of the epitopes are numbered P+1, P+2 and the like.
The term "NKT cell epitope" refers to a portion of an antigen protein that is specifically recognized and bound by a receptor on the cell surface of NKT cells. In particular, the NKT cell peptide epitope is an epitope bound by a CDld molecule, having the motif [ FWHY ] -XX- [ ILMV ] -XX- [ fvthy ] [ SEQ ID NO:51] or a more restrictive form thereof, e.g., [ FVV ] -XX- [ ILMV ] -XX- [ FVV ] [ SEQ ID NO:52]. In this motif, F represents phenylalanine, W represents tryptophan, H represents histidine, Y represents tyrosine, I represents isoleucine, L represents leucine, M represents methionine, V represents valine, and X represents any amino acid. FWHY indicates that F, W, H or Y can occupy the first anchor residue (P1), P4 can be occupied by I, L, M or V, and P7 can be occupied by F, W, H or Y. It will be clear to those skilled in the art that various combinations of these amino acid residues are possible.
The term "NKT cell" refers to a cell of the innate immune system, characterized by the fact that it carries receptors such as NK1.1 and NKG2D and recognizes peptide epitopes presented by CD1D molecules. In the context of the present invention, NKT cells may belong to a subset of type 1 (unchanged) or type 2, or to any of the less characterized NKT cells having more polymorphic T cell receptors than type 1 or type 2 NKT cells.
"CD1d molecule" refers to a molecule of non-MHC origin consisting of a group of 3 alpha chains and antiparallel beta chains expressed on the surface of various APCs, said beta chains being arranged to be open on both sides and capable of presenting lipids, glycolipids or hydrophobic peptides into the deep hydrophobic groove of an NKT cell.
The present invention provides methods for producing antigen-specific cytolytic cd4+ T cells in vivo or in vitro, and methods for distinguishing cytolytic cd4+ T cells from other cell populations, such as foxp3+ tregs, based on characteristic expression data, independent thereof.
An immunogenic peptide as defined herein comprises the oxidoreductase motif of the following general amino acid sequence: z as defined in aspect 2 m -[CST]-X n -C- (SEQ ID NOS: 1 to 25) or Z m -C-X n -[CST]- (SEQ ID NOS: 26 to 50) selected from the amino acid motifs:
(a) Z as defined in aspect 2 m -[CST]-X n -C-or Z m -C-X n -[CST]-, wherein n is 0, and wherein
Wherein m is an integer selected from 0 to 3,
wherein Z is any amino acid, preferably a basic amino acid selected from the group consisting of: H. k, R and a non-natural basic amino acid as defined herein, such as L-ornithine, preferably K or R, most preferably K.
In some preferred embodiments of motif (a), m is 1 or 2, and Z is a basic amino acid selected from the group consisting of: H. k, R and a non-natural basic amino acid as defined herein, such as L-ornithine, preferably K or R, most preferably K.
Some particularly preferred but non-limiting examples of such motifs are:
CC, KCC, KKCC (SEQ ID NO: 53), RCC, RRCC (SEQ ID NO: 54), RKCC (SEQ ID NO: 55), or KRCC (SEQ ID NO: 56).
(b) Z as defined in aspect 2 m -[CST]-X n -C-or Z m -C-X n -[CST]-, wherein n is 1,
wherein X is any amino acid, preferably a basic amino acid selected from the group consisting of: H. k, R and unnatural basic amino acids such as L-ornithine, preferably K or R, most preferably K,
wherein m is an integer selected from 0 to 3,
wherein Z is any amino acid, preferably a basic amino acid selected from the group consisting of: H. k, R and a non-natural basic amino acid as defined herein, such as L-ornithine, preferably K or R, most preferably K.
In some preferred embodiments of motif (b), m is 1 or 2, and Z is a basic amino acid selected from the group consisting of: H. k, R and a non-natural basic amino acid as defined herein, such as L-ornithine, preferably K or R, most preferably K.
Some particularly preferred but non-limiting examples of such motifs are:
CRC, CKC, KCXC (SEQ ID NO: 57), KKCXC (SEQ ID NO: 58), RCXC (SEQ ID NO: 59), RRCXC (SEQ ID NO: 60), RKCXC (SEQ ID NO: 61), KCXC (SEQ ID NO: 52), KCKCKCKC (SEQ ID NO: 63), KKKCKC (SEQ ID NO: 64), KCRC (SEQ ID NO: 65), KKCCRC (SEQ ID NO: 66), RCRC (SEQ ID NO: 67), RRCRC (SEQ ID NO: 68), RKCKC (SEQ ID NO: 69), or KCKC (SEQ ID NO: 70).
(c) Z as defined in aspect 2 m -[CST]-X n -C-or Z m -C-X n -[CST]-, where n is 2, thereby producing an internal X within the oxidoreductase motif 1 X 2 Amino acid conjugate, wherein m is an integer selected from 0 to 3, wherein Z is any amino acid, preferably a basic amino acid selected from the group consisting of: H. k, R and a non-natural basic amino acid as defined herein, such as L-ornithine, preferably K or R, most preferably K. Preferred are motifs wherein m is 1 or 2.
In some preferred embodiments, m is 1 and Z is a basic amino acid selected from the group consisting of: H. k or R or a non-natural basic amino acid as defined herein, such as L-ornithine, preferably K or R, most preferably K.
In some preferred embodiments, X 1 And X 2 Each independently may be any amino acid selected from the group consisting of: g, A, V, L, I, M, F, W, P, S, T, C, Y, N, Q, D, E, K, R, and H, or an unnatural amino acid. Preferably, X in the motif 1 And X 2 Is any amino acid other than C, S or T. In a specific embodiment, X in the motif 1 Or X 2 At least one of which is a basic amino acid selected from the group consisting of: H. k or R or a non-natural basic amino acid as defined herein, such as L-ornithine. In another embodiment, X in the motif 1 Or X 2 At least one of which is P or Y. Internal X within the oxidoreductase motif 1 X 2 Some specific examples of amino acid conjugates are: PY, HY, KY, RY, PH, PK, PR, HG, KG, RG, HH, HK, HR, GP, HP, KP, RP, GH, GK, GR, GH, KH, and RH.
Some particularly preferred motifs of this type are HCPYC, KCPYC, RCPYC, HCGHC, KCGHC, and RCGHC (corresponding to SEQ ID NOS: 71 to 76).
(d) Z as defined in aspect 2 m -[CST]-X n -C-or Z m -C-X n -[CST]-, where n is 3, thereby producing an internal X within the oxidoreductase motif 1 X 2 X 3 Amino acid segment, wherein m is an integer selected from 0 to 3, wherein Z is any amino acid, preferably a basic amino acid selected from the group consisting of: H. k, R and a non-natural basic amino acid as defined herein, such as L-ornithine, preferably K or R, most preferably K. Excellent (excellent)Selected are motifs wherein m is 1 or 2.
In some embodiments, X 1 、X 2 And X 3 Each independently may be any amino acid selected from the group consisting of: g, A, V, L, I, M, F, W, P, S, T, C, Y, N, Q, D, E, K, R, and H, or an unnatural amino acid. Preferably, X in the motif 1 、X 2 And X 3 Is any amino acid other than C, S or T. In a specific embodiment, X in the motif 1 、X 2 Or X 3 At least one of which is a basic amino acid selected from the group consisting of: H. k or R or a non-natural basic amino acid as defined herein, such as L-ornithine.
Internal X within the oxidoreductase motif 1 X 2 X 3 Some specific examples of amino acid segments are: XPY, PXY and PYX, where X may be any amino acid, preferably a basic amino acid such as K, R or H or a non-natural basic amino acid such as L-ornithine.
Some non-limiting examples are:
KPY, RPY, HPY, GPY, APY, VPY, LPY, IPY, MPY, FPY, WPY, PPY, SPY, TPY, CPY, YPY, NPY, QPY, DPY, EPY, and KPY; or alternatively
PKY, PRY, PHY, PGY, PAY, PVY, PLY, PIY, PMY, PFY, PWY, PPY, PSY, PTY, PCY, PYY, PNY, PQY, PDY, PEY, and PLY; or alternatively
PYK, PYR, PYH, PYG, PYA, PYV, PYL, PYI, PYM, PYF, PYW, PYP, PYS, PYT, PYC, PYY, PYN, PYQ, PYD, PYE, and PYL;
XHG, HXG and HGX, wherein X may be any amino acid, for example in the following:
KHG, RHG, HHG, GHG, AHG, VHG, LHG, IHG, MHG, FHG, WHG, PHG, SHG, THG, CHG, YHG, NHG, QHG, DHG, EHG, and KHG; or alternatively
HKG, HRG, HHG, HGG, HAG, HVG, HLG, HIG, HMG, HFG, HWG, HPG, HSG, HTG, HCG, HYG, HNG, HQG, HDG, HEG, and HLG; or alternatively
HGK, HGR, HGH, HGG, HGA, HGV, HGL, HGI, HGM, HGF, HGW, HGP, HGS, HGT, HGC, HGY, HGN, HGQ, HGD, HGE, and HGL;
XGP, GXP, and GPX, where X can be any amino acid, for example in the following:
KGP, RGP, HGP, GGP, AGP, VGP, LGP, IGP, MGP, FGP, WGP, PGP, SGP, TGP, CGP, YGP, NGP, QGP, DGP, EGP, and KGP; or alternatively
GKP, GRP, GHP, GGP, GAP, GVP, GLP, GIP, GMP, GFP, GWP, GPP, GSP, GTP, GCP, GYP, GNP, GQP, GDP, GEP, and GLP; or alternatively
GPK, GPR, GPH, GPG, GPA, GPV, GPL, GPI, GPM, GPF, GPW, GPP, GPS, GPT, GPC, GPY, GPN, GPQ, GPD, GPE, and GPL;
XGH, GXH and GHX, wherein X may be any amino acid, for example in the following:
KGH, RGH, HGH, GGH, AGH, VGH, LGH, IGH, MGH, FGH, WGH, PGH, SGH, TGH, CGH, YGH, NGH, QGH, DGH, EGH, and KGH; or alternatively
GKH, GRH, GHH, GGH, GAH, GVH, GLH, GIH, GMH, GFH, GWH, GPH, GSH, GTH, GCH, GYH, GNH, GQH, GDH, GEH, and GLH; or alternatively
GHK, GHR, GHH, GHG, GHA, GHV, GHL, GHI, GHM, GHF, GHW, GHP, GHS, GHT, GHC, GHY, GHN, GHQ, GHD, GHE, and GHL;
XGF, GXF, and GFX, where X may be any amino acid, for example, in the following:
KGF, RGF, HGF, GGF, AGF, VGF, LGF, IGF, MGF, FGF, WGF, PGF, SGF, TGF, CGF, YGF, NGF, QGF, DGF, EGF, and KGF; or alternatively
GKF, GRF, GHF, GGF, GAF, GVF, GLF, GIF, GMF, GFF, GWF, GPF, GSF, GTF, GCF, GYF, GNF, GQF, GDF, GEF, and GLF; or alternatively
GFK, GFR, GFH, GFG, GFA, GFV, GFL, GFI, GFM, GFF, GFW, GFP, GFS, GFT, GFC, GFY, GFN, GFQ, GFD, GFE, and GFL;
XRL, RXL and RLX, where X can be any amino acid, for example in the following:
KRL, RRL, HRL, GRL, ARL, VRL, LRL, IRL, MRL, FRL, WRL, PRL, SRL, TRL, CRL, YRL, NRL, QRLRL, DRL, ERL, and KRL; or alternatively
GKF, GRF, GHF, GGF, GAF, GVF, GLF, GIF, GMF, GFF, GVVF, GPF, GSF, GTF, GCF, GYF, GNF, GQF, GDF, GEF, and GLF; or alternatively
RLK, RLR, RLH, RLG, RLA, RLV, RLL, RL1, RLM, RLF, RLW, RLP, RLS, RLT, RLC, RLY, RLN, RLQ, RLD, RLE, and RLL;
XHP, HXP and HPX, where X can be any amino acid, for example in the following:
KHP, RHP, HHP, GHP, AHP, VHP, LHP, IHP, MHP, FHP, WHP, PHP, SHP, THP, CHP, YHP, NHP, QHP, DHP, EHP, and KHP; or alternatively
HKP, HRP, HHP, HGP, HAF, HVF, HLF, HIF, HMF, HFF, HWF, HPF, HSF, HTF, HCF, HYP, HNF, HQF, HDF, HEF, and HLP; or alternatively
HPK, HPR, HPH, HPG, HPA, HPV, HPL, HPI, HPM, HPF, HPW, HPP, HPS, HPT, HPC, HPY, HPN, HPQ, HPD, HPE, and HPL;
some particularly preferred examples are:
CRPYC, KCRPYC, KHCRPYC, RCRPYC, HCRPYC, CPRYC, KCPRYC, RCPRYC, HCPRYC, CPYRC, KCPYRC, RCPYRC, HCPYRC, CKPYC, KCKPYC, RCKPYC, HCKPYC, CPKYC, KCPKYC, RCPKYC, HCPKYC, CPYKC, KCPYKC, RCPYKC, and HCPYKC
(corresponding to SEQ ID NOS: 83 to 107).
(e) Z as defined in aspect 2 m -[CST]-X n -C-or Z m -C-X n -[CST]-, where n is 4, thereby producing an internal X within the oxidoreductase motif 1 X 2 X 3 X 4 Amino acid segment, wherein m is an integer selected from 0 to 3, wherein Z is any amino acid, preferably a basic amino acid selected from the group consisting of: H. k, R and a non-natural basic amino acid as defined herein, such as L-ornithine, preferably K or R, most preferably K. Preferred are motifs wherein m is 1 or 2. X is X 1 、X 2 、X 3 And X 4 Each independently may be any amino acid selected from the group consisting of: g, A, V, L, I, M, F, W, P, S, T, C, Y, N, Q, D, E, K, R, and H, or an unnatural amino acid as defined herein. Preferably, X in the motif 1 、X 2 、X 3 And X 4 Is any amino acid other than C, S or T. In a specific embodiment, X in the motif 1 、X 2 、X 3 Or X 4 At least one of which is a basic amino acid selected from the group consisting of: H. k or R or a non-natural basic amino acid as defined herein.
Some specific examples are: LAVL (SEQ ID NO: 108), TVQA (SEQ ID NO: 109) or GAVH (SEQ ID NO: 110), and variants thereof, for example: x is X 1 AVL,LX 2 VL,LAX 3 L, or LAVX 4 ;X 1 VQA,TX 2 QA,TVX 3 A, or TVQX 4 ;X 1 AVH,GX 2 VH,GAX 3 H, or GAVX 4 (corresponding to SEQ ID NOS: 112 to 122); wherein X is 1 、X 2 、X 3 And X 4 Each independently may be any amino acid selected from the group consisting of: g, A, V, L, I, M, F, W, P, S, T, C, Y, N, Q, D, E, K, R, and H, or a non-natural basic amino acid as defined herein.
(f) Z as defined in aspect 2 m -[CST]-X n -C-or Z m -C-X n -[CST]-, where n is 5, thereby producing an internal X within the oxidoreductase motif 1 X 2 X 3 X 4 X 5 (SEQ ID NO: 125) amino acid segment, wherein m is an integer selected from 0 to 3, wherein Z is any amino acid, preferably a basic amino acid selected from the group consisting of: H. k, R and a non-natural basic amino acid as defined herein, such as L-ornithine, preferably K or R, most preferably K. Preferred are motifs wherein m is 1 or 2. X is X 1 、X 2 、X 3 、X 4 And X 5 Each independently may be any amino acid selected from the group consisting of: g, A, V, L, I, M, F, W, P, S, T, C, Y, N, Q, D, E, K, R, and H,or an unnatural amino acid. Preferably, X in the motif 1 、X 2 、X 3 、X 4 And X 5 Is any amino acid other than C, S or T. In a specific embodiment, X in the motif 1 、X 2 、X 3 、X 4 Or X 5 At least one of which is a basic amino acid selected from the group consisting of: H. k or R or a non-natural basic amino acid as defined herein.
Some specific examples are: PAFPL (SEQ ID NO: 123) or DQGGE (SEQ ID NO: 124), and variants thereof, for example: x is X 1 AFPL,PX 2 FPL,PAX 3 PL,PAFX 4 L, or PAFPX 5 ;X 1 QGGE,DX 2 GGE,DQX 3 GE,DQGX 4 E, or DQGGX 5 (corresponding to SEQ ID NOS: 138 to 143), wherein X 1 、X 2 、X 3 、X 4 And X 5 Each independently may be any amino acid selected from the group consisting of: g, A, V, L, I, M, F, W, P, S, T, C, Y, N, Q, D, E, K, R, and H, or an unnatural amino acid as defined herein.
(g) Z as defined in aspect 2 m -[CST]-X n -C-or Z m -C-X n -[CST]-, where n is 6, thereby producing an internal X within the oxidoreductase motif 1 X 2 X 3 X 4 X 5 X 6 (SEQ ID NO: 137) amino acid segment, wherein m is an integer selected from 0 to 3, wherein Z is any amino acid, preferably a basic amino acid selected from the group consisting of: H. k, R and a non-natural basic amino acid as defined herein, such as L-ornithine, preferably K or R, most preferably K. Preferred are motifs wherein m is 1 or 2. X is X 1 、X 2 、X 3 、X 4 、X 5 And X 6 Each independently may be any amino acid selected from the group consisting of: g, A, V, L, I, M, F, W, P, S, T, C, Y, N, Q, D, E, K, R, and H, or an unnatural amino acid. Preferably, X in the motif 1 、X 2 、X 3 、X 4 、X 5 And X 6 Is any amino acid other than C, S or T.
In a specific embodiment, X in the motif 1 、X 2 、X 3 、X 4 、X 5 Or X 6 At least one of which is a basic amino acid selected from the group consisting of: H. k or R or a non-natural basic amino acid as defined herein.
Some specific examples are: DIADKY (SEQ ID NO: 136), or variants thereof, for example: x is X 1 IADKY,DX 2 ADKY,DIX 3 DKY,DIAX 4 KY,DIADX 5 Y, or DIADKX 6 (corresponding to SEQ ID NOS: 138 to 143), wherein X 1 、X 2 、X 3 、X 4 、X 5 And X 6 Each independently may be any amino acid selected from the group consisting of: g, A, V, L, I, M, F, W, P, S, T, C, Y, N, Q, D, E, K, R, and H, or a non-natural basic amino acid as defined herein.
(h)Z m -[CST]-X n -C-or Z m -C-X n -[CST]-, where n is 0 to 6 and where m is 0, and where C or [ CST ]]One of the residues has been modified to carry an acetyl, methyl, ethyl or propionyl group on the N-terminal amide or on the C-terminal carboxyl group of the amino acid residue of the motif (SEQ ID NOS: 144 to 163).
In some preferred embodiments of such motifs, n is 2 and m is 0, wherein internal X 1 X 2 Each independently may be any amino acid selected from the group consisting of: g, A, V, L, I, M, F, W, P, S, T, C, Y, N, Q, D, E, K, R, and H, or an unnatural amino acid. Preferably, X in the motif 1 And X 2 Is any amino acid other than C, S or T. In another example, X in the motif 1 Or X 2 At least one of which is a basic amino acid selected from the group consisting of: H. k or R or a non-natural basic amino acid as defined herein, such as L-ornithine. In another example of a motif, X in the motif 1 Or X 2 At least one of which is P or Y. Internal X within the oxidoreductase motif 1 X 2 Some specific non-limiting examples of amino acid conjugates are: PY, HY, KY, RY, PH, PK, PR, HG, KG, RG, HH, HK, HR, GP, HP, KP, RP, GH, GK, GR, GH, KH, and RH. Preferably, the modification results in N-terminal acetylation of the first cysteine (N-acetyl-cysteine) in the motif.
The oxidoreductase motif is placed next to the epitope sequence within the peptides of the invention or separated from the T cell or NKT cell epitope by a linker. More particularly, the linker comprises an amino acid sequence of 0 to 7 amino acids, i.e. O, 1, 2, 3, 4, 5, 6, 7, 8, 9 or 10 amino acids. Most particularly, the linker comprises an amino acid sequence of 0 to 4 amino acids, i.e. 0, 1, 2, 3 or 4 amino acids. Alternatively, the linker may comprise 5, 6, 7, 8, 9 or 10 amino acids.
In addition to peptide linkers, other organic compounds may be used as linkers to link portions of the peptide to each other (e.g., an oxidoreductase motif to a T cell or NKT cell epitope sequence).
The peptides of the invention may also comprise additional short amino acid sequences at the N-or C-terminus of the (artificial) sequence comprising a T-cell or NKT-cell epitope and an oxidoreductase motif. Such amino acid sequences are generally referred to herein as "flanking sequences". Flanking sequences may be located between the epitope and the endosomal targeting sequence and/or between the oxidoreductase motif and the endosomal targeting sequence. In other embodiments that do not include an endosomal targeting sequence, a short amino acid sequence may be present in the peptide at the N-terminus and/or C-terminus of the oxidoreductase motif and/or epitope sequence. More particularly, the flanking sequences are sequences of 1 to 7 amino acids, most particularly 2 amino acids.
The sequence comprising the T cell epitope and the reducing compound within the peptide may be further linked to an amino acid sequence (or another organic compound) that facilitates uptake of the peptide into late endosomes for processing and presentation in MHC class II or CD1d determinants. Late endosomal targeting is mediated by signals present in the cytoplasmic tail of the protein and corresponds to a putative peptide motif. Late endosomal targeting is mediated by signals present in the cytoplasmic tail of the protein and corresponds to putative peptide motifs, such as the dual leucine-based [ DE ] XXXL [ LI ] (SEQ ID NO: 192) or the DXXLL motif (SEQ ID NO: 193) (e.g., DXXXL, SEQ ID NO 194)), the tyrosine-based YXX motif or the so-called acidic cluster motif (SEQ ID NO: 195). Symbol 0 represents an amino acid residue with a large hydrophobic side chain, such as Phe, tyr and Trp. Late endosomal targeting sequences allow antigen-derived T cell epitopes to be processed and presented efficiently by MHC class II or CDld molecules. Such endosomal targeting sequences are contained, for example, within gp75 protein (Vijayasaradi et al (1995) J.cell. Biol.130, 807-820), human CD3 gamma protein, HLA-BM 11 (Copier et al (1996) J.1mM nol.157, 1017-1027), DEC205 receptor cytoplasmic tail (Mahnke et al (2000) J.cell biol.151, 673-683). Further examples of peptides that function as sorting signals for endosomes are disclosed in reviews by Bonifacio and Traub (2003) Annu.Rev.biochem.72, 395-447. Alternatively, the sequence may be a sequence from a subdominant or minor T cell epitope of the protein that promotes uptake in late endosomes but does not overcome T cell responses to the antigen. Advanced endosomal targeting sequences may be located at the amino-or carboxy-terminal end of the antigen-derived peptide for efficient uptake and processing, and may also be coupled through flanking sequences, such as peptide sequences of up to 10 amino acids. When a minor T cell epitope is used for targeting purposes, the latter is typically located at the amino-terminal end of the antigen-derived peptide.
The term "fumarate-related disorders" encompasses all disorders or diseases that benefit from treatment with a fumarate. Some preferred examples of such diseases or conditions are autoimmune conditions, demyelinating diseases, graft rejection and cancer. Some preferred examples of such diseases and conditions are: multiple Sclerosis (MS), psoriasis, neuromyelitis optica (NMO), rheumatoid Arthritis (RA), polyarthritis, asthma, atopic dermatitis, scleroderma, ulcerative colitis, juvenile diabetes, thyroiditis, grave's disease, systemic Lupus Erythematosus (SLE), sjogren's syndrome, pernicious anemia, chronic active hepatitis, transplant rejection and cancer. Some preferred examples are MOG autoantigen related diseases and disorders, such as MS and NMO.
The term "demyelination" as used herein within the framework of a demyelinating disease or disorder refers to the damage and/or degeneration of the myelin sheath surrounding the axons of neurons, with the result of the formation of lesions or plaques. Due to demyelination, signaling along the affected nerve is impaired and may cause neurological symptoms such as sensory, motor, cognitive and/or other neurological deficits. The specific symptoms of a patient suffering from a demyelinating disease will vary depending on the disease and the state of disease progression. These may include: blurred vision and/or vision (double vision), ataxia, clonic contracture, dysarthria, fatigue, clumsiness, hand paralysis, hemiparalysis, genital anesthesia, movement disorders, paresthesia, eye paralysis, impaired muscle coordination, muscle weakness, sensory loss, vision impairment, neurological symptoms, unstable walking patterns (gait), spastic paraplegia (spastic paraparesis), incontinence, hearing problems, speech problems, and the like. Demyelinating diseases can be classified into central nervous system demyelinating diseases and peripheral nervous system. Alternatively, demyelinating diseases may be categorized according to the cause of demyelination: damage to myelin sheath (demyelinating demyelination (demyelinating myelinoclastic)), or abnormal and degenerated myelin sheath (demyelinating leukodystrophy (dysmyelinating leukodystrophic)). MS is known in the art as a demyelinating disorder of the central nervous system (Lubetzki and stankoff. (2014). Handb Clin neurol.122, 89-99). Other specific but non-limiting examples of such demyelinating diseases and disorders include: neuromyelitis optica (NMO), acute inflammatory demyelinating polyneuropathy (acute inflammatory demyelinating polyneuropathy, AIDP), chronic inflammatory demyelinating polyneuropathy (Chronic inflammatory demyelinating polyneuropathy, CIDP), acute transverse myelitis, progressive Multifocal Leukoencephalopathy (PML), acute disseminated encephalomyelitis (acute disseminated encephalomyelitis, ADEM), or other hereditary demyelinating disorders.
The term "multiple sclerosis", abbreviated herein and in the art as "MS", refers to autoimmune disorders affecting the central nervous system. MS is considered the most common non-traumatic disabling disease in young adults (Dobson and Giovannoni, (2019) eur.j. Neurol.26 (1), 27-40), and the most common autoimmune disorder affecting the central nervous system (Berer and Krishnamoorthy (2014) FEBS lett.588 (22), 4207-4213). MS can manifest itself in subjects by a number of different symptoms ranging from physical to psychological to mental problems. Typical symptoms include blurred or blurred vision, muscle weakness, blindness of one eye, and difficulties in coordination and sensation. In most cases, MS can be considered as a two-stage disease, where early inflammation is the cause of relapsing remitting disease and delayed neurodegeneration results in non-relapsing progression, i.e., secondary and primary progressive MS. Despite advances in this area, the decisive underlying cause of disease has remained elusive to date, and more than 150 single nucleotide polymorphisms have been associated with MS susceptibility (International Multiple Sclerosis Genetics Consortium Nat genet. (2013). 45 (11): 1353-60). Vitamin D deficiency, smoke inhalation, ultraviolet B (UVB) exposure, childhood obesity and infection by EB virus have all been reported to contribute to disease development (aschorio (2013) experert Rev neuron.13 (12 journal), 3-9).
Thus, MS can be considered to be a single disease that exists in a range extending from recurrent (where inflammation is a dominant feature) to progressive (neurodegenerative dominant). It is therefore evident that the term multiple sclerosis as used herein encompasses any type of multiple sclerosis that belongs to any type of disease course classification. In particular, the present invention is contemplated as an effective therapeutic strategy for patients diagnosed with or suspected of having Clinically Isolated Syndrome (CIS), relapsing Remitting MS (RRMS), secondary Progressive MS (SPMS), primary Progressive MS (PPMS), and even Radiological Isolated Syndrome (RIS) of suspected MS. While the course of disease of MS is not strictly considered, RIS is used to classify subjects showing abnormalities on magnetic resonance imaging (Magnetic Resonance Imaging, MRI) of the brain and/or spinal cord that correspond to MS lesions and cannot be explained initially by other diagnostics. CIS is the first onset of neurological symptoms (by definition, lasting more than 24 hours) caused by inflammation and demyelination in the central nervous system. Subjects classified according to RIS, CIS may or may not continue to develop MS, with subjects displaying MS-like lesions on brain MRI being more likely to develop MS. RRMS is the most common disease course of MS, with 85% of subjects with MS diagnosed with RRMS. In view of the present invention, RRMS diagnosed patients are a preferred patient group. RRMS are characterized by new or increased attacks of neurological symptoms, either recurrent or worsening. In RRMS, the recurrence is followed by a periodic or partial or complete remission of symptoms, and no disease progression is experienced and/or observed during these remissions. RRMS can be further classified as active RRMS (evidence of relapse and/or new MRI activity), inactive RRMS, worsening RRMS (increased disability within a specific period of time after relapse), or non-worsening RRMS. A fraction of RRMS diagnosed subjects will progress to the progression of SPMS disease, characterized by progressive deterioration of neurological function over time, i.e., accumulation of disability. SPMS can be sub-classified, e.g., active (recurrent and/or new MRI activity), inactive, progressive (worsening of disease over time) or non-progressive SPMS. Finally, PPMS is the course of MS disease characterized by worsening neurological function and thus accumulation of disability from the onset of symptoms without early recurrence or remission. Other subgroups of PPMS can be formed, such as active PPMS (occasional relapse and/or new MRI activity), inactive PPMS, progressive PPMS (evidence of disease deterioration over time, regardless of new MRI activity), and non-progressive PPMS. In general, the course of MS disease is characterized by substantial inter-subject variability in both severity (in the case of relapse) and duration in terms of relapse and remission.
Several disease modifying treatments are available for MS and thus the present invention may be used as an alternative treatment strategy or in combination with these existing treatments. Some non-limiting examples of active pharmaceutical ingredients include: interferon beta-1 a, interferon beta-1 b, glatiramer acetate (glatiramer acetate), glatiramer acetate, polyethylene glycol interferon beta-1 a, teriflunomide (teriflunomide), fingolimod (fingolimod), cladribine (cladribine), cilnimod (siponimod), dimethyl fumarate, duloxetine fumarate, ozagrimod (ozanimod), alemtuzumab (alemtuzumab), mitoxantrone (mitoxantrone), omelizumab (ocrelizumab), and natalizumab (natalizumab). Alternatively, the present invention may be used in combination with therapies or drugs intended for relapse management, such as, but not limited to, methylprednisolone, prednisone, and adrenocorticotropic hormone (adrenocorticotropic hormone, ACTH). Furthermore, the present invention may be used in combination with therapies aimed at alleviating specific symptoms. Some non-limiting examples include drugs intended to ameliorate or avoid symptoms selected from the group consisting of: bladder problems, bowel dysfunction, depression, dizziness, mood changes, fatigue, itching, pain, sexual problems, cramps, tremors and walking difficulties.
The MS is characterized by three interleaved signature features: 1) lesion formation in the central nervous system, 2) inflammation, and 3) degeneration of neuronal myelin sheaths. Although traditionally thought to be a demyelinating disease of the central nervous system and white matter, recent reports indicate that demyelination of cortical and deep gray matter can exceed that of white matter (Kutzelnigg et al (2005). Brain.128 (11), 2705-2712). Two main assumptions have been assumed about how to induce MS at the molecular level. The generally accepted "outside-in-hypothesis" is based on activation of the peripheral autoreactive effector cd4+ T cells that migrate to the central nervous system and initiate the course of the disease. Once in the central nervous system, the T cells are locally reactivated by APC and recruit additional T cells and macrophages to establish inflammatory lesions. Notably, MS lesions have been shown to contain cd8+ T cells that reside primarily at the edges of the lesion as well as cd4+ T cells that reside at the center of disease modification. These cells are thought to cause demyelination, oligodendrocyte destruction, and axonal injury, leading to neurological dysfunction. In addition, the immune modulatory network is triggered to limit inflammation and initiate repair, which results in at least partial remyelination reflected by clinical remission. However, without adequate treatment, further attacks often lead to disease progression.
The onset of MS is believed to begin well before the first clinical symptoms are detected, as evidenced by the typical appearance of older and inactive lesions apparent on the patient's MRI. Due to advances in the development of diagnostic methods, MS can now be detected even before the clinical manifestation of the disease (i.e., pre-symptomatic MS). In the context of the present invention, "treatment of MS" and similar expressions contemplate treatment and therapeutic strategies for both symptomatic and pre-symptomatic MS. In particular, when immunogenic peptides and/or the resulting cytolytic cd4+ T cells are useful in treating pre-symptomatic MS patients, the disease stops at such an early stage that clinical manifestations can be partially or even completely avoided. MS in which the subject is not fully responsive to treatment with interferon beta is also encompassed by the term "MS".
The terms "neuromyelitis optica" or "NMO" and "NMO lineage disorders (NMO Spectrum Disorder, NMOSD)" also known as "Devic's disease" refer to autoimmune disorders in which leukocytes and antibodies attack predominantly the optic nerve and spinal cord but may also attack the brain (reviewed in Wingerchuk 2006,Int MS J.2006May;13 (2): 42-50). Injury to the optic nerve produces swelling and inflammation that causes pain and vision loss; injury to the spinal cord causes weakness or paralysis of the legs or arms, sensory loss, and problems with bladder and bowel function. NMO is a relapsing remitting disease. During recurrence, new injuries to the optic nerve and/or spinal cord can lead to cumulative disability. Unlike MS, there is no progressive phase of the disease. Thus, prevention of attacks is critical for good long-term outcomes. In the case of anti-MOG antibodies, it is believed that anti-MOG antibodies may trigger an attack on myelin, causing demyelination. In most cases, the cause of NMO is due to specific attack on self-antigens. Up to one third of subjects may be positive for autoantibodies against myelin components called Myelin Oligodendrocyte Glycoprotein (MOG). Patients with anti-MOG-related NMO similarly have episodes of transverse myelitis and optic neuritis. Particularly contemplated within the framework of the present application are NMO induced by MOG autoantigens and/or NMO caused by anti-MOG antibodies.
The term "rheumatoid arthritis" or "RA" is an autoimmune inflammatory disease that results in pain, swelling, stiffness, and loss of function in a variety of joints (most commonly found in the hands, wrists, and knees). The corresponding joint lining becomes inflamed, resulting in tissue damage, chronic pain, instability, and deformity. Disease progression is typically in bilateral/symmetric mode (e.g., both hands or knees are affected). RA can also affect extra-articular sites including the eye, mouth, lungs, and heart. Patients may experience an acute exacerbation of their symptoms (known as onset), but symptoms may be ameliorated over time with early intervention and appropriate treatment (reviewed by Sana Iqbal et al, 2019,US Pharm.2019;44 (1) (Specialty & Oncology suppl): 8-11). Antigens that are attacked by the immune system and cause disease are diverse, but some examples are: GRP78, HSP60, 60kDa chaperonin 2, gelsolin, chitinase-3-like protein 1, cathepsin S, serum albumin and cathepsin D.
The term "psoriasis" refers to chronic inflammatory skin diseases with a strong genetic predisposition and autoimmune pathogenic traits. Global prevalence is about 2%, but varies depending on region. It shows lower prevalence in asia and some african populations, and up to 11% in the Caucasian and scandinavia (Scandinavian) populations. The dermatological manifestations of psoriasis are different; psoriasis vulgaris is also known as plaque psoriasis and is the most common type. The terms psoriasis and psoriasis vulgaris are used interchangeably in the scientific literature; however, there is an important distinction between the different clinical subtypes. Psoriasis vulgaris (about 90% of cases of psoriasis) is chronic plaque psoriasis. Typical clinical manifestations are a distinct demarcation, erythema, and itchy plaque overlaid on silvery scales. Plaques may merge (coalesce) and cover a large area of skin. Common locations include the torso, extensor surfaces of the limbs, and the scalp. Other types are: inverse psoriasis, also known as flexor psoriasis, affects the location of the rugate interweaving (intrortricignius), and is clinically characterized by mild erosive erythema (plaque) and plaque (patch); spot psoriasis, which is a variant in the event of an acute episode of small erythema plaque. It usually affects children or adolescents and is usually triggered by group a streptococcal infection of the tonsils. About one third of patients with punctiform psoriasis will develop plaque psoriasis throughout their adulthood; pustular psoriasis, characterized by multiple, pooled sterile pustules. Pustular psoriasis may be localized or generalized. Two different local phenotypes have been described: palmoplantar pustular psoriasis (psoriasis pustulosa palmoplantaris, PPP) and halopeau's continuous acrodermatitis. Both of which affect the hand and A foot; PPP is localized to the palm and sole, while ACS is located farther from the tips of the fingers and toes and affects the nail organs (nail apparatus). The generalized pustule psoriasis manifests as an acute and rapidly progressive course characterized by diffuse redness and subcorneal pustules and is often accompanied by systemic symptoms. Psoriasis is marked by persistent inflammation that leads to uncontrolled keratinocyte proliferation and dysfunctional differentiation. The histology of psoriatic plaques shows a thickening of the stratum spinosum (epidermal hyperplasia) which covers the inflammatory infiltrate consisting of dermal dendritic cells, macrophages, T cells and neutrophils. Neovascularization is also a significant feature. The active inflammatory pathway in plaque psoriasis overlaps with that in the rest of the clinical variants, but also shows different differences leading to different phenotypes and therapeutic outcomes. (by render and)Int J Mol sci.2019mar;20 (6) reviewed in 1475).
When referring to peptides, the term "natural" refers to the fact that: the sequence is identical to a fragment of a naturally occurring protein (wild type or mutant). In contrast, the term "artificial" refers to sequences that are not themselves present in nature. Artificial sequences are obtained from natural sequences by limited modifications, such as altering/deleting/inserting one or more amino acids within the naturally occurring sequence or by adding/removing N-terminal or C-terminal amino acids of the naturally occurring sequence. The choice of antigen on which to design an epitope of an immunogenic peptide or tolerogenic peptide as described herein will depend on the fumarate-related disease.
Some exemplary antigens may be:
in the case of MS, myelin antigens, neuronal antigens and antigens of astrocyte origin, for example: myelin Oligodendrocyte Glycoprotein (MOG), myelin Basic Protein (MBP), proteolipid protein (PLP), oligodendrocyte-specific protein (OSP), myelin-associated antigen (MAG), myelin-associated oligodendrocyte basic protein (MOBP), and 2',3' -cyclic nucleotide 3' -phosphodiesterase (CNPase), S100 beta protein, or transaldolase H autoantigen (Riedhammer and Weissert,2015;Front Immunol.2015;6:322), preferably MOG, MBP, PLP and MOBP.
ADAMTS L5, PLA2G4D, keratins such as keratin 14 or keratin 17, antigens from wheat, pso p27, antimicrobial peptides, cetroroph defensin 1 and LL37, preferably LL37 (Jason E.Hawkes et al, volume 2017:Current Dermatology Reports, volume 6, pages 104 to 112).
In rheumatoid arthritis, N-acetylglucosamine-6-sulfatase (GNS), filamin a (FLNA), focal adhesion protein, GRP78, HSP60, 60kDa chaperonin 2, gelsolin, chitinase-3-like protein 1, cathepsin S, serum albumin and cathepsin D.
In the case of asthma, allergens, such as those derived from pollen, spores, dust mites and pet dander.
In the case of cancer, tumor or cancer associated antigens, such as oncogenes, proto-oncogenes, viral proteins, survival factors or clonotype or idiotype determinants. Some specific examples are: shows the MAGE (melanoma-associated gene) products spontaneously expressed by tumor cells in the context of MHC class I determinants and recognized by cd8+ cytolytic T cells as such. However, MAGE derived antigens such as MAGE-3 are also expressed in CD4+ specific T cells and MHC class II determinants that have been cloned from melanoma patients (Schutz et al (2000) Cancer Research 60:6272-6275; schulter-Thurer et al (2002) J.exp. Med.195:1279-1288). Peptides presented by MHC class II determinants are known in the art. Further examples include gp100 antigen expressed by P815 mastocytoma and by melanoma cells (Lapointe (2001; J.Immunol.167:4758-4764;Cochlovius et al. (1999) int.J.cancer, 83:547-554.) the protooncogene includes a number of polypeptides and proteins that are preferentially expressed in tumor cells but only minimally expressed in healthy tissue cyclin D1 is a cell cycle regulator involved in the conversion of G1 to S. High expression of cyclin D1 has been demonstrated in renal cell carcinoma, parathyroid carcinoma and multiple myeloma. Peptides encompassing residues 198 to 212 have been shown to carry T cell epitopes recognized in the context of MHC class II determinants (Dengiel et al (2004) Eur.J.of Immunol.34); 3644-3651) survivin is an example of a factor that inhibits apoptosis and thus confers the advantage of survivin expression cell expansion survivin is abnormally expressed in human cancers of epithelial and hematopoietic origin and not expressed in healthy adult tissues other than thymus, testis and placenta and in growth hormone stimulated hematopoietic progenitor and endothelial cells, it is interesting that survivin-specific CD8+ T cells can be detected in the Blood of melanoma patients survivin is expressed by a wide variety of malignant cell lines including kidney cancer, breast cancer and multiple myeloma, but also in acute myelogenous leukemia as well as acute and chronic lymphocytic leukemia (Schmidt (2003) Blood 102:571-576) other examples of inhibitors for apoptosis are Bcl2 and spi6 idiotype determinants presented by B cells in follicular lymphomas, multiple myelomas and some forms of leukemia, and by T cell lymphomas and some T cell leukemias. The idiotype determinant is part of an antigen specific receptor of a B Cell Receptor (BCR) or a T Cell Receptor (TCR). Such determinants are essentially encoded by hypervariable regions of the receptor corresponding to complementarity-determining region (CDR) regions of VH or VL regions in B cells, or CDR3 of β chains in T cells. Since the receptor is generated by random rearrangement of genes, it is unique to each individual. Peptides derived from idiotype determinants are present in MHC class II determinants (Baskar et al (2004) J.Clin. Invest. 113:1498-1510). Some tumors are associated with expression of virus-derived antigens. Thus, some forms of Hodgkin disease express antigens from Epstein-Barr virus (EBV). Such antigens are expressed in both class I and class II determinants. Cd8+ cytolytic T cells specific for EBV antigen can eliminate hodgkin's lymphoma cells (Bollard et a/(2004) j.exp. Med. 200:1623-1633). Antigenic determinants such as LMP-1 and LMP-2 are presented by MHC class II determinants.
In the case of graft rejection, the graft-specific antigen will obviously depend on the type of tissue or organ being transplanted. Some examples may be: tissue, such as cornea, skin, bone (bone fragments), blood vessels, or fascia; organs, such as kidney, heart, liver, lung, pancreas or intestine; or even individual cells such as islet cells, alpha cells, beta cells, muscle cells, cartilage cells, heart cells, brain cells, blood cells, bone marrow cells, kidney cells and liver cells. Some specific exemplary antigens involved in graft rejection are minor histocompatibility antigens, major histocompatibility antigens or tissue specific antigens. When the alloantigen protein is the major histocompatibility antigen, it is an MHC class I antigen or an MHC class II antigen. An important point to remember is the variability in the mechanism by which alloantigen-specific T cells recognize cognate peptides at the surface of APCs. The alloreactive T cells may recognize alloantigenic determinants of the MHC molecule itself, alloantigenic peptides bound to autologous or allogeneic derived MHC molecules, or a combination of residues located within the alloantigen derived peptides and MHC molecules (the latter being autologous or allogeneic derived). Some examples of minor histocompatibility antigens are those derived from proteins encoded by the HY chromosome (H-Y antigen), such as Dby. Further examples can be found in, for example, goulmy E, current Opinion in Immunology, volume 8, 75-81,1996 (see in particular Table 3 therein). It must be noted that many minor histocompatibility antigens in humans are detected by their presentation into MHC class I determinants using cytolytic cd8+ T cells. However, such peptides are derived by processing proteins that also contain MHC class II restricted T cell epitopes, thereby providing the feasibility of designing the peptides of the invention. Tissue specific alloantigens can be identified using the same procedure. An example of this is an MHC class I restriction epitope derived from a protein expressed in the kidney but not in the spleen and capable of eliciting CD8+ T cells with cytotoxic activity against kidney cells (Poindex et al Journal of Immunology,154:3880-3887,1995).
The term myelin oligodendrocyte glycoprotein refers to a human protein encoded by the MOG gene. The term MOG (protein) or myelin oligodendrocyte glycoprotein as used herein is defined by the amino acid sequence (SEQ ID NO: 184) corresponding to NCBI gene 4340 and UniProtKB identifier Q16653 (mog_human):
myelin oligodendrocyte glycoprotein is a membrane protein expressed on the oligodendrocyte cell surface and myelin outermost surface, and is the primary target antigen involved in immune-mediated demyelination. This protein may be involved in myelin completion and maintenance and intercellular communication. Alternative splice transcriptional variants have been identified that encode different isoforms. Thus, it is contemplated that the MOG epitope incorporated into the immunogenic or tolerogenic peptides of the invention may be an epitope present in a typical MOG amino acid sequence (SEQ ID NO: 184) and/or one or more MOG protein isoforms. In the context of the present invention, a suitable MOG epitope is one comprising FLRVPCWKI (SEQ ID NO: 164) or consisting of FLRVPCWKI (SEQ ID NO: 164). SEQ ID NOs of human and mouse MOG proteins: portion 164 is characterized by 100% sequence identity. Alternatively, the sequence of SEQ ID NO:164. alternatively, a point mutation may be introduced into the MOG epitope SEQ ID NO:164 to form the amino acid sequence FLRVPSWKI (SEQ ID NO: 165), which is a preferred MOG epitope in the context of the present invention. Further suitable MOG epitopes in the context of the present invention are MOG epitopes comprising VVHLYRNGK (SEQ ID NO: 170) or consisting of VVHLYRNGK (SEQ ID NO: 170). SEQ ID NOs of human and mouse MOG proteins: portion 164 is characterized by 100% sequence identity. The term "treatment" or variations thereof encompasses therapeutic treatment of a fumarate-related disease or disorder that has occurred. The term "prevention" refers to a prophylactic or preventative measure wherein the object is to prevent or reduce the chance of developing a fumarate-related disease or disorder. Beneficial or desired clinical results can include, but are not limited to, alleviation of one or more symptoms or one or more biomarkers, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, and the like. The term "treatment" or variant thereof may also mean prolonged survival compared to the expected survival if not treated.
The immunogenic peptide as defined herein may be adsorbed on an adjuvant suitable for administration to a mammal, such as aluminium hydroxide (alum). Typically, 50 μg of alum-adsorbed peptide was injected 3 times at 2 week intervals by subcutaneous route. It will be apparent to those skilled in the art that other routes of administration are also possible, including but not limited to oral, intranasal, or intramuscular. Furthermore, the number of injections and the amount of injections may vary depending on the severity of the condition to be treated and other parameters, such as the age, weight, general health, sex and diet of the patient. In addition, other adjuvants besides alum can be used, provided that they promote peptide presentation in MHC class II or CD1d and T cell or NKT cell activation. Thus, although the immunogenic peptide may be administered without any adjuvant, it is typically present as a pharmaceutical formulation. Formulations for both veterinary use and for human use comprise at least one immunogenic peptide as described above together with one or more pharmaceutically acceptable carriers.
The terms "peptide-encoding polynucleotide (or nucleic acid)" and "peptide-encoding polynucleotide (or nucleic acid)" as used herein refer to nucleotide sequences that, when expressed in a suitable environment, result in the production of the relevant peptide sequence or derivative or homologue thereof. Such polynucleotides or nucleic acids include normal sequences encoding the peptide, as well as derivatives and fragments of these nucleic acids that are capable of expressing peptides having the desired activity. A nucleic acid encoding a peptide or fragment thereof according to the invention is a sequence encoding a peptide or fragment thereof (most particularly a human peptide fragment) derived from or corresponding to a mammal. Such polynucleotides or nucleic acid molecules can be readily prepared using automated synthesizers and known codon-amino acid relationships of the genetic code. Such polynucleotides or nucleic acids may be incorporated into expression vectors, including plasmids, suitable for expression of the polynucleotide or nucleic acid and production of the polypeptide in a suitable host, such as a bacterium (e.g., escherichia coli)), yeast cell, human cell, animal cell, or plant cell.
For therapeutic approaches, polynucleotides encoding the immunogenic or tolerogenic peptides disclosed herein may be part of an expression system, cassette, plasmid, or vector system (e.g., viral and non-viral expression systems). Viral vectors known for therapeutic purposes are adenoviruses, adeno-associated viruses (AAV), lentiviruses and retroviruses. Non-viral vectors may also be used and some non-limiting examples include: transposon-based vector systems, such as those derived from Sleeping Beauty (SB) or PiggyBac (PB). Nucleic acids may also be delivered by other carriers (e.g., without limitation, nanoparticles, cationic lipids, liposomes, etc.).
The term "pharmaceutically acceptable carrier" as used herein with respect to a dosage form comprising a tolerogenic or immunogenic peptide as defined herein means any material or substance formulated with the immunogenic or tolerogenic peptide to facilitate its application or spreading at the site to be treated, for example by dissolving, dispersing or diffusing the composition, and/or to facilitate its storage, transport or handling without compromising its efficacy. Pharmaceutically acceptable carriers include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents (e.g., phenols, sorbic acid, chlorobutanol), isotonic agents (e.g., sugars or sodium chloride), and the like. In order to control the duration of action of the immunogenic or tolerogenic peptide in the pharmaceutical formulation, additional ingredients may be included. The pharmaceutically acceptable carrier may be a solid or liquid or a gas which has been compressed to form a liquid, i.e. the formulation may suitably be used as a concentrate, emulsion, solution, granule, dust (durt), spray, aerosol, suspension, ointment, cream, tablet, pill or powder. Suitable pharmaceutical carriers for pharmaceutical formulations of peptides are well known to those skilled in the art and their choice is not particularly limited within the present invention. Pharmaceutically acceptable carriers may also include additives such as wetting agents, dispersing agents, stickers, adhesives, emulsifying agents, solvents, coatings, antibacterial and antifungal agents (e.g., phenols, sorbic acid, chlorobutanol), isotonic agents (e.g., sugars or sodium chloride), and the like, provided that they are consistent with pharmaceutical practices, i.e., carriers and additives that do not cause permanent damage to mammals. Pharmaceutical formulations of the immunogenic or tolerogenic peptides may be prepared in any known manner, for example by homogeneously mixing the active ingredient with the selected carrier material and, if appropriate, further additives (e.g. surfactants), coating and/or grinding it in one or more operations. They can also be prepared by micronization (micronization), for example, considering that they are obtained in the form of microspheres, generally having a diameter of about 1 to 10 μm, i.e. for the preparation of microcapsules for the controlled or sustained release of immunogenic or tolerogenic peptides.
The pharmaceutical composition may comprise a therapeutically or prophylactically effective amount of the or each immunogenic or tolerogenic peptide, and optionally a pharmaceutically acceptable carrier, diluent or excipient. Furthermore, in the pharmaceutical compositions of the invention, the or each immunogenic or tolerogenic peptide may be admixed with any suitable binder, lubricant, suspending agent, coating agent or solubilising agent.
Suitable surfactants (also known as emulsifiers) or emulsifying agents (emulgators) for pharmaceutical formulations of immunogenic or tolerogenic peptides are nonionic, cationic and/or anionic materials with good emulsifying, dispersing and/or wetting properties. Suitable anionic surfactants include both water-soluble soaps and water-soluble synthetic surfactants. Suitable soaps are higher fatty acids (C 10 To C 22 ) An unsubstituted or substituted ammonium salt, such as the sodium or potassium salt of oleic or stearic acid, or the sodium or potassium salt of a mixture of natural fatty acids obtainable from coconut oil or tallow oil (tall oil). Synthetic surfactants include sodium or calcium salts of polyacrylic acid; fatty sulfonates and sulfates; sulfonated benzimidazole derivatives and alkylaryl sulfonates. Fatty sulfonates or sulfates are typically in the following form: alkali metal salts or alkaline earth metal salts, unsubstituted ammonium salts or ammonium salts substituted by alkyl or acyl groups having 8 to 22 carbon atoms, for example sodium or calcium salts of lignin sulfonic acid (lignosulphonic acid) or dodecyl sulfonic acid, or mixtures of fatty alcohol sulfates obtained from natural fatty acids, alkali metal salts or alkaline earth metals of sulfates or sulfonates Salts (e.g., sodium lauryl sulfate) and sulfonic acids of fatty alcohol/ethylene oxide adducts. Suitable sulphonated benzimidazole derivatives generally contain from 8 to 22 carbon atoms. Some examples of alkylaryl sulfonates are sodium, calcium or alkanolamine salts of dodecylbenzenesulfonic acid or dibutyl-naphthalenesulfonic acid or naphthalene-sulfonic acid/formaldehyde condensation products. Also suitable are the corresponding phosphates, for example the adducts of phosphoric acid esters and p-nonylphenols with ethylene oxide and/or propylene oxide, or the salts of phospholipids. Suitable phospholipids for this purpose are natural (of animal or plant cell origin) or synthetic phospholipids of the cephalin or lecithin type, such as phosphatidylethanolamine, phosphatidylserine, phosphatidylglycerol, lysolecithin, cardiolipin, dioctyl phosphatidylcholine, dipalmitoyl phosphatidylcholine, and mixtures thereof. Suitable nonionic surfactants include polyethoxylated and polypropoxylated derivatives of alkylphenols, fatty alcohols, fatty acids, aliphatic amines or amides containing at least 12 carbon atoms in the molecule, alkylaromatic sulfonates and dialkylsulfosuccinates, such as polyethylene glycol ether derivatives of aliphatic and cycloaliphatic alcohols, saturated and unsaturated fatty acids and alkylphenols, said derivatives typically containing 3 to 10 glycol ether groups and 8 to 20 carbon atoms in the (aliphatic) hydrocarbon moiety and 6 to 18 carbon atoms in the alkyl moiety of the alkylphenol. Further suitable nonionic surfactants are water-soluble adducts of polyethylene oxide with polypropylene glycol, ethylenediamine-based polypropylene glycols (ethylenediaminopolypropylene glycol) containing from 1 to 10 carbon atoms in the alkyl chain, which adducts contain from 20 to 250 glycol ether groups and/or from 10 to 100 propylene glycol ether groups. Such compounds typically comprise 1 to 5 ethylene glycol units/propylene glycol units. Representative examples of nonionic surfactants are nonylphenol-polyethoxyethanol, castor oil polyethoxyethanol ether (castor oil polyglycolic ether), polypropylene/polyethylene oxide adducts, tributylphenoxy polyethoxyethanol, polyethylene glycol, and octylphenoxy polyethoxyethanol. The following are also suitable nonionic surfactants: fatty acid esters of polyethylene sorbitan (e.g. polyoxyethylene sorbitan trioleate), glycerol, sorbitan, sucrose Sugar and pentaerythritol. Suitable cationic surfactants include quaternary ammonium salts, particularly halides, having 4 hydrocarbyl groups optionally substituted with halogen, phenyl, substituted phenyl or hydroxy groups; for example comprising at least one C as N-substituent 8 C 22 Alkyl groups (e.g., cetyl, lauryl, palmityl, myristyl, oleyl, etc.), and as additional substituents, quaternary ammonium salts of unsubstituted or halogenated lower alkyl, benzyl and/or hydroxy-lower alkyl.
Pharmaceutical dosage forms or formulations of immunogenic or tolerogenic peptides suitable for injectable use include sterile aqueous solutions or dispersions; formulations including sesame oil, peanut oil or aqueous propylene glycol; sterile powders for the preparation of sterile injectable solutions or dispersions of ready-to-use (extemporaneous) formulations. In all cases, the form must be sterile and must be fluid to the extent that easy injectability is achieved. It must be stable under the conditions of preparation and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier may also be a solvent or dispersion medium comprising, for example, water, ethanol, polyols (e.g., glycerol, propylene glycol, and liquid polyethylene glycols, and the like), suitable mixtures thereof, and vegetable oils. Proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersions and by the use of surfactants. Prevention of microbial action can be brought about by a variety of antibacterial and antifungal agents, such as parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the composition of delayed absorption agents, for example, aluminum monostearate and gelatin.
Sterile injectable solutions are prepared by incorporating the immunogenic or tolerogenic peptide in the required amount in the appropriate solvent with various other ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating a sterilized immunogenic or tolerogenic peptide into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the immunogenic or tolerogenic peptide plus any additional desired ingredient from a previously sterile-filtered solution thereof.
In formulation, the pharmaceutical formulation as defined herein or the peptide as defined herein or the fumarate compound as defined herein may be administered in a manner compatible with the dosage formulation and in such amounts as to be therapeutically effective.
Administration of tolerogenic peptides should preferably be performed in soluble form in the absence of an adjuvant. The tolerogenic peptide of the invention or a pharmaceutical composition comprising such a tolerogenic peptide as defined herein is preferably administered by: delivery via mucous membranes, for example, by nasal, oral, buccal, pulmonary, ocular, vaginal or rectal delivery; or by intradermal, transdermal or subcutaneous injection. Studies have shown that tolerogenic peptides can induce T cell tolerance when administered intraperitoneally (i.p.), intravenously (i.v.), or intranasally (i.n.), or orally, in soluble form (e.g., as Anderton and Wraith (1998); as Liu and Wraith (1995); metzler and Wraith (1999) Immunology 97:257-263). A dose escalation regimen may be followed in which multiple doses of tolerogenic peptide are administered to a patient at increasing concentrations, as have been successfully tested in the case of bee venom (bee venom) allergies (Muller et al (1998) j. Allergy Clin immunol.101:747-754 and Akdis et al (1998) j. Clin. Invest. 102:98-106) and as disclosed in WO 2018127828. In one embodiment, the tolerogenic peptide may be formulated according to techniques known in the art and exemplified, for example, in patent application WO2013160865 A1.
The immunogenic peptide of the invention or a pharmaceutical composition comprising such an immunogenic peptide as defined herein is preferably administered by subcutaneous or intramuscular administration. Preferably, the peptide or pharmaceutical composition comprising such peptide may be injected Subcutaneously (SC) into the region of the lateral part of the upper arm intermediate the elbow and shoulder. When two or more separate injections are required, they may be concomitantly administered in both arms.
The immunogenic peptide according to the invention or the pharmaceutical composition comprising such immunogenic peptide is administered in a therapeutically effective dose. Some exemplary but non-limiting dosage regimens are 50 to 1500 μg, preferably 100 to 1200 μg. Some more specific dosage regimens may be 50 to 250 μg,250 to 450 μg or 850 to 1300 μg depending on the condition of the patient and the severity of the disease. The dosage regimen may comprise administration in a single dose or simultaneously or consecutively in 2, 3, 4, 5 or more doses.
In certain embodiments, the treatment may be repeated several times throughout the disease in the subject. Such continuous treatment may be performed daily or at intervals of 1 to 10 days, such as every 5 to 9 days (e.g., about every 7 days), for example.
Alternatively, the treatment may be repeated weekly, biweekly, monthly, bi-monthly, or tri-to-tetra month.
Some exemplary non-limiting administration regimens are as follows:
a low dose regimen comprising SC administration of 50 μg of peptide, performed as two separate injections of 25 μg each (100 μl each); three subsequent injections of 25 μg of the immunogenic peptide were performed as two separate injections of 12.5 μg each (50 μl each).
A medium dose regimen comprising SC administration of 150 μg of peptide, performed as two separate injections of 75 μg each (300 μl); three subsequent consecutive administrations of 75 μg of the immunogenic peptide were performed with two separate injections of 37.5 μg (150 μl each) of the immunogenic peptide.
A high dose regimen comprising SC administration of 450 μg of peptide, in two separate injections of 225 μg each (900 μl each); three subsequent consecutive administrations of 225. Mu.g of the immunogenic peptide were performed with two separate injections of 112.5. Mu.g each (450. Mu.L each).
Other exemplary non-limiting administration regimens are as follows:
dose regimen comprising 6 SC administrations (2 weeks apart) of 450 μg of immunogenic peptide, said administrations of 450 μg of immunogenic peptide being performed in two separate injections of 225 μg each.
Dose regimen comprising 6 SC administrations (2 weeks apart) of 1350 μg of immunogenic peptide, said administrations being carried out in two separate injections of 675 μg each.
The immunogenic peptide or tolerogenic peptide formulation is readily administered in a variety of dosage forms (e.g., the types of injectable solutions described above), but drug release capsules and the like may also be used. For example, for parenteral administration in aqueous solution, the solution should be buffered appropriately if necessary, and the liquid diluent first isotonic with sufficient saline or glucose. These particular aqueous solutions are particularly suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration. In this regard, sterile aqueous media that may be employed will be known to those of skill in the art in light of the present disclosure. For example, a dose may be dissolved in 1ml of isotonic NaCl solution and added to 1000ml of subcutaneous infusion fluid, or injected at the proposed infusion site. Depending on the condition of the subject being treated, some variation in the dosage will necessarily occur. In any event, the person responsible for administration will determine the appropriate dosage for the individual subject.
Administration of the immunogenic peptide should preferably be performed in soluble form in the presence of an adjuvant.
The immunogenic or tolerogenic peptide, homologue or derivative thereof according to the invention (and physiologically acceptable salts or pharmaceutical compositions thereof, all included in the term "active ingredient") may be administered by any route suitable for the condition to be treated and for the compound (herein proteins and fragments to be administered). Possible routes include regional, systemic, oral (solid form or inhalation), rectal, nasal, topical (including ocular, buccal and sublingual), vaginal and parenteral (including subcutaneous, intramuscular, intravenous, intradermal, intraarterial, intrathecal and epidural). The preferred route of administration may vary depending upon, for example, the condition of the recipient or the disease to be treated. As described herein, the carrier is optimally "acceptable" in the sense of being compatible with the other ingredients of the formulation and not deleterious to the recipient thereof. Such formulations include those suitable for oral, rectal, nasal, topical (including buccal and sublingual), vaginal and parenteral (including subcutaneous, intramuscular, intravenous, intradermal, intraarterial, intrathecal and epidural) administration.
Formulations suitable for parenteral administration include: aqueous and non-aqueous sterile injection solutions which may contain antioxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient; and aqueous and non-aqueous sterile suspensions, which may include suspending agents and thickening agents. The formulations may be presented in unit-dose or multi-dose containers (e.g., sealed ampoules and vials) and may be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid carrier (e.g., water for injection) immediately prior to use. Ready-to-use injection solutions and suspensions can be prepared from sterile powders, granules and tablets of the type previously described.
Typical unit dosage formulations are those containing a daily dose or unit daily sub-dose of the active ingredient as hereinbefore described, or an appropriate fraction thereof. It will be appreciated that in addition to the ingredients specifically mentioned above, the formulations of the present invention may comprise other agents conventional in the art, such as those suitable for oral administration, which may comprise flavoring agents, in view of the type of formulation in question. The peptides according to the invention, homologues or derivatives thereof may be used to provide controlled release pharmaceutical formulations comprising one or more compounds of the invention as active ingredient ("controlled release formulations"), wherein the release of the active ingredient may be controlled and regulated to allow for less frequent administration or to improve the pharmacokinetic profile or toxicity profile of a given compound of the invention. Controlled release formulations suitable for oral administration may be prepared according to conventional methods, wherein the discrete units comprise one or more compounds of the invention. Additional ingredients may be included in order to control the duration of action of the active ingredients in the composition. Thus, the controlled release composition may be obtained by selecting a suitable polymeric carrier such as, for example, polyesters, polyamino acids, polyvinylpyrrolidone, ethylene-vinyl acetate copolymers, methylcellulose, carboxymethylcellulose, protamine sulfate and the like. The rate of drug release and duration of action can also be controlled by incorporating the active ingredient into particles of the polymer (e.g., microcapsules, microspheres, microemulsions, nanoparticles, nanocapsules, etc.). Depending on the route of administration, the pharmaceutical composition may require a protective coating. Pharmaceutical forms suitable for injection include sterile aqueous solutions or dispersions and sterile powders for the ready-to-use preparation thereof. Typical carriers for this purpose therefore include biocompatible aqueous buffers, ethanol, glycerol, propylene glycol, polyethylene glycol and the like, and mixtures thereof. In view of the following facts: when several active ingredients are used in combination, they do not necessarily exert their combined therapeutic effect directly in the mammal to be treated at the same time, and thus the corresponding composition may also be in the form of a medical kit or package containing the two ingredients in separate but adjacent reservoirs or chambers. Thus, in the latter case, each active ingredient may be formulated in a manner suitable for a different route of administration than that of the other ingredient, for example, one of them may be in the form of an oral or parenteral formulation, while the other is in the form of an ampoule or aerosol for intravenous injection.
The invention is further illustrated by the following examples, which are not intended to limit the scope of the invention in any way.
Examples
Example 1: with dimethyl fumarate (BG-12, TECFIDERA) TM ) Combined comprising MOG 35 to 55 Prophylactic administration of immunogenic peptides of MHCII T cell epitopes and HCPYC oxidoreductase motifs has an effect on the occurrence of experimental autoimmune encephalomyelitis (experimental auto-immune encephalomyelitis, EAE) in mice.
Groups of mice and administration
A total of 64 female C57BL/6 mice (Taconic Biosciences, 10 weeks of age at day 0) were used for the study. Mice were acclimatized for 8 days prior to the first injection. At the beginning of the study, mice were assigned to groups in a balanced manner to achieve similar average body weights between groups. Table 1 below shows the treatments applied to each group.
TABLE 1 treatment protocol
Treatment 1 was administered once per day at a volume s.c. of 0.05 mL/site shown in table 1, with each mouse receiving injections at two sites, for a total of 0.1 mL/mouse/dosing day, corresponding to 100 μg of peptide.
Treatment 2 was administered at a volume p.o.BID of 10mL/kg over the days indicated in Table 1. BG-12 was administered at 100 mg/kg. All administrations were carried out at the same time (+/-1 hour) per day of administration. For the BID group, dosing intervals were no less than 10 hours and no more than 14 hours.
Preparation of Compounds
For treatment 1, 0.9% NaCl solution was prepared on each dosing day. Immediately prior to use, lyophilized immunogenic peptide IMCY-0189 having sequence HCPYCGWYRSPFSRVVHLYR (SEQ ID NO: 185) comprising the oxidoreductase motif HCPYC (SEQ ID NO: 71), linker GW, murine Myelin Oligodendrocyte Glycoprotein (MOG) 35 to 55 ) MHCII T cell epitope YRSPFSRVV (SEQ ID NO: 169) and flanking sequence HLYR (SEQ ID NO: 186) (Smart Bioscience). Lyophilized IMCY-0189 was thawed at room temperature for 10 minutes, resuspended in 50mM sodium acetate buffer pH 5.4, and incubated at room temperature for 5 minutes. The reconstituted peptide was then combined with an Imject prior to injection TM Alum adjuvant.
For treatment 2, the vehicle was 0.5% HPMC, 0.2% tween 20 and 50mM citrate buffer at pH 4. Preparation of BG-12 (or TECFIDERA) once a week TM From Santa Cruz Biotechnology, catalog No. sc-239774). At each preparation, the desired amount of BG-12 was weighed into a mortar and ground with a pestle. The loading agent is then added in small increments and mixed until the final volume is reached. The material was then vortexed and sonicated in a water bath until a uniform suspension was obtained. The prepared BG-12 was stored at 4℃with continuous stirring.
EAE induction
EAE was induced in all mice as follows:
day 0, hour 0-with amino acids 35 to 55 of MOG (MOG) 35 to 55 ) Immunization with CFA-corresponding peptides
Day 0, 2 hours-pertussis (pertussis) toxin injection
Day 1, hours 0-2 pertussis toxin (24 hours after initial immunization) was injected.
Hooke Kit was subcutaneously injected at two sites on the back of mice TM MOG 35 to 55 Emulsion component (containing MOG) of CFA emulsion PTX (catalog number EK-2110 (batch number 127, hooke laboratories, lawrence Mass.) 35 to 55 ). One injection site was located about 1cm posterior to the neck line in the upper back region. The second site is at the lower back region, approximately 2cm of the skull (crannial) at the base of the tail. The injection volume at each site was 0.1mL. Each mouse received 200 μg MOG 35 to 55
The pertussis toxin component in the kit is administered intraperitoneally within 2 hours of the injection of the emulsion, and then the pertussis toxin component in the kit is administered intraperitoneally again 24 hours after the injection of the emulsion. Pertussis toxin (batch number 1008, hooke laboratories) was administered at 100 ng/dose for two injections, and the volume per injection was 0.1mL.
EAE scoring
Animals were scored daily from day 7 to the end of the study. Scoring was performed blindly by persons blinded to both treatment and prior scoring of each mouse. EAE were scored on a scale of 0 to 5 as shown in table 2 below. An intermediate score is specified when a clinical sign falls between two or more defined scores.
TABLE 2 EAE scoring criteria
Scoring of Clinical observations
0 The exercise function has no obvious change
1 Tail fatigue soft (limp tail)
2 Fatigue and weakness of the rear legs
3 Fatigue and complete paralysis of the hind legs
4 The tail is soft, the rear leg is completely paralyzed and the front leg is partially paralyzed
5 Complete paralysis of the hind legs and complete paralysis of the front legs, or death due to paralysis
Plasma neurofilament level determination
On day 28, blood was collected from all mice into tubes containing K2EDTA and gently mixed. The blood was then centrifuged at about 10000g for 5 minutes. The plasma was transferred to Eppendorf tubes and stored at-80℃until shipment to Quantix TM . UsingNF-light dominance kits (digital immunoassays for quantitative determination of NF-L in serum, plasma and CSF) quantify plasma neurofilament light (NF-L) protein levels. Antibodies used (Uman Diagnostics, ">Sweden) also cross-reacts with NF-L epitopes from mice, cattle and rhesus, and therefore, the assay Can be used for research using these species. All samples were tested in duplicate at a dilution factor of 40 x.
Terminal collection
At the end of the study, all mice were euthanized and the spinal column (spin) was collected and placed in 10% buffered formalin for histological analysis.
Histological examination
For each spinal column, one H & E stained slide and one anti MBP stained slide were prepared and analyzed. Each slide contained sections with samples (3 samples) from spinal cord of the waist, chest and neck. All analyses were performed by pathologists blinded to the experimental group and all clinical reads.
Inflammatory lesions were counted for about 20 cells in each H & E stained section. When inflammatory infiltrates consisted of more than 20 cells, an estimate was made of how many 20-cell lesions were present.
Demyelination was scored in each anti-MBP (using immunohistochemistry) stained section. In anti-MBP sections, demyelination was observed as a distinct undyed region in white matter tracts and correlated with the presence of large vacuoles. The demyelination score represents an estimate of the demyelination area of each slice as follows:
0-demyelination free (less than 5% demyelination area)
1-5% to 20% demyelination area
Area of demyelination of 2-20% to 40%
3-40% to 60% demyelination area
Area of demyelination of 4-60% to 80%
Area of demyelination of 5-80% to 100%
Statistical analysis
Quantitative data for AUC, MMS, inflammation and demyelination and NF-L levels were analyzed by performing a common one-way ANOVA. The Holm-Sidak method was used for multiplexing adjustment. The significance differences were referenced as follows: * p <0.05, < p <0.01, < p <0.001, < p <0.0001.
Results and data interpretation
EAE scoring
EAE occurrence was assessed by comparing clinical EAE readout of all groups with the negative control (saline/vehicle) group. EAE score, AUC (area under the curve) and MMS (average max score) are given in figures 1, 2 and 3.
For this model, mice of the saline/vehicle group (negative control) developed typical EAE. Two (2) mice in this group died from severe EAE.
Mice treated with BG-12 (saline/BG-12 group) showed delayed onset of disease and reduced endpoint scores, as well as statistically significant reductions in AUC and MMS compared to the negative control group. No mice in this group died.
Mice treated with IMCY-0189 (IMCY-0189/vehicle group) also showed delayed onset and reduced endpoint scores, as well as statistically significantly reduced AUC and MMS, compared to the negative control group. These clinical results were shown to be similar to those observed with BG-12 treatment. Two (2) mice in this group died. One (1) mouse died from severe EAE. The death of the other mouse was shown not to be due to EAE and therefore the mouse was excluded from the analysis.
All clinical readouts (onset of disease, endpoint scores, AUC and MMS) were statistically significantly improved in mice treated with both IMCY-0189 and BG-12 compared to negative control groups, and also especially compared to mice treated with either IMCY-0189 alone (IMCY-0189/vehicle group) or BG-12 alone (saline/BG-12 group). One (1) mouse in this group died, but the death of this mouse was shown not to be due to EAE and was therefore excluded from the analysis. The interaction between IMCY-0189 and BG-12 treatments was analyzed by two-way ANOVA. The trend of synergy was assessed by a p-value near 0.2 (0.2373) for MMS data.
Histological examination
Histological readout was assessed by comparing the inflammation and demyelination levels of all groups with the negative control (saline/vehicle) group. Inflammation and demyelination data are given in figures 4 and 5.
The histological results of the saline/vehicle group (negative control) were consistent with the clinical findings and as expected for this model.
Mice treated with BG-12 (saline/BG-12, positive control) showed similar demyelination levels and reduced inflammation levels compared to the negative control group, but were not statistically significant.
Similarly, mice treated with IMCY-0189 (IMCY-0189/vehicle group) showed reduced levels of both inflammation and demyelination, but were not statistically significant.
Histological readout of mice treated with IMCY-0189 and BG-12 was also statistically significantly improved compared to negative control groups, and also especially compared to mice treated with IMCY-0189 alone (IMCY-0189/vehicle group) or BG-12 alone (saline/BG-12 group). The interaction between IMCY-0189 and BG-12 treatments was analyzed by two-way ANOVA. Synergy between the two treatments was assessed by an interaction p-value <0.05 for demyelinated data.
Plasma neurofilament level
Neurofilament light (NF-L) is a 68kDa cytoskeletal silk protein expressed in neurons as one of the major components of the neuronal cytoskeleton that provides structural support to axons. The nerve filaments may be released after axonal injury or degeneration of neurons. NF-L has been shown to be associated with neurodegenerative diseases such as multiple sclerosis.
The experiments described in table 1 were repeated to evaluate axonal damage by comparing NF-L levels of all groups with negative control (saline/vehicle) groups. The data are given in figure 6.
NF-L levels of saline/vehicle group (negative control) were consistent with clinical findings and as expected for this model.
Mice treated with BG-12 (saline/BG-12 group, positive control) showed similar NF-L levels compared to the negative control group.
Mice treated with IMCY-0189 (IMCY-0189/vehicle group) and mice treated with both IMCY-0189 and BG-12 (IMCY-0189/BG-12 group) showed reduced NF-L levels.
Example 2: with dimethyl fumarate (BG-12, TECFIDERA) TM ) Combined inclusion with HCPYMOG with or without C oxidoreductase motif 35 to 55 Therapeutic administration of immunogenic peptides of MHCII T cell epitopes has an effect on Experimental Autoimmune Encephalomyelitis (EAE) development in mice.
Groups of mice and administration
A total of 96 female C57BL/6 mice (Taconic Biosciences, 10 weeks of age at day 0) were used for the study. Mice were acclimatized for 14 days prior to the first injection. At the beginning of the study, mice were assigned to groups in a balanced manner to achieve similar average body weights between groups. Table 3 below shows the treatments applied to each group.
TABLE 3 treatment protocol
Treatment 1 was administered once per day at a volume s.c. of 0.05 mL/site shown in table 3, with each mouse receiving injections at two sites for a total of 0.1 mL/mouse/dosing day. IMCY-0189 or MOG 35 to 55 The total dose of peptide was 30 μg per administration.
Treatment 2 was administered at a volume p.o.BID of 10mL/kg over the days indicated in Table 3. BG-12 was administered at 100 mg/kg.
All administrations were carried out at the same time (+/-1 hour) per day of administration. For the BID group, dosing intervals were no less than 10 hours and no more than 14 hours.
Preparation of Compounds
For saline treatment, 0.9% NaCl solution was prepared on each dosing day.
Treatment of 1-MOG 35 to 55 Peptide preparation:
freeze-dried mouse MOG having the sequence MEVGWYRSPFSRVVVVHLYRNGK (SEQ ID NO: 205) was solubilized immediately before use 35 to 55 A peptide comprising mouse Myelin Oligodendrocyte Glycoprotein (MOG) 35 to 55 ) MHCII T cell epitope YRSPFSRVV (SEQ ID NO: 1)69). MOG to be lyophilized 35 to 55 Peptides were thawed for 10 min at room temperature, resuspended in 50mM NaCl 0.9%pH 5.4 sodium acetate buffer, and incubated for 10 min at room temperature. The reconstituted MOG is then subjected to injection prior to injection 35 to 55 Peptides and immject TM Alum adjuvant. Note that positions 35 to 55 refer to the mature protein defined by SEQ ID NO. 208, i.e.the mature protein after cleavage of the signal peptide (AA 1 to 29). In the full-length amino acid sequence of MOG (SEQ ID NO: 184), the peptide will be at positions 64 to 84.
Treatment 1-IMCY-0189 peptide preparation:
IMCY-0189 has the sequence described in example 1. Lyophilized IMCY-0189 was thawed at room temperature for 10 minutes, resuspended in 50mM NaCl 0.9%pH 5.4 sodium acetate buffer and incubated at room temperature for 10 minutes. The reconstituted peptide was then combined with an Imject prior to injection TM Alum adjuvant.
Process 2
For treatment 2, the vehicle was 0.5% HPMC, 0.2% tween 20 and 50mM citrate buffer at pH 4. And BG-12 (Santa Cruz Biotechnology, sc-239774) was prepared at least every 2 weeks. At each preparation, the desired amount of BG-12 was weighed into a mortar and ground with a pestle. The loading agent is then added in small increments and mixed until the final volume is reached. The material was then vortexed and sonicated in a water bath until a uniform suspension was obtained. The prepared BG-12 was stored at 4℃with continuous stirring.
EAE induction, scoring and statistical analysis were performed as described in example 1.
Results and data interpretation
EAE scoring
EAE occurrence was assessed by comparing clinical EAE readout of all groups with the negative control (saline/vehicle) group. EAE score, AUC (area under the curve) and MMS (average max score) are given in fig. 7, 8 and 9.
For this model, mice of the saline/vehicle group (negative control) developed typical EAE. Two (2) mice in this group died from severe EAE.
Mice treated with BG-12 (saline/BG-12 group) showed delayed onset of disease and reduced endpoint scores, as well as statistically significant reductions in AUC and MMS compared to the negative control group. No mice in this group died.
Compared with the negative control group, the MOG is used 35 to 55 Peptide (MOG) 35 to 55 Vehicle group) or IMCY-0189 (IMCY-0189/vehicle group) also shows delayed onset and reduced endpoint scores, as well as statistically significantly reduced AUC and MMS. MOG (metal oxide gate) 35 to 55 No mice died in the vehicle group, whereas one (1) mice died in the IMCY-0189/vehicle group, but the mice' death was shown not to be due to EAE and was therefore excluded from analysis.
MOG was used compared to mice treated with peptide alone or BG-12 alone 35 to 55 All clinical readouts (onset of disease, endpoint score, AUC and MMS) were statistically significantly improved for both peptide and BG-12 or mice treated with both IMCY-0189 and BG-12. MOG (metal oxide gate) 35 to 55 Two (2) and one (1) mice died in the vehicle and IMCY-0189/vehicle groups, respectively, but the death of these mice was shown not to be due to EAE and was therefore excluded from analysis. IMCY-0189 and BG-12 treatments or MOG 35 to 55 And BG-12 treatment by two factor ANOVA analysis. The trend of synergy was assessed by a p-value of approximately 0.1 (0.1306 and 0.1574, respectively) for MMS data.
Example 3: with dimethyl fumarate (BG-12, TECFIDERA) TM ) Combined MOG comprising a sequence linked to a KCRC (SEQ ID NO: 65) or KCRPYC (SEQ ID NO: 84) oxidoreductase motif 35 to 55 Therapeutic administration of immunogenic peptides of MHCII T cell epitopes has an effect on Experimental Autoimmune Encephalomyelitis (EAE) development in mice.
Groups of mice and administration
A total of 128 female C57BL/6 mice (Taconic Biosciences, 9 weeks of age at day 0) were used for the study. Mice were acclimatized for 7 days prior to the first injection. At the beginning of the study, mice were assigned to groups in a balanced manner to achieve similar average body weights between groups. Table 4 below shows the treatments applied to each group.
TABLE 4 treatment protocol
Treatment 1 was administered once per day at a volume s.c. of 0.05 mL/site shown in table 4, with each mouse receiving injections at two sites for a total of 0.1 mL/mouse/dosing day. The total dose of IMCY-0189 (CXXC oxidoreductase motif), IMCY-0453 (CXC oxidoreductase motif) or IMCY-0455 (CXXXC oxidoreductase motif) peptide was 30 μg per administration.
Treatment 2 was administered at a volume p.o.BID of 10mL/kg over the days indicated in Table 4. BG-12 was administered at 100 mg/kg.
All administrations were carried out at the same time (+/-1 hour) per day of administration. For the BID group, dosing intervals were no less than 10 hours and no more than 14 hours.
Preparation of Compounds
For saline treatment, 0.9% NaCl solution was prepared on each dosing day.
Treatment 1-IMCY-0189 peptide preparation:
IMCY-0189 has the sequence described in example 1 and is prepared as described in example 2.
Treatment 1-IMCY-0453 and IMCY-0455 peptides preparation:
immediately prior to use, lyophilized immunogenic peptide IMCY-0453 having sequence KCRCGWYRSPFSRVVHLYR (SEQ ID NO: 266) comprising the oxidoreductase motif KCRC (SEQ ID NO: 65), linker GW, murine Myelin Oligodendrocyte Glycoprotein (MOG) 35 to 55 ) MHCII T cell epitope YRSPFSRVV (SEQ ID NO: 169) and flanking sequence HLYR (SEQ ID NO: 186) (Smart Bioscience). Lyophilized IMCY-0453 was thawed at room temperature for 10 minutes, resuspended in 50mM NaCl 0.9%pH 5.4 sodium acetate buffer, and incubated at room temperature for 10 minutes. The reconstituted peptide was then combined with an Imject prior to injection TM Alum adjuvant.
Immediately prior to use, lyophilized immunogenic peptide IMCY-0455 having sequence KCRPYCGWYRSPFSRVVHLYR (SEQ ID NO: 268) comprising the oxidoreductase motif KCRPYC (SEQ ID NO: 84), linker GW, murine Myelin Oligodendrocyte Glycoprotein (MOG) 35 to 55 ) MHCII T cell epitope YRSPFSRVV (SEQ ID NO: 169) and flanking sequence HLYR (SEQ ID NO: 186) (Smart Bioscience). Lyophilized IMCY-0455 was thawed at room temperature for 10 minutes, resuspended in 50mM NaCl 0.9%pH 5.4 sodium acetate buffer, and incubated at room temperature for 10 minutes. The reconstituted peptide was then combined with an Imject prior to injection TM Alum adjuvant.
Process 2 was prepared as described in example 2.
Serum neurofilament level determination
On day 28, blood was collected from all mice into gel clot activator tubes and allowed to clot at room temperature for about 30 minutes. The blood was then centrifuged at about 10000g for 5 minutes. The serum was transferred to Eppendorf tubes and stored at-80℃until shipment to Quantix TM . Use as described in example 1NF-light dominance kits quantify serum neurofilament light (NF-L) protein levels.
EAE induction, scoring and statistical analysis were performed as described in example 2.
Results and data interpretation
EAE scoring
EAE occurrence was assessed by comparing clinical EAE readout of all groups with the negative control (saline/vehicle) group. EAE score, AUC (area under the curve) and MMS (average max score) are given in figures 10, 11 and 12.
Mice of the saline/vehicle group (negative control) developed slightly milder EAE, but were still within the expected range of the model. The group was free of mice death.
Mice treated with BG-12 (saline/BG-12 group) showed delayed onset of disease and reduced endpoint scores, as well as statistically significant reductions in AUC and MMS compared to the negative control group. Mice treated with IMCY-0189 (IMCY-0189/vehicle group) showed a similar profile. No mice in both groups died.
All clinical readouts (onset, endpoint score, AUC and MMS) were statistically significantly improved in mice treated with IMCY-0453 or IMCY-0455 compared to the negative control group. Co-administration of BG-12 with IMCY-0453 or IMCY-0455 was shown to be most effective, and no mice in these groups developed EAE. One (1) mice of IMCY-453/vehicle group, one (1) mice of IMCY-455/vehicle group and one (1) mice of IMCY-453/BG-12 group died, but the death of these mice showed not to be due to EAE and thus was excluded from the analysis.
Serum neurofilament level
Axonal injury was also assessed by comparing NF-L levels in all groups with negative control (saline/vehicle) groups. The data are given in figure 13.
NF-L levels of saline/vehicle group (negative control) were consistent with clinical findings and as expected for this model.
Mice treated with IMCY-0189, IMCY-0453 and IMCY-0455 or with BG-12 (saline/BG-12 group, positive control) showed statistically significantly reduced NF-L levels compared to the negative control group.
NF-L levels were almost eliminated in mice treated with both IMCY-0189, IMCY-0453, IMCY-0455 and BG-12, as compared to the negative control group.
Example 4: with dimethyl fumarate (BG-12, TECFIDERA) TM ) Combined human MOG comprising a KHCPYC oxidoreductase motif linked thereto 201 to 212 Therapeutic administration of immunogenic peptides of MHCII T cell epitopes has an effect on Experimental Autoimmune Encephalomyelitis (EAE) development in mice.
Groups of mice and administration
A total of 96 female C57BL/6 mice (Taconic Biosciences, 9 weeks of age at day 0) were used for the study. Mice were acclimatized for 7 days prior to the first injection. At the beginning of the study, mice were assigned to groups in a balanced manner to achieve similar average body weights between groups. Table 5 below shows the treatments applied to each group.
TABLE 5 treatment protocol
Treatment 1 was administered once per day at a volume s.c. of 0.05 mL/site shown in table 5, with each mouse receiving injections at two sites for a total of 0.1 mL/mouse/dosing day. The total dose of IMCY-0189 or P4 peptide was 30 μg per administration.
Treatment 2 was administered at a volume p.o.BID of 10mL/kg over the days shown in Table 5. BG-12 was administered at 100 mg/kg.
All administrations were carried out at the same time (+/-1 hour) per day of administration. For the BID group, dosing intervals were no less than 10 hours and no more than 14 hours.
Preparation of Compounds
For saline treatment, 0.9% NaCl solution was prepared on each dosing day.
Treatment 1-IMCY-0189 peptide preparation:
IMCY-0189 has the sequence described in example 1 and is prepared as described in example 2.
Treatment 1-P4 peptide preparation:
immediately prior to use, lyophilized immunogenic peptide P4 having sequence KHCPYCVRYFLRVPSWKITLFKK (SEQ ID NO: 176) comprising the oxidoreductase motif KHCPYC (SEQ ID NO: 77), linker VRY, human Myelin Oligodendrocyte Glycoprotein (MOG) 201 to 212 ) MHCII T cell epitope FLRVPSWKI (SEQ ID NO: 165) and flanking sequence TLEKK (SEQ ID NO: 267) (Smart Bioscience). Lyophilized P4 was thawed at room temperature for 10 minutes, resuspended in 50mM NaCl 0.9%pH 5.4 sodium acetate buffer, and incubated at room temperature for 10 minutes. The reconstituted peptide was then combined with an Imject prior to injection TM Alum adjuvant.
Process 2 was prepared as described in example 2.
Serum neurofilament levels were quantified as described in example 3.
EAE induction, scoring and statistical analysis were performed as described in example 2.
Results and data interpretation
EAE scoring
EAE occurrence was assessed by comparing clinical EAE readout of all groups with the negative control (saline/vehicle) group. EAE score, AUC (area under the curve) and MMS (average max score) are given in figures 14, 15 and 16.
Mice of the saline/vehicle group (negative control) developed slightly milder EAE, but were still within the expected range of the model. The group was free of mice death.
Mice treated with BG-12 (saline/BG-12 group), IMCY-0189 (IMCY-0189/vehicle group) and P4 (P4/vehicle group) showed delayed onset of disease and reduced endpoint scores, as well as statistically significantly reduced AUC and MMS, compared to the negative control group. None of the mice in these three groups died.
All clinical readouts (onset, endpoint score, AUC and MMS) were statistically significantly improved in mice treated with either both IMCY-0189 and BG-12 or with both P4 and BG-12 compared to negative control groups, and also especially compared to mice treated with either peptide alone or BG-12 alone. No mice in both groups died. Interaction between either IMCY-0189 and BG-12 treatments or P4 and BG-12 treatments was analyzed by two-way ANOVA. The trend of synergy was assessed by a p-value of approximately 0.2 (0.2340 and 0.2392, respectively) for AUC data.
Serum neurofilament level
Axonal injury was also assessed by comparing NF-L levels in all groups with negative control (saline/vehicle) groups. The data are given in fig. 17.
NF-L levels of saline/vehicle group (negative control) were consistent with clinical findings and as expected for this model.
Mice treated with IMCY-0189, P4 or with BG-12 (saline/BG-12 group, positive control) showed statistically significantly reduced NF-L levels compared to the negative control group.
Mice treated with both IMCY-0189 and BG-12 or with both P4 and BG-12 showed highly statistically significantly reduced NF-L levels compared to the negative control group.
Standard definition and abbreviation
Area under AUC curve
BG-12 dimethyl fumarate, DMF
BID twice daily
Degree centigrade
CFA complete Freund's adjuvant
EAE experimental autoimmune encephalomyelitis
H & E hematoxylin and eosin
HPMC hydroxypropyl methylcellulose
MBP myelin basic protein
MMS average maximum score
MOG myelin oligodendrocyte glycoprotein
Na sodium
NF-L neurofilament light protein
p.o. oral administration
PTX pertussis toxin
s.c. subcutaneous
Standard deviation of SD
Standard error of SEM mean
Sequence listing
<110> IMCYSE SA
<120> combination therapy for fumarate-related diseases
<130> IMCY-028-PCT
<150> 20173209.6
<151> 2020-05-06
<150> 20179019.3
<151> 2020-06-09
<160> 255
<170> PatentIn version 3.5
<210> 1
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> peptide
<222> (1)..(1)
<223> C, S or T
<220>
<221> misc_feature
<222> (2)..(3)
<223> Xaa can be any naturally occurring amino acid
<400> 1
Xaa Xaa Xaa Cys
1
<210> 2
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> peptide
<222> (1)..(1)
<223> C, S or T
<220>
<221> misc_feature
<222> (2)..(4)
<223> Xaa can be any naturally occurring amino acid
<400> 2
Xaa Xaa Xaa Xaa Cys
1 5
<210> 3
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> peptide
<222> (1)..(1)
<223> C, S or T
<220>
<221> misc_feature
<222> (2)..(5)
<223> Xaa can be any naturally occurring amino acid
<400> 3
Xaa Xaa Xaa Xaa Xaa Cys
1 5
<210> 4
<211> 7
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> peptide
<222> (1)..(1)
<223> C, S or T
<220>
<221> misc_feature
<222> (2)..(6)
<223> Xaa can be any naturally occurring amino acid
<400> 4
Xaa Xaa Xaa Xaa Xaa Xaa Cys
1 5
<210> 5
<211> 8
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> peptide
<222> (1)..(1)
<223> C, S or T
<220>
<221> misc_feature
<222> (2)..(7)
<223> Xaa can be any naturally occurring amino acid
<400> 5
Xaa Xaa Xaa Xaa Xaa Xaa Xaa Cys
1 5
<210> 6
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(1)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (2)..(2)
<223> C, S or T
<220>
<221> misc_feature
<222> (3)..(3)
<223> Xaa can be any naturally occurring amino acid
<400> 6
Xaa Xaa Xaa Cys
1
<210> 7
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(1)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (2)..(2)
<223> C, S or T
<220>
<221> misc_feature
<222> (3)..(4)
<223> Xaa can be any naturally occurring amino acid
<400> 7
Xaa Xaa Xaa Xaa Cys
1 5
<210> 8
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(1)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (2)..(2)
<223> C, S or T
<220>
<221> misc_feature
<222> (3)..(5)
<223> Xaa can be any naturally occurring amino acid
<400> 8
Xaa Xaa Xaa Xaa Xaa Cys
1 5
<210> 9
<211> 7
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(1)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (2)..(2)
<223> C, S or T
<220>
<221> misc_feature
<222> (3)..(6)
<223> Xaa can be any naturally occurring amino acid
<400> 9
Xaa Xaa Xaa Xaa Xaa Xaa Cys
1 5
<210> 10
<211> 8
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(1)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (2)..(2)
<223> C, S or T
<220>
<221> misc_feature
<222> (3)..(7)
<223> Xaa can be any naturally occurring amino acid
<400> 10
Xaa Xaa Xaa Xaa Xaa Xaa Xaa Cys
1 5
<210> 11
<211> 9
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(1)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (2)..(2)
<223> C, S or T
<220>
<221> misc_feature
<222> (3)..(8)
<223> Xaa can be any naturally occurring amino acid
<400> 11
Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Cys
1 5
<210> 12
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(2)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (3)..(3)
<223> C, S or T
<400> 12
Xaa Xaa Xaa Cys
1
<210> 13
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(2)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (3)..(3)
<223> C, S or T
<220>
<221> misc_feature
<222> (4)..(4)
<223> Xaa can be any naturally occurring amino acid
<400> 13
Xaa Xaa Xaa Xaa Cys
1 5
<210> 14
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(2)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (3)..(3)
<223> C, S or T
<220>
<221> misc_feature
<222> (4)..(5)
<223> Xaa can be any naturally occurring amino acid
<400> 14
Xaa Xaa Xaa Xaa Xaa Cys
1 5
<210> 15
<211> 7
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(2)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (3)..(3)
<223> C, S or T
<220>
<221> misc_feature
<222> (4)..(6)
<223> Xaa can be any naturally occurring amino acid
<400> 15
Xaa Xaa Xaa Xaa Xaa Xaa Cys
1 5
<210> 16
<211> 8
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(2)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (3)..(3)
<223> C, S or T
<220>
<221> misc_feature
<222> (4)..(7)
<223> Xaa can be any naturally occurring amino acid
<400> 16
Xaa Xaa Xaa Xaa Xaa Xaa Xaa Cys
1 5
<210> 17
<211> 9
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(2)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (3)..(3)
<223> C, S or T
<220>
<221> misc_feature
<222> (4)..(8)
<223> Xaa can be any naturally occurring amino acid
<400> 17
Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Cys
1 5
<210> 18
<211> 10
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(2)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (3)..(3)
<223> C, S or T
<220>
<221> misc_feature
<222> (4)..(9)
<223> Xaa can be any naturally occurring amino acid
<400> 18
Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Cys
1 5 10
<210> 19
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(3)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (4)..(4)
<223> C, S or T
<400> 19
Xaa Xaa Xaa Xaa Cys
1 5
<210> 20
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(3)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (4)..(4)
<223> C, S or T
<220>
<221> misc_feature
<222> (5)..(5)
<223> Xaa can be any naturally occurring amino acid
<400> 20
Xaa Xaa Xaa Xaa Xaa Cys
1 5
<210> 21
<211> 7
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(3)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (4)..(4)
<223> C, S or T
<220>
<221> misc_feature
<222> (5)..(6)
<223> Xaa can be any naturally occurring amino acid
<400> 21
Xaa Xaa Xaa Xaa Xaa Xaa Cys
1 5
<210> 22
<211> 8
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(3)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (4)..(4)
<223> C, S or T
<220>
<221> misc_feature
<222> (5)..(7)
<223> Xaa can be any naturally occurring amino acid
<400> 22
Xaa Xaa Xaa Xaa Xaa Xaa Xaa Cys
1 5
<210> 23
<211> 9
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(3)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (4)..(4)
<223> C, S or T
<220>
<221> misc_feature
<222> (5)..(8)
<223> Xaa can be any naturally occurring amino acid
<400> 23
Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Cys
1 5
<210> 24
<211> 10
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(3)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (4)..(4)
<223> C, S or T
<220>
<221> misc_feature
<222> (5)..(9)
<223> Xaa can be any naturally occurring amino acid
<400> 24
Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Cys
1 5 10
<210> 25
<211> 11
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(3)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (4)..(4)
<223> C, S or T
<220>
<221> misc_feature
<222> (5)..(10)
<223> Xaa can be any naturally occurring amino acid
<400> 25
Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa Cys
1 5 10
<210> 26
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (2)..(3)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (4)..(4)
<223> C, S or T
<400> 26
Cys Xaa Xaa Xaa
1
<210> 27
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (2)..(4)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (5)..(5)
<223> C, S or T
<400> 27
Cys Xaa Xaa Xaa Xaa
1 5
<210> 28
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (2)..(5)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (6)..(6)
<223> C, S or T
<400> 28
Cys Xaa Xaa Xaa Xaa Xaa
1 5
<210> 29
<211> 7
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (2)..(6)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (7)..(7)
<223> C, S or T
<400> 29
Cys Xaa Xaa Xaa Xaa Xaa Xaa
1 5
<210> 30
<211> 8
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (2)..(7)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (8)..(8)
<223> C, S or T
<400> 30
Cys Xaa Xaa Xaa Xaa Xaa Xaa Xaa
1 5
<210> 31
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(1)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> misc_feature
<222> (3)..(3)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (4)..(4)
<223> C, S or T
<400> 31
Xaa Cys Xaa Xaa
1
<210> 32
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(1)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> misc_feature
<222> (3)..(4)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (5)..(5)
<223> C, S or T
<400> 32
Xaa Cys Xaa Xaa Xaa
1 5
<210> 33
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(1)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> misc_feature
<222> (3)..(5)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (6)..(6)
<223> C, S or T
<400> 33
Xaa Cys Xaa Xaa Xaa Xaa
1 5
<210> 34
<211> 7
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(1)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> misc_feature
<222> (3)..(6)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (7)..(7)
<223> C, S or T
<400> 34
Xaa Cys Xaa Xaa Xaa Xaa Xaa
1 5
<210> 35
<211> 8
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(1)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> misc_feature
<222> (3)..(7)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (8)..(8)
<223> C, S or T
<400> 35
Xaa Cys Xaa Xaa Xaa Xaa Xaa Xaa
1 5
<210> 36
<211> 9
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(1)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> misc_feature
<222> (3)..(8)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (9)..(9)
<223> C, S or T
<400> 36
Xaa Cys Xaa Xaa Xaa Xaa Xaa Xaa Xaa
1 5
<210> 37
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(2)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (4)..(4)
<223> C, S or T
<400> 37
Xaa Xaa Cys Xaa
1
<210> 38
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(2)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> misc_feature
<222> (4)..(4)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (5)..(5)
<223> C, S or T
<400> 38
Xaa Xaa Cys Xaa Xaa
1 5
<210> 39
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(2)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> misc_feature
<222> (4)..(5)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (6)..(6)
<223> C, S or T
<400> 39
Xaa Xaa Cys Xaa Xaa Xaa
1 5
<210> 40
<211> 7
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(2)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> misc_feature
<222> (4)..(6)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (7)..(7)
<223> C, S or T
<400> 40
Xaa Xaa Cys Xaa Xaa Xaa Xaa
1 5
<210> 41
<211> 8
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(2)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> misc_feature
<222> (4)..(7)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (8)..(8)
<223> C, S or T
<400> 41
Xaa Xaa Cys Xaa Xaa Xaa Xaa Xaa
1 5
<210> 42
<211> 9
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(2)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> misc_feature
<222> (4)..(8)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (9)..(9)
<223> C, S or T
<400> 42
Xaa Xaa Cys Xaa Xaa Xaa Xaa Xaa Xaa
1 5
<210> 43
<211> 10
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(2)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> misc_feature
<222> (4)..(9)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (10)..(10)
<223> C, S or T
<400> 43
Xaa Xaa Cys Xaa Xaa Xaa Xaa Xaa Xaa Xaa
1 5 10
<210> 44
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(3)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (5)..(5)
<223> C, S or T
<400> 44
Xaa Xaa Xaa Cys Xaa
1 5
<210> 45
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(3)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> misc_feature
<222> (5)..(5)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (6)..(6)
<223> C, S or T
<400> 45
Xaa Xaa Xaa Cys Xaa Xaa
1 5
<210> 46
<211> 7
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(3)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> misc_feature
<222> (5)..(6)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (7)..(7)
<223> C, S or T
<400> 46
Xaa Xaa Xaa Cys Xaa Xaa Xaa
1 5
<210> 47
<211> 8
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(3)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> misc_feature
<222> (5)..(7)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (8)..(8)
<223> C, S or T
<400> 47
Xaa Xaa Xaa Cys Xaa Xaa Xaa Xaa
1 5
<210> 48
<211> 9
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(3)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> misc_feature
<222> (5)..(8)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (9)..(9)
<223> C, S or T
<400> 48
Xaa Xaa Xaa Cys Xaa Xaa Xaa Xaa Xaa
1 5
<210> 49
<211> 10
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(3)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> misc_feature
<222> (5)..(9)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (10)..(10)
<223> C, S or T
<400> 49
Xaa Xaa Xaa Cys Xaa Xaa Xaa Xaa Xaa Xaa
1 5 10
<210> 50
<211> 11
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(3)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> misc_feature
<222> (5)..(10)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (11)..(11)
<223> C, S or T
<400> 50
Xaa Xaa Xaa Cys Xaa Xaa Xaa Xaa Xaa Xaa Xaa
1 5 10
<210> 51
<211> 7
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> peptide
<222> (1)..(1)
<223> F, W, H or Y
<220>
<221> misc_feature
<222> (2)..(3)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (4)..(4)
<223> I, L, M or V
<220>
<221> misc_feature
<222> (5)..(6)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (7)..(7)
<223> F, W, T, H or Y
<400> 51
Xaa Xaa Xaa Xaa Xaa Xaa Xaa
1 5
<210> 52
<211> 7
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> peptide
<222> (1)..(1)
<223> F or W
<220>
<221> misc_feature
<222> (2)..(3)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (4)..(4)
<223> I, L or M
<220>
<221> misc_feature
<222> (5)..(6)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (7)..(7)
<223> F or W
<400> 52
Xaa Xaa Xaa Xaa Xaa Xaa Xaa
1 5
<210> 53
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 53
Lys Lys Cys Cys
1
<210> 54
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 54
Arg Arg Cys Cys
1
<210> 55
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 55
Arg Lys Cys Cys
1
<210> 56
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 56
Lys Arg Cys Cys
1
<210> 57
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (3)..(3)
<223> Xaa can be any naturally occurring amino acid
<400> 57
Lys Cys Xaa Cys
1
<210> 58
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (4)..(4)
<223> Xaa can be any naturally occurring amino acid
<400> 58
Lys Lys Cys Xaa Cys
1 5
<210> 59
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (3)..(3)
<223> Xaa can be any naturally occurring amino acid
<400> 59
Arg Cys Xaa Cys
1
<210> 60
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (4)..(4)
<223> Xaa can be any naturally occurring amino acid
<400> 60
Arg Arg Cys Xaa Cys
1 5
<210> 61
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (4)..(4)
<223> Xaa can be any naturally occurring amino acid
<400> 61
Arg Lys Cys Xaa Cys
1 5
<210> 62
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (4)..(4)
<223> Xaa can be any naturally occurring amino acid
<400> 62
Lys Arg Cys Xaa Cys
1 5
<210> 63
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 63
Lys Cys Lys Cys
1
<210> 64
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 64
Lys Lys Cys Lys Cys
1 5
<210> 65
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 65
Lys Cys Arg Cys
1
<210> 66
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 66
Lys Lys Cys Arg Cys
1 5
<210> 67
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 67
Arg Cys Arg Cys
1
<210> 68
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 68
Arg Arg Cys Arg Cys
1 5
<210> 69
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 69
Arg Lys Cys Lys Cys
1 5
<210> 70
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 70
Lys Arg Cys Lys Cys
1 5
<210> 71
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 71
His Cys Pro Tyr Cys
1 5
<210> 72
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 72
Lys Cys Pro Tyr Cys
1 5
<210> 73
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 73
Arg Cys Pro Tyr Cys
1 5
<210> 74
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 74
His Cys Gly His Cys
1 5
<210> 75
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 75
Lys Cys Gly His Cys
1 5
<210> 76
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 76
Arg Cys Gly His Cys
1 5
<210> 77
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 77
Lys His Cys Pro Tyr Cys
1 5
<210> 78
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 78
Lys Lys Cys Pro Tyr Cys
1 5
<210> 79
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 79
Lys Arg Cys Pro Tyr Cys
1 5
<210> 80
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 80
Lys His Cys Gly His Cys
1 5
<210> 81
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 81
Lys Lys Cys Gly His Cys
1 5
<210> 82
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 82
Lys Arg Cys Gly His Cys
1 5
<210> 83
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 83
Cys Arg Pro Tyr Cys
1 5
<210> 84
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 84
Lys Cys Arg Pro Tyr Cys
1 5
<210> 85
<211> 7
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 85
Lys His Cys Arg Pro Tyr Cys
1 5
<210> 86
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 86
Arg Cys Arg Pro Tyr Cys
1 5
<210> 87
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 87
His Cys Arg Pro Tyr Cys
1 5
<210> 88
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 88
Cys Pro Arg Tyr Cys
1 5
<210> 89
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 89
Lys Cys Pro Arg Tyr Cys
1 5
<210> 90
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 90
Arg Cys Pro Arg Tyr Cys
1 5
<210> 91
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 91
His Cys Pro Arg Tyr Cys
1 5
<210> 92
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 92
Cys Pro Tyr Arg Cys
1 5
<210> 93
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 93
Lys Cys Pro Tyr Arg Cys
1 5
<210> 94
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 94
Arg Cys Pro Tyr Arg Cys
1 5
<210> 95
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 95
His Cys Pro Tyr Arg Cys
1 5
<210> 96
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 96
Cys Lys Pro Tyr Cys
1 5
<210> 97
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 97
Lys Cys Lys Pro Tyr Cys
1 5
<210> 98
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 98
Arg Cys Lys Pro Tyr Cys
1 5
<210> 99
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 99
His Cys Lys Pro Tyr Cys
1 5
<210> 100
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 100
Cys Pro Lys Tyr Cys
1 5
<210> 101
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 101
Lys Cys Pro Lys Tyr Cys
1 5
<210> 102
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 102
Arg Cys Pro Lys Tyr Cys
1 5
<210> 103
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 103
His Cys Pro Lys Tyr Cys
1 5
<210> 104
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 104
Cys Pro Tyr Lys Cys
1 5
<210> 105
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 105
Lys Cys Pro Tyr Lys Cys
1 5
<210> 106
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 106
Arg Cys Pro Tyr Lys Cys
1 5
<210> 107
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 107
His Cys Pro Tyr Lys Cys
1 5
<210> 108
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 108
Leu Ala Val Leu
1
<210> 109
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 109
Thr Val Gln Ala
1
<210> 110
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 110
Gly Ala Val His
1
<210> 111
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(4)
<223> Xaa can be any naturally occurring amino acid
<400> 111
Xaa Xaa Xaa Xaa
1
<210> 112
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(1)
<223> Xaa can be any naturally occurring amino acid
<400> 112
Xaa Ala Val Leu
1
<210> 113
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (2)..(2)
<223> Xaa can be any naturally occurring amino acid
<400> 113
Leu Xaa Val Leu
1
<210> 114
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (3)..(3)
<223> Xaa can be any naturally occurring amino acid
<400> 114
Leu Ala Xaa Leu
1
<210> 115
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (4)..(4)
<223> Xaa can be any naturally occurring amino acid
<400> 115
Leu Ala Val Xaa
1
<210> 116
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(1)
<223> Xaa can be any naturally occurring amino acid
<400> 116
Xaa Val Gln Ala
1
<210> 117
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (2)..(2)
<223> Xaa can be any naturally occurring amino acid
<400> 117
Thr Xaa Gln Ala
1
<210> 118
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (4)..(4)
<223> Xaa can be any naturally occurring amino acid
<400> 118
Thr Val Gln Xaa
1
<210> 119
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(1)
<223> Xaa can be any naturally occurring amino acid
<400> 119
Xaa Ala Val His
1
<210> 120
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (2)..(2)
<223> Xaa can be any naturally occurring amino acid
<400> 120
Gly Xaa Val His
1
<210> 121
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (3)..(3)
<223> Xaa can be any naturally occurring amino acid
<400> 121
Gly Ala Xaa His
1
<210> 122
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (4)..(4)
<223> Xaa can be any naturally occurring amino acid
<400> 122
Gly Ala Val Xaa
1
<210> 123
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 123
Pro Ala Phe Pro Leu
1 5
<210> 124
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 124
Asp Gln Gly Gly Glu
1 5
<210> 125
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(5)
<223> Xaa can be any naturally occurring amino acid
<400> 125
Xaa Xaa Xaa Xaa Xaa
1 5
<210> 126
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(1)
<223> Xaa can be any naturally occurring amino acid
<400> 126
Xaa Ala Phe Pro Leu
1 5
<210> 127
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (2)..(2)
<223> Xaa can be any naturally occurring amino acid
<400> 127
Pro Xaa Phe Pro Leu
1 5
<210> 128
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (3)..(3)
<223> Xaa can be any naturally occurring amino acid
<400> 128
Pro Ala Xaa Pro Leu
1 5
<210> 129
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (4)..(4)
<223> Xaa can be any naturally occurring amino acid
<400> 129
Pro Ala Phe Xaa Leu
1 5
<210> 130
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (5)..(5)
<223> Xaa can be any naturally occurring amino acid
<400> 130
Pro Ala Phe Pro Xaa
1 5
<210> 131
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(1)
<223> Xaa can be any naturally occurring amino acid
<400> 131
Xaa Gln Gly Gly Glu
1 5
<210> 132
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (2)..(2)
<223> Xaa can be any naturally occurring amino acid
<400> 132
Asp Xaa Gly Gly Glu
1 5
<210> 133
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (3)..(3)
<223> Xaa can be any naturally occurring amino acid
<400> 133
Asp Gln Xaa Gly Glu
1 5
<210> 134
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (4)..(4)
<223> Xaa can be any naturally occurring amino acid
<400> 134
Asp Gln Gly Xaa Glu
1 5
<210> 135
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (5)..(5)
<223> Xaa can be any naturally occurring amino acid
<400> 135
Asp Gln Gly Gly Xaa
1 5
<210> 136
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 136
Asp Ile Ala Asp Lys Tyr
1 5
<210> 137
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(6)
<223> Xaa can be any naturally occurring amino acid
<400> 137
Xaa Xaa Xaa Xaa Xaa Xaa
1 5
<210> 138
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (1)..(1)
<223> Xaa can be any naturally occurring amino acid
<400> 138
Xaa Ile Ala Asp Lys Tyr
1 5
<210> 139
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (2)..(2)
<223> Xaa can be any naturally occurring amino acid
<400> 139
Asp Xaa Ala Asp Lys Tyr
1 5
<210> 140
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (3)..(3)
<223> Xaa can be any naturally occurring amino acid
<400> 140
Asp Ile Xaa Asp Lys Tyr
1 5
<210> 141
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (4)..(4)
<223> Xaa can be any naturally occurring amino acid
<400> 141
Asp Ile Ala Xaa Lys Tyr
1 5
<210> 142
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (5)..(5)
<223> Xaa can be any naturally occurring amino acid
<400> 142
Asp Ile Ala Asp Xaa Tyr
1 5
<210> 143
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (6)..(6)
<223> Xaa can be any naturally occurring amino acid
<400> 143
Asp Ile Ala Asp Lys Xaa
1 5
<210> 144
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> peptide
<222> (1)..(1)
<223> acetylation, methylation, ethylation, or propionylation C, S or T
<220>
<221> misc_feature
<222> (2)..(3)
<223> Xaa can be any naturally occurring amino acid
<400> 144
Xaa Xaa Xaa Cys
1
<210> 145
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> peptide
<222> (1)..(1)
<223> acetylation, methylation, ethylation, or propionylation C, S or T
<220>
<221> misc_feature
<222> (2)..(4)
<223> Xaa can be any naturally occurring amino acid
<400> 145
Xaa Xaa Xaa Xaa Cys
1 5
<210> 146
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> peptide
<222> (1)..(1)
<223> acetylation, methylation, ethylation, or propionylation C, S or T
<220>
<221> misc_feature
<222> (2)..(5)
<223> Xaa can be any naturally occurring amino acid
<400> 146
Xaa Xaa Xaa Xaa Xaa Cys
1 5
<210> 147
<211> 7
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> peptide
<222> (1)..(1)
<223> acetylation, methylation, ethylation, or propionylation C, S or T
<220>
<221> misc_feature
<222> (2)..(6)
<223> Xaa can be any naturally occurring amino acid
<400> 147
Xaa Xaa Xaa Xaa Xaa Xaa Cys
1 5
<210> 148
<211> 8
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> peptide
<222> (1)..(1)
<223> acetylation, methylation, ethylation, or propionylation C, S or T
<220>
<221> misc_feature
<222> (2)..(7)
<223> Xaa can be any naturally occurring amino acid
<400> 148
Xaa Xaa Xaa Xaa Xaa Xaa Xaa Cys
1 5
<210> 149
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (2)..(3)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (4)..(4)
<223> acetylation, methylation, ethylation, or propionylation C, S or T
<400> 149
Cys Xaa Xaa Xaa
1
<210> 150
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (2)..(4)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (5)..(5)
<223> acetylation, methylation, ethylation, or propionylation C, S or T
<400> 150
Cys Xaa Xaa Xaa Xaa
1 5
<210> 151
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (2)..(5)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (6)..(6)
<223> acetylation, methylation, ethylation, or propionylation C, S or T
<400> 151
Cys Xaa Xaa Xaa Xaa Xaa
1 5
<210> 152
<211> 7
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (2)..(6)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (7)..(7)
<223> acetylation, methylation, ethylation, or propionylation C, S or T
<400> 152
Cys Xaa Xaa Xaa Xaa Xaa Xaa
1 5
<210> 153
<211> 8
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (2)..(7)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (8)..(8)
<223> acetylation, methylation, ethylation, or propionylation C, S or T
<400> 153
Cys Xaa Xaa Xaa Xaa Xaa Xaa Xaa
1 5
<210> 154
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> peptide
<222> (1)..(1)
<223> C, S or T
<220>
<221> misc_feature
<222> (2)..(3)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (4)..(4)
<223> acetylated, methylated, ethylated, or propionylated C
<400> 154
Xaa Xaa Xaa Xaa
1
<210> 155
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> peptide
<222> (1)..(1)
<223> C, S or T
<220>
<221> misc_feature
<222> (2)..(4)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (5)..(5)
<223> acetylated, methylated, ethylated, or propionylated C
<400> 155
Xaa Xaa Xaa Xaa Xaa
1 5
<210> 156
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> peptide
<222> (1)..(1)
<223> C, S or T
<220>
<221> misc_feature
<222> (2)..(5)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (6)..(6)
<223> acetylated, methylated, ethylated, or propionylated C
<400> 156
Xaa Xaa Xaa Xaa Xaa Xaa
1 5
<210> 157
<211> 7
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> peptide
<222> (1)..(1)
<223> C, S or T
<220>
<221> misc_feature
<222> (2)..(6)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (7)..(7)
<223> acetylated, methylated, ethylated, or propionylated C
<400> 157
Xaa Xaa Xaa Xaa Xaa Xaa Xaa
1 5
<210> 158
<211> 8
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> peptide
<222> (1)..(1)
<223> C, S or T
<220>
<221> misc_feature
<222> (2)..(7)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (8)..(8)
<223> acetylated, methylated, ethylated, or propionylated C
<400> 158
Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa
1 5
<210> 159
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> peptide
<222> (1)..(1)
<223> acetylated, methylated, ethylated, or propionylated C
<220>
<221> misc_feature
<222> (2)..(3)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (4)..(4)
<223> C, S or T
<400> 159
Xaa Xaa Xaa Xaa
1
<210> 160
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> peptide
<222> (1)..(1)
<223> acetylated, methylated, ethylated, or propionylated C
<220>
<221> misc_feature
<222> (2)..(4)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (5)..(5)
<223> C, S or T
<400> 160
Xaa Xaa Xaa Xaa Xaa
1 5
<210> 161
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> peptide
<222> (1)..(1)
<223> acetylated, methylated, ethylated, or propionylated C
<220>
<221> misc_feature
<222> (2)..(5)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (6)..(6)
<223> C, S or T
<400> 161
Xaa Xaa Xaa Xaa Xaa Xaa
1 5
<210> 162
<211> 7
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> peptide
<222> (1)..(1)
<223> acetylated, methylated, ethylated, or propionylated C
<220>
<221> misc_feature
<222> (2)..(6)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (7)..(7)
<223> C, S or T
<400> 162
Xaa Xaa Xaa Xaa Xaa Xaa Xaa
1 5
<210> 163
<211> 8
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> peptide
<222> (1)..(1)
<223> acetylated, methylated, ethylated, or propionylated C
<220>
<221> misc_feature
<222> (2)..(7)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (8)..(8)
<223> C, S or T
<400> 163
Xaa Xaa Xaa Xaa Xaa Xaa Xaa Xaa
1 5
<210> 164
<211> 9
<212> PRT
<213> Homo sapiens (Homo sapiens)
<400> 164
Phe Leu Arg Val Pro Cys Trp Lys Ile
1 5
<210> 165
<211> 9
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 165
Phe Leu Arg Val Pro Ser Trp Lys Ile
1 5
<210> 166
<211> 12
<212> PRT
<213> Chile person
<400> 166
Phe Leu Arg Val Pro Cys Trp Lys Ile Thr Leu Phe
1 5 10
<210> 167
<211> 12
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 167
Phe Leu Arg Val Pro Ser Trp Lys Ile Thr Leu Phe
1 5 10
<210> 168
<211> 9
<212> PRT
<213> Chile person
<400> 168
Tyr Arg Pro Pro Phe Ser Arg Val Val
1 5
<210> 169
<211> 9
<212> PRT
<213> mice (Mus musculus)
<400> 169
Tyr Arg Ser Pro Phe Ser Arg Val Val
1 5
<210> 170
<211> 9
<212> PRT
<213> Chile person
<400> 170
Val Val His Leu Tyr Arg Asn Gly Lys
1 5
<210> 171
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 171
Arg Cys Lys Cys
1
<210> 172
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 172
Cys Pro Tyr Cys
1
<210> 173
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 173
Cys Pro Arg Tyr Cys
1 5
<210> 174
<211> 20
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 174
His Cys Pro Tyr Cys Val Arg Tyr Phe Leu Arg Val Pro Ser Trp Lys
1 5 10 15
Ile Thr Leu Phe
20
<210> 175
<211> 20
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 175
His Cys Pro Tyr Cys Val Arg Tyr Phe Leu Arg Val Pro Cys Trp Lys
1 5 10 15
Ile Thr Leu Phe
20
<210> 176
<211> 23
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 176
Lys His Cys Pro Tyr Cys Val Arg Tyr Phe Leu Arg Val Pro Ser Trp
1 5 10 15
Lys Ile Thr Leu Phe Lys Lys
20
<210> 177
<211> 23
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 177
Lys His Cys Pro Tyr Cys Val Arg Tyr Phe Leu Arg Val Pro Cys Trp
1 5 10 15
Lys Ile Thr Leu Phe Lys Lys
20
<210> 178
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> peptide
<222> (1)..(1)
<223> basic amino acid
<220>
<221> peptide
<222> (2)..(2)
<223> C, S or T
<220>
<221> misc_feature
<222> (3)..(3)
<223> Xaa can be any naturally occurring amino acid
<400> 178
Xaa Xaa Xaa Cys
1
<210> 179
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> peptide
<222> (1)..(1)
<223> basic amino acid
<220>
<221> misc_feature
<222> (3)..(3)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (4)..(4)
<223> C, S or T
<400> 179
Xaa Cys Xaa Xaa
1
<210> 180
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> peptide
<222> (1)..(1)
<223> basic amino acid
<220>
<221> peptide
<222> (2)..(2)
<223> C, S or T
<220>
<221> misc_feature
<222> (3)..(4)
<223> Xaa can be any naturally occurring amino acid
<400> 180
Xaa Xaa Xaa Xaa Cys
1 5
<210> 181
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> peptide
<222> (1)..(1)
<223> basic amino acid
<220>
<221> misc_feature
<222> (3)..(4)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (5)..(5)
<223> C, S or T
<400> 181
Xaa Cys Xaa Xaa Xaa
1 5
<210> 182
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> peptide
<222> (1)..(1)
<223> basic amino acid
<220>
<221> peptide
<222> (2)..(2)
<223> C, S or T
<220>
<221> misc_feature
<222> (3)..(5)
<223> Xaa can be any naturally occurring amino acid
<400> 182
Xaa Xaa Xaa Xaa Xaa Cys
1 5
<210> 183
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> peptide
<222> (1)..(1)
<223> basic amino acid
<220>
<221> misc_feature
<222> (3)..(5)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (6)..(6)
<223> C, S or T
<400> 183
Xaa Cys Xaa Xaa Xaa Xaa
1 5
<210> 184
<211> 247
<212> PRT
<213> Chile person
<400> 184
Met Ala Ser Leu Ser Arg Pro Ser Leu Pro Ser Cys Leu Cys Ser Phe
1 5 10 15
Leu Leu Leu Leu Leu Leu Gln Val Ser Ser Ser Tyr Ala Gly Gln Phe
20 25 30
Arg Val Ile Gly Pro Arg His Pro Ile Arg Ala Leu Val Gly Asp Glu
35 40 45
Val Glu Leu Pro Cys Arg Ile Ser Pro Gly Lys Asn Ala Thr Gly Met
50 55 60
Glu Val Gly Trp Tyr Arg Pro Pro Phe Ser Arg Val Val His Leu Tyr
65 70 75 80
Arg Asn Gly Lys Asp Gln Asp Gly Asp Gln Ala Pro Glu Tyr Arg Gly
85 90 95
Arg Thr Glu Leu Leu Lys Asp Ala Ile Gly Glu Gly Lys Val Thr Leu
100 105 110
Arg Ile Arg Asn Val Arg Phe Ser Asp Glu Gly Gly Phe Thr Cys Phe
115 120 125
Phe Arg Asp His Ser Tyr Gln Glu Glu Ala Ala Met Glu Leu Lys Val
130 135 140
Glu Asp Pro Phe Tyr Trp Val Ser Pro Gly Val Leu Val Leu Leu Ala
145 150 155 160
Val Leu Pro Val Leu Leu Leu Gln Ile Thr Val Gly Leu Ile Phe Leu
165 170 175
Cys Leu Gln Tyr Arg Leu Arg Gly Lys Leu Arg Ala Glu Ile Glu Asn
180 185 190
Leu His Arg Thr Phe Asp Pro His Phe Leu Arg Val Pro Cys Trp Lys
195 200 205
Ile Thr Leu Phe Val Ile Val Pro Val Leu Gly Pro Leu Val Ala Leu
210 215 220
Ile Ile Cys Tyr Asn Trp Leu His Arg Arg Leu Ala Gly Gln Phe Leu
225 230 235 240
Glu Glu Leu Arg Asn Pro Phe
245
<210> 185
<211> 20
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 185
His Cys Pro Tyr Cys Gly Trp Tyr Arg Ser Pro Phe Ser Arg Val Val
1 5 10 15
His Leu Tyr Arg
20
<210> 186
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 186
His Leu Tyr Arg
1
<210> 187
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (2)..(3)
<223> Xaa can be any naturally occurring amino acid
<400> 187
Cys Xaa Xaa Cys
1
<210> 188
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (2)..(3)
<223> Xaa can be any naturally occurring amino acid
<400> 188
Cys Xaa Xaa Ser
1
<210> 189
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (2)..(3)
<223> Xaa can be any naturally occurring amino acid
<400> 189
Cys Xaa Xaa Thr
1
<210> 190
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (2)..(3)
<223> Xaa can be any naturally occurring amino acid
<400> 190
Ser Xaa Xaa Cys
1
<210> 191
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (2)..(3)
<223> Xaa can be any naturally occurring amino acid
<400> 191
Thr Xaa Xaa Cys
1
<210> 192
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> peptide
<222> (1)..(1)
<223> D or E
<220>
<221> misc_feature
<222> (2)..(4)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (6)..(6)
<223> L or I
<400> 192
Xaa Xaa Xaa Xaa Leu Xaa
1 5
<210> 193
<211> 5
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (2)..(3)
<223> Xaa can be any naturally occurring amino acid
<400> 193
Asp Xaa Xaa Leu Leu
1 5
<210> 194
<211> 6
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (2)..(4)
<223> Xaa can be any naturally occurring amino acid
<400> 194
Asp Xaa Xaa Xaa Leu Leu
1 5
<210> 195
<211> 4
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (2)..(3)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> peptide
<222> (4)..(4)
<223> F, Y or W
<400> 195
Tyr Xaa Xaa Xaa
1
<210> 196
<211> 14
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (2)..(3)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> misc_feature
<222> (5)..(5)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> misc_feature
<222> (7)..(8)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> misc_feature
<222> (10)..(10)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> misc_feature
<222> (12)..(13)
<223> Xaa can be any naturally occurring amino acid
<400> 196
Cys Xaa Xaa Cys Xaa Cys Xaa Xaa Cys Xaa Cys Xaa Xaa Cys
1 5 10
<210> 197
<211> 12
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (2)..(3)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> misc_feature
<222> (6)..(7)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> misc_feature
<222> (10)..(11)
<223> Xaa can be any naturally occurring amino acid
<400> 197
Cys Xaa Xaa Cys Cys Xaa Xaa Cys Cys Xaa Xaa Cys
1 5 10
<210> 198
<211> 10
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (2)..(3)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> misc_feature
<222> (5)..(6)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> misc_feature
<222> (8)..(9)
<223> Xaa can be any naturally occurring amino acid
<400> 198
Cys Xaa Xaa Cys Xaa Xaa Cys Xaa Xaa Cys
1 5 10
<210> 199
<211> 10
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<220>
<221> misc_feature
<222> (2)..(2)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> misc_feature
<222> (5)..(5)
<223> Xaa can be any naturally occurring amino acid
<220>
<221> misc_feature
<222> (8)..(8)
<223> Xaa can be any naturally occurring amino acid
<400> 199
Cys Xaa Cys Cys Xaa Cys Cys Xaa Cys Cys
1 5 10
<210> 200
<211> 21
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 200
Tyr Arg Pro Pro Phe Ser Arg Val Val His Leu Tyr Arg Asn Gly Lys
1 5 10 15
Asp Gln Asp Gly Asp
20
<210> 201
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 201
Arg Pro Pro Phe Ser Arg Val Val His Leu Tyr Arg Asn Gly Lys
1 5 10 15
<210> 202
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 202
Pro Phe Ser Arg Val Val His Leu Tyr Arg Asn Gly Lys Asp Gln
1 5 10 15
<210> 203
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 203
Phe Ser Arg Val Val His Leu Tyr Arg Asn Gly Lys Asp Gln Asp
1 5 10 15
<210> 204
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 204
Ser Arg Val Val His Leu Tyr Arg Asn Gly Lys Asp Gln Asp Gly
1 5 10 15
<210> 205
<211> 21
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 205
Met Glu Val Gly Trp Tyr Arg Ser Pro Phe Ser Arg Val Val His Leu
1 5 10 15
Tyr Arg Asn Gly Lys
20
<210> 206
<211> 21
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 206
Met Glu Val Gly Trp Tyr Arg Pro Pro Phe Ser Arg Val Val His Leu
1 5 10 15
Tyr Arg Asn Gly Lys
20
<210> 207
<211> 277
<212> PRT
<213> Chile person
<400> 207
Met Gly Leu Leu Glu Cys Cys Ala Arg Cys Leu Val Gly Ala Pro Phe
1 5 10 15
Ala Ser Leu Val Ala Thr Gly Leu Cys Phe Phe Gly Val Ala Leu Phe
20 25 30
Cys Gly Cys Gly His Glu Ala Leu Thr Gly Thr Glu Lys Leu Ile Glu
35 40 45
Thr Tyr Phe Ser Lys Asn Tyr Gln Asp Tyr Glu Tyr Leu Ile Asn Val
50 55 60
Ile His Ala Phe Gln Tyr Val Ile Tyr Gly Thr Ala Ser Phe Phe Phe
65 70 75 80
Leu Tyr Gly Ala Leu Leu Leu Ala Glu Gly Phe Tyr Thr Thr Gly Ala
85 90 95
Val Arg Gln Ile Phe Gly Asp Tyr Lys Thr Thr Ile Cys Gly Lys Gly
100 105 110
Leu Ser Ala Thr Val Thr Gly Gly Gln Lys Gly Arg Gly Ser Arg Gly
115 120 125
Gln His Gln Ala His Ser Leu Glu Arg Val Cys His Cys Leu Gly Lys
130 135 140
Trp Leu Gly His Pro Asp Lys Phe Val Gly Ile Thr Tyr Ala Leu Thr
145 150 155 160
Val Val Trp Leu Leu Val Phe Ala Cys Ser Ala Val Pro Val Tyr Ile
165 170 175
Tyr Phe Asn Thr Trp Thr Thr Cys Gln Ser Ile Ala Phe Pro Ser Lys
180 185 190
Thr Ser Ala Ser Ile Gly Ser Leu Cys Ala Asp Ala Arg Met Tyr Gly
195 200 205
Val Leu Pro Trp Asn Ala Phe Pro Gly Lys Val Cys Gly Ser Asn Leu
210 215 220
Leu Ser Ile Cys Lys Thr Ala Glu Phe Gln Met Thr Phe His Leu Phe
225 230 235 240
Ile Ala Ala Phe Val Gly Ala Ala Ala Thr Leu Val Ser Leu Leu Thr
245 250 255
Phe Met Ile Ala Ala Thr Tyr Asn Phe Ala Val Leu Lys Leu Met Gly
260 265 270
Arg Gly Thr Lys Phe
275
<210> 208
<211> 29
<212> PRT
<213> Chile person
<400> 208
Met Ala Ser Leu Ser Arg Pro Ser Leu Pro Ser Cys Leu Cys Ser Phe
1 5 10 15
Leu Leu Leu Leu Leu Leu Gln Val Ser Ser Ser Tyr Ala
20 25
<210> 209
<211> 26
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 209
His Glu Ala Leu Thr Gly Thr Glu Lys Leu Ile Glu Thr Tyr Phe Ser
1 5 10 15
Lys Asn Tyr Gln Asp Tyr Glu Tyr Leu Ile
20 25
<210> 210
<211> 28
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 210
Thr Trp Thr Thr Cys Gln Ser Ile Ala Phe Pro Ser Lys Thr Ser Ala
1 5 10 15
Ser Ile Gly Ser Leu Cys Ala Asp Ala Arg Met Tyr
20 25
<210> 211
<211> 28
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 211
Gly Val Leu Pro Trp Asn Ala Phe Pro Gly Lys Val Cys Gly Ser Asn
1 5 10 15
Leu Leu Ser Ile Cys Lys Thr Ala Glu Phe Gln Met
20 25
<210> 212
<211> 19
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 212
Leu Thr Gly Thr Glu Lys Leu Ile Glu Thr Tyr Phe Ser Lys Asn Tyr
1 5 10 15
Gln Asp Tyr
<210> 213
<211> 19
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 213
Trp Thr Thr Cys Gln Ser Ile Ala Phe Pro Ser Lys Thr Ser Ala Ser
1 5 10 15
Ile Gly Ser
<210> 214
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 214
Val Leu Pro Trp Asn Ala Phe Pro Gly Lys Val Cys Gly Ser Asn
1 5 10 15
<210> 215
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 215
Leu Thr Gly Thr Glu Lys Leu Ile Glu Thr Tyr Phe Ser Lys Asn
1 5 10 15
<210> 216
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 216
Thr Glu Lys Leu Ile Glu Thr Tyr Phe Ser Lys Asn Tyr Gln Asp
1 5 10 15
<210> 217
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 217
Glu Lys Leu Ile Glu Thr Tyr Phe Ser Lys Asn Tyr Gln Asp Tyr
1 5 10 15
<210> 218
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 218
Trp Thr Thr Cys Gln Ser Ile Ala Phe Pro Ser Lys Thr Ser Ala
1 5 10 15
<210> 219
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 219
Thr Thr Cys Gln Ser Ile Ala Phe Pro Ser Lys Thr Ser Ala Ser
1 5 10 15
<210> 220
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 220
Thr Cys Gln Ser Ile Ala Phe Pro Ser Lys Thr Ser Ala Ser Ile
1 5 10 15
<210> 221
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 221
Cys Gln Ser Ile Ala Phe Pro Ser Lys Thr Ser Ala Ser Ile Gly
1 5 10 15
<210> 222
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 222
Gln Ser Ile Ala Phe Pro Ser Lys Thr Ser Ala Ser Ile Gly Ser
1 5 10 15
<210> 223
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 223
Val Leu Pro Trp Asn Ala Phe Pro Gly Lys Val Cys Gly Ser Asn
1 5 10 15
<210> 224
<211> 26
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 224
His Glu Ala Leu Thr Gly Thr Glu Lys Leu Ile Glu Thr Tyr Phe Ser
1 5 10 15
Lys Asn Tyr Gln Asp Tyr Glu Tyr Leu Ile
20 25
<210> 225
<211> 28
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 225
Thr Trp Thr Thr Cys Gln Ser Ile Ala Phe Pro Ser Lys Thr Ser Ala
1 5 10 15
Ser Ile Gly Ser Leu Cys Ala Asp Ala Arg Met Tyr
20 25
<210> 226
<211> 28
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 226
Gly Val Leu Pro Trp Asn Ala Phe Pro Gly Lys Val Cys Gly Ser Asn
1 5 10 15
Leu Leu Ser Ile Cys Lys Thr Ala Glu Phe Gln Met
20 25
<210> 227
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 227
Pro Arg His Arg Asp Thr Gly Ile Leu Asp Ser Ile Gly Arg Phe
1 5 10 15
<210> 228
<211> 17
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 228
Glu Asn Pro Val Val His Phe Phe Lys Asn Ile Val Thr Pro Arg Thr
1 5 10 15
Pro
<210> 229
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 229
Arg Ala Ser Asp Tyr Lys Ser Ala His Lys Gly Phe Lys Gly Val
1 5 10 15
<210> 230
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 230
Gly Phe Lys Gly Val Asp Ala Gln Gly Thr Leu Ser Lys Ile Phe
1 5 10 15
<210> 231
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 231
Leu Gly Gly Arg Asp Ser Arg Ser Gly Ser Pro Met Ala Arg Arg
1 5 10 15
<210> 232
<211> 20
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 232
Thr Gln Asp Glu Asn Pro Val Val His Phe Phe Lys Asn Ile Val Thr
1 5 10 15
Pro Arg Thr Pro
20
<210> 233
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 233
Thr Gln Asp Glu Asn Pro Val Val His Phe Phe Lys Asn Ile Val
1 5 10 15
<210> 234
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 234
Gln Asp Glu Asn Pro Val Val His Phe Phe Lys Asn Ile Val Thr
1 5 10 15
<210> 235
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 235
Asp Glu Asn Pro Val Val His Phe Phe Lys Asn Ile Val Thr Pro
1 5 10 15
<210> 236
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 236
Glu Asn Pro Val Val His Phe Phe Lys Asn Ile Val Thr Pro Arg
1 5 10 15
<210> 237
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 237
Asn Pro Val Val His Phe Phe Lys Asn Ile Val Thr Pro Arg Thr
1 5 10 15
<210> 238
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 238
Pro Val Val His Phe Phe Lys Asn Ile Val Thr Pro Arg Thr Pro
1 5 10 15
<210> 239
<211> 28
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 239
Ala Ser Asp Tyr Lys Ser Ala His Lys Gly Phe Lys Gly Val Asp Ala
1 5 10 15
Gln Gly Thr Leu Ser Lys Ile Phe Lys Leu Gly Gly
20 25
<210> 240
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 240
Ala Ser Asp Tyr Lys Ser Ala His Lys Gly Phe Lys Gly Val Asp
1 5 10 15
<210> 241
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 241
Ser Asp Tyr Lys Ser Ala His Lys Gly Phe Lys Gly Val Asp Ala
1 5 10 15
<210> 242
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 242
Asp Tyr Lys Ser Ala His Lys Gly Phe Lys Gly Val Asp Ala Gln
1 5 10 15
<210> 243
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 243
Tyr Lys Ser Ala His Lys Gly Phe Lys Gly Val Asp Ala Gln Gly
1 5 10 15
<210> 244
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 244
Lys Ser Ala His Lys Gly Phe Lys Gly Val Asp Ala Gln Gly Thr
1 5 10 15
<210> 245
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 245
Ser Ala His Lys Gly Phe Lys Gly Val Asp Ala Gln Gly Thr Leu
1 5 10 15
<210> 246
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 246
Ala His Lys Gly Phe Lys Gly Val Asp Ala Gln Gly Thr Leu Ser
1 5 10 15
<210> 247
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 247
His Lys Gly Phe Lys Gly Val Asp Ala Gln Gly Thr Leu Ser Lys
1 5 10 15
<210> 248
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 248
Lys Gly Phe Lys Gly Val Asp Ala Gln Gly Thr Leu Ser Lys Ile
1 5 10 15
<210> 249
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 249
Gly Phe Lys Gly Val Asp Ala Gln Gly Thr Leu Ser Lys Ile Phe
1 5 10 15
<210> 250
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 250
Phe Lys Gly Val Asp Ala Gln Gly Thr Leu Ser Lys Ile Phe Lys
1 5 10 15
<210> 251
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 251
Lys Gly Val Asp Ala Gln Gly Thr Leu Ser Lys Ile Phe Lys Leu
1 5 10 15
<210> 252
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 252
Gly Val Asp Ala Gln Gly Thr Leu Ser Lys Ile Phe Lys Leu Gly
1 5 10 15
<210> 253
<211> 15
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 253
Val Asp Ala Gln Gly Thr Leu Ser Lys Ile Phe Lys Leu Gly Gly
1 5 10 15
<210> 254
<211> 14
<212> PRT
<213> artificial sequence
<220>
<223> synthetic peptides
<400> 254
Leu Ser Arg Phe Ser Trp Gly Ala Glu Gly Gln Arg Pro Gly
1 5 10
<210> 255
<211> 171
<212> PRT
<213> Chile person
<400> 255
Met Ala Ser Gln Lys Arg Pro Ser Gln Arg His Gly Ser Lys Tyr Leu
1 5 10 15
Ala Thr Ala Ser Thr Met Asp His Ala Arg His Gly Phe Leu Pro Arg
20 25 30
His Arg Asp Thr Gly Ile Leu Asp Ser Ile Gly Arg Phe Phe Gly Gly
35 40 45
Asp Arg Gly Ala Pro Lys Arg Gly Ser Gly Lys Asp Ser His His Pro
50 55 60
Ala Arg Thr Ala His Tyr Gly Ser Leu Pro Gln Lys Ser His Gly Arg
65 70 75 80
Thr Gln Asp Glu Asn Pro Val Val His Phe Phe Lys Asn Ile Val Thr
85 90 95
Pro Arg Thr Pro Pro Pro Ser Gln Gly Lys Gly Arg Gly Leu Ser Leu
100 105 110
Ser Arg Phe Ser Trp Gly Ala Glu Gly Gln Arg Pro Gly Phe Gly Tyr
115 120 125
Gly Gly Arg Ala Ser Asp Tyr Lys Ser Ala His Lys Gly Phe Lys Gly
130 135 140
Val Asp Ala Gln Gly Thr Leu Ser Lys Ile Phe Lys Leu Gly Gly Arg
145 150 155 160
Asp Ser Arg Ser Gly Ser Pro Met Ala Arg Arg
165 170

Claims (25)

1. A kit comprising:
a) One or more dosage forms of a fumarate compound selected from the group consisting of: dimethyl fumarate (DMF), monomethyl fumarate (MMF), compounds that are metabolizable to MMF in vivo, and combinations thereof; and
b) An immunogenic or tolerogenic peptide in one or more dosage forms comprising a T cell epitope of an antigenic protein involved in a fumarate-related disease or disorder.
2. The kit of claim 1, wherein the peptide is an immunogenic peptide having an oxidoreductase motif linked to the T cell epitope, the oxidoreductase motif having a sequence of the general formula:
Z m -[CST]-X n -C- (SEQ ID NOS: 1 to 25) or Z m -C-X n -[CST]- (SEQ ID NO:26 to 50), wherein n is an integer selected from 2, 1, 3 or 0, wherein m is an integer selected from 0 to 3, wherein X is any amino acid, wherein Z is any amino acid, wherein [ CST ] ]Represents any one of cysteine (C), serine (S) or threonine (T);
wherein the oxidoreductase motif and the T cell epitope are separated by a linker having 0 to 7 amino acids,
wherein hyphen (-) in the oxidoreductase motif represents the point of attachment of the oxidoreductase motif to the N-terminal end of the linker or the epitope or to the C-terminal end of the linker or the T cell epitope.
3. The kit of claim 1 or 2, wherein the fumarate compound is dimethyl fumarate, monomethyl fumarate, or a combination of dimethyl fumarate and monomethyl fumarate, or a prodrug, deuterated form, clathrate, solvate, tautomer, stereoisomer, or pharmaceutically acceptable salt thereof.
4. The kit of claim 1, 2 or 3, wherein the compound is selected from the group consisting of dimethyl fumarate (formula (II) below), monomethyl fumarate (formula (III) below), duloxethyl fumarate (formula (IV) below), and tepilamide fumarate (formula (V) below):
or any one or more thereof, or any one or more deuterated forms, clathrates, solvates, tautomers, stereoisomers, or pharmaceutically acceptable salts thereof, or a combination of any one of the foregoing.
5. The kit of any one of claims 1-4, wherein the fumarate-related disease or disorder is an autoimmune disorder, a demyelinating disorder, a graft rejection, or a cancer.
6. The kit of any one of claims 1 to 5, wherein the fumarate-related disease or disorder is MS, and wherein the autoantigen is selected from the group consisting of: myelin Oligodendrocyte Glycoprotein (MOG), myelin Basic Protein (MBP), proteolipid protein (PLP), myelin-associated antigen (MAG), oligodendrocyte-specific protein (OSP), myelin-associated oligodendrocyte basic protein (MOBP), and 2',3' -cyclic nucleotide 3' -phosphodiesterase (CNPase), S100 beta protein, and transaldolase H; or alternatively
Wherein the fumarate-related disease or disorder is NMO, and wherein the autoantigen is MOG; or alternatively
Wherein the fumarate-related disease or disorder is psoriasis, and wherein the autoantigen is selected from the group consisting of: ADAMTSL5, PLA2G4D, keratins such as keratin 14 or keratin 17, antigens from wheat (Triticum aestivum), pso p27, antimicrobial peptides, cetrorofil defensin 1 and LL37; or alternatively
Wherein the fumarate-related disease or disorder is Rheumatoid Arthritis (RA), and wherein the antigenic protein is selected from the group comprising: GRP78, HSP60, 60kDa chaperonin 2, gelsolin, chitinase-3-like protein 1, cathepsin S, serum albumin, focal adhesion proteins and cathepsin D.
7. The kit according to any one of claims 1 to 6, comprising an immunogenic peptide, wherein the (self) antigen involved in a fumarate-related disease or disorder does not naturally comprise an oxidoreductase motif within the 11 amino acids of the N-or C-terminus adjacent to the epitope, and/or wherein the epitope does not naturally comprise an oxidoreductase motif in its sequence.
8. The kit of any one of claims 1 to 7, wherein the T cell epitope is an MHC class II T cell epitope or a NKT cell epitope.
9. The kit according to any one of claims 1 to 8, wherein the T cell epitope is a NKT cell epitope having the amino acid motif [ FWHY ] -XX- [ ILMV ] -XX- [ FWHY ] (SEQ ID NO: 51), preferably the amino acid motif [ FW ] -XX- [ ILMV ] -XX- [ FW ] (SEQ ID NO: 52).
10. The kit of any one of claims 1 to 9, wherein the oxidoreductase motif in the immunogenic peptide is located N-terminal to the epitope.
11. The kit according to any one of claims 1 to 10, wherein in the immunogenic peptide the epitope is 7 to 30 amino acids in length, preferably 7 to 25 amino acids, more preferably 7 to 20 amino acids.
12. The kit according to any one of claims 1 to 11, wherein the tolerogenic or immunogenic peptide is 9 to 50 amino acids in length, preferably 9 to 40 amino acids, more preferably 9 to 30 amino acids.
13. The kit according to any one of claims 2 to 12, wherein in the immunogenic peptide the oxidoreductase motif is selected from the following amino acid motifs:
(a)Z m -[CST]-X n -C-or Z m -C-X n -[CST]-,
Wherein n is 0, and wherein m is O, 1 or 2,
wherein Z is a basic amino acid, preferably selected from H, K, R and unnatural basic amino acids as defined herein, e.g. L-ornithine, more preferably K or H;
(b)Z m -[CST]-X n -C-or Z m -C-X n -[CST]-,
Wherein n is 1, wherein X is any amino acid, preferably a basic amino acid, selected from H, K, R and unnatural basic amino acids such as L-ornithine, preferably K or R,
wherein m is O, 1 or 2,
wherein Z is a basic amino acid, preferably selected from H, K, R and unnatural basic amino acids as defined herein, e.g. L-ornithine, preferably K or H;
(c)Z m -[CST]-X n -C-or Z m -C-X n -[CST]-,
Wherein n is 2, thereby producing an internal X within the oxidoreductase motif 1 X 2 Amino acid conjugate, wherein m is 0, 1 or 2, wherein Z is a basic amino acid, preferably selected from H, K, R and unnatural basic amino acids as defined herein, e.g. L-ornithine, preferably K or H; or alternatively
(d)Z m -[CST]-X n -C-or Z m -C-X n -[CST]-,
Wherein n is 3, thereby producing an internal X within the oxidoreductase motif 1 X 2 X 3 An amino acid segment wherein m is 0, 1 or 2, wherein Z is a basic amino acid, preferably selected from H, K, R and unnatural basic amino acids as defined herein, e.g. L-ornithine, preferably K or H.
14. The kit of any one of claims 1 to 13, wherein the immunogenic peptide has an oxidoreductase motif comprising the sequence: CC, KCC, RCC, CRC, CKC, KCRC (SEQ ID NO: 65), KCKCKC (SEQ ID NO: 63), RCKC (SEQ ID NO: 171), RCRC (SEQ ID NO: 67), CPYC (SEQ ID NO: 172), HCPYC (SEQ ID NO: 71), KCPYC (SEQ ID NO: 72), RCPYC (SEQ ID NO: 73), CRPYC (SEQ ID NO: 83), CPRYC (SEQ ID NO: 88), CPYRC (SEQ ID NO: 92), CKPYC (SEQ ID NO: 96), CPKYC (SEQ ID NO: 100), CPYKC (SEQ ID NO: 104), RCRPYC (SEQ ID NO: 86), RCPRYC (SEQ ID NO: 90), RCPYC (SEQ ID NO: 94), RCKPYC (SEQ ID NO: 98), RCYC (SEQ ID NO: 102), RCPYC (SEQ ID NO: 106), RPYC (SEQ ID NO: 84), CPKYC (SEQ ID NO: 89), CPKCKC (SEQ ID NO: 101), PKKC (KC ID NO: 93), or PKKC (SEQ ID NO: 105).
15. The kit according to any one of claims 1 to 14, wherein the tolerogenic or immunogenic peptide comprises a T cell epitope derived from the Myelin Oligodendrocyte Glycoprotein (MOG) antigen amino acid sequence, preferably selected from the group comprising:
YRPPFSRVVHLYRNGKDQDGD(SEQ ID NO:200),
RPPFSRVVHLYRNGK(SEQ ID NO:201),
PFSRVVHLYRNGKDQ(SEQ ID NO:202),
FSRVVHLYRNGKDQD(SEQ ID NO:203),
SRVVHLYRNGKDQDG(SEQ ID NO:204),
FLRVPCWKI(SEQ ID NO:164),
FLRVPSWKI(SEQ ID NO:165),
VVHLYRNGK(SEQ ID NO:170),
MEVGWYRSPFSRVVVHLYRNGK (mouse SEQ ID NO: 205),
MEVGWYRPPFSRVVHLYRNGK (human SEQ ID NO: 206),
YRSPFSRVV (mouse SEQ ID NO: 169), and
YRPPFSRVV (human SEQ ID NO: 168), or a combination thereof;
wherein the immunogenic peptide comprises a T cell epitope derived from the myelin proteolipid protein (PLP) antigenic amino acid sequence, preferably selected from the group comprising:
HEALTGTEKLIETYFSKNYQDYEYLI(SEQ ID NO:209),
TVVTTCQSIAFPSKTSASIGSLCADARMY(SEQ ID NO:210),
GVLPWNAFPGKVCGSNLLSICKTAEFQM(SEQ ID NO:211),
LTGTEKLIETYFSKNYQDY(SEQ ID NO:212),
WTTCQSIAFPSKTSASIGS(SEQ ID NO:213),
VLPWNAFPGKVCGSN(SEQ ID NO:214),
LTGTEKLIETYFSKN(SEQ ID NO:215),
TEKLIETYFSKNYQD(SEQ ID NO:216),
EKLIETYFSKNYQDY(SEQ ID NO:217),
WTTCQSIAFPSKTSA(SEQ ID NO:218),
TTCQSIAFPSKTSAS(SEQ ID NO:219),
TCQSIAFPSKTSASI(SEQ ID NO:220),
CQSIAFPSKTSASIG(SEQ ID NO:221),
QSIAFPSKTSASIGS(SEQ ID NO:222),
VLPWNAFPGKVCGSN(SEQ ID NO:223),
HEALTGTEKLIETYFSKNYQDYEYLI(SEQ ID NO:224),
TWTTCQSIAFPSKTSASIGSLCADARMY (SEQ ID NO: 225), and
GVLPWNAFPGKVCGSNLLSICKTAEFQM(SEQ ID NO:226),
or a combination thereof;
wherein the immunogenic peptide comprises a T cell epitope derived from the Myelin Basic Protein (MBP) antigen amino acid sequence, preferably selected from the group comprising: PRHRDTGILDSIGRF (SEQ ID NO: 227)
ENPVVHFFKNIVTPRTP(SEQ ID NO:228)
RASDYKSAHKGFKGV(SEQ ID NO:229)
GFKGVDAQGTLSKIF(SEQ ID NO:230)
LGGRDSRSGSPMARR(SEQ ID NO:231)
TQDENPVVHFFKNIVTPRTP(SEQ ID NO:232)
TQDENPVVHFFKNIV(SEQ ID NO:233)
QDENPVVHFFKNIVT(SEQ ID NO:234)
DENPVVHFFKNIVTP(SEQ ID NO:235)
ENPVVHFFKNIVTPR(SEQ ID NO:236)
NPWHFFKNIVTPRT(SEQ ID NO:237)
PVVHFFKNIVTPRTP(SEQ ID NO:238)
ASDYKSAHKGFKGVDAQGTLSKIFKLGG(SEQ ID NO:239)
ASDYKSAHKGFKGVD(SEQ ID NO:240)
SDYKSAHKGFKGVDA(SEQ ID NO:241)
DYKSAHKGFKGVDAQ(SEQ ID NO:242)
YKSAHKGFKGVDAQG(SEQ ID NO:243)
KSAHKGFKGVDAQGT(SEQ ID NO:244)
SAHKGFKGVDAQGTL(SEQ ID NO:245)
AHKGFKGVDAQGTLS(SEQ ID NO:246)
HKGFKGVDAQGTLSK(SEQ ID NO:247)
KGFKGVDAQGTLSKI(SEQ ID NO:248)
GFKGVDAQGTLSKIF(SEQ ID NO:249)
FKGVDAQGTLSKIFK(SEQ ID NO:250)
KGVDAQGTLSKIFKL(SEQ ID NO:251)
GVDAQGTLSKIFKLG(SEQ ID NO:252)
VDAQGTLSKIFKLGG (SEQ ID NO: 253), and
LSRFSWGAEGQRPG(SEQ ID NO:254),
or a combination thereof, preferably SEQ ID NO:227 to 230.
16. The kit according to any one of claims 1 to 15, wherein the immunogenic or tolerogenic peptide comprises a T cell epitope derived from Myelin Oligodendrocyte Glycoprotein (MOG) antigen amino acid sequence selected from the group consisting of YRSPFSRVV (SEQ ID NO: 169) and YRPPFSRVV (human SEQ ID NO: 168), and comprises the amino acid sequence GW as linker and the amino acid sequence HLYR (SEQ ID NO: 186) as flanking.
17. The kit according to any one of claims 1 to 16, wherein the immunogenic or tolerogenic peptide comprises a T cell epitope derived from a Myelin Oligodendrocyte Glycoprotein (MOG) antigen amino acid sequence selected from the group consisting of FLRVPCWKI (SEQ ID NO: 164) and FLRVPSWKI (SEQ ID NO: 165), and comprises amino acid sequence VRY (SEQ ID NO: 271) as a linker and comprises as a flanking amino acid sequence selected from the group consisting of: TLF (SEQ ID NO: 269), TLFK (SEQ ID NO: 270), or TLFKK (SEQ ID NO: 267).
18. The kit of any one of claims 2 to 15, wherein the immunogenic peptide is selected from the group consisting of:
KHCPYCVRYFLRVPSWKITLFKK(SEQ ID NO:176),
KHCPYCVRYFLRVPCWKITLFKK(SEQ ID NO:177).
HCPYCVRYFLRVPSWKITLF(SEQ ID NO:174),
HCPYCVRYFLRVPCWKITLF(SEQ ID NO:175),
HCPYCGWYRSPFSRVVHLYR(SEQ ID NO:185),
KCRCGWYRSPFSRVVHLYR (SEQ ID NO: 266), and
KCRPYCGWYRSPFSRVVHLYR(SEQ ID NO:268)。
19. the kit of any one of aspects 1 to 18, wherein the immunogenic peptide or tolerogenic peptide is present in the form of one or more nucleic acid molecules encoding the immunogenic peptide or tolerogenic peptide, preferably wherein the nucleic acid is selected from the group consisting of: isolated deoxyribonucleic acid (DNA), plasmid DNA (pDNA), coding DNA (cDNA), ribonucleic acid (RNA), messenger RNA (mRNA), or modified forms thereof.
20. The kit according to any one of claims 1 to 19 for use in the treatment of a fumarate-related disease or disorder, the prevention of a fumarate-related disease or disorder and/or the amelioration of symptoms of a fumarate-related disease or disorder.
21. A kit for use according to claim 20 for treating, preventing and/or ameliorating the following symptoms: autoimmune disorders, for example, psoriasis, rheumatoid Arthritis (RA), asthma, atopic dermatitis, scleroderma, ulcerative colitis; demyelinating disorders, e.g., multiple Sclerosis (MS), neuromyelitis optica (NMO); transplant rejection; or cancer.
22. Kit for use according to claim 20 or 21, wherein the immunogenic peptide or tolerogenic peptide is administered to a subject as a peptide or as a nucleic acid encoding the immunogenic peptide or tolerogenic peptide, preferably selected from isolated deoxyribonucleic acid (DNA), plasmid DNA (pDNA), encoding DNA (cDNA), ribonucleic acid (RNA), messenger RNA (mRNA), or modified forms thereof.
23. A method of treating a fumarate-related disease or disorder, ameliorating symptoms of a fumarate-related disease or disorder, and/or preventing a fumarate-related disease or disorder in a patient in need thereof comprising the step of administering an effective amount of a kit according to any one of aspects 1 to 19.
24. The kit for use according to claim 21, 22 or 23, or the method according to claim 23, wherein the fumarate compound and the immunogenic or tolerogenic peptide are administered simultaneously, sequentially and/or separately.
25. The kit for use according to claim 21, 22 or 23, or the method according to claim 23 or 24, wherein the fumarate compound is administered orally once or twice daily, and/or wherein the immunogenic peptide or tolerogenic peptide is administered by subcutaneous injection.
CN202180047680.0A 2020-05-06 2021-05-06 Combination therapy for fumarate-related diseases Pending CN117320710A (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
EP20173209.6 2020-05-06
EP20179019.3 2020-06-09
EP20179019 2020-06-09
PCT/EP2021/061972 WO2021144478A2 (en) 2020-05-06 2021-05-06 Combination treatment for fumarate-related diseases

Publications (1)

Publication Number Publication Date
CN117320710A true CN117320710A (en) 2023-12-29

Family

ID=71083405

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202180047680.0A Pending CN117320710A (en) 2020-05-06 2021-05-06 Combination therapy for fumarate-related diseases

Country Status (1)

Country Link
CN (1) CN117320710A (en)

Similar Documents

Publication Publication Date Title
ES2270870T3 (en) PEPTIDES DERIVED FROM MUC-1.
EP1951281B1 (en) Wt1 hla class ii-binding peptides and compositions and methods comprising same
US10617748B2 (en) Immunogenic control of tumours and tumour cells
US9132178B2 (en) Cytotoxic T lymphocyte inducing immunogens for prevention treatment and diagnosis of cancer
US20170100466A1 (en) Strategies to prevent and/or treat immune responses to soluble allofactors
US9044507B2 (en) Elimination of immune responses to viral vectors
RU2761653C2 (en) Peptides and diabetes treatment methods
CN115702162A (en) Peptides and methods for treating multiple sclerosis
US20230172894A1 (en) Combination treatment for fumarate-related diseases
US20150132329A1 (en) Novel melanoma antigen peptide and uses thereof
US20230181638A1 (en) Immunogenic peptides with new oxidoreductase motifs
CN117320710A (en) Combination therapy for fumarate-related diseases
JP2023525276A (en) Immunogenic peptides with extended oxidoreductase motifs
US9573975B2 (en) Melanoma antigen peptide and uses thereof
CN114746111A (en) Method for stratifying diabetic patients
CN117651711A (en) Improved methods of treatment using immunogenic peptides
US20150202273A1 (en) Prostate-specific tumor antigens and uses thereof
CN117729932A (en) Peptides and methods for treating neuromyelitis optica
CA3220605A1 (en) Methods of treating or preventing autoimmune diseases
TW202322845A (en) Neoantigens and methods of their use

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