TYPE 1 INTERFERON NEUTRALIZING FC-FUSION PROTEIN AND USE THEREOF

Information

  • Patent Application
  • 20220089684
  • Publication Number
    20220089684
  • Date Filed
    December 20, 2019
    4 years ago
  • Date Published
    March 24, 2022
    2 years ago
Abstract
The present invention relates to a type 1 interferon neutralizing FC-fusion protein and a use thereof and, more specifically, to: a dimer-type polypeptide to which a monomer comprising an interferon receptor fragment or an antibody Fc fragment is bound; a preparation method there for; and a pharmaceutical composition comprising same. The type 1 interferon neutralizing FC-fusion protein of the present invention blocks binding between type 1 interferon and an interferon receptor, and has an excellent ability of inhibiting the signaling and biological activities of interferon, thereby enabling diseases mediated by a type 1 interferon to be effectively treated.
Description
TECHNICAL FIELD

This application claims priority to Korean Patent Application No. 10-2018-0168801 filed on Dec. 24, 2018, and the entire specifications of which are incorporated herein by reference in their entireties.


The present invention relates to a type 1 interferon neutralizing Fc-fusion protein and use thereof, more specifically, it relates to a dimeric polypeptide in which a monomer comprising an interferon receptor fragment or an antibody Fc fragment is bound, and a method for preparing the same, and a composition for preventing or treating diseases related to abnormal expression of type 1 interferon or interferon-inducible gene comprising the same.


BACKGROUND OF THE INVENTION

Type I interferons (IFNs) (IFN-α, IFN-β, INF-ω, IFN-τ) are a group of structurally related cytokines with antiviral, antitumor and immunomodulatory effects. The human IFNa locus has two subgroups. The first subgroup has 14 non-alleles and 4 pseudogenes with at least 80% identity. The second subgroup, aII or omega (ω), has 1 functional gene and 5 pseudogenes showing 70% identity to the IFNα gene. The subtypes of IFNα have different specific activities, but show the same biological spectrum and have the same cellular receptors. Interferon β (IFN β) is encoded by a single gene with nearly 50% identity to the IFNα gene. Interferon γ produced by activated lymphocytes has no identity with alpha/beta interferon and does not react with their receptors.


All human type I IFNs bind to a cell surface receptor (IFN alpha receptor, IFNAR), which consists of two transmembrane proteins, IFNAR-1 and IFNAR-2. IFNAR-1 is essential for the high affinity binding and different specificity of the IFNAR complex. Although the difference in the function of each type I IFN subtype has not been elucidated, it is thought that each exhibits a different action with the IFNAR receptor component, potentially leading to various signaling.


Early studies of the function of type I IFN have focused on innate defenses against viral infection. However, more recent studies relate to type I IFNs as potent immunoregulatory cytokines in adaptive immune response. In particular, type I IFN has been shown to facilitate the mutation of native T cells in the Th1 pathway, increase antibody production, and support the functional activity and survival of memory T cells.


Several groups of recent studies have suggested that IFN-α can enhance the maturation or activation of dendritic cells (DCs). Moreover, increased expression of type I interferon has been described in a number of autoimmune diseases related literature.


The most studied example of this is insulin-dependent diabetes mellitus (IDDM) (Paulis et al. (1987) Lancet 2:1423), systemic lupus erythematosus (SLE) (Hooks et al. (1982) Arthritis Rheum. 25:396), Sjogren's syndrome (Lee Hong-Yau et al. (2013) Autoimmun Rev. 12(5):558-66), inflammatory myositis (Beckler et al. (2007) Mol Med. 13 (1-2): 59-68.) and rheumatoid arthritis (RA) in which IFN-β plays a more important role (Herzzog et al. (1988) Clin. Immunol. Immunopathol. 48:192, Hopkins and Meggle (1988) Clin. Exp. Immunol. 73:88, Alvin and Miller (1984) Arthritis Rheum. 27:582), which is all associated with elevated levels of IFNα.


Moreover, it has been reported that administration of interferon α, a representative type of type 1 interferon, exacerbates the underlying disease in patients with psoriasis and multiple sclerosis and induces SLE-like symptoms in patients without prior experience of autoimmune disease. Interferon α has also been shown to induce glomerulonephritis in normal mice and accelerate the development of concomitant autoimmune disease in NZB/W mice. Furthermore, IFN-α therapy has been shown to induce undesirable side effects including fever and neurological disorders in some cases.


Although it has the diversity of type 1 IFN, and in recent studies on SLE patients, not only IFN-α but also IFN-β acts as the etiology for mucosal skin diseases, there is a problem in that the treatment for type 1 interferon-mediated disease is concentrated on IFN-α alone, such as a vaccine (IFN-kinoid) or anti-IFN-α monoclonal neutralizing antibody (Sifalimumab, Rontalizumab, AGS-009) treatment. In addition, the problem of such neutralizing antibodies is that they cannot neutralize all the parts that bind to two different receptors of interferon, and have binding ability only to IFNAR1 or IFNAR2. In order to compensate for this problem, Anifrolumab (MAbs. 2015 March-April; 7(2): 428-439.) that specifically binds to IFNAR1, a type I IFN receptor, and exhibits neutralizing ability is being developed. However, the characteristics of these neutralizing antibody therapeutics in various signaling mechanisms of type 1 interferon are expected that there will be a limitation in inability to remove type 1 interferon from the body and complex signaling mechanisms such as non-canonical pathways mediated by IFNAR1 (Nat Immunol. 2013 September; 14(9):901-7.) or IFNAR2 (Sci Signal. 2014 May 27; 7 (327):ra50.; PLoS One. 2017; 12 (8): e0182866.) other than the canonical pathways mediated by IFNAR1 and IFNAR2.


DETAILED DESCRIPTION OF THE INVENTION
Technical Problem

The inventors of the present invention have completed the present invention, while developing a treatment for type 1 interferon-mediated disease, as it confirmed that the dimer-type polypeptide comprising the interferon receptor 1 fragment and the interferon receptor 2 fragment not only binds to type 1 interferon, but also significantly inhibits the inhibition of signaling mechanism initiation and biological activity during treatment.


Therefore, the object of the present invention is to provide a dimeric polypeptide in which a monomer comprising an interferon receptor fragment or an antibody Fc fragment is bounded, Wherein the monomer is a polypeptide selected from the group consisting of (i), (ii) and (iii):


(i) a monomer comprising an interferon receptor 1 (IFNAR1) fragment and an antibody Fc fragment;


(ii) a monomer comprising an interferon receptor 2 (IFNAR2) fragment and an antibody Fc fragment; and


(iii) an antibody Fc fragment.


Another object of the present invention is to provide a polynucleotide encoding the polypeptide.


Another object of the present invention is to provide a vector comprising the polynucleotide.


Another object of the present invention is to provide a host cell transformed with the vector.


Another object of the present invention is to provide a method for producing the polypeptide comprising:


(a) providing the host cell;


(b) culturing the provided cells; and


(c) preparing the polypeptide by recovering the polypeptide from the cell or culture medium.


Another object of the present invention is


to provide a pharmaceutical composition for preventing or treating a type 1 interferon-mediated disease or disorder comprising a polypeptide as an active ingredient.


also, to provide a pharmaceutical composition for preventing or treating a type 1 interferon-mediated disease or disorder consisting of a polypeptide as an active ingredient.


also, to provide a pharmaceutical composition for preventing or treating a type 1 interferon-mediated disease or disorder consisting essentially of a polypeptide as an active ingredient.


Another object of the present invention is to provide a use of the polypeptide for the preparation of an agent for the prevention or treatment of a type 1 interferon-mediated disease or disorder.


Another object of the present invention is to provide a method of treating a type 1 interferon-mediated disease or disorder, comprising administering to a subject in need thereof an effective amount of a composition comprising the polypeptide as an active ingredient


Technical Solution

Accordingly, the present invention in order to achieve the object of the present invention provides,


a dimeric polypeptide in which a monomer comprising an interferon receptor fragment or an antibody Fc fragment is bounded,


Wherein the monomer is a polypeptide selected from the group consisting of (i), (ii) and (iii):


(i) a monomer comprising an interferon receptor 1 (IFNAR1) fragment and an antibody Fc fragment;


(ii) a monomer comprising an interferon receptor 2 (IFNAR2) fragment and an antibody Fc fragment; and


(iii) an antibody Fc fragment.


In order to achieve another object of the present invention, the present invention provides a polynucleotide encoding the polypeptide.


In order to achieve another object of the present invention, the present invention provides a vector comprising the polynucleotide.


In order to achieve another object of the present invention, the present invention provides a host cell transformed with the vector.


In order to achieve another object of the present invention, the present invention provides a method for producing the polypeptide comprising:


(a) providing the host cell;


(b) culturing the provided cells; and


(c) preparing the polypeptide by recovering the polypeptide from the cell or culture medium.


In order to achieve another object of the present invention, the present invention, provides a pharmaceutical composition for prevention or treatment of type 1 interferon-mediated diseases or disorders comprising the polypeptide as an active ingredient.


In addition, provides a pharmaceutical composition for preventing or treating a type 1 interferon-mediated disease or disorder consisting of a polypeptide as an active ingredient.


In addition, provides a pharmaceutical composition for preventing or treating a type 1 interferon-mediated disease or disorder consisting essentially of a polypeptide as an active ingredient.


In order to achieve another object of the present invention, the present invention provides a use of the polypeptide for the preparation of an agent for the prevention or treatment of a type 1 interferon-mediated disease or disorder.


In order to achieve another object of the present invention, the present invention provides a method of treating a type 1 interferon-mediated disease or disorder, comprising administering to a subject in need thereof an effective amount of a composition comprising the polypeptide as an active ingredient.


Hereinafter, the present invention will be described in detail.


Therefore, the present invention is a dimeric polypeptide in which a monomer comprising an interferon receptor fragment or an antibody Fc fragment is bounded, wherein the monomer provide a polypeptide selected from the group consisting of (i), (ii) and (iii):


(i) a monomer comprising an interferon receptor 1 (IFNAR1) fragment and an antibody Fc fragment;


(ii) a monomer comprising an interferon receptor 2 (IFNAR2) fragment and an antibody Fc fragment; and


(iii) an antibody Fc fragment.


In the present specification including the claims, the term “interferon receptor fragment” is meant to include all polypeptides having human interferon receptor activity while having all or part of an amino acid sequence derived from a native human interferon receptor.


The terms “interferon receptor 1” “IFNAR 1” and “IFNAR-1 antigen” are used interchangeably, and variants, isoforms, species isoforms of human IFNAR 1 and analogs having at least one common epitope of IFNAR 1 are included. Thus, the polypeptides of the invention may in some cases cross-react with IFNAR 1 from other non-human species or other proteins structurally associated with human IFNAR 1 (e.g., human IFNAR-1 homologs). In other cases, the polypeptide may be intactly specific for human IFNAR-1 and does not represent a species or other type of cross-reactivity. The complete cDNA sequence of human IFNAR 1 has genbank accession number XM_005260964.2, NM_000629.2, or XM_011529552.2.


The nucleotide sequence used in the present invention may include the extracellular domain (28-436a.a.; P17181-1) of the sequence of IFNAR 1 Isoform 1 (genebank accession number: NM_000629.2), and may include a part of the nucleotide sequence. The IFNAR 1 fragment of the present invention may preferably be an interferon receptor 1 comprising the amino acid sequence represented by SEQ ID NO: 4, more preferably, it may be an interferon receptor 1 consisting of the amino acid sequence represented by SEQ ID NO: 4.


In one embodiment of the present invention, the polypeptide was prepared using the interferon receptor 1 comprising the amino acid sequence (Polynucleotide sequence represented by SEQ ID NO: 3—The polynucleotide sequence may include a stop codon. Hereinafter, it is the same for all DNA sequences) represented by SEQ ID NO: 4.


The terms “interferon receptor 2” “IFNAR 2” and “IFNAR-2 antigen” are used interchangeably, variants, isoforms, species isoforms of human IFNAR 2 and analogs having at least one common epitope of IFNAR 2 are included. Thus, the polypeptides of the invention may in some instances cross-react with IFNAR 2 from other non-human species or other proteins structurally related to human IFNAR 2 (e.g., human IFNAR-2 homologs). In other cases, the polypeptide may be intactly specific for human IFNAR-2 and does not represent a species or other type of cross-reactivity. The complete cDNA sequence of human IFNAR 2 has genbank accession number NM_000874.4, NM_001289125.1, or NM_001289126.1. The nucleotide sequence used in the present invention may include the extracellular domain (27-243a.a.; P48551-1) of the sequence of Isoform 1 of human IFNAR2 (genebank accession number: NM_001289125.1), may include a part of the nucleotide sequence. The IFNAR 2 fragment of the present invention may preferably be an interferon receptor 2 comprising the amino acid sequence represented by SEQ ID NO: 6, more preferably, it may be an interferon receptor 2 consisting of the amino acid sequence represented by SEQ ID NO: 6.


In one embodiment of the present invention, the polypeptide was prepared using an interferon receptor 2 fragment comprising the amino acid sequence (Polynucleotide sequence represented by SEQ ID NO: 5) represented by SEQ ID NO: 6.


In addition, the term “polypeptide having all or part of an amino acid sequence derived from an interferon receptor fragment” is meant to include a polypeptide comprising all or a substantial part of the amino acid sequence of SEQ ID NO: 4 or SEQ ID NO: 6, which is the amino acid sequence of a native interferon receptor fragment, or a polypeptide substantially similar to such a polypeptide.


Here, the “polypeptide comprising a substantial portion of the entire amino acid sequence of SEQ ID NO: 4 or SEQ ID NO: 6” is defined as a polypeptide comprising having an activity equal to or higher than that of the native interferon receptor having the amino acid sequence of SEQ ID NO: 4 or SEQ ID NO: 6, or a portion of the amino acid sequence of SEQ ID NO: 4 or SEQ ID NO: 6 comprising one or more substituted amino acids while still retaining the activity of the interferon receptor, although low in activity. The “polypeptide substantially similar to all or a substantial part of the amino acid sequence set forth in SEQ ID NO: 4 or SEQ ID NO: 6” is defined as a polypeptide comprising having an activity equal to or higher than that of the interferon receptor of SEQ ID NO: 4 or SEQ ID NO: 6, or the whole or a substantial portion of the amino acid sequence of SEQ ID NO: 4 or SEQ ID NO: 6 comprising one or more substituted amino acids while still retaining the activity of the interferon receptor, although low in activity.


There may be cases in which an N-terminal portion and/or a C-terminal portion are deleted in a polypeptide comprising the amino acid sequence set forth in SEQ ID NO: 4 or SEQ ID NO: 6 as a polypeptide comprising a substantial portion of the entire amino acid sequence set forth in SEQ ID NO: 4 or SEQ ID NO: 6, and there may be cases that even if one or more amino acids are substituted, if the amino acid before the substitution is chemically equivalent to the substituted amino acid, for example, when alanine, which is a hydrophobic amino acid, is substituted with another hydrophobic amino acid, in particular, a case where it is substituted with a more hydrophobic amino acid, for example, valine, leucine or isoleucine as a polypeptide substantially similar to all or a substantial part of the amino acid sequence set forth in SEQ ID NO: 4 or SEQ ID NO: 6.


In the present invention, “fusion” refers to integrating two molecules with different or the same function or structure, and may be fusion by any physical, chemical or biological method capable of binding the antibody Fc portion or each monomer to the interferon receptor.


The fusion is preferably by a linker peptide, which may bind to, for example, the C-terminus of an Fc fragment of an antibody.


“Antibody Fc fragment” may be an Fc fragment of an IgA, IgM, IgE, IgD, or IgG antibody, or a modification thereof. In one embodiment, the fragment is an Fc fragment (e.g., Fc fragment of an IgG1, IgG2a, IgG2b, IgG3, or IgG4 antibody) of an IgG antibody. In addition, the polypeptide comprising the Fc fragment of the present invention may be partially or entirely glycosylated or non-glycosylated. In addition, the polypeptide may include one or more regions derived from an antibody in addition to the Fc fragment. Additionally, the polypeptide may include an antibody-derived antigen binding domain, and a plurality of polypeptides may form an antibody or antibody-like protein.


In the present specification, amino acid residue number of antibody Fc fragments is according to the Kabat numbering system commonly used in the art (EU index number as in Kabat et al., in of Proteins of Immunological Interest 5th Ed., U.S. Department of Health and Human Services, NIH Publication No. 91-3242, 1991).


According to an embodiment of the present invention, it may include the wild-type Fc fragment of the present invention as well as a substituted Fc variant.


The wild-type Fc fragment sequence of the present invention may comprise the amino acid represented by SEQ ID NO: 8, and preferably, may consist of the amino acid represented by SEQ ID NO: 8.


The substituted Fc fragment of the present invention comprises one or more amino acid substitutions selected from the group consisting of Q347R, K360E, D399V, F405T and K409W according to the Kabat numbering system.


The substituted Fc fragment of the present invention may be one comprising K360E and K409W amino acid substitutions, preferably, it may comprise the amino acid sequence represented by SEQ ID NO: 12, more preferably, it may consist of the amino acid sequence represented by SEQ ID NO: 12.


The substituted Fc fragment of the present invention may be one comprising Q347R, D399V, and F405T amino acid substitutions, preferably, it may consist of the amino acid sequence represented by SEQ ID NO: 14, more preferably, it may consist of SEQ ID NO: 14.


In addition, the antibody Fc fragment as a monomer forming a dimer comprises SEQ ID NO: 8, SEQ ID NO: 12 or SEQ ID NO: 14, and a linker or other short fragment peptide may be attached to the N-terminus or C-terminus. The linker may be a linker comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 10, SEQ ID NO: 34, and SEQ ID NO: 53 to SEQ ID NO: 76. The antibody Fc fragment as a monomer may consist of an amino acid sequence selected from the group consisting of SEQ ID NO: 8, SEQ ID NO: 12, SEQ ID NO: 14, SEQ ID NO: 16, SEQ ID NO: 18, SEQ ID NO: 20, SEQ ID NO: 36, SEQ ID NO: 38 and SEQ ID NO: 40.


“Monomer comprising interferon receptor 1 (IFNAR1) fragment and antibody Fc fragment” of the present invention preferably may one comprising i) an interferon receptor 1 (IFNAR1) fragment comprising the amino acid sequence represented by SEQ ID NO: 4 and ii) an antibody Fc fragment comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 8, SEQ ID NO: 12 and SEQ ID NO: 14, more preferably, it may one comprising i) an interferon receptor 1 (IFNAR1) fragment comprising the amino acid sequence represented by SEQ ID NO: 4, ii) a linker comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 10, SEQ ID NO: 34, and SEQ ID NO: 53 to SEQ ID NO: 76, and iii) an antibody Fc fragment comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 8, SEQ ID NO: 12 and SEQ ID NO: 14, most preferably, it may be a monomer comprising or consisting of an amino acid selected from the group consisting of SEQ ID NO: 22, SEQ ID NO: 24, SEQ ID NO: 26, SEQ ID NO: 42, SEQ ID NO: 44 and SEQ ID NO: 46.


The “monomer comprising the interferon receptor 2 (IFNAR2) fragment and the antibody Fc fragment” of the present invention is preferably may one comprising i) an interferon receptor 2 (IFNAR2) fragment comprising the amino acid sequence represented by SEQ ID NO: 6 and ii) an antibody Fc fragment comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 8, SEQ ID NO: 12 and SEQ ID NO: 14, more preferably, it may one comprising i) an interferon receptor 2 (IFNAR2) fragment comprising the amino acid sequence represented by SEQ ID NO: 6, ii) a linker comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 10, SEQ ID NO: 34, SEQ ID NO: 53 to SEQ ID NO: 76 and iii) an antibody Fc fragment comprising an amino acid sequence selected from the group consisting of SEQ ID NO: 8, SEQ ID NO: 12 and SEQ ID NO: 14, most preferably, it may be a monomer comprising or consisting of amino acids selected from the group consisting of SEQ ID NO: 28, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 48, SEQ ID NO: 50 and SEQ ID NO: 52.


The polypeptide of the present invention may be one that neutralizes type I interferon, and thus can be used as a type I interferon neutralizing antibody.


As used herein, the term “type I interferon” is intended to refer to the multiple class I interferon group (ie, multiple type I interferon groups of molecules capable of binding to IFNAR 1 or IFNAR 2) of molecules that are ligands of IFNAR 1 and IFNAR 2. Examples of type I interferon ligands are interferon alpha 1, 2a, 2b, 4, 5, 6, 7, 8, 10, 14, 16, 17, 21, interferon beta, interferon omega and interferon epsilon.


A person skilled in the art can use the above-described polypeptides without limitation in practicing the present invention.


The present invention also provides a dimeric polypeptide wherein the monomer is linked to the antibody or fragment thereof by a peptide linker. A peptide linker refers to a molecule that connects two or more separate substances to each other as a short fragment of amino acids or amino acid analogs in which amino acids or amino acid-like substances are linked to each other by peptide bonds. Glycine, serine, and alanine are used as major constituent amino acids, so a glycine-serine linker, a glycine-serine-alanine linker, etc. can be used, and according to a preferred embodiment of the present invention, the linker may consist of or comprise an amino acid sequence represented by SEQ ID NO: 8 and SEQ ID NO: 10.


Polypeptides of the present invention may preferably comprise a flexible linker sequence inserted between the monomers.


The linker refers to a naturally-derived peptide linker or a synthetically-derived peptide linker. The peptide linker consists of a linear amino acid chain, wherein the 20 naturally occurring amino acids are monomeric building blocks. The linker may have a repetitive amino acid sequence or may have the sequence of a naturally occurring polypeptide, for example, a polypeptide having a hinge function. Since all peptide linkers can be encoded by nucleic acid molecules, they can be expressed in a recombinant method. Since the linker is itself a peptide, each monomer can be linked to the linker via a peptide bond to form a dimer.


The linker consists of amino acids linked together by peptide bonds, preferably 1 to 20 amino acids linked together by peptide bonds, in this case, the amino acid is preferably selected from among 20 natural amino acids. One or more of these amino acids are glycosylated as understood by one of ordinary skill in the art. Preferably, but not limited to, 1 to 20 amino acids are selected from among glycine, alanine, proline, asparagine, glutamine and lysine.


Suitable linkers include, for example, cleavable linkers and non-cleavable linkers. Cleavable linkers are typically readily cleaved under intracellular conditions. Suitable cleavable linkers include, for example, peptide linkers cleavable by intracellular proteases such as lysosomal proteases or endosomal proteases.


The linker is, for example, the N-terminus of the linker is connected to the C-terminus of the interferon receptor. Linking to the C-terminus of the interferon receptor can preferably be directly linked to the antibody expressed by the expression vector as the nucleotide sequence encoding the linker sequence is linked to the expression vector expressing the protein of the present invention so that the protein expression frame coincides. In addition, the N-terminus of the Fc portion of the antibody of the present invention is linked to the C-terminus of the linker.


The peptide linker of the present invention may be a peptide linker known in the art, but preferably a glycine-serine linker, a helix-forming peptide linker, or a peptide linker comprising or consisting of the amino acid sequence of SEQ ID NO: 8 or SEQ ID NO: 10.


The liker is, preferably, a gly-ser linker, for example (GlyxSery)z type (x is an integer from 1 to 5, y is an integer from 1 to 2, z is an integer from 1 to 6), for example (gly4ser1)3 or (gly3ser2)3, in the case of a helix-forming peptide linker (or rigid helical linker), it may be A(EAAAK)nA (n=an integer from 2 to 5).


Preferably, the peptide linker of the present invention may be GGGGS, (GGGGS)3, (GGGGS)n (n=1, 2, 4), (Gly)6, (Gly)S, AEAAAKEAAAKA, A(EAAAK)4ALEA(EAAAK)4A, (EAAAK)3, EAAAK, (EAAAK)2, (Ala -Pro)n (n is 5 to 17, length from 10 to 34 aa), PAPAP, and may be a peptide linker consisting of an amino acid sequence selected from SEQ ID NO: 10, SEQ ID NO: 34, and SEQ ID NO: 53 to SEQ ID NO: 76.


In the present invention, the peptide linker may be suitably constructed by overlapping the above-exemplified peptide linkers or in combination with other linkers, if necessary.


For heterodimerization of Fc in the present invention, Fc heterodimerization techniques known in the art may be used. Fc heterodimerization technology is shown in Table 1 below.











TABLE 1





strategy
CH3 domain 1
CH3 domain 2







knobs-into-holes (Y-T)
T366Y
Y407T


knobs-into-holes (CW-CSAV)
S354C, T366W
Y349C, T366S, L368A,




Y407V


HA-TF
S364H, F405A
Y349T, T394F


ZW1 (VYAV-VLLW)
T350V, L351Y, F405A,
T350V, T366L, K392L,



Y407V
T394W


CH3 charge pairs (DD-KK)
K392D, K409D
E356K, D399K


IgG1 hinge/CH3 charge pairs
IgG1: D221E, P228E, L368E
IgG1: D221R, P228R, K409R


(EEE-RRR)


IgG2 hinge/CH3 charge pairs
IgG2: C223E, P228E, L368E
IgG2: C223R, E225R,


(EEE-RRRR)

P228R, K409R


EW-RVT
K360E, K409W,
Q347R, D399V, F405T


EW-RVTS-S
K360E, K409W, Y349C
Q347R, D399V, F405T,




S354C


Biclonic
366K (+351K)
351D or E or D at 349, 368,




349, or 349 + 355


DuoBody (L-R)
F405L
K409R


SEEDbody
IgG/A chimera
IgG/A chimera


BEAT
residues from TCRα interface
residues from TCRβ interface


7.8.60 (DMA-RRVV)
K360D, D399M, Y407A
E345R, Q347R, T366V,




K409V


20.8.34 (SYMV-GDQA)
Y349S, K370Y, T366M,
E356G, E357D, S364Q,



K409V
Y407A









The present invention also provides a polypeptide comprising the amino acid sequence of SEQ ID NO: 4 or SEQ ID NO: 6, respectively or all of the polypeptide.


Also, the present invention provides


(a) a monomer comprising an interferon receptor 1 fragment and an antibody Fc fragment represented by the amino acid sequence of SEQ ID NO: 4;


(b) a dimeric polypeptide comprising a monomer comprising an interferon receptor 2 fragment and antibody Fc fragment represented by the amino acid sequence of SEQ ID NO: 6.


The monomer (a) comprises an interferon receptor 1 fragment and an antibody Fc fragment represented by the amino acid sequence of SEQ ID NO: 4, preferably, it may consist of or comprise an amino acid sequence selected from the group consisting of SEQ ID NO: 22, SEQ ID NO: 24, SEQ ID NO: 26, SEQ ID NO: 42, SEQ ID NO: 44 and SEQ ID NO: 46.


The monomer (b) comprises an interferon receptor 2 fragment and an antibody Fc fragment represented by the amino acid sequence of SEQ ID NO: 6, preferably, it may consist of or comprise an amino acid sequence selected from the group consisting of SEQ ID NO: 28, SEQ ID NO: 30, SEQ ID NO: 32, SEQ ID NO: 48, SEQ ID NO: 50 and SEQ ID NO: 52.


The polypeptide of the present invention may preferably be an antibody that mediates type 1 interferon.


The present invention also provides a polynucleotide encoding the polypeptide.


(i) In the present invention, ‘polynucleotide’ or ‘nucleic acid’ refers to deoxyribonucleic acid (DNA) or ribonucleic acid (RNA) in the form of single or double strands. Unless otherwise limited, known analogs of natural nucleotides that hybridize to nucleic acids in a manner analogous to naturally occurring nucleotides are also included.


A monomer comprising an interferon receptor 1 fragment and an antibody Fc fragment represented by the amino acid sequence of SEQ ID NO: 4, preferably, it may consist of or comprise a DNA sequence selected from the group consisting of SEQ ID NO: 21, SEQ ID NO: 23, SEQ ID NO: 25, SEQ ID NO: 41, SEQ ID NO: 43 and SEQ ID NO: 45.


A monomer comprising an interferon receptor 2 fragment and an antibody Fc fragment represented by the amino acid sequence of SEQ ID NO: 6, preferably, it may consist of or comprise a DNA sequence selected from the group consisting of SEQ ID NO: 27, SEQ ID NO: 29, SEQ ID NO: 31, SEQ ID NO: 47, SEQ ID NO: 49, and SEQ ID NO: 51.


The polynucleotide may be used without limitation as long as it encodes the polypeptide of the present invention, and includes all of DNA, cDNA and RNA sequences. It refers to a polynucleotide encoding a peptide having an amino acid sequence represented by the sequence or having an amino acid sequence having at least 70% homology with the amino acid sequence, it can be isolated from nature or prepared by genetic engineering methods known in the art.


The present invention also provides a vector comprising the polynucleotide.


The vector refers to an expression vector prepared by those skilled in the art to express the polypeptide of the present invention by inserting the polynucleotide of the present invention into the vector according to any method known in the art and using appropriate transcription/translation control sequences.


The polynucleotide sequence cloned according to the present invention may be operably linked to an appropriate expression control sequence, and the operably linked gene sequence and expression control sequence may be included in one expression vector including a selection marker and a replication origin. ‘Operably linked’ is meant that the polynucleotide sequence is linked to expression control sequences in such a way as to enable gene expression. The ‘expression control sequence’ refers to a DNA sequence that controls the expression of an operably linked polynucleotide sequence in a specific host cell. Such regulatory sequences may comprise one or more selected from the group consisting of a promoter for effecting transcription, an optional operator sequence for regulating transcription, sequences encoding suitable mRNA liposome binding sites, and sequences controlling the termination of transcription and translation and the like.


The vector used as the parent vector of the expression vector is not particularly limited, all plasmids, viruses or other mediators commonly used for expression in microorganisms used as host cells in the art to which the present invention pertains can be used. For example, the plasmid includes E. coli-derived plasmids (pBR322, pBR325, pUC118 and pUC119, pET-22b(+)), Bacillus subtilis-derived plasmids (pUB110 and pTP5) and yeast-derived plasmids (YEp13, YEp24 and YCp50) and the like, the virus may be used by an animal virus such as retrovirus, adenovirus or vaccinia virus, insect viruses such as baculoviruses and the like, but is not limited thereto.


The present invention also provides a host cell transformed with the vector.


The host cell may choose to modulate the expression of the inserted sequence or to process the gene product in the particular manner desired. Different host cells have characteristic and specific mechanisms for translation and post-translational processing and modification of proteins. Suitable cell lines or host systems can be selected that provide for the desired modification and processing of the expressed heterologous protein. Expression in yeast can produce biologically active products. Expression in eukaryotic cells may increase the likelihood of “native” folding.


As a host cell capable of stably and continuously cloning and expressing the vector of the present invention may be used in any host cell known in the art, for example, E. coli JM109, E. coli BL21DE, E. coli DHS, E. coli RR1, E. coli LE392, E. coli B, E. coli X 1776, E. coli W3110 and the like may be used, in addition, strains of the genus Agrobacterium, such as Agrobacterium A4, Bacilli, such as Bacillus subtilis, another intestinal Bacteria and various strains of the genus Pseudomonas such as Salmonella typhimurium, or Serratia marcescens can be used as host cells.


In addition, when the vector of the present invention is transfected into eukaryotic cells, as host cells, yeast (Saccharomyces cerevisiae), insect cells and human cells (eg, CHO cell lines (Chinese hamster ovary), Expi293, W138, BHK, COS-7, 293, HepG2, 3T3, RIN and MDCK cell lines) and the like can be used.


In the present invention, the host cell may preferably be Expi293 or CHO cell line.


The method for transfecting the host cell by delivering the vector into the host cell may be any known method and is not particularly limited. For example, transfection can be performed by gene bombardment, polycation, and receptor-mediated transfection using calcium phosphate precipitation, DEAE-dextran method, electroporation method, direct microinjection method, DNA-loaded liposome method, lipofectamine-DNA complex method, cell sonication method (cell sonication), high velocity microprojectile. Some of these techniques can be improved for in vivo or ex vivo use.


The present invention also provides a method for producing a dimeric polypeptide comprising the step of (a) providing a host cell, (b) culturing the provided cell, and (c) preparing the polypeptide of the present invention by recovering the polypeptide from the cell or culture medium.


Culturing of the transformed cells is carried out under appropriate conditions allowing expression of the fusion protein (or fusion polypeptide), these conditions can be carried out according to methods well known to those skilled in the art. Transformed cells can be cultured in large quantities by conventional culture methods. As the culture medium, a medium composed of a carbon source, a nitrogen source, vitamins and minerals may be used, for example, 2XYT medium may be used. Cell culture is possible under normal cell culture conditions, for example, it can be cultured for 10 hours to 40 hours at a temperature range of 15° C. to 45° C. In order to remove the cells in the culture medium and recover only the culture medium, centrifugation or filtration may be performed. These steps can be performed by those skilled in the art as needed. The culture medium (filtrate) from which the cells have been removed can be refrigerated according to a conventional method and stored for a short time so as not to lose its activity.


The fusion protein expressed in the transformed cell (or transformant) may be purified in a conventional manner, for example, the fusion protein of the present invention may be purified by applying techniques such as salting out (e.g., ammonium sulfate precipitation, sodium phosphate precipitation), column chromatography and ultrafiltration such as solvent precipitation (precipitation of protein fraction using acetone, ethanol, etc.), dialysis, gel filtration, ion exchange, reverse phase column chromatography, affinity chromatography alone or in combination.


The interferon receptor of the present invention is a hyper-glycosylated protein containing a large amount of glycosylation during post-translational modification, and microbial culture may not be suitable because the corresponding sugar causes structural stability problems. Accordingly, the polypeptide of the present invention may preferably be a host cell of an animal cell.


Accordingly, the present invention provides a pharmaceutical composition for preventing or treating a type 1 interferon-mediated disease or disorder comprising a polypeptide as an active ingredient.


“Type 1 interferon-mediated disease or disorder” according to the present invention refers to a disease or disorder mediated by type 1 interferon or a disease associated with abnormal expression (e.g., overexpression or inhibition of expression) of an interferon-inducible gene, examples include, but are not limited to, systemic lupus erythematosus (SLE, J Immunol. 2014 Jun. 15; 192(12): 5459-5468.) Sjogren's Syndrome (Autoimmun Rev. 2013 March; 12(5):558-66.), Insulin Dependent Diabetes Mellitus (IDDM), Inflammatory Bowel Disease (IBD) (including Crohn's Disease, Ulcerative Colitis and Celiac Disease), Multiple Sclerosis (MS), psoriasis, autoimmune thyroiditis, rheumatoid arthritis (RA, Front Immunol. 2017; 8: 2007.), inflammatory myositis (Arthritis Rheum 2009; 60:181524.; Mol Med 2007; 13:5968.) and glomerulonephritis. Moreover, the composition of the present invention can be used for the inhibition or prevention of transplant rejection or for the treatment of graft-versus-host reaction (GVHD) or for the treatment of HIV infection/AIDS.


Meanwhile, the pharmaceutical composition according to the present invention may be provided by formulating the polypeptide in a pure form or in a suitable form together with a pharmaceutically acceptable carrier. ‘Pharmaceutically acceptable’ refers to a non-toxic composition that is physiologically acceptable and does not normally cause allergic reactions such as gastrointestinal disorders, dizziness, or similar reactions when administered to humans. The carrier includes all kinds of solvents, dispersion media, oil-in-water or water-in-oil emulsions, aqueous compositions, liposomes, microbeads and microsomes.


Meanwhile, the pharmaceutical composition according to the present invention may be formulated with a suitable carrier depending on the route of administration. The route of administration of the pharmaceutical composition according to the present invention is not limited thereto, but may be administered orally or parenterally. Parenteral routes of administration include multiple routes such as, for example, transdermal, nasal, intraperitoneal, intramuscular, subcutaneous, or intravenous.


When the pharmaceutical composition of the present invention is orally administered, the pharmaceutical composition of the present invention may be formulated in the form of powder, granules, tablets, pills, dragees, capsules, liquids, gels, syrups, suspensions, wafers and the like according to a method known in the art together with a suitable carrier for oral administration. Examples of suitable carriers may be included in sugars including lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol and maltitol and the like, and starches including corn starch, wheat starch, rice starch and potato starch and the like, celluloses including cellulose, methyl cellulose, sodium carboxymethylcellulose and hydroxypropylmethyl-cellulose and the like, and the like, fillers such as gelatin, polyvinylpyrrolidone, and the like. In addition, cross-linked polyvinylpyrrolidone, agar, alginic acid or sodium alginate may be added as a disintegrant if necessary. Furthermore, the pharmaceutical composition may further include an anti-aggregating agent, a lubricant, a wetting agent, a flavoring agent, an emulsifying agent, and a preservative and the like.


In addition, when administered parenterally, the pharmaceutical composition of the present invention may be formulated according to methods known in the art in the form of injections, transdermal administrations and nasal inhalants together with suitable parenteral carriers. In the case of the injection, it must be sterilized and protected from contamination of microorganisms such as bacteria and fungi. For injection, examples of suitable carriers may include, but are not limited to, water, ethanol, polyol (e.g., glycerol, propylene glycol and liquid polyethylene glycol, etc.), mixtures thereof and/or a solvent or dispersion medium containing vegetable oil. More preferably, as a suitable carrier, Hanks' solution, Ringer's solution, an isotonic solution such as phosphate buffered saline (PBS) or sterile water for injection, 10% ethanol, 40% propylene glycol and 5% dextrose with triethanolamine, and the like can be used. In order to protect the injection from microbial contamination, it may further include various antibacterial and antifungal agents such as parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In addition, in most cases, the injection may further contain an isotonic agent such as sugar or sodium chloride.


In the case of transdermal administration, forms such as ointment, cream, lotion, gel, external solution, pasta, liniment, and air are included. As used herein, “transdermal administration” means that an effective amount of the active ingredient contained in the pharmaceutical composition is delivered into the skin by topically administering the pharmaceutical composition to the skin. These formulations are described in formularies commonly known in pharmaceutical chemistry.


For administration by inhalation, the compounds for use according to the invention may be conveniently delivered in the form of an aerosol spray from a pressurized pack or nebulizer using a suitable propellant, for example dichlorofluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol, the dosage unit may be determined by providing a valve to deliver a metered amount. For example, gelatin capsules and cartridges used in inhalers or insufflators may be formulated to contain a powder mixture of the compound and a suitable powder base such as lactose or starch. As other pharmaceutically acceptable carriers, those known in the art may be referred to.


In addition, the pharmaceutical composition according to the present invention may further comprise one or more buffers (e.g., saline or PBS), Carbohydrate (e.g., glucose, mannose, sucrose or dextran), stabilizer (sodium bisulfite, sodium sulfite or ascorbic acid) antioxidant, bacteriostatic, chelating agent (e.g., EDTA or glutathione), adjuvants (e.g., aluminum hydroxide), suspending agents, thickening agents and/or preservatives (benzalkonium chloride, methyl- or propyl-paraben and chlorobutanol).


In addition, the pharmaceutical compositions of the present invention may be formulated using methods known in the art to provide rapid, sustained or delayed release of the active ingredient after administration to a mammal.


The pharmaceutical composition formulated in the above manner may be administered in an effective amount through various routes including oral, transdermal, subcutaneous, intravenous or intramuscular. As used herein, the term ‘effective amount’ refers to an amount of a compound or extract that enables tracking of diagnostic or therapeutic effects when administered to a patient. The dosage of the pharmaceutical composition according to the present invention may be appropriately selected according to the route of administration, the administration target, the target disease and its severity, age, sex, body weight, individual differences and disease state. Preferably, the pharmaceutical composition comprising the polypeptide of the present invention may vary the content of the active ingredient depending on the severity of the disease, but, in general, based on an adult, it may be repeatedly administered several times a day at an effective dose of 10 μg to 10 mg when administered once.


In addition, the present invention provides a vaccine comprising a polypeptide as an active ingredient, and the vaccine according to the present invention is a vaccine for preventing or treating a type 1 interferon-mediated disease or disorder.


As used herein, the term “vaccine” refers to an immunogen or antigenic substance that induces immunity in a living body by injecting or orally administering to a person or animal for the prevention of infection as a biological agent containing an antigen that gives immunity to a living body. In vivo immunity is largely divided into automatic immunity, which is obtained automatically after infection with a pathogen, and passive immunity, which is obtained by an externally injected vaccine. While autoimmunity is characterized by a long period of production of antibodies related to immunity and continuous immunity, passive immunity by vaccine works immediately for the treatment of infection, but has the disadvantage of poor durability.


The vaccine composition of the present invention may include a pharmaceutically acceptable carrier. It refers to any component suitable for delivery of an antigenic material to a site in vivo, examples include, but are not limited to, water, saline, phosphate buffered saline, Ringer's solution, dextrose solution, serum-containing solution, Hans' solution, other aqueous physiologically equilibrated solutions, oils, esters and glycols, and the like.


The carrier of the present invention may contain suitable auxiliary ingredients and preservatives to enhance chemical stability and isotonicity, stabilizers such as trehalose, glycine, sorbitol, lactose or monosodium glutamate (MSG) may be included to protect the vaccine composition against temperature changes or lyophilization. The vaccine composition of the present invention may contain a suspension liquid such as sterile water or saline (preferably buffered saline).


The vaccine composition of the present invention may contain any adjuvant in an amount sufficient to enhance the immune response to the immunogen. Suitable adjuvants are known in the art, examples include, but are not limited to, aluminum salts (aluminum phosphate or aluminum hydroxide), Squalene mixture (SAF-1), muramyl peptide, saponin derivative, mycobacterium cell wall preparation, monophosphoryl lipid A, mycolic acid derivative, Nonionic block copolymer surfactants, Quil A, cholera toxin B subunit, polyphosphazene and derivatives, and immun-stimulating complexes (ISCOMs).


As with all other vaccine compositions, the immunologically effective amount of the immunogen must be determined empirically, in which case factors that may be considered include immunogenicity, route of administration, and frequency of administration of the immune system. In addition, it can be adjusted according to the patient's disease progression and metastasis status, the type of formulation, the patient's age, sex, weight, health status, diet, administration time, and administration method.


The dimeric polypeptide in the vaccine composition of the present invention may be present in various concentrations in the composition of the present invention, but, typically, the antigen material is included in a concentration necessary to induce the formation of an appropriate level of antibody in vivo.


As used herein, the term “administration” means introducing a predetermined substance into a patient by any suitable method, and the administration route of the vaccine of the present invention may be administered through any general route as long as they can reach the target tissue.


The vaccine composition of the present invention can be used to treat type 1 interferon-mediated diseases or disorders by administration via the systemic or mucosal route.


Administration of the vaccine composition may include, but is not limited to, injection via an intramuscular, intraperitoneal, intradermal or subcutaneous route, oral/meal, respiratory, and mucosal administration to the genitourinary tract.


In addition, in order to increase the efficacy of the vaccine, when the vaccine is injected, cytokines that help activate T cells, such as IL-12, are co-administered, or a vaccine transfected with these cytokine genes may be used.


Cells containing the dimer-type polypeptide, which is an active ingredient of the vaccine produced by the present invention, can also be depleted of cell proliferation in order to increase safety, because the human body is inoculated as a therapeutic vaccine. For example, in order to be selectively used as a cell vaccine more safely, it can be treated with heat treatment, radiation treatment, or mitomycin C (MMC) treatment, and the proliferation can be eliminated, while leaving the function as a vaccine. For example, when using X-ray irradiation, it can be irradiated with a total radiation dose of 1000 to 3300 Rad. In the mitomycin C treatment method, for example, 25 to 50 μg/ml of mitomycin C is added to the polypeptide, and heat treatment can be performed at 37° C. for 30 minutes to 60 minutes. In the cell treatment method by heat, for example, heat treatment can be performed at 50° C. to 65° C. for 20 minutes.


As used herein, ‘treatment’ refers to a clinical procedure intended to alter the natural process of a subject or cell to be treated, it can also be performed for the prevention of clinical pathology. Desirable effects of treatment include suppression of occurrence or recurrence of disease, alleviation of symptoms, reduction of any direct or indirect pathological consequences of disease, reducing the rate of disease progression, ameliorating, improving, alleviating, or improving prognosis and the like. Also, the term ‘prevention’ refers to any action that suppresses the onset or delays the progression of a disease.


As used herein, the term ‘comprising’ is used with the same meaning as ‘including’ or ‘characterized by’, in the composition or method according to the present invention, additional components or steps of the method not specifically mentioned are not excluded. In addition, the term ‘consisting of’ means excluding additional elements, steps, or ingredients that are not separately described. The term ‘essentially consisting of’ means that, in the scope of the composition or method, it may include substances or steps that do not substantially affect the basic properties thereof in addition to the substances or steps described.


The present invention also provides a use of the polypeptide for the preparation of an agent for preventing or treating a type 1 interferon-mediated disease or disorder.


In addition, the present invention provides a method for treating a type 1 interferon-mediated disease or disorder, comprising administering to a subject in need thereof an effective amount of a composition comprising the polypeptide as an active ingredient.


The ‘effective amount’ of the present invention refers to an amount that, when administered to an individual, exhibits an effect of improving, treating, preventing, detecting, diagnosing, or inhibiting or reducing a type 1 interferon-mediated disease or disorder, or a type 1 interferon-mediated disease or disorder, and the ‘subject’ may be an animal, preferably an animal, including a mammal, particularly a human, and a cell, tissue, organ, or the like derived from an animal. The subject may be a patient in need of the effect.


The ‘treatment’ of the present invention refers generically to ameliorating a type 1 interferon mediated disease or disorder, or a symptom of a type 1 interferon mediated disease or disorder, and this may include curing, substantially preventing, or ameliorating the condition, and including alleviating, curing or preventing one or most of the symptoms resulting from the disease, but is not limited thereto.


Advantageous Effect

The type 1 interferon-neutralizing Fc-fusion protein of the present invention blocks the binding of type 1 interferon to the interferon receptor, and has excellent initiation and biological activity inhibition of signaling mechanisms, thereby effectively treating diseases mediated by type 1 interferon.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 shows a schematic diagram of the preparation of an expression vector for the production of type 1 interferon Fc fusion receptor protein.



FIG. 2a is a result of comparing the size of each protein through size exclusion chromatography.



FIG. 2b is a result confirming the binding of IFNAR1/2 hetero Fc fusion protein and human IFN 13 under native conditions and reducing conditions.



FIG. 2c is a schematic diagram showing the type 1 interferon Fc fusion receptor protein.



FIG. 3 is a diagram showing the results of SPR analysis of the type 1 interferon Fc fusion receptor protein.



FIGS. 4a to 4c are results confirming the type 1 interferon neutralizing ability of the type 1 interferon Fc fusion receptor protein of the present invention.



FIG. 4a is a result showing the cell viability in Daudi cells treated with human IFN β-1a alone or type 1 interferon Fc fusion receptor protein.



FIG. 4b is a result showing the IC50 value treated with human IFN β-1a alone or type 1 interferon Fc fusion receptor protein.



FIG. 4c is a result showing the cell viability of Daudi cells treated with human IFN β-1a according to the concentration of the protein of the present invention.



FIG. 5 confirms the neutralizing ability of the IFNAR1/2-Fc heterodimer (4GS*3) protein of the present invention for the biological activity of type 1 interferon (IFN-α 1, IFN-α 2a, IFN-α 2b, IFN-α 5, IFN-α 8, IFN-α 10, IFN-β 1a, IFN-ω), and the cell viability of Daudi cells in the group to which type 1 interferon was added and the group in which each interferon was treated with IFNAR1/2-Fc heterodimer (4GS*3) protein was shown.



FIGS. 6a and 6b are the results of confirming the neutralizing ability of the IFNAR1/2-Fc heterodimer (4GS*3) protein of the present invention for the signaling mechanism of type 1 interferon, and the results show the phosphorylation change of STAT1 protein in Daudi cells of the group to which type 1 interferon (IFN-α 1, IFN-α 2a, IFN-α 2b, IFN-α 5, IFN-α 8, IFN-α 10, IFN-β 1a, IFN-ω, IFN-ε) was added and the group in which each interferon was treated with IFNAR1/2-Fc heterodimer (4GS*3) protein.





MODE FOR CARRYING OUT INVENTION

Hereinafter, the present invention will be described in detail.


However, the following examples are only illustrative of the present invention, and the content of the present invention is not limited to the following examples.


Example 1: Preparation of Expression Vector for Production of Type 1 Interferon Fc Fusion Receptor Protein

Human IFNAR1 (P17181, 28-436 aa.), IFNAR2 (P48552, 27-243 aa.) and IGHG (P01857, 100-330 aa) amino acid sequences were used to develop an Fc fusion protein capable of binding or neutralizing type 1 interferon protein. As shown in FIG. 1 (left), Fc were labeled with a polypeptide linker (L; IEGRMD) at the C-terminus of the expression vector in all Fc fusion protein expression vectors, it is linked by IFNAR1 or IFNAR2 between the C-terminus and the N-terminus including the Kozac sequence (1) and the extracellular secretion inducing signal sequence (S), or directly linked.


As shown in FIG. 1 (right), the sequence including each gene was linked between restriction enzymes NheI and XhoI. Expression vectors were named pIFNAR1-FcWT, pIFNAR2-FcWT, pIFNAR1-FcA, pIFNAR2-FcB, pIFNAR1-FcB and pLeader-FcA, respectively.


Codon optimization of the gene comprising the polynucleotide sequence (SEQ ID NO: 1), the extracellular domain of IFNAR1 (SEQ ID NO: 3), the extracellular domain of IFNAR2 (SEQ ID NO: 5), a polypeptide linker (SEQ ID NO: 7), and the Fc portion of human IGHG (SEQ ID NO: 11) and variants (SEQ ID NO: 13, SEQ ID NO: 15) comprising the Kozac sequence and the signal sequence was performed to synthesize the gene to prepare the nucleotide sequence fragments e to g of FIG. 1. An expression vector was prepared using T4 DNA ligase (RBC) between NheI and Xho I among the restriction enzyme sites of the expression vector (pOptiVec-TOPO) for each nucleotide sequence fragment.


In addition, the expression vector was prepared in the same manner except that the linker of SEQ ID NO: 33 was used instead of the polypeptide linker of SEQ ID NO: 7. In other words, the gene comprising the polynucleotide sequence (SEQ ID NO: 1), the extracellular domain of IFNAR1 (SEQ ID NO: 3), the extracellular domain of IFNAR2 (SEQ ID NO: 5), a polypeptide linker (SEQ ID NO: 33), and the Fc portion of human IGHG (SEQ ID NO: 11) and variants (SEQ ID NO: 13, SEQ ID NO: 13) number 15) comprising the Kozac sequence and the signal sequence was codon-optimized to synthesize the gene to prepare the nucleotide sequence fragments e to g of FIG. 1. An expression vector was prepared using T4 DNA ligase (RBC) between NheI and Xho I among the restriction enzyme sites of the expression vector (pOptiVec-TOPO) for each nucleotide sequence fragment.


Example 2: Protein Expression Purification, Structure Confirmation and Binding Confirmation

The following expression and purification processes were performed in order to produce a protein having the structure shown in FIG. 2c using expression vectors into which the nucleotide sequence fragments of each combination were inserted.


In order to maintain the native post-translational modification (PTM) of each type 1 interferon receptor, all recombinant protein expression use Expi293TM expression medium in 300-600 mL according to the protocol of the transient expression system, using the human-derived expression cell, Expi293 cell line (Thermo Fisher Scientific). Depending on the final protein, one or two expression vectors were transfected into cells, and then Enhancers 1 and 2 were added according to the manufacturer's protocol. Thereafter, the culture medium cultured for 6 days was centrifuged at 6580 rpm, 20 minutes, and 8° C., and then filtered using a 0.22 μm polytyrene filter (Corning). All proteins were purified in the AKTA avant purification system (GE healthcare Life Sciences), and the column filled with MabSelect SuRe (GE healthcare Life Science) was equilibrated as much as 2 CV using 20 mM sodium phosphate, pH 7.2 and 150 mM NaCl buffer at a rate of 5 ml/min, and then the culture solution was flowed to bind to the resin.


Thereafter, the recombinant protein was eluted with 100 mM citrate buffer, pH 3.5 buffer after a column wash process using 5 CV of 35 mM sodium phosphate, pH 7.2, 500 mM NaCl buffer and 1 CV of 20 mM sodium phosphate, pH 7.2 buffer. The eluted protein was concentrated to 3 ml using an Amicon Ultra Centrifugal filter (50K-cut off, Merck Millipore) after dialysis for 12 hours and 3 times using 4L of 1×PBS buffer (Biosesang). The concentrated recombinant protein was additionally separated from the target protein using the same buffer used for dialysis on a Hiload Superdex 200 pg (GE healthcare Life Sciences) column.


As a result, as shown in FIG. 2a, the size of each Fc fusion protein was compared through size exclusion chromatography, and it was expected to have the protein structure shown in FIG. 2c.


In addition, as shown in FIG. 2b, binding between the IFNAR1/2 hetero Fc fusion protein and human IFN β-1a was confirmed, respectively in native conditions (7.5% Mini-PROTEIN® TGXTM Precase protein gels), reducing conditions (Any kDTM Mini-PROTEIN® TGXTM Precast protein gel, Bio-rad)


The FC portion of the amino acid sequence of the present invention includes the HINGE portion of IGHG1 (100-330a.a.), and a disulfide bond is formed by amino acids 109-109 and 112-112 to form a dimer. In more detail, the formation of a heterodimer occurs dominantly from a homodimer to a heterodimer due to a change in the specific amino acid sequence of the CH3 portion of FC.


Example 3: SPR Analysis Using Fc Fusion Protein

Binding affinity and kinetics of Fc fusion proteins (IFNAR2-Fc heterodimer, IFNAR1/2-Fc heterodimer, IFNAR1-Fc heterodimer) expressed/purified through the expression vector combination of FIG. 1 and hIFN β-1a among Type I IFNs were analyzed.


In this experiment, (IFNAR1-Fc EW+-Fc RVT), (IFNAR2-Fc EW+-Fc RVT) and (IFNAR1-Fc EW+IFNAR2Fc RVT) were used to analyze binding strength and kinetics. When expressing (IFNAR1-Fc RVT+IFNAR2-Fc EW), as it was confirmed that the problem that the homodimer form appeared more than (IFNAR1-Fc EW+IFNAR2-Fc RVT) was confirmed, so (IFNAR1-Fc RVT+IFNAR2-Fc EW) was not used.


Based on Biacore T200 (GE Healthcare Life Sciences), 25° C., 30 μl/min conditions and 0.005% DPBST (DPBS/modified, Hyclone and Tween20, Signal) running buffer were used, and the gold sensor chip was amine-coupled with an anti-human Fcγ capture antibody (AffiniPure Goat Anti-Human IgG, Fcγ fragment specific, Jackson ImmunoResearch). Each analyte was tested under 180 seconds, association and 600 seconds, and dissociation conditions.


As shown in FIG. 3, the curve fitting was performed according to hIFNb-1a-IFNAR2 (1:1 fitting model), hIFNb-1a-IFNAR1/2 (heterogeneous ligand model), hIFNb-1a-IFNAR1 (two-state binding models) sensorgram shape and the docking model prediction. In particular, the IFNAR1/2 hetero Fc fusion protein showed a low KD value. (See Table 2 and Table 3)
















TABLE 2







k1a × 105
k1d × 10−3
K1D
k2a × 107
k2d × 10−3
K2D


Ligand
Analyte
(M−1 s−1)
(s−1)
(nM)
(M−1 s−1)
(s−1)
(pM)






















IFNAR2-Fc
IFN-β-1aa
75.45
3.556
0.47





heterodimer
IFN-β-1ab


4.26





IFNAR1/2-Fc
IFN-β-1ac
76.85
0.0044
0.0006
1.57
3.109
198.6


heterodimer
IFN-β-1ab


4.775





IFNAR1-Fc
IFN-β-1aa
0.2368
0.9723
41.11





heterodimer
IFN-β-1ab


81.51






aSensorgrams fit with one to one binding model (A + B  custom-character   AB)




bthe steady state equilibrium analysis were estimated using each parameter at 179.04 sec.




cSensorgrams fit with heterogeneous ligand model (A + B  custom-character   AB + C  custom-character   ABC)




















TABLE 3







k1a × 105
k1d × 10−3
k2a × 10−3
k2d × 10−3
K2D


Ligand
Analyte
(M−1 s−1)
(s−1)
(s−1)
(s−1)
(nM)





















IFNAR1-Fc heterodimer
IFN-β-1aa
5.426
129.7
1.776
45.75
48.97


IFNAR2-Fc heterodimer
IFN-β-1aa
2.23E+06
8.81E+05
22.04
3.444
0.5342






aSensorgrams fit with two-state binding model (A + B  custom-character   AB  custom-character   AB*)







Example 4: Confirmation of Biological Activity Neutralizing Ability by Ligand of Fc Fusion Protein

The following experiment was conducted to confirm the neutralizing ability of the Fc fusion protein to the biological activity caused by the ligand. As in Example 3, (IFNAR1-Fc EW+-Fc RVT), (IFNAR2-Fc EW+-Fc RVT) and (IFNAR1-Fc EW+IFNAR2Fc RVT) were used in this experiment.


All experiments were designed based on the anti-proliferative effect of hIFN-β using the Ez-cytox cell viability assay kit (Deaillab) in Daudi cells, where the expression of each receptor is high. This test method showed test results that well reflect the biophysical characteristics of the ligand, similar to the results of the kinetics analysis of SPR.


Specifically, in a 96-well plate (SPL), the concentration of hIFN-β-1a in cells of 3×103 cells was determined by concentration (10 nM of IFNAR1-Fc heterodimer, IFNAR2-Fc heterodimer, IFNAR1/2-Fc heterodimer; 6 pM of IFNAR1-Fc heterodimer; IFNAR1/2-Fc heterodimer), and then the reaction was carried out in a cell incubator at 37° C. and 5% CO2 for 72 hours. Then, after adding the Ex-cytox reagent according to the manufacturer's protocol, after an additional 3 hours of reaction, it was measured at 450 nm with a Microplate reader (Genios Pro, Tecan), and IC50 values were compared and analyzed using nonlinear regression analysis (GraphPad Prism version 7.0 software, san diego, Ca, USA). The neutralizing ability of each protein of the present invention was confirmed for statistical significance through One-way ANOVA, Bonferroni's multiple comparisons post hoc test.


As shown in FIG. 4a, it was confirmed that anti-cell proliferation by interferon was significantly reduced as compared to the case of a fusion protein that blocks IFNAR1 and IFNAR2, respectively, when interferon is treated with an Fc fusion protein that blocks both IFNAR1 and IFNAR2 binding.


As shown in FIG. 4b, the IC50 value was the highest when the Fc fusion protein blocking both IFNAR1 and IFNAR2 according to the present invention was added. Therefore, it was confirmed that anti-cell proliferation by interferon was superior to cell viability when blocking IFNAR1 or IFNAR2, respectively.


As shown in FIG. 4c, it was confirmed that the cell viability increased in a concentration-dependent manner when the IFNAR1/2-Fc heterodimer with the highest neutralizing ability was treated.


Example 5: Confirmation of Signal Mechanism Activity Neutralizing Ability by Ligand of IFNAR1/2-Fc Heterodimer

In the same manner as in Example 4, except for the IFNAR1/2-Fc heterodimer mentioned below, the ability to neutralize the biological activity by the ligand of the IFNAR1/2-Fc heterodimer was confirmed.


Specifically, the group added IFNs (IFN-α 1 (pbl assay science_cat #11125-1), IFN-α 2a (pbl assay science_cat #11100-1), IFN-α 2b (pbl assay science_cat #11105-1), IFN-α 5 (pbl assay science_cat #11135-1), IFN-α 8 (pbl assay science_cat #11115-1), IFN-α 10 (pbl assay science_cat #11120-1), IFN-β 1a (pbl assay science_cat #11415-1), IFN-ω (pbl assay science_cat #11395) -One) at a concentration of 1 nM and the group that each interferon was treated with a type 1 interferon Fc fusion receptor protein at a concentration of 10 nm in cells of 3×103 cells were reacted in a cell incubator at 37° C. and 5% CO2 for 72 hours. Then, after adding the Ex-cytox reagent according to the manufacturer's protocol, and after an additional 2 hours of reaction, it was measured at 450 nm with a Microplate reader (Genios Pro, Tecan), and IC50 values were compared and analyzed using nonlinear regression analysis (GraphPad Prism version 7.0 software, san diego, Ca, USA).


As a result, as shown in FIG. 5, it was confirmed that the cell viability was significantly superior as compared with the cell viability treated only with interferon, when IFN-α 1, IFN-α 2a, IFN-α 2b, IFN-α 5, IFN-α 8, IFN-α 10, IFN-β 1a, IFN-ω was treated with an IFNAR1/2-Fc heterodimer (using 4GS*3 as a linker) that blocks both the binding of IFNAR1 and IFNAR2.


Through these results, it was found that the regulation of excessive interferon signals for cells is possible by the dimer-type polypeptide to which the monomer (monomer) comprising the interferon receptor fragment or antibody Fc fragment of the present invention is bound, and through this, it was confirmed that it would show excellent pharmacological effects on patients with type 1 interferon-mediated diseases such as systemic lupus erythematosus, Sjogren's syndrome, systemic sclerosis, myositis and rheumatoid arthritis in which interferon-inducible genes expressed in response to excessive interferon signals are high.


Example 6: Confirmation of Chemical Signal Inhibition Ability by Ligand of IFNAR1/2-Fc Heterodimer

All experiments were performed to confirm changes in STAT1 and its phosphorylated pSTAT1 protein by Type I IFNs using Western blot in Daudi cells, in which the expression of each receptor is high.


Specifically, in a 6-well plate (SPL_cat #30006), the group added IFNs (IFN-α 1 (pbl assay science_cat #11125-1), IFN-α 2a (pbl assay science_cat #11100-1), IFN-α 2b (pbl assay science_cat #11105-1), IFN-α 5 (phi assay science_cat #11135-1), IFN-α 8 (01 assay science_cat #11115-1), IFN-α 10 (phi assay science_cat #11120-1), IFN-β 1a (phi assay science_cat #11415-1), IFN-ω (phi assay science_cat #11395-) 1), IFN-ε (R&D systems_cat #9667-ME) at a concentration of 1 nM and the group that each interferon was treated with IFNAR1/2-Fc heterodimer protein at a concentration of 10 nM in the cells of 2×106 cells were reacted in a cell incubator at 37° C. and 5% CO2 for 72 hours. Thereafter, cells were collected from the plate, total protein was extracted and determined with BCA protein assay kit (Thermo scientific_cat #23227). Total protein was separated on a 10% gel by sodium-dodecylsulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and transcribed onto a PVDF membrane (BIO RAD_cat #1620177). The membrane was blocked with 5% skim milk, 3% bovine serum albumin, 10 mmol/L Tris-HCL (pH 8.0), 150 mmol/L NaCl, and 0.05% Tween-20 for 1 hour at room temperature. The blocked membrane was treated with STAT1 antibody (cell signaling_cat #06-501), pSTAT1 antibody (cell signaling_cat #58D6), and beta actin antibody (santa cruz_cat #sc 47778) as primary antibodies (1:3000 dilution) overnight at 4° C. Thereafter, a secondary HRP-conjugated antibody, goat anti rabbit antibody (Invitrogen_cat #31460) and goat anti mouse antibody (invitrogen_cat #G21040), was treated at room temperature for 1 hour. After treating the membrane with ECL solution (BIO RAD_cat #1705061), it was visualized on a film (Agfa healthcare_cat #EA8EC) using a developer and fixer from poohung.


As a result, as shown in FIGS. 6a and 6b, it was confirmed that phosphorylation of the pSTAT1 protein was significantly reduced as compared with the high phosphorylation of pSTAT1 protein when only interferon was treated, when the IFNAR1/2-Fc heterodimer (4GS*3) protein, which blocks both the binding of IFNAR1 and IFNAR2, was treated with interferon (IFN-α 1, IFN-α 2a, IFN-α 2b, IFN-α 5, IFN-α 8, IFN-α 10, IFN-β 1a, IFN-ω, IFN-ε). This indicates that the IFNAR1/2-Fc heterodimer protein binds to type 1 interferon and exhibits neutralizing ability, indicating that interferon-related signaling in the cell is inhibited.


INDUSTRIAL APPLICABILITY

The type 1 interferon neutralizing Fc-fusion protein of the present invention has excellent industrial applicability as while blocking the binding of type 1 interferon to the interferon receptor, has excellent initiation and biological activity inhibition of signaling mechanisms, so it can be very usefully used in the development of therapeutic agents for preventing or treating type 1 interferon-mediated diseases.

Claims
  • 1. A dimeric polypeptide in which a monomer comprising an interferon receptor fragment or an antibody Fc fragment is bound, wherein the monomer is a polypeptide selected from the group consisting of (i), (ii) and (iii):(i) a monomer comprising an interferon receptor 1 (IFNAR1) fragment and an antibody Fc fragment;(ii) a monomer comprising an interferon receptor 2 (IFNAR2) fragment and an antibody Fc fragment; and(iii) an antibody Fc fragment.
  • 2. The dimeric polypeptide according to claim 1, wherein the polypeptide neutralizes type 1 interferon.
  • 3. The dimeric polypeptide according to claim 1, wherein the polypeptide comprises (i) and (ii).
  • 4. The dimeric polypeptide according to claim 1, wherein the interferon receptor 1 fragment comprises the amino acid sequence of SEQ ID NO: 4.
  • 5. The dimeric polypeptide according to claim 1, wherein the interferon receptor 2 fragment comprises the amino acid sequence of SEQ ID NO: 6.
  • 6. The dimeric polypeptide according to claim 1, wherein the antibody Fc fragment comprises the amino acid sequence of SEQ ID NO: 12.
  • 7. A polynucleotide encoding the polypeptide of claim 1.
  • 8. A vector comprising the polynucleotide of claim 7.
  • 9. A host cell transformed with the vector of claim 8.
  • 10. A method for producing the polypeptide of claim 1 comprising: (a) providing a host cell transformed with a vector comprising a polynucleotide encoding the polypeptide of claim 1;(b) culturing the provided cells; and(c) preparing the polypeptide by recovering the polypeptide from the cell or culture medium.
  • 11. A pharmaceutical composition for prevention or treatment of type 1 interferon-mediated diseases or disorders comprising the polypeptide of claim 1 as an active ingredient.
  • 12. The pharmaceutical composition according to claim 11, wherein the type 1 interferon-mediated disease or disorder is selected from the group consisting of systemic lupus erythematosus, Sjogren's syndrome, systemic sclerosis, insulin-dependent diabetes mellitus (IDDM), inflammatory bowel disease (IBD) (including Crohn's disease, ulcerative colitis and celiac disease), multiple sclerosis (MS), psoriasis, autoimmune thyroiditis, rheumatoid arthritis, inflammatory Myositis, glomerulonephritis HIV infection, AIDS, transplant rejection and graft-versus-host reaction (GVHD).
  • 13. Use of the polypeptide of claim 1 for the preparation of an agent for the prevention or treatment of a type 1 interferon-mediated disease or disorder.
  • 14. A method of treating a type 1 interferon-mediated disease or disorder, comprising administering to a subject in need thereof an effective amount of a composition comprising the polypeptide of claim 1 as an active ingredient.
Priority Claims (1)
Number Date Country Kind
10-2018-0168801 Dec 2018 KR national
PCT Information
Filing Document Filing Date Country Kind
PCT/KR2019/018233 12/20/2019 WO 00