NOVEL HYALURONIDASE VARIANTS AND PHARMACEUTICAL COMPOSITION COMPRISING THE SAME

Information

  • Patent Application
  • 20210155913
  • Publication Number
    20210155913
  • Date Filed
    July 25, 2019
    4 years ago
  • Date Published
    May 27, 2021
    3 years ago
Abstract
The present invention is related to the field of protein engineering technology which increases the enzymatic activity and thermal stability of human hyaluronidase which is an enzyme that hydrolyzes hyaluronic acid; and more particularly to hyaluronidase PH20 variants or fragments thereof, which comprise one or more amino acid residue substitutions in the region corresponding to the alpha-helix region and its linker region in the amino acid sequence of wild-type PH20 of SEQ ID NO: 1 and in which one or more amino acid residues at the N-terminus and/or the C-terminus are selectively cleaved additionally.
Description
TECHNICAL FIELD

The present invention relates to novel human hyaluronidase variants having increased enzymatic activity and thermal stability compared to human hyaluronidase which is an enzyme that hydrolyzes hyaluronic acid, and more particularly to hyaluronidase PH20 variants or fragments thereof, which comprise one or more amino acid residue substitutions in the region corresponding to the alpha-helix region and/or its linker region in wild-type PH20 having the amino acid sequence of SEQ ID NO: 1, preferably mature wild-type PH20 consisting of amino acid residues L36 to S490, and in which one or more of the N-terminal or C-terminal amino acid residues are selectively deleted, a method for producing the same, and a pharmaceutical composition comprising the same.


BACKGROUND ART

The human skin is composed of epidermis, dermis and a subcutaneous fat layer, and there are six types of glycosaminoglycans in the skin. These glycosaminoglycans include hyaluronic acid, chondroitin sulfate, dermatan sulfate, heparan sulfate, heparin, and keratin sulfate.


These glycosaminoglycans are composed of repeating disaccharide sugar units. The number of the disaccharide sugar units is different among the glycosaminoglycans, but ranges from several hundreds to thousands. Among the glycosaminoglycans, hyaluronic acid is present in the skin more than half of the amount in the body. Hyaluronic acid is synthesized by hyaluronan synthase present in the cell membrane, is present alone without binding to proteoglycans, and is the only glycosaminoglycan having no sulfate group. Other glycosaminoglycans bind to proteoglycans and have a sulfate group. Hyaluronic acid consists of glucuronic acid and N-acetylglucosamine linked via alternating β-1,4 and β-1,3 bonds, and is composed of about 5,000 repeating units of these disaccharides. It is known that about one-third (5 g) of hyaluronic acid in the human body is turned over every day.


Hyaluronidases are enzymes that degrade hyaluronic acid present in the extracellular matrix. It is known that there are six types of hyaluronidases in humans: they are Hyal1, Hyal2, Hyal3, Hyal4, HyalPS1, and PH20/SPAM1. Human Hyal1 and Hyal2 are expressed in most of the tissues. PH20/SPAM1 (hereinafter referred to as PH20) is expressed in the sperm plasma membrane and the acrosomal membrane. However, HyalPS1 is not expressed because it is a pseudogene. Hyaluronidases are divided into, according to a method of cleaving hyaluronic acid, three types: enzymes (EC 3.2.1.35) that cleave β-1,4 bonds between N-acetylglucosamine and glucuronic acid by the use of H2O; enzymes (EC 3.2.1.36) that cleave β-1,3 bonds between N-acetylglucosamine and glucuronic acid by the use of H2O; and bacterial hyaluronidases (EC 4.2.99.1) that cleave β-1,4 bonds without using H2O.


The catalytic amino acids of Hyal1 are D129 and E131, which hydrolyze hyaluronic acid by substrate-assisted catalysis. Hyal1 exhibits the optimum activity at an acidic pH of 3 to 4, and has no enzymatic activity at pH 4.5 or higher. In contrast to Hyal1, PH20 exhibits enzymatic activity at a wide pH range of 3 to 8.


Arming et al. identified that the catalytic amino acids of PH20 are D111 and E113 (Arming et al., 1997). Arming et al. labelled Leu as the first amino acid of the mature protein, and thus the catalytic amino acids of the full-length PH20 with the signal peptide correspond to D146 and E148, respectively.


Hyaluronidase hydrolyzes hyaluronic acid, thereby reducing the viscosity of hyaluronic acid in the extracellular matrix and increasing the permeability thereof into tissue (skin). The subcutaneous area of the skin has a neutral pH of about 7.0 to 7.5. Thus, among the various types of hyaluronidases, PH20 is widely used in clinical practice (Bookbinder et al., 2006). In examples in which PH20 is used in clinical practice, PH20 is used as an eye relaxant and an anesthetic additive in ophthalmic surgery, and is also co-administered with an antibody therapeutic agent which is injected subcutaneously (Bookbinder et al., 2006). In addition, based on the property of hyaluronic acid that is overexpressed in tumor cells, PH20 is used to hydrolyze hyaluronic acid in the extracellular matrix of tumor cells, thereby increasing the access of an anticancer therapeutic agent to the tumor cells. In addition, it is also used to promote resorption of body fluids and blood, which are excessively present in tissue.


PH20 was first identified in guinea pig sperm by Lathrop et al., and is also known to be expressed in sperms of different species. Human PH20 gene was cloned by Lin et al. and Gmachl et al. Human PH20 has the amino acid sequence of SEQ ID NO: 1 which consists of 509 amino acid residues, and exhibits 60% amino acid identity with guinea pig PH20 gene. Human PH20 enzyme is encoded from SPAM1 (sperm adhesion molecule-1) gene, and Ser490 of PH20 is present as binding to the glycosylphosphatidylinositol (GPI) on the surface of the sperm plasma membrane and in the acrosomal membrane. Sperm hydrolyzes hyaluronic acid using PH20 when it penetrates oocytes through the hyaluronan-rich cumulus layer of the oocytes. PH20 is present in the amount corresponding to 1% or less of the amount of proteins in sperm, and has six N-glycosylation sites (N82, N166, N235, N254, N368, and N393).


Currently commercially available PH20 is obtained by extraction from the testes of cattle or sheep. Examples thereof include Amphadase® (bovine hyaluronidase) and Vitrase® (sheep hyaluronidase).


Bovine testicular hyaluronidase (BTH) is obtained by removing a signal peptide and 56 amino acids on the C-terminal from bovine wild-type PH20 during post-translational modification. BTH is also a glycoprotein, and has a mannose content of 5% and a glucosamine content of 2.2%, based on the total components including amino acids. When animal-derived hyaluronidase is repeatedly administered to the human body at a high dose, a neutralizing antibody can be produced. Since animal-derived hyaluronidase contains other biomaterials in addition to PH20, it may cause an allergic reaction when administered to the human body (Bookbinder et al., 2006). In particular, the production and the use of PH20 extracted from cattle can be limited due to concerns of mad cow disease. In order to overcome this problem, studies on the recombinant protein of human PH20 have been conducted.


Recombinant protein of human PH20 has been reported to be expressed in yeast (P. pastoris), DS-2 insect cells, and animal cells. The recombinant PH20 proteins produced in insect cells and yeast differ from human PH20 in terms of the pattern of N-glycosylation during post-translational modification.


Among hyaluronidases, only three dimensional structures of Hyal1 (PDB ID: 2PE4) (Chao et al., 2007) and bee venom hyaluronidase (PDB ID: 1FCQ, 1FCU, 1FCV) are determined. Hyal1 is composed of two domains, a catalytic domain and an EGF-like domain. The catalytic domain is in the form of (β/α)8 in which an alpha-helix and a beta-strand, which characterize the secondary structure of the protein, are each repeated eight times (Chao et al., 2007). The EGF-like domain is completely conserved in variants in which the C-terminus of Hyal1 is spliced differently. The amino acid sequences of Hyal1 and PH20 are 35.1% identical, and the protein structure of PH20 has not yet been found.


A recombinant protein of human PH20 was developed by Halozyme Therapeutic, Inc. and has been sold under the trade name Hylenex® (Bookbinder et al., 2006; Frost, 2007).


When D146 and E148, which are the catalytic amino acids of PH20, were mutated to asparagine (D146N) and glutamine (E148Q), respectively, there was no enzymatic activity (Arming et al., 1997). In addition, when R246 of PH20 was substituted with glycine, the enzymatic activity was reduced by 90%, and when E319 was substituted with glutamine and R322 was substituted with threonine, the enzymatic activity disappeared. A variant in which 36 amino acids at the C-terminus of PH20 were removed (474-509 amino-acid truncation) showed a 75% reduction in enzymatic activity compared to wild-type PH20. This variant was not secreted extracellularly and remained in HeLa cells. When C-terminal 134 amino acids were removed from PH20, PH20 had no enzymatic activity and was not secreted extracellularly. According to Frost et al., the C-terminal 477-483 region of PH20 is essential for soluble expression (Frost, 2007). The activity of full-length PH20 (1 to 509) or a PH20 variant having a C-terminus truncated at position 467 was merely 10% of a PH20 variant having a C-terminus truncated at one of positions 477 to 483 (Frost, 2007).


Meanwhile, recombinant PH20 still has insufficient thermal stability or expression levels in the recombinant cells. Therefore, there is a great demand in industry for a recombinant hyaluronidase having further improved biological and physico-chemical characteristics.


DISCLOSURE OF INVENTION
Technical Problem

It is an object of the present invention to provide a hyaluronidase PH20 variant or fragment thereof which is improved in thermal stability, enzyme activity and expression level, compared to wild-type PH20, preferably mature wild-type PH20.


Another object of the present invention is to provide a composition for treating cancer, comprising the above-described hyaluronidase PH20 variant or fragment thereof, and a method of treating cancer using the same.


Technical Solution

To achieve the above objects, the present invention provides a hyaluronidase PH20 variant or fragment thereof, which comprises one or more amino acid residue substitutions in the region corresponding to an alpha-helix region and/or its linker region in the amino acid sequence of wild-type PH20, preferably mature wild-type PH20, and in which one or more of the N-terminal or C-terminal amino acid residues are selectively deleted.


The present invention also provides a composition for treating cancer, comprising the above-described hyaluronidase PH20 variant or fragment thereof, and a method of treating cancer using the same.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 shows the protein tertiary structure model of PH20. The protein tertiary structure of PH20 was modeled using, as a template, Hyal1 (PDB ID: 2PE4) (Chao et. al., 2007) whose protein crystal structure was found, in Swiss-Model server (https://swissmodel.expasy.org/).



FIG. 1A shows the protein tertiary structure model of PH20, and indicates D146 and E148 which are catalytic amino acids. The protein tertiary structure model of PH20 is composed of eight repeats of a beta-strand and an alpha-helix.



FIG. 1B shows an eta (η) 8 loop in which alpha-helix 8 of PH20 and G340 to I344 which form a linker region in the N-terminal region of alpha-helix 8 are located. G340, T341, L342, S343 and I344 residues are each shown.



FIG. 1C shows amino acid residues (C351, Y357, and N363) which interact with the adjacent secondary structure, among the amino acids located in alpha-helix 8 of PH20. C351 forms a disulfide bond with C60 located in alpha-helix 1, Y357 hydrophobically interacts with F315 located between beta-strand 7 and alpha-helix 7, and N363 forms a hydrogen bond with a D69 residue located in alpha-helix 1.



FIG. 2 compares the protein expression levels of WT (wild type) and variants constructed in the present invention. WT and variants were expressed by transient transfection in ExpiCHO cells. WT had an expression level of 16.1 mg/L. The protein expression levels of variants based on the variants HM1 and HM6 were higher than that of WT, and the protein expression levels of HM4 and HM7 were the highest. The protein expression level of HM11 obtained by introducing additional amino acid substitutions (Y365F and I367L) into the variant HM6 decreased to 6.4 mg/mL.



FIG. 3 shows experimental results for the variants HM1 and HM6.



FIG. 3A shows the results of SDS-PAGE after purification of WT and the variants HM1 and HM6. Purification was performed using a HisTrap column and a Q Sepharose column. WT and the variants HM1 and HM6 had a molecular weight of ˜70 kDa (figure legend: M, molecular weight marker; CS, supernatant; FT, flow-through; and Elution, elution fractions).



FIG. 3B shows the enzymatic activity values of WT and the variants HM1 and HM6, measured by turbidimetric assay at pH 7.0. In the present invention, the enzymatic activity value measured by the turbidimetric assay was expressed as specific activity.



FIG. 3C shows the enzymatic activities of WT and the variants HM1 and HM6, measured by substrate-gel assay. After SDS was removed with 2.5% Triton X-100 (w/v) at 4° C., an enzymatic reaction was performed at 37° C. for 1 to 4 h. The variant HM6 renatured faster than WT and the variant HM1 and thus more rapidly hydrolyzed hyaluronic acid on polyacrylamide gel. The white band shows hyaluronic acid degraded by WT and the variant protein.



FIG. 3D shows the enzymatic activities of WT and the variant HM1, measured by substrate-gel assay at pH of 5 to 8. WT and the variant HM1 exhibit activity in the pH range of 5 to 8, and show the highest enzymatic activity at pH 5.0. The variant HM1 has the signal peptide of human serum albumin or human Hyal1. The white band shows hyaluronic acid degraded by WT and the variant protein.



FIG. 3E shows the results of separating WT and the variants HM1 and HM6 by a phenyl column. The variants were eluted faster than WT from the phenyl column.



FIG. 4 shows the results of analyzing the final product of hyaluronic acid, degraded by WT and the variant HM6, after 10 min and 1 h by means of an Amide-80 column.



FIG. 5 shows experimental results for G340 to I344 amino acid mutations of PH20.



FIG. 5A shows SDS-PAGE results after HisTrap column purification for variants HM7, HM8, HM9, HM10 and HM21.



FIG. 5B shows the results of measuring the enzymatic activities of WT and variants HM6, HM8, HM9, HM10, HM21 and HM7 by turbidimetric assay at pH 7.0.



FIG. 5C shows the results of measuring the enzymatic activities of WT and variants HM6, HM8, HM9, HM10, HM21 and HM7 by substrate-gel assay. The bar graph at the bottom of FIG. 5C shows the degree of enzymatic activity obtained by quantifying the band after staining the gel with Alcian blue. The white band shows hyaluronic acid degraded by WT and the variant protein.



FIG. 5D shows the results of analyzing WT and variants HM8, HM9, HM10, HM21 and HM7 by phenyl column chromatography.



FIG. 5E shows the results of separating WT and variants HM6, HM8, HM9, HM10, HM21 and HM7 depending on their isoelectric points at the pH of 3 to 7 by means of IEF gel.



FIG. 6 shows experimental results for the variant HM11.



FIG. 6A shows the results of purifying protein by HisTrap column chromatography for the variant HM11.



FIG. 6B shows the results of measuring the enzymatic activities of WT and the variant HM11 at pH 7.0 by turbidimetric assay.



FIG. 7 shows experimental results for N-terminally truncated PH20 variants HM40, HM13, HM41, HM24, HM42, and HM25.



FIG. 7A shows the results of purifying protein by HisTrap column chromatography for the PH20 variants HM40, HM13, HM41, HM24, HM42 and HM25.



FIG. 7B shows the expression levels of the PH20 variants HM40, HM13, HM41, HM24, HM42, HM25, HP61 and HP62 in ExpiCHO cells.



FIG. 7C shows the enzymatic activities of the PH20 variants HM40, HM13, HM41, HM24, HM42 HM25, HP61 and HP62 measured at pH 7.0 by turbidimetric assay and expressed as specific activities.



FIG. 7D shows the enzymatic activities of the PH20 variants HM40, HM13, HM41, HM24, and HM42, measured by substrate-gel assay. The white band shows hyaluronic acid degraded by WT and the variant protein.



FIG. 7E shows the results of analyzing WT and the variants HM40, HM13, HM41, HM24 and HM42 by phenyl column chromatography.



FIG. 7F shows the change in particle size with increasing temperature for the PH20 variants HM40, HM13, HM41 and HM42.



FIG. 8 shows experimental results for C-terminally truncated variants HM14, HM15 and HM16 constructed using HM6 as a template.



FIG. 8A shows the results of SDS-PAGE after HisTrap purification for the variants HM14, HM15 and HM16. As controls, WT and the variant HM6 were included.



FIG. 8B shows the results of measuring the enzymatic activities of WT and the variants HM6, HM14, HM15, and HM16 at pH 7.0 by turbidimetric assay.



FIG. 8C shows the results of measuring the enzymatic activities of WT and the variants HM6, HM14, HM15, and HM16 for 1, 2 and 4 h by substrate-gel assay. The right graph of FIG. 8C is a bar graph showing enzymatic activities measured after staining with Alcian-blue after 1 h of enzymatic reaction. The white band shows hyaluronic acid degraded by WT and the variant protein.



FIG. 8D shows the results of analyzing WT and the variants HM6, HM14, HM15, and HM16 by phenyl column chromatography.



FIG. 8E shows the results of separating the variants HM14, HM15, and HM16 depending on their isoelectric points at the pH of 3 to 7 by IEF gel.



FIG. 9 shows experimental results for PH20 variants HM19 and HM20 constructed using HM10 as a template.



FIG. 9A shows the results of purified protein by HisTrap column chromatography for the PH20 variants HM19 and HM20.



FIG. 9B shows the results of comparing the enzymatic activities of the PH20 variant HM19 and HM20 at pH 7.0 by turbidimetric assay.



FIG. 9C shows the results of staining SDS gel with Alcian blue dye after 1 h of enzymatic reaction at 37° C. by substrate-gel assay for WT and the variants HM10, HM19, and HM20. The white band shows hyaluronic acid degraded by WT and the variant protein.



FIG. 10 shows the results of measuring the aggregation temperatures of WT and the PH20 variants by dynamic light scattering (hereinafter referred to as DLS) system. The measurements were performed in triplicate and expressed as mean±S.E. values.



FIG. 11 shows a Stern-Volmer plot obtained after measuring the change in fluorescence of tryptophan residues of WT and PH20 variants by addition of acrylamide (0 to 0.5 M). Among amino acids, tryptophan is excited at 295 nm and emits maximum fluorescence at 340 nm. Acrylamide is a small molecule that can penetrate a protein structure and quench the fluorescence emission of tryptophan. As the protein structure is more flexible, the quenching of fluorescence by acrylamide is greater. F0 is the fluorescence value in the absence of acrylamide, and F is the fluorescence value in the presence of acrylamide (0 to 0.5 M). The change in the fluorescence value measured was expressed as the ratio F0/F.



FIG. 11A is a Stern-Volmer plot for WT and variants HM1, HM4, HM6 and HM7.



FIG. 11B is a Stern-Volmer plots for WT and variants HM14, HM15 and HM16.



FIG. 12 shows the expression levels of HM10-based PH20 variants in ExpiCHO cells.



FIG. 12A graphically shows the expression levels of respective variants.



FIG. 12B shows the expression levels of respective variants in the table. WT and the PH20 variants had a 6×His-tag at the C-terminus, and the protein expression levels after HisTrap column purification were expressed in mg/L. HM30 to HM33 variants were not expressed in ExpiCHO cells.



FIG. 13 shows Western blot results for cell cultures of variants HM29, HM30, HM31, HM32 and HM33. The C-termini of HM10-based variants HM29, HM30, HM31, HM32 and HM33 were cleaved after A467, C464, D461, C358, or C455, respectively. C-terminally cleaved HM29 was expressed in ExpiCHO cells, but variants having a C-terminus cleaved at C464 or shorter in length was not expressed in ExpiCHO cells. Primary antibody was rabbit anti-PH20 polyclonal antibody (Abcam) diluted at 1:500. Secondary antibody was Goat anti-rabbit IgG HRP diluted at 1:2,000.



FIG. 14 shows experimental results for C-terminally truncated variants constructed using HM10 as a template.



FIG. 14A shows the results of measuring enzymatic activities at pH 7.0 by turbidimetric assay for C-terminally cleaved variants constructed using HM10 as a template.



FIG. 14B compares enzymatic activities depending on the C-terminal cleavage sites of 17 PH20 variants (HM43, HM44, HM45, HM20, HM19, HM35, HM36, HM37, HM38, HM39, HM47, HM48, HM49, HM50, HM51, HM52 and HM10) constructed using HM10 as a template.



FIG. 14C shows the results of staining SDS gel with Alcian blue dye after 1 h of enzymatic reaction at 37° C. by substrate-gel assay for some (HM29, HM35, HM36, HM37, HM38, HM39, HM43, HM44 and HM45) of PH20 variants constructed using HM10 as a template. The white band shows hyaluronic acid degraded by WT and the variant protein.



FIG. 15 shows SDS gel after passage through a final column during protein purification for the HP34 (FIG. 15A) and HP46 (FIG. 15B) expressed in ExpiCHO cells. HP34 was subjected to a four-step chromatography purification procedure consisting of Q Sepharose, Butyl HP, Heparin and Blue Sepharose columns, and SDS gel is a result obtained after Blue Sepharose column chromatography. HP46 was subjected to a three-step chromatography purification procedure consisting of Q Sepharose, Butyl HP and Heparin columns, and SDS gel is a result obtained after Heparin column chromatography.



FIG. 16 shows the enzymatic activities of 6×His-tag-free PH20 variants HP34 and HP46 constructed using HM21 as a template.



FIG. 16A shows the results of measuring the enzymatic activities of WT and the variants HM21, HP34 and HP46 at pH 7.0 by turbidimetric assay.



FIG. 16B shows the results of measuring the enzymatic activities of WT (HW2) and the variants HM21, HP34 and HP46 at pH 5.3 by Morgan-Elson assay (Km: Michaelis-Menten constant, kcat: turnover number, and kcat/Km: catalytic efficiency).



FIG. 17 shows the results of characterization of HM21-Based PH20 Variants.



FIG. 17A shows the results of measuring aggregation temperatures by DLS for 6×His-tag-free PH20 variants HP34 and HP46 constructed using HM21 as a template. As controls, the aggregation temperatures of HW2 and HM21 are shown.



FIG. 17B shows the results of measuring enzymatic activities for 1 h by substrate-gel assay for HW2 and the variants PH20 (HP20, HP34 and HP46).



FIG. 17C shows the results of performing a substrate-gel assay after allowing variant (HW2 and HP46) samples to stand at pH 3.0 and pH 7.0 for 14 h. After SDS-PAGE, SDS was removed with 2.5% Triton X-100 (w/v), and an enzymatic reaction was performed at 37° C. for 1 h.



FIG. 17D shows the expression levels of PH20 variants HM21, HM53, HM54, HM55, HM56, HP59 and HP60 in ExpiCHO cells.



FIG. 17E shows the expression of the enzymatic activity value of PH20 variants HM21, HM53, HM54, HM55, HM56, HP59 and HP60, measured by the turbidimetric assay, as specific activity at pH 5.3.



FIG. 18 shows the results of measuring the stimulating index of CD4+ T cells in treatment with PH20 and PH20 variant at concentrations of 1.5 ng/mL and 15 ng/mL, respectively.



FIG. 19 shows the results of measuring the stimulating index of CD8+ T cells in treatment with PH20 and PH20 variant at concentrations of 1.5 ng/mL and 15 ng/mL, respectively.





BEST MODE FOR CARRYING OUT THE INVENTION

Unless defined otherwise, all the technical and scientific terms used herein have the same meaning as those generally understood by one of ordinary skill in the art to which the invention pertains. Generally, the nomenclature used herein and the experiment methods, which will be described below, are those well-known and commonly employed in the art.


The present invention provides a hyaluronidase PH20 variant or fragment thereof, which comprises one or more amino acid residue substitutions in the region corresponding to an alpha-helix region and/or its linker region, preferably an alpha-helix 8 region (S347 to C381) and/or a linker region (A333 to R346) between alpha-helix 7 and alpha-helix 8, in the amino acid sequence of wild-type PH20, preferably mature wild-type PH20, and in which one or more of the N-terminal or C-terminal amino acid residues are selectively cleaved and deleted.


In the present invention, the positions of amino acid residues in each variant correspond to the amino acid positions of wild-type PH20 having the sequence of SEQ ID NO: 1.


In addition, in the present invention, “mature wild-type PH20” means a protein consisting of amino acid residues L36 to S490 of SEQ ID NO: 1, which lack M1 to T35, which form a signal peptide, and A491 to L509, which are not related to the substantial enzymatic function of PH20, in the amino acid sequence of wild-type PH20 having the sequence of SEQ ID NO: 1.









TABLE 1





Amino acid sequence of wild-type PH20


(SEQ ID NO: 1) 















MGVLKFKHIFFRSFVKSSGVSQIVFTFLLIPCCLTLNFRAPPVIPNVPFL





WAWNAPSEFCLGKFDEPLDMSLFSFIGSPRINATGQGVTIFYVDRLGYYP





YIDSITGVTVNGGIPQKISLQDHLDKAKKDITFYMPVDNLGMAVIDWEEW





RPTWARNWKPKDVYKNRSIELVQQQNVQLSLTEATEKAKQEFEKAGKDFL





VETIKLGKLLRPNHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKRNDDLS





WLWNESTALYPSIYLNTQQSPVAATLYVRNRVREAIRVSKIPDAKSPLPV





FAYTRIVFTDQVLKFLSQDELVYTFGETVALGASGIVIWGTLSIMRSMKS





CLLLDNYMETILNPYIINVTLAAKMCSQVLCQEQGVCIRKNWNSSDYLHL





NPDNFAIQLEKGGKFTVRGKPTLEDLEQFSEKFYCSCYSTLSCKEKADVK





DTDAVDVCIADGVCIDAFLKPPMETEEPQIFYNASPSTLSATMFIVSILF





LIISSVASL









Specifically, the PH20 variant or fragment thereof according to the present invention may comprise one or more mutations, preferably amino acid residue substitutions selected from the group consisting of T341A, T341C, T341G, S343E, M345T, K349E, L353A, L354I, N356E and I361T, more preferably selected from the group consisting of T341A, T341C, L354I and N356E in wild-type PH20 having the amino acid sequence of SEQ ID NO: 1.


In the present invention, the term “PH20 variant” is intended to include mutation of one or more amino acid residues, preferably substitution of one or more amino acid residues in the amino acid sequence of wild-type PH20, as well as occurrence of deletion of one or more amino acid residues at N-terminus or C-terminus together with substitution of the amino acid residues, and is used as substantially the same meaning as the expression “PH20 variant or fragment thereof”.


In the present invention, the protein tertiary structure of PH20 located outside the active site was studied through the protein structure modeling of human PH20 on the basis of Hyal1 (SEQ ID NO: 2) which is a human hyaluronidase whose protein tertiary structure is known. As a result, amino acids located in the alpha-helix 8 region of PH20 were selected and substituted with the amino acid sequence of alpha-helix 8 of Hyal1, thereby attempting to enhance the thermal stability of the protein structure without affecting the catalytic activity of the enzyme. In particular, because alpha-helix 8 is located in the outer portion of the protein tertiary structure of PH20, there is less interaction with the adjacent alpha-helix or beta-strand than the other alpha-helices of PH20. According to the present invention, it has been found that when the amino acid sequence of the alpha-helix 8 region of human PH20 and a linker region between alpha-helix 7 and alpha-helix 8 is partially substituted with the amino acid sequence of the alpha-helix 8 region of highly hydrophilic Hyal1 and a linker region between alpha-helix 7 and alpha-helix 8 of Hyal1, the enzymatic activity at neutral pH and the protein aggregation temperature (Tagg.) increase. Based on these experimental results, it has been found that a novel PH20 variant or fragment thereof, which has increased enzymatic activity and thermal stability compared to wild-type PH20, can be provided.


Thus, the PH20 variant according to the present invention comprises one or more amino acid residue substitutions selected from the group consisting of T341A, T341C, T341G, S343E, M345T, K349E, L353A, L354I, N356E and I361T, preferably selected from the group consisting of T341A, T341C, L354I and N356E in the amino acid sequence of wild-type PH20 (having the amino acid sequence of SEQ ID NO: 1), preferably mature wild-type PH20 (having a sequence consisting of amino acid residues L36 to S490 in the amino acid sequence of SEQ ID NO: 1).


The PH20 variant according to the present invention also comprises one or more amino acid residue substitutions in the region corresponding to an alpha-helix region and/or its linker region, preferably an alpha-helix 8 region (S347 to C381) and/or the linker region (A333 to R346) between alpha-helix 7 and alpha-helix 8, more preferably T341 to N363, T341 to I361, L342 to I361, S343 to I361, I344 to I361, M345 to I361, or M345 to N363 in wild-type PH20 having the amino acid sequence of SEQ ID NO: 1.


In particular, in the PH20 variant according to the present invention, the alpha-helix 8 region (S347 to C381) and/or the linker region (A333 to R346) between alpha-helix 7 and alpha-helix 8 of the wild-type PH20, preferably the mature wild-type PH20 may be substituted with one or more amino acid residues of the corresponding region (see Tables 2 and 3) of Hyal1 having the sequence of SEQ ID NO: 2, but is not limited thereto.









TABLE 2





Amino acid sequence of wild-type Hyal1


(SEQ ID NO: 2) 















MAAHLLPICALFLTLLDMAQGFRGPLLPNRPFTTVWNANTQWCLERHGVD





VDVSVFDVVANPGQTFRGPDMTIFYSSQLGTYPYYTPTGEPVFGGLPQNA





SLIAHLARTFQDILAAIPAPDFSGLAVIDWEAWRPRWAFNWDTKDIYRQR





SRALVQAQHPDWPAPQVEAVAQDQFQGAARAWMAGTLQLGRALRPRGLWG





FYGFPDCYNYDFLSPNYTGQCPSGIRAQNDQLGWLWGQSRALYPSIYMPA





VLEGTGKSQMYVQHRVAEAFRVAVAAGDPNLPVLPYVQIFYDTTNHFLPL





DELEHSLGESAAQGAAGVVLWVSWENTRTKESCQAIKEYMDTTLGPFILN





VTSGALLCSQALCSGHGRCVRRTSHPKALLLLNPASFSIQLTPGGGPLSL





RGALSLEDQAQMAVEFKCRCYPGWQAPWCERKSMW
















TABLE 3







Comparison of alpha-helices and amino


acid sequence between PH20 and Hyal1












amino acid
amino acid



alpha-helix
sequences of PH20
sequences of Hyal1







alpha-helix 1

P56~D65

N39~G48



alpha-helix 3
 S119~M135
S101~I117 



alpha-helix 4′
K161~N176
K144~H159



alpha-helix 4
S180-R211

P163~R194




alpha-helix 5
F239~S256
P222~S239



alpha-helix 6
A274~D293
K257~G277



alpha-helix 7

S317~G332


P299~G314




alpha-helix 8

S347~C381

T329~C363










More specifically, the novel PH20 variant or fragment thereof according to the present invention preferably comprises an amino acid residue substitution of L354I and/or N356E in the amino acid sequence of wild-type PH20, preferably mature wild-type PH20,


and further comprises at least the amino acid residue substitution at one or more positions selected from among T341 to N363, particularly one or more positions selected from the group consisting of T341, L342, S343, I344, M345, S347, M348, K349, L352, L353, D355, E359, I361 and N363, but is not limited thereto.


More preferably, the amino acid residue substitution at one or more positions selected from the group consisting of T341, L342, S343, I344, M345, S347, M348, K349, L352, L353, D355, E359, I361 and N363 may be one or more amino acid residue substitutions selected from the group consisting of T341A, T341C, T341D, T341G, T341S, L342W, S343E, I344N, M345T, S347T, M348K, K349E, L352Q, L353A, D355K, E359D, I361T and N363G, but is not limited thereto.


Preferably, the novel PH20 variant or fragment thereof according to the present invention may comprise one or more amino acid residue substitutions selected from the group consisting of M345T, S347T, M348K, K349E, L352Q, L353A, L354I, D355K, N356E, E359D and I361T,


and may further comprise one or more amino acid residue substitutions selected from the group consisting of T341A, T341C, T341D, T341G, T341S, L342W, S343E, I344N and N363G, but is not limited thereto.


More preferably, the novel PH20 variant or fragment thereof according to the present invention may comprise any one amino acid residue substitution selected from the following amino acid residue substitution groups, but is not limited thereto:


(a) T341S, L342W, S343E, I344N, M345T, S347T, M348K, K349E, L352Q, L353A, L354I, D355K, N356E, E359D and I361T;


(b) L342W, S343E, I344N, M345T, S347T, M348K, K349E, L352Q, L353A, L354I, D355K, N356E, E359D and I361T;


(c) M345T, S347T, M348K, K349E, L352Q, L353A, L354I, D355K, N356E, E359D, I361T and N363G;


(d) T341G, L342W, S343E, I344N, M345T, S347T, M348K, K349E, L352Q, L353A, L354I, D355K, N356E, E359D and I361T;


(e) T341A, L342W, S343E, I344N, M345T, S347T, M348K, K349E, L352Q, L353A, L354I, D355K, N356E, E359D and I361T;


(f) T341C, L342W, S343E, I344N, M345T, S347T, M348K, K349E, L352Q, L353A, L354I, D355K, N356E, E359D and I361T;


(g) T341D, L342W, S343E, I344N, M345T, S347T, M348K, K349E, L352Q, L353A, L354I, D355K, N356E, E359D and I361T;


(h) I344N, M345T, S347T, M348K, K349E, L352Q, L353A, L354I, D355K, N356E, E359D and I361T; and


(i) S343E, I344N, M345T, S347T, M348K, K349E, L352Q, L353A, L354I, D355K, N356E, E359D and I361T.


In the present invention, an expression described by one-letter amino acid residue code together with numbers, such as “S347”, means the amino acid residue at each position in the amino acid sequence of SEQ ID NO: 1.


For example, “S347” means that the amino acid residue at position 347 in the amino acid sequence of SEQ ID NO: 1 is serine.


In addition, “S347T” means that serine at position 347 of SEQ ID NO: 1 is substituted with threonine.


The PH20 variant according to the present invention is interpreted as including variants or fragments thereof in which the amino acid residue at the specific amino acid residue position is conservatively substituted.


As used herein, the term “conservative substitution” refers to modifications of a PH20 variant that involves the substitution of one or more amino acids for amino acids having similar biochemical properties that do not result in loss of the biological or biochemical function of the PH20 variant.


A “conservative amino acid substitution” is one in which the amino acid residue is replaced with an amino acid residue having a similar side chain. Families of amino acid residues having similar side chains have been defined and are well known in the art to which the present invention pertains. These families include amino acids with basic side chains (e.g., lysine, arginine and histidine), amino acids with acidic side chains (e.g., aspartic acid and glutamic acid), amino acids with uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, and cysteine), amino acids with nonpolar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, and tryptophan), amino acids with beta-branched side chains (e.g., threonine, valine, and isoleucine), and amino acids with aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, and histidine).


It is envisioned that the PH20 variant or fragments thereof of the present invention may still retain the activity although it has conservative amino acid substitutions.


In addition, the PH20 variant or fragment thereof according to the present invention is interpreted to include PH20 variants or fragments thereof having substantially the same function and/or effect with those/that of the PH20 variant or fragment thereof according to the present invention, and having an amino acid sequence homology of at least 80% or 85%, preferably at least 90%, more preferably at least 95%, most preferably at least 99% to the PH20 variant or fragment thereof according to the present invention.


The PH20 variants according to the present invention have increased expression levels and protein refolding rate, thereby increasing high thermal stability, compared to mature wild-type PH20. Furthermore, the enzymatic activity of the PH20 variants was more increased than or similar to that of mature wild-type PH20 despite an increase in the thermal stability.


Meanwhile, even if the mature wild type PH20 variants with the C-terminal deletion showed the decrease in enzymatic activities, based on the present invention, the C-terminal deleted PH20 variants showed the similar or increased enzymatic activities due to the more rapid protein refolding and thermal stabilities. In addition, PH20 variants in this present invention maintained the enzymatic activities when the N-terminal amino acids were deleted up to five amino acid residues. This indicated that for the protein expression and enzyme activities P41 of the N-terminus was important.


Accordingly, the PH20 variant or fragment thereof according to the present invention is characterized in that it comprises one or more amino acid residue substitutions in the alpha-helix 8 region (S347 to C381) and/or the linker region (A333 to R346) between alpha-helix 7 and alpha-helix 8 in the amino acid sequence of wild-type PH20, and one or more of the N-terminal and/or C-terminal amino acid residues are additionally deleted, but is not limited thereto.


In one aspect, the PH20 variant or fragment thereof according to the present invention may be one in which cleavage occurs before an amino acid residue selected from the group consisting of M1 to P42 of the amino acid sequence of SEQ ID NO: 1, preferably before an amino acid residue L36, N37, F38, R39, A40, P41, or P42 at the N-terminus, so that one or more amino acid residues at the N-terminus are deleted, and/or cleavage occurs after an amino acid residue selected from the group consisting of V455 to L509, preferably after an amino acid residue selected from the group consisting of V455 to S490, most preferably after an amino acid residue V455, C458, D461, C464, I465, D466, A467, F468, K470, P471, P472, M473, E474, T475, E476, P478, I480, Y482, A484, P486, T488, or S490 at the C-terminus, so that one or more amino acid residues at the C-terminus are deleted.


The expression “cleavage occurs before an amino acid residue selected from the group consisting of M1 to P42 at the N-terminus” means that an amino acid residue immediately before an amino acid residue selected from among M1 to P42 at the N-terminus is cleaved and deleted.


For example, the expression “cleavage occurs before an amino acid residue L36, N37, F38, R39, A40, P41, or P42” respectively means that all amino acid residues from M1 to T35 immediately before L36, all amino acid residues from M1 to L36 immediately before N37, all amino acid residues from M1 to N37 immediately before F38, all amino acid residues from M1 to F38 immediately before R39, all amino acid residues from M1 to R39 immediately before A40, all amino acid residues from M1 to A40 immediately before P41, or all amino acid residues from M1 to P41 immediately before P42 in the amino acid sequence of SEQ ID NO: 1 is cleaved and removed, respectively.


In addition, the expression “cleavage occurs after an amino acid residue selected from the group consisting of V455 to L509 at the C-terminus” means that an amino acid residue immediately after an amino acid residue selected from among V455 to L509 at the C-terminus is cleaved and deleted.


For example, the expression “cleavage occurs after an amino acid residue V455, C458, D461, C464, I465, D466, A467, F468, K470, P471, P472, M473, E474, T475, E476, P478, I480, Y482, A484, P486, T488 or S490 at the C-terminus” means that an amino acid residue after the amino acid residue V455, C458, D461, C464, I465, D466, A467, F468, K470, P471, P472, M473, E474, T475, E476, P478, I480, Y482, A484, P486, T488 or S490 in the amino acid sequence of SEQ ID NO: 1 is cleaved and removed.


Preferably, the novel PH20 variant or fragment thereof according to the present invention may be selected from the group consisting of amino acid sequences of SEQ ID NOs: 60 to 115, but is not limited thereto.


Most preferably, the novel PH20 variant or fragment thereof according to the present invention may have the amino acid sequence of SEQ ID NO: 99. The novel PH20 variant or fragment thereof having an amino acid sequence of SEQ ID NO: 99 may comprise 15 amino acid substitutions of T341S, L342W, S343E, I344N, M345T, S347T, M348K, K349E, L352Q, L353A, L354I, D355K, N356E, E359D and I361T, and deletion before F38 at the N-terminus, and truncation after F468 at the C-terminus.


The sequences of the substituted or cleaved amino acids in the PH20 variants constructed in the specific embodiment according to the present invention are as shown in Table 11.


A study focused on increasing the enzymatic activity and thermal stability of PH20 by amino acid substitution of an alpha-helix and its linker region, which are secondary structures forming the tertiary structure of the protein, as disclosed in the present invention, has not been previously reported. Previous studies reported that the enzymatic activity of wild-type PH20 changes depending on the cleavage positions of amino acid residues located at the C-terminal region. However, in the present invention, a specific alpha-helix forming the secondary structure of PH20 was substituted with the alpha-helix of other human hyaluronidase, thereby constructing PH20 variants having higher stability than wild-type PH20. These variants may be variants in which the interaction of the substituted alpha-helix domain with portions forming other secondary structures of PH20 shows a pattern different from that of wild-type PH20, indicating that the variants have consistent enzymatic activity regardless of the C-terminal cleavage position.


In specific embodiment, the novel PH20 variant or fragment thereof according to the present invention, which has increased enzymatic activity and thermal stability compared to mature wild-type PH20, may be one which comprises one or more amino acid residue substitutions selected from the group consisting of T341A, T341C, T341G, S343E, M345T, K349E, L353A, L354I, N356E and I361T, and in which one or more amino acids located in an alpha-helix 8 region (S347 to C381) and/or the linker region (A333 to R346) between alpha-helix 7 and alpha-helix 8 in the amino acid sequence of wild-type PH20, preferably mature wild-type PH20, are substituted with other amino acids.


Specifically, the amino acid substitution in the linker region between alpha-helix 7 and alpha-helix 8 comprises the substitution of one or more amino acid residues in the region consisting of amino acid residues T341 to N363, T341 to I361, L342 to I361, L342 to I361, S343 to I361, I344 to I361, M345 to I361, or M345 to N363.


In order to examine the effect of C-terminal truncation in PH20 variants in which alpha-helix 8 and a linker region between alpha-helix 7 and alpha-helix 8 are substituted, three PH20 variants (HM6, HM10 and HM21) were selected as templates.


HM6 is a variant in which amino acids in M345 to N363 region are substituted with the amino acid sequence of Hyal1 (M345T, S347T, M348K, K349E, L352Q, L353A, L354I, D355K, N356E, E359D and I361T in SEQ ID NO: 1 are substituted). In addition, HM6 is a variant in which the substitution of alpha-helix 8 and a linker region between alpha-helix 7 and alpha-helix 8 is the least substituted variant among the PH20 variants according to the present invention, which do not comprise additional C-terminal cleavage (that is, a form in which the C-terminal amino acid residue is S490, like mature wild-type PH20).


HM10 is a variant in which amino acids in L342 to I361 region are substituted with the amino acid sequence of Hyal1 (L342W, S343E, I344N, M345T, S347T, M348K, K349E, L352Q, L353A, L354I, D355K, N356E, E359D and I361T in SEQ ID NO: 1 are substituted), and which has the highest thermal stability while having an enzymatic activity similar to that of mature wild-type PH20 among the PH20 variants according to the present invention, which do not comprise additional C-terminal truncation.


HM21 is a variant in which amino acids in T341 to I361 region are substituted with the amino acid sequence of Hyal1 (T341S, L342W, S343E, I344N, M345T, S347T, M348K, K349E, L352Q, L353A, L354I, D355K, N356E, E359D and I361T in SEQ ID NO: 1 are substituted), and which has an enzymatic activity which is about two times higher than that of wild-type PH20 at pH 7.0 among the PH20 variants according to the present invention, which do not comprise additional C-terminal truncation.


The HM6-based PH20 variants constructed in the present invention have the N-terminus starting at L36 and the C-terminus terminating at I465, F468, or P471 as shown in Table 4 below.









TABLE 4







C-terminal amino acid truncated PH20 variants


constructed using HM6 as a template














Number of
Number of



Total
Substituted
substituted
truncated


Variant
length
region
amino acids
amino acids














HM6
36~490
M345~I361
11
0


HM14
36~465

11
25


HM15
36~468

11
22


HM16
36~471

11
19









The HM10-based PH20 variants commonly have the N-terminus cleaved before the F38 residue and the C-terminus cleaved after the V455, C458, D461, C464, I465, D466, A467, F468, K470, P472, M473, E474, T475, E476, P478, I480, Y482, A484, P486, or T488 residue as shown in Table 5 below.









TABLE 5







C-terminal amino acid truncated PH20 variants


constructed using HM10 as a template














Number of
Number of



Total
Substituted
substituted
truncated


Variant
length
region
amino acids
amino acids














HM10
36~490
L342~I361
14
0


HM52
38~488

14
4


HM51
38~486

14
6


HM50
38~484

14
8


HM49
38~482

14
10


HM48
38~480

14
12


HM47
38~478

14
14


HM39
38~476

14
16


HM38
38~475

14
17


HM37
38~474

14
18


HM36
38~473

14
19


HM35
38~472

14
20


HM19
38~470

14
22


HM20
38~468

14
24


HM45
38~467

14
25


HM29
36~467

14
23


HM44
38~466

14
26


HM43
38~465

14
27


HM30
36~464

14
26


HM31
36~461

14
29


HM32
36~458

14
32


HM33
36~455

14
35


HP19
38~470

14
22


HP20
38~468

14
24









As shown in the examples of variants which comprise amino acid substitutions in the L342 to I361 region corresponding to the alpha-helix 8 region and the linker region between alpha-helix 7 and alpha-helix 8 of HM10 as a template and in which the N-terminus was cleaved before the F38 residue and the C-terminus was cleaved at I465, D466, A467, F468, K470, P472, M473, E474, T475, E476, P478, I480, Y482, A484, P486, or T488, the PH20 variants according to the present invention exhibited an enzymatic activity similar to that of mature wild-type PH20 regardless of the C-terminal cleavage position.


As shown in Table 6 below, two HM21-based PH20 variants commonly have the N-terminus cleaved before the F38 residue and the C-terminus cleaved after the F468 or K470 residue.









TABLE 6







C-terminal amino acid truncated PH20 variants


constructed using HM21 as a template














Number of
Number of



Total
Substituted
substituted
truncated


Variant
length
region
amino acids
amino acids














HM21
38~490
T341~I361
15
2


HP34
38~470

15
22


HP46
38~468

15
24









Variants were constructed using, as a template, HM21 having an enzymatic activity which is about two times higher than that of mature wild-type PH20. These variants are those which comprise amino acid substitutions in the T341 to I361 region corresponding to the alpha-helix 8 region and the linker region between alpha helix 7 and alpha-helix 8 and in which the N-terminus was cleaved before the F38 residue and the C-terminus was cleaved after F468 or K470. Surprisingly, these variants maintained the high enzymatic activity of HM21 regardless of the C-terminal cleavage position.


In the study conducted by Frost et al., when the length of PH20 is shorter due to the cleavage before the amino acid position of 477, the enzymatic activity decreased to about 10% of a variant having a C-terminus cleaved after the position 477. However, in the present invention, when one or more amino acids in alpha-helix 8 of PH20 and its linker region were substituted, the enzymatic activity was maintained due to the increase in the stability of the protein regardless of the C-terminal cleavage position. This result is very significant in that it solves the problem that the enzymatic activity of wild-type PH20 is reduced due to the C-terminal truncation of wild-type PH20.


In addition, in the present invention, the effect of the N-terminal amino acids of PH20, which has not been known previously, was studied.


In order to examine the effects of N-terminal cleavage sites in HM6 variants in which one or more amino acid residues in the region (M345 to I361) corresponding to the alpha-helix region of wild-type PH20 and the linker region between alpha-helix 7 and alpha-helix 8 (substituted with M345T, S347T, M348K, K349E, L352Q, L353A, L354I, D355K, N356E, E359D or I361T in SEQ ID NO: 1 and comprising no additional C-terminal cleavage) were substituted with amino acid residues of the corresponding alpha-helix 8 region of Hyal1 and the linker region between alpha-helix 7 and alpha-helix 8, variants, in which amino acid residues L36 to V47 in SEQ ID NO: 1 were substituted with FRGPLLPNR or amino acid residues L36 to A52 in SEQ ID NO: 1 were substituted with FRGPLLPNRPFTTV, were constructed using HM6 as a template. In addition, using HM6 as a template, the variants HM40, HM13, HM41, HM24, HM42 and HM25 were constructed in which the N-terminus in the amino acid sequence of SEQ ID NO: 1 was cleaved before the N37, F38, R39, A40, P41 or P42 residue (see Table 7).









TABLE 7







N-terminal amino acid cleaved variants based on HM6














Number of
Number of



Total
Substituted
substituted
truncated


Variant
length
region
amino acids
amino acids





HM6
36~490
M345~I361
11
0


HM40
37~490

11
1


HM13
38~490

11
2


HM41
39~490

11
3


HM24
40~490

11
4


HM42
41~490

11
5


HM25
42~490

11
6


HM17

 L36~V47,
23




M345~I361


HM18

 L36~A52,
28




M345~I361









As a result, it was shown that when the N-terminus of HM6 was cleaved before the N37, F38, R39, A40 or P41 residue, the enzymatic activity was not greatly influenced; however, when the N-terminus was cleaved before the P42 residue, the enzymatic activity significantly decreased, indicating that the N-terminal region of PH20, located after P41, is important for protein expression and enzymatic activity. In addition, when one or more amino acids in the N-terminal L36 to V47 or L36 to A52 region of HM6 were substituted with the amino acids of Hyal1, the variant protein was not expressed in ExpiCHO cells, indicating that the N-terminal region is important for protein expression.


In addition, in the present invention, it was attempted to increase the expression of a recombinant PH20 protein in animal cells by using the signal peptide of other proteins, instead of using the original signal peptide of PH20.


Therefore, in another aspect, the novel PH20 variant according to the present invention may be one in which the N-terminus further comprises a human growth hormone signal peptide having an amino acid sequence MATGSRTSLLLAFGLLCLPWLQEGSA of SEQ ID NO: 3, a human serum albumin signal peptide having an amino acid sequence MKWVTFISLLFLFSSAYS of SEQ ID NO: 4, or a human Hyal1 signal peptide having an amino acid sequence MAAHLLPICALFLTLLDMAQG of SEQ ID NO: 5 as shown in Table 8 below, instead of the signal peptide of wild-type PH20, which consists of M1 to T35, but is not limited thereto.


The expression “instead of the signal peptide of wild-type PH20, which consists of M1 to T35” means a case in which the signal peptide of wild-type PH20 is partially or completely deleted; thus it does not perform its function. In addition, the expression is meant to include a case in which a portion of the N-terminus is further deleted, for example, a case in which cleavage occurs before the N37, F38, R39, A40, P41 or P42 residue occurs so that an additional deletion of the N-terminus together with the deletion of the signal peptide of wild-type PH20 occurs.









TABLE 8







Amino acid sequences of signal peptide


of human growth hormone, human serum


albumin or human Hyal1 










Amino acid sequences
SEQ NO. 





human growth
MATGSRTSLLLAFGLLCLPWLQEGSA
3 


hormone 







human serum
MKWVTFISLLFLFSSAYS
4 


albumin 







human Hyal1
MAAHLLPICALFLTLLDMAQG
5 









In another aspect, the present invention provides a composition for treating cancer comprising the novel PH20 variant according to the present invention, and a method for treating cancer using the same.


Cancers or carcinomas that can be treated by the novel PH20 variant according to the present invention are not limited particularly, but includes both solid cancers and blood cancers. The cancer may be selected from the group consisting of liver cancer, hepatocellular carcinoma, gastric cancer, breast cancer, lung cancer, ovarian cancer, bronchial cancer, nasopharyngeal cancer, laryngeal cancer, pancreatic cancer, bladder cancer, colorectal cancer, colon cancer, uterine cervical cancer, brain cancer, prostate cancer, bone cancer, thyroid cancer, parathyroid cancer, renal cancer, esophageal cancer, biliary tract cancer, testis cancer, rectal cancer, head and neck cancer, ureteral cancer, osteosarcoma, neurocytoma, fibrosarcoma, rhabdomyosarcoma, astrocytoma, neuroblastoma and neuroglioma, but is not limited thereto. Preferably, cancers that can be treated by the composition according to the present invention may be selected from the group consisting of colorectal cancer, breast cancer, lung cancer and renal cancer, but is not limited thereto.


The composition of the present invention may be a pharmaceutical composition. The pharmaceutical composition may further comprise a pharmaceutically acceptable component. The component that is typically used in the formulation of drugs may be one or more selected from the group consisting of, but not limited to, lactose, dextrose, sucrose, sorbitol, mannitol, starch, gum acacia, calcium phosphate, alginate, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, syrups, methyl cellulose, methylhydroxybenzoate, propylhydroxybenzoate, talc, magnesium stearate, and mineral oil. In addition, the pharmaceutical composition may further comprise one or more selected from the group consisting of diluents, excipients, lubricants, wetting agents, sweeteners, aromatics, emulsifiers, suspensions, and preservatives.


The pharmaceutical composition of the present invention may be administered orally or parenterally. The parenteral administration is carried out by intravenous injection, subcutaneous injection, intramuscular injection, intraperitoneal injection, endothelial administration, topical administration, intranasal administration, intrapulmonary administration, rectal administration, and the like. For the oral administration, the active ingredient in the composition needs to be formulated into a coated dosage form or into a dosage form which can protect the active ingredient from being disintegrated in stomach considering that peptides and proteins are digested in stomach. Alternatively, the present composition may be administered via any device by which the active ingredient can move to the target cell of interest.


The pharmaceutical composition may be formulated in the form of solutions, suspensions, syrups or emulsions in oils or aqueous media, or in the form of extracts, grains, suppositories, powders, granules, tablets or capsules, and may additionally include dispersing or stabilizing agents for the purpose of formulation.


In particular, the composition for treating cancer according to the present invention may be used for combined treatment with other anticancer drugs.


An anticancer drug that can be used for combined treatment with the novel PH20 variant according to the present invention is preferably a chemical anticancer drug, an antibody-based anticancer drug, a biological anticancer drug, an RNAi, or a cell therapeutic agent, but is not limited thereto.


Preferably, the anticancer drug that can be used for combined treatment with the novel PH20 variant according to the present invention is preferably an immuno-oncologic agent, more preferably an immune checkpoint inhibitor, but is not limited thereto.


In another aspect, the present invention is directed to a nucleic acid encoding the PH20 variant or fragment thereof.


The nucleic acids, as used herein, may be present in cells, in the cell lysate, or in the partially purified or substantially pure form. “Isolated” or “substantially pure”, when referring to nucleic acids, refer to those that have been purified away from other cellular components or other contaminants, e.g., other cellular nucleic acids or proteins, by standard techniques, including alkaline/SDS treatment, CsCl banding, column chromatography, agarose gel electrophoresis, and others well known in the art. The nucleic acids of the present invention may be DNA or RNA.


In still another aspect, the present invention is directed to recombinant expression vector comprising the nucleic acid. For expression of the PH20 variant or fragment thereof according to the present invention, a DNA encoding the PH20 variant can be obtained by standard molecular biology techniques (e.g., PCR amplification or cDNA cloning using a hybridoma that expresses the PH20 variant), and the DNA can be inserted into an expression vector such that it is “operatively linked” to transcriptional and translational control sequences.


As used herein, the term “operatively linked” is intended to mean that a gene encoding the PH20 variant or fragment thereof is ligated into a vector such that transcriptional and translational control sequences serve their intended function of regulating the transcription and translation of the gene encoding the PH20 variant or fragment thereof. The expression vector and expression control sequences are chosen to be compatible with the expression host cell used. The genes encoding the PH20 are inserted into the expression vector by standard methods (e.g., ligation of complementary restriction enzyme sites on a fragment of the gene encoding the PH20 variant or fragment thereof and vector, or blunt end ligation if no restriction enzyme sites are present).


In addition, the recombinant expression vectors carry regulatory sequences that control the expression of a gene encoding the PH20 variant in the host cell. The term “regulatory sequence” is intended to include promoters, enhancers and other expression control elements (e.g., polyadenylation signals) that control the transcription or translation of the genes encoding the PH20 variant or fragment thereof. It will be appreciated by those skilled in the art that the design of the expression vector, including the selection of regulatory sequences, may depend on such factors as the choice of the host cell to be transformed, the level of expression of protein desired, etc.


In yet another aspect, the present invention is directed to a host cell comprising the nucleic acid or the vector. The host cell according to the present invention is preferably selected from the group consisting of animal cells, plant cells, yeasts, E. coli., and insect cells, but is not limited thereto.


Specifically, the host cell according to the present invention include prokaryotic cells such as E. coli, Bacillus subtilis, Streptomyces sp., Pseudomonas sp., Proteus mirabilis or Staphylococcus sp., fungi such as Aspergillus sp., yeasts such as Pichia pastoris, Saccharomyces cerevisiae, Schizosaccharomyces sp. and Neurospora crassa, and eukaryotic cells such as lower eukaryotic cells, and higher other eukaryotic cells such as insect cells.


In addition, the host cells that can be used in the present invention may be derived from plants or mammals. Preferably, examples of the host cells include, but are not limited to, monkey kidney cells (COS7), NSO cells, SP2/0, Chinese hamster ovary (CHO) cells, W138, baby hamster kidney (BHK) cells, MDCK, myeloma cells, HuT 78 cells and HEK293 cells. More preferably, CHO cells may be used.


The nucleic acid or the vector is transfected into a host cell. Transfection can be performed using various techniques that are generally used to introduce foreign nucleic acid (DNA or RNA) into prokaryotic or eukaryotic cells, for example, electrophoresis, calcium phosphate precipitation, DEAE-dextran transfection or lipofection. In order to express the PH20 variant or fragment thereof of the present invention, various combinations of recombinant expression vectors and host cells can be employed. The preferred expression vector for eukaryotic cells comprises gene expression regulatory sequences derived from, but not limited to, SV40, bovine papillomavirus, adenovirus, adeno-associated virus, cytomegalovirus and retrovirus. The expression vector, which can be used for bacterial hosts, comprises bacterial plasmids, such as, pET, pRSET, pBluescript, pGEX2T, pUC vector, col E1, pCR1, pBR322, pMB9 and the derivatives thereof, obtained from E. coli; a plasmid having broad host range, such as, RP4; phage DNAs exemplified by various phage lambda derivatives, such as, λgt10, λgt11 and NM989; and other DNA phages, such as, M13 and filamentous single-stranded DNA phage. The expression vector available for yeast cells may be 2-μm plasmid and its derivatives. The expression vector for insect cells includes pVL941.


In a further aspect, the present invention is directed to a method for producing an PH20 variant or fragment thereof, the method comprising a step of culturing the host cell and expressing the PH20 variant or fragment thereof according to the present invention.


When a recombinant expression vector capable of expressing the PH20 variant or fragment thereof is introduced into mammalian host cells, the PH20 variant or fragment thereof can be produced by culturing the host cells for a period of time such that the PH20 variant or fragment thereof is expressed in the host cells, preferably a period of time such that the PH20 variant is secreted into the medium during culture of the host cells.


In some cases, the expressed PH20 variant can be isolated and purified from the host cells. Isolation or purification of the PH20 variant can be performed by conventional isolation/purification methods (e.g., chromatography) that are used for proteins. The chromatography may include one or more combinations selected from affinity chromatography, ion exchange chromatography, and hydrophobic chromatography, but is not limited thereto. In addition to the chromatography, a combination of filtration, ultrafiltration, salting out, dialysis, and the like may be used to isolate and purify the antibody.


Examples

Hereinafter, the present invention will be described in further detail with reference to examples. It will be obvious to a person having ordinary skill in the art that these examples are illustrative purposes only and are not to be construed to limit or change the scope of the present invention.


Example 1. Construction of PH20 Variants

For construction of PH20 variants, the cDNA (clone ID: hMU002604) of wild-type PH20 was purchased from the Korean Human Gene Bank. Wild-type PH20 encodes amino acids from L36 to S490. The PH20 gene was amplified by polymerase chain reaction (hereinafter referred to as PCR) and inserted into the XhoI and NotI restriction enzyme sites of a pcDNA3.4-TOPO vector. For expression in ExpiCHO cells, the signal peptide of human growth hormone, human serum hormone or human Hyal1 was used as a signal peptide instead of the original signal peptide of PH20. For protein purification using a HisTrap column, the DNA sequence of a 6×His-tag was located at the 3′-end of the PH20 cDNA. The amino acid substitution of PH20 variants was performed using PCR method, and the amino acid substitution was confirmed by DNA sequencing.


The list of primers used in cloning of the PH20 variants are summarized in Table 9 below, and the specific sequences of the primers are summarized in Table 10 below.









TABLE 9







List of primers used in cloning of PH20 variants










Primer











Clone
1
2
3





cB4202
ALB-SP-Xho
ALB-PH20-MR
SPAM1-6H-Not


cB4203-HM1
ALB-SP-Xho
PH20_M345-364-F
SPAMl-6H-not


cB4203-HM2
ALB-SP-Xho
PH20_Y365-L380-F
SPAM1-7H-not


cB4203-HM3
ALB-SP-Xho
PH20_M345-L380-F
SPAM1-8H-not


cB4203-HM4
ALB-SP-Xho
B4203-HM4-F
SPAM1-9H-not


cB4203-HM5
ALB-SP-Xho
B4203-HM5-F
SPAM1-10H-not


cB4203-HM6
ALB-SP-Xho
PH20-G363N
SPAM1-6H-not


cB4203-HM7
ALB-SP-Xho
PH20-G363N
SPAM1-12H-not


cB4203-HM8
ALB-SP-Xho
cB4203-HM8-M
SPAM1-13H-not


cB4203-HM9
ALB-SP-Xho
cB4203-HM9-M
SPAM1-14H-not


cB4203-HM10
ALB-SP-Xho
cB4203-HM10-M
SPAM1-6H-Not


cB4203-HM11
ALB-SP-Xho
4203-HM11
SPAM1-16H-not


cB4203-HM12
ALB-SP-Xho
4203-HM12
SPAM1-17H-not


cB4203-HM13
ALB-SP-Xho
SASP-LN-del-R
SPAM1-18H-not


cB4203-HM14
ALB-SP-Xho

I465-6H-not


cB4203-HM15
ALB-SP-Xho

F468-6H-not


cB4203-HM16
ALB-SP-Xho

P471-6H-not


cB4203-HM17
ALB-SP-Xho
PH20-HM17
SPAM1-22H-not


cB4203-HM18
ALB-SP-Xho
PH20-HM18
SPAM1-23H-not


cB4203-HM19
ALB-SP-Xho
SASP-LN-del-R
K470-6H-not


cB4003-HP19
ALB-SP-Xho
K470-not


cB4203-HM20
ALB-SP-Xho
SASP-LN-del-R
F468-6H-not


cB4003-HP20
ALB-SP-Xho
F468-not


cB4203-HM21
ALB-SP-Xho
M21-mega-F
SPAM1-6H-Not


cB4203-HM24
ALB-SP-Xho
M24-R
SPAM1-6H-not


cB4203-HM25
ALB-SP-Xho
M25-R
SPAM1-6H-not


cB4203-HM26
ALB-SP-Xho
B4-HM26
SPAM1-6H-not


cB4203-HM27
ALB-SP-Xho
B4-HM27
SPAM1-6H-not


cB4203-HM28
ALB-SP-Xho
B4-HM28
SPAM1-6H-not


cB4203-HM29
ALB-SP-Xho
B4-HM29


cB4203-HM30
ALB-SP-Xho
B4-HM30


cB4203-HM31
ALB-SP-Xho
B4-HM31


cB4203-HM32
ALB-SP-Xho
B4-HM32


cB4203-HM33
ALB-SP-Xho
B4-m33


cB4003-HP34
ALB-SP-Xho
M21-mega-F
K470-not


cB4203-HM35
ALB-SP-Xho
SASP-LN-del-R
P472-6H-not


cB4003-HP35
ALB-SP-Xho
P472-not


cB4203-HM36
ALB-SP-Xho
SASP-LN-del-R
M473-6H-not


cB4003-HP36
ALB-SP-Xho
M473-not


cB4203-HM37
ALB-SP-Xho
SASP-LN-del-R
E474-6H-not


cB4003-HP37
ALB-SP-Xho
E474-not


cB4203-HM38
ALB-SP-Xho
SASP-LN-del-R
T475-6H-not


cB4003-HP38
ALB-SP-Xho
T475-not


cB4203-HM39
ALB-SP-Xho
SASP-LN-del-R
E476-6H-not


cB4003-HP39
ALB-SP-Xho
SASP-LN-del-R
E476-not


cB4203-HM40
ALB-SP-Xho
M40-mega
SPAM1-6H-Not


cB4203-HM41
ALB-SP-Xho
M41-mega
SPAM1-6H-Not


cB4203-HM42
ALB-SP-Xho
m42-mega
SPAM1-6H-not


cB4203-HM43
ALB-SP-Xho
I465-6H-not


cB4203-HM44
ALB-SP-Xho
D466-6H-not


cB4203-HM45
ALB-SP-Xho
B4-HM29


cB4003-HP46
ALB-SP-Xho
F468-Not


cB4203-HM47
ALB-SP-Xho
P478-H-Not


cB4203-HM48
ALB-SP-Xho
I480-H-Not


cB4203-HM49
ALB-SP-Xho
Y482-H-Not


cB4203-HM50
ALB-SP-Xho
A484-H-not


cB4203-HM51
ALB-SP-Xho
P4 8 6-H-not


cB4203-HM52
ALB-SP-Xho
SASP-LN-del-R
T488-H-not
















TABLE 10







Primer sequences used in cloning of PH20 variants









Primer
SEQ NO.
Nucleotide Sequences (5′ -> 3′)





ALB-SP-Xho
 6
GAA TAT CTC GAG GCC ACC ATG AAG TGG GTT




ACA





SPAM1-6H-Not
 7
CTA ATT GCG GCC GCT CAT TAG TGG TGA TGG




TGA TGA TGG AG AAA CCA ATT CTG C





ALB-PH20-MR
 8
TAA CAG GAG GTG CTC TGA AAT TCA GAG AGT




AAG CAG AGG AG





PH20_M345-P364-F
 9
ATG GGG AAC CCT CAG TAT AAC AAG AAC CAA




GGA ATC ATG TCA GGC CAT CAA GGA GTA TAT




GGA CAC TAC ACT GGG GCC CTA CAT AAT CAA




CGT CAC AC





PH20_Y365-L380-F
10
ATG GAG ACT ATA CTG AAT CCT TTC ATC CTG




AAC GTG ACC AGT GGG GCC CTT CTC TGC AGT




CAA GCC CTG TGC CAG GAG CAA GGA GTG TG





PH20_M345-L380-F*
11
ATG GAC ACT ACA CTG GGG CCC TTC ATC CTG




AAC GTG ACC AGT GGG GCC CTT CTC TGC AGT




CAA GCC CTG TGC CAG GAG CAA GGA GTG TG





B4203-HM4-F
12
ACT GTT GCT CTG GGT GCT TCT GGA ATT GTA




ATA TGG GTA AGC TGG GAA AAT ACA AGA ACC




AG GAA TCA TGT CA





B4203-HM5-F
13
AGC AAG GAG TGT GTA TAA GGA AAA CCA GCC




ACC CAA AAG ACT ATC TTC ACC TCA ACC CAG




A





PH20-G363N
14
AGT ATA TGG ACA CTA CAC TGA ACC CCT ACA




TAA TCA ACG TCA C





cB4203-HM8-M
15
ATT GTA ATA TGG GGA ACC CTC AGT AAT ACA




AGA ACC AG GAA TC





cB4203-HM9-M
16
ATT GTA ATA TGG GGA ACC CTC GAA AAT ACA




AGA ACC AAG GAA TC





cB4203-HM10-M
17
ATT GTA ATA TGG GGA ACC TGG GAA AAT ACA




AGA ACC AG GAA TC





4203-HM11
18
ACA CTA CAC TGA ACC CCT TCA TAC TCA ACG




TCA CCC TAG CAG CCA





4203-HM12
19
ACA CTA CAC TGA ACC CCT TCA TAC TCA ACG




TCA CCC TAT CAG GCA AAA TGT GTA GCC AAG




TGC





SASP-LN-del-R
20
TAA CAG GAG GTG CTC TGA AAG AGT AAG CAG




AGG AG





I465-6H-not
21
CTA ATT GCG GCC GCT CAT TAG TGG TGA TGG




TGA TGA TGT ATA CAG ACA CCA TCA GC





F468-6H-not
22
CTA ATT GCG GCC GCT CAT TAG TGG TGA TGG




TGA TGA TGA AAA GCA TCT ATA CAG ACA CC





P471-6H-not
23
CTA ATT GCG GCC GCT CAT TAG TGG TGA TGG




TGA TGA TGA GGT TTT AGA AAA GCA TCT ATA




C





PH20-HM17
24
TCC AGG CCC AGA GGA AAG GCC GGT TGG GTA




GCA AGG GGC CCC TAA AAG AGT AAG CAG AGG




AG





PH20-HM18
25
TCA CTT GGG GCA TTC CAG ACG GTG GTG AAG




GGC CGG TTG GGT AGC AAG GGG CCC CTA AAA




GAG TAA GCA GAG GAG





K470-not
26
CTA ATT GCG GCC GCT CAT TAG TGG TGA TGG




TGA TGA TGT TTT AGA AAA GCA TCT ATA C





F468-not
27
CTA ATT GCG GCC GCT CAT TAA AAA GCA TCT




ATA CAG ACA CC





M21-mega-F
28
AAT TGT AAT ATG GGG AAG CTG GGA AAA TAC




AAG AA





M24-R
29
TGG AAT AAC AGG AGG TGC AGA GTA AGC AGA




GGA GA





M25-R
30
TTT GGA ATA ACA GGA GAG TAA GCA GAG GAG




A





B4-HM26
31
AGT TTT GAA ATT CCT TTC TCT GGA TGA GCT




GGA GCA CAG CCT GGG GGA GAG TGC GGC CCA




GGG TGC TTC TGG AAT TG





B4-HM27
32
ATG AGC TGG AGC ACA GCT TTG GGG AGA GTG




CGG CCC AG





B4-HM28
33
ATT CCT TTC TCA AGA TGA ACT TGA GCA CAG




CTT TGG CGA AAC TGT TGC





B4-HM29
34
CTA ATT GCG GCC GCT CAT TAG TGG TGA TGG




TGA TGA TGA GCA TCT ATA CAG ACA CC





B4-HM30
35
CTA ATT GCG GCC GCT CAT TAG TGG TGA TGG




TGA TGA TGA CAG ACA CCA TCA GCA ATA C





B4-HM31
36
CTA ATT GCG GCC GCT CAT TAG TGG TGA TGG




TGA TGA TGA TCA GCA ATA CAC ACA TC





B4-HM32
37
CTA ATT GCG GCC GCT CAT TAG TGG TGA TGG




TGA TGA TGA CAC ACA TCA ACA GCA TC





B4-HM33
38
CTA ATT GCG GCC GCT CAT TAG TGG TGA TGG




TGA TGA TGA ACA GCA TCA GTG TCT TTT AC





P472-not
39
GTT ATA GCG GCC GCT CAT TAG GGA GGT TTT




AGA AAA GCA TC





M473-not
40
GTT ATA GCG GCC GCT CAT TAC ATG GGA GGT




TTT AGA AAA GCA TC





E474-not
41
GTT ATA GCG GCC GCT CAT TAC TCC ATG GGA




GGT TTT AGA AAA GC





T475-not
42
GTT ATA GCG GCC GCT CAT TAT GTC TCC ATG




GGA GGT TTT AG





M40-mega
43
TAA CAG GAG GTG CTC TGA AAT TAG AGT AAG




CAG AGG AG





M41-mega
44
TGG AAT AAC AGG AGG TGC TCT AGA GTA AGC




AGA GGA G





M42-mega
45
TTT GGA ATA ACA GGA GGA GAG TAA GCA GAG




GAG





D466-6H-not
46
CTA ATT GCG GCC GCT CAT TAG TGG TGA TGG




TGA TGA TGA TCT ATA CAG ACA CCA TCA GC





P478-H-Not
47
GAT AAT GCG GCC GCT CAT TAG TGG TGA TGG




TGA TGA TGA GGT TCT TCT GTC TCC ATG GG





I480-H-Not
48
GAT AAT GCG GCC GCT CAT TAG TGG TGA TGG




TGA TGA TGA ATT TGA GGT TCT TCT GTC TCC





Y482-H-Not
49
GAT AAT GCG GCC GCT CAT TAG TGG TGA TGG




TGA TGA TGG TAG AAA ATT TGA GGT TCT TCT




G





A484-H-not
50
GAT AAT GCG GCC GCT CAT TAG TGG TGA TGG




TGA TGA TGA GCA TTG TAG AAA ATT TGA GGT




TC





P486-H-not
51
GAT AAT GCG GCC GCT CAT TAG TGG TGA TGG




TGA TGA TGG GGT GAA GCA TTG TAG AAA ATT




TGA GG





T488-H-not
52
GAT AAT GCG GCC GCT CAT TAG TGG TGA TGG




TGA TGA TGT GTG GAG GGT GAA GCA TTG TAG





K470-6H-not
53
CTA ATT GCG GCC GCT CAT TAG TGG TGA TGG




TGA TGA TGT TTT AGA AAA GCA TCT ATA C





P472-6H-not
54
GTT ATA GCG GCC GCT CAT TAG TGG TGA TGG




TGA TGA TGG GGA GGT TTT AGA AAA GCA TC





M473-6H-not
55
GTT ATA GCG GCC GCT CAT TAG TGG TGA TGG




TGA TGA TGC ATG GGA GGT TTT AGA AAA GCA




TC





E474-6H-not
56
GTT ATA GCG GCC GCT CAT TAG TGG TGA TGG




TGA TGA TGC TCC ATG GGA GGT TTT AGA AAA




GC





T475-6H-not
57
GTT ATA GCG GCC GCT CAT TAG TGG TGA TGG




TGA TGA TGT GTC TCC ATG GGA GGT TTT AG





E476-6H-not
58
GTT ATA GCG GCC GCT CAT TAG TGG TGA TGG




TGA TGA TGT TCT GTC TCC ATG GGA GG





E476-not
59
GTT ATA GCG GCC GCT CAT TAT TCT GTC TCC




ATG GGA GG









After finding the PH20 variant with increased enzymatic activity and thermal stability, the 6×His-tag-free cDNA of the PH20 variant was also constructed.


When the cell density of ExpiCHO cells reached 6×106/mL, a plasmid comprising the wild-type or variant PH20 cDNA inserted in the pcDNA3.4-TOPO vector was transfected into the ExpiCHO cells by ExpiFectamine CHO reagent. As a cell culture medium, ExpiCHO expression medium (100 to 500 mL) was used. After transfection, the ExpiCHO cells were shake-cultured at 130 rpm for total 6 days, during which the cells were cultured at 37° C. for 1 day and further cultured at lower temperature of 32° C. for 5 days. After completion of the culture, the cell supernatant was collected by centrifugation at 10,000 rpm for 30 min.


The recombinant proteins of the C-terminal 6×His-attached wild-type PH20 and variant PH20, produced in the ExpiCHO cells, were purified in three steps (performed using a HisTrap column, a Q Sepharose column and a Phenyl column, respectively) by an AKTA prime system.


For protein purification using the HisTrap column, buffer A (20 mM sodium phosphate, pH 7.5, 0.5 M NaCl) and buffer B (20 mM sodium phosphate, pH 7.5, 0.5 M NaCl, 0.5 M imidazole) were prepared. The protein was bound to the HisTrap column, and the column was flushed with 5 column volumes (CV) of buffer A to remove non-specifically bound proteins. It was confirmed that the conductivity was maintained at constant level, the column was flushed with 5 CV of 20% buffer B to elute the protein. The eluted protein was dialyzed with dialysis buffer (20 mM sodium phosphate, pH 7.5, 50 mM NaCl). For protein purification using the Q Sepharose column, buffer A (20 mM sodium phosphate, pH 7.5) and buffer B (20 mM sodium phosphate, pH 7.5, 0.5 M NaCl) were prepared. The protein was bound to the Q Sepharose column, and the column was flushed with 5 CV of buffer A to remove nonspecifically bound proteins, and then 5 CV of buffer B was flushed at concentration gradient of 0 to 100% to elute the protein.


For protein purification using the phenyl column, buffer A (20 mM sodium phosphate, pH 7.0, 1.5 M (NH4)2SO4) and buffer B (20 mM sodium phosphate, pH 7.0) were prepared. The protein was bound to the phenyl column, and the column was flushed with 5 CV of buffer A to remove nonspecifically bound proteins, and then 5 CV of buffer B was flushed at concentration gradient of 0 to 100% to elute the protein.


The enzymatic activities of wild-type PH20 and variant PH20 were measured by turbidimetric assay, substrate-gel assay, and Morgan-Elson assay.


The turbidimetric assay is a method of measuring the absorbance in the precipitate produced when hyaluronic acid is mixed with albumin (BSA). When hyaluronic acid is hydrolyzed by PH20, the absorbance of the precipitate produced when mixed with albumin decreases. Hyaluronidase PH20 (Sigma) was diluted to 1, 2, 5, 7.5, 10, 15, 20, 30, 50 and 60 units/mL, and prepared in each tube. The purified protein sample was dissolved in enzyme diluent buffer (20 mM Tris.HCl, pH 7.0, 77 mM NaCl, 0.01% (w/v) bovine serum albumin) and diluted to 100×, 300×, 600×, 1200× and 2400× and prepared in each tube. In fresh tubes, the hyaluronic acid solution having a concentration of 3 mg/mL was diluted 10-fold to a concentration of 0.3 mg/mL so that the volume of each tube became 180 μL. 60 μL of enzyme was added to and mixed with the diluted hyaluronic acid solution and allowed to react at 37° C. for 45 min. After completion of the reaction, 50 μL of the reacted enzyme and 250 μL of acidic albumin solution were added to each well of a 96-well plate and shaken for 10 min, and then the absorbance was measured at 600 nm by spectrophotometer.


In the substrate-gel assay, the protein was electrophoresed on 10% SDS gel (including 0.17 mg/mL hyaluronic acid) for 1 h, and SDS was removed with 2.5% Triton X-100 (w/v) at 4° C. for 2 h. Thereafter, an enzymatic reaction was performed in the buffer (50 mM sodium phosphate, pH 7.0, 150 mM NaCl) at 37° C. (which is the optimum temperature for PH20) for 1 to 4 h, and the protein was stained with 0.5% Alcian blue reagent. Alcian blue reagent not bound to the hyaluronic acid was removed using de-staining solution. The SDS gel stained with Alcian blue was imaged, and then the band was quantified.


The thermal stability of the protein was measured by a method of measuring the aggregation temperature by dynamic light scattering (DLS), a method of measuring the melting temperature (Tm) in real-time PCR using Sypro-Orange dye, a method of measuring the enzymatic activity after leaving the protein to stand at predetermined temperature for a predetermined time, etc. In the method of measuring the aggregation temperature by DLS, the aggregation of molecules is measured using light scattering, and thus the sensitivity is high and the aggregation temperature is generally lower than the melting temperature of the protein.


The sequences of the substituted or cleaved amino acids in the PH20 variants constructed in the present invention are shown in Table 11 below.


Among the variants according to the present invention, the variant having the 6×His-tag attached to the C-terminus of PH20 was named HM; the variant free of the 6×His-tag was named HP; mature PH20 (L36 to S490) having the 6×His-tag attached to the C-terminus was named WT; and mature wild-type PH20 (L36 to Y482) having the C-terminus cleaved after Y482 while being free of the 6×His-tag was named HW2.









TABLE 11







Amino acid sequences of PH20 variants according


to the present invention and substitution/cleavage 


characteristics thereof 











SEQ




Name
NO.
Substitution
Amino Acid Sequence













HM1
60
12 amino acids
LNFRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDM




substitution of
SLFSFIGSPRINATGQGVTIFYVDRLGYYPYIDSI




M345T, S347T,
TGVIVNGGIPQKISLQDHLDKAKKDITFYMPVDNL




M348K, K349E,
GMAVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQ




L352Q, L353A,
NVQLSLTEATEKAKQEFEKAGKDFLVETIKLGKLL




L354I, D355K,
RPNHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKR




N356E, E359D,
NDDLSWLWNESTALYPSIYLNTQQSPVAATLYVRN




I361T and
RVREAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKF




N363G.
LSQDELVYTFGETVALGASGIVIWGILSITRTKES





CQAIKEYMDTTLGPYIINVTLAAKMCSQVLCQEQG





VCIRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGK





PTLEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAV





DVCIADGVCIDAFLKPPMETEEPQIFYNASPSTLS





HM2
61
7 amino acids
LNFRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDM




substitution of
SLFSFIGSPRINATGQGVTIFYVDRLGYYPYIDSI




Y365F, I367L,
TGVIVNGGIPQKISLQDHLDKAKKDITFYMPVDNL




L371S, A372G,
GMAVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQ




K374L, M375L
NVQLSLTEATEKAKQEFEKAGKDFLVETIKLGKLL




and V379A
RPNHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKR





NDDLSWLWNESTALYPSIYLNTQQSPVAATLYVRN





RVREAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKF





LSQDELVYTFGETVALGASGIVIWGILSITRTKES





CQAIKEYMDTTLNPFILNVISGALLCSQALCQEQG





VCIRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGK





PTLEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAV





DVCIADGVCIDAFLKPPMETEEPQIFYNASPSTLS





HM3
62
19 amino acids
LNFRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDM




substitution of
SLFSFIGSPRINATGQGVTIFYVDRLGYYPYIDSI




M345T, S347T,
TGVIVNGGIPQKISLQDHLDKAKKDITFYMPVDNL




M348K, K349E,
GMAVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQ




L352Q, L353A,
NVQLSLTEATEKAKQEFEKAGKDFLVETIKLGKLL




L354I, D355K,
RPNHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKR




N356E, E359D,
NDDLSWLWNESTALYPSIYLNTQQSPVAATLYVRN




I361T, N363G,
RVREAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKF




Y365F, I367L,
LSQDELVYTFGETVALGASGIVIWGILSITRTKES




L371S, A372G,
CQAIKEYMDTTLGPFILNVISGALLCSQALCQEQG




K374L, M375L
VCIRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGK




and V379A
PTLEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAV





DVCIADGVCIDAFLKPPMETEEPQIFYNASPSTLS





HM4
63
17 amino acids
LNFRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDM




substitution of
SLFSFIGSPRINATGQGVTIFYVDRLGYYPYIDSI




G340V, T341S,
TGVIVNGGIPQKISLQDHLDKAKKDITFYMPVDNL




L342W, S343E,
GMAVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQ




I344N, M345T,
NVQLSLTEATEKAKQEFEKAGKDFLVETIKLGKLL




S347T, M348K,
RPNHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKR




K349E, L352Q,
NDDLSWLWNESTALYPSIYLNTQQSPVAATLYVRN




L353A, L354I,
RVREAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKF




D355K, N356E,
LSQDELVYTFGETVALGASGIVIWVSWENTRTKES




E359D, I361T
CQAIKEYMDTTLGPYIINVTLAAKMCSQVLCQEQG




and N363G
VCIRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGK





PTLEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAV





DVCIADGVCIDAFLKPPMETEEPQIFYNASPSTLS





HM6
64
11 amino acids
LNFRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDM




substitution of
SLFSFIGSPRINATGQGVTIFYVDRLGYYPYIDSI




M345T, S347T,
TGVTVNGGIPQKISLQDHLDKAKKDITFYMPVDNL




M348K, K349E,
GMAVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQ




L352Q, L353A,
NVQLSLTEATEKAKQEFEKAGKDFLVETIKLGKLL




L354I, D355K,
RPNHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKR




N356E, E359D
NDDLSWLWNESTALYPSIYLNTQQSPVAATLYVRN




and I361T
RVREAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKF





LSQDELVYTFGETVALGASGIVIWGTLSITRTKES





CQAIKEYMDTTLNPYIINVTLAAKMCSQVLCQEQG





VCIRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGK





PTLEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAV





DVCIADGVCIDAFLKPPMETEEPQIFYNASPSTLS





HM7
65
16 amino acids
LNFRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDM




substitution of
SLFSFIGSPRINATGQGVTIFYVDRLGYYPYIDSI




G340V, T341S
TGVTVNGGIPQKISLQDHLDKAKKDITFYMPVDNL




L342W, S343E,
GMAVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQ




I344N, M345T,
NVQLSLTEATEKAKQEFEKAGKDFLVETIKLGKLL




S347T, M348K,
RPNHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKR




K349E, L352Q,
NDDLSWLWNESTALYPSIYLNTQQSPVAATLYVRN




L353A, L354I,
RVREAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKF




D355K, N356E,
LSQDELVYTFGETVALGASGIVIWVSWENTRTKES




E359D and
CQAIKEYMDTTLNPYIINVTLAAKMCSQVLCQEQG




I361T
VCIRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGK





PTLEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAV





DVCIADGVCIDAFLKPPMETEEPQIFYNASPSTLS





HM8
66
12 amino acids
LNFRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDM




substitution of
SLFSFIGSPRINATGQGVTIFYVDRLGYYPYIDSI




I344N, M345T,
TGVTVNGGIPQKISLQDHLDKAKKDITFYMPVDNL




S347T, M348K,
GMAVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQ




K349E, L352Q,
NVQLSLTEATEKAKQEFEKAGKDFLVETIKLGKLL




L353A, L354I,
RPNHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKR




D355K, N356E,
NDDLSWLWNESTALYPSIYLNTQQSPVAATLYVRN




E359D and
RVREAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKF




I361T
LSQDELVYTFGETVALGASGIVIWGTLSNTRTKES





CQAIKEYMDTTLNPYIINVTLAAKMCSQVLCQEQG





VCIRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGK





PTLEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAV





DVCIADGVCIDAFLKPPMETEEPQIFYNASPSTLS





HM9
67
13 amino acids
LNFRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDM




substitution of
SLFSFIGSPRINATGQGVTIFYVDRLGYYPYIDSI




S343E, I344N,
TGVTVNGGIPQKISLQDHLDKAKKDITFYMPVDNL




M345T, S347T,
GMAVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQ




M348K, K349E,
NVQLSLTEATEKAKQEFEKAGKDFLVETIKLGKLL




L352Q, L353A,
RPNHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKR




L354I, D355K,
NDDLSWLWNESTALYPSIYLNTQQSPVAATLYVRN




N356E, E359D
RVREAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKF




and I361T
LSQDELVYTFGETVALGASGIVIWGTLENTRTKES





CQAIKEYMDTTLNPYIINVTLAAKMCSQVLCQEQG





VCIRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGK





PTLEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAV





DVCIADGVCIDAFLKPPMETEEPQIFYNASPSTLS





HM10
68
14 amino acids
LNFRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDM




substitution of
SLFSFIGSPRINATGQGVTIFYVDRLGYYPYIDSI




L342W, S343E,
TGVTVNGGIPQKISLQDHLDKAKKDITFYMPVDNL




I344N, M345T,
GMAVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQ




S347T, M348K,
NVQLSLTEATEKAKQEFEKAGKDFLVETIKLGKLL




K349E, L352Q,
RPNHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKR




L353A, L354I,
NDDLSWLWNESTALYPSIYLNTQQSPVAATLYVRN




D355K, N356E,
RVREAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKF




E359D and
LSQDELVYTFGETVALGASGIVIWGTWENTRTKES




I361T
CQAIKEYMDTTLNPYIINVTLAAKMCSQVLCQEQG





VCIRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGK





PTLEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAV





DVCIADGVCIDAFLKPPMETEEPQIFYNASPSTLS





HM11
69
13 amino acids
LNFRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDM




substitution of
SLFSFIGSPRINATGQGVTIFYVDRLGYYPYIDSI




M345T, S347T,
TGVTVNGGIPQKISLQDHLDKAKKDITFYMPVDNL




M348K, K349E,
GMAVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQ




L352Q, L353A,
NVQLSLTEATEKAKQEFEKAGKDFLVETIKLGKLL




L354I, D355K,
RPNHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKR




N356E, E359D,
NDDLSWLWNESTALYPSIYLNTQQSPVAATLYVRN




I361T, Y365F
RVREAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKF




and I367L
LSQDELVYTFGETVALGASGIVIWGTLSITRTKES





CQAIKEYMDTTLNPFILNVTLAAKMCSQVLCQEQG





VCIRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGK





PTLEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAV





DVCIADGVCIDAFLKPPMETEEPQIFYNASPSTLS





HM12
70
15 amino acids
LNFRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDM




substitution of
SLFSFIGSPRINATGQGVTIFYVDRLGYYPYIDSI




M345T, S347T,
TGVTVNGGIPQKISLQDHLDKAKKDITFYMPVDNL




M348K, K349E,
GMAVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQ




L352Q, L353A,
NVQLSLTEATEKAKQEFEKAGKDFLVETIKLGKLL




L354I, D355K,
RPNHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKR




N356E, E359D,
NDDLSWLWNESTALYPSIYLNTQQSPVAATLYVRN




I361T, Y365F,
RVREAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKF




I367L, L371S
LSQDELVYTFGETVALGASGIVIWGTLSITRTKES




and A372G
CQAIKEYMDTTLNPFILNVISGAKMCSQVLCQEQG





VCIRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGK





PTLEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAV





DVCIADGVCIDAFLKPPMETEEPQIFYNASPSTLS





HM13
71
11 amino acids
FRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDMSL




substitution of
FSFIGSPRINATGQGVTIFYVDRLGYYPYIDSITG




M345T, S347T,
VTVNGGIPQKISLQDHLDKAKKDITFYMPVDNLGM




M348K, K349E,
AVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQNV




L352Q, L353A,
QLSLTEATEKAKQEFEKAGKDFLVETIKLGKLLRP




L354I, D355K,
NHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKRND




N356E, E359D
DLSWLWNESTALYPSIYLNTQQSPVAATLYVRNRV




and I361T,
REAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKFLS




and truncation
QDELVYTFGETVALGASGIVIWGTLSITRTKESCQ




before F38


AIKE
YMDTTLNPYIINVTLAAKMCSQVLCQEQGVC





at the
IRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGKPT




N-terminus
LEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAVDV





CIADGVCIDAFLKPPMETEEPQIFYNASPSTLS





HM14
72
11 amino acids
LNFRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDM




substitution of
SLFSFIGSPRINATGQGVTIFYVDRLGYYPYIDSI




M345T, S347T,
TGVTVNGGIPQKISLQDHLDKAKKDITFYMPVDNL




M348K, K349E,
GMAVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQ




L352Q, L353A,
NVQLSLTEATEKAKQEFEKAGKDFLVETIKLGKLL




L354I, D355K,
RPNHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKR




N356E, E359D
NDDLSWLWNESTALYPSIYLNTQQSPVAATLYVRN




and I361T
RVREAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKF




and truncation 
LSQDELVYTFGETVALGASGIVIWGTLSITRTKES




after I465
CQAIKEYMDTTLNPYIINVTLAAKMCSQVLCQEQG




at the
VCIRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGK




C-terminus
PTLEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAV





DVCIADGVCI





HM15
73
11 amino acids
LNFRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDM




substitution of
SLFSFIGSPRINATGQGVTIFYVDRLGYYPYIDSI




M345T, S347T,
TGVTVNGGIPQKISLQDHLDKAKKDITFYMPVDNL




M348K, K349E,
GMAVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQ




L352Q, L353A,
NVQLSLTEATEKAKQEFEKAGKDFLVETIKLGKLL




L354I, D355K,
RPNHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKR




N356E, E359D
NDDLSWLWNESTALYPSIYLNTQQSPVAATLYVRN




and I361T,
RVREAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKF




and truncation
LSQDELVYTFGETVALGASGIVIWGTLSITRTKES




after F468
CQAIKEYMDTTLNPYIINVTLAAKMCSQVLCQEQG




at the
VCIRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGK




C-terminus
PTLEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAV





DVCIADGVCIDAF





HM16
74
11 amino acids
LNFRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDM




substitution of
SLFSFIGSPRINATGQGVTIFYVDRLGYYPYIDSI




M345T, S347T,
TGVTVNGGIPQKISLQDHLDKAKKDITFYMPVDNL




M348K, K349E,
GMAVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQ




L352Q, L353A,
NVQLSLTEATEKAKQEFEKAGKDFLVETIKLGKLL




L354I, D355K,
RPNHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKR




N356E, E359D
NDDLSWLWNESTALYPSIYLNTQQSPVAATLYVRN




and I361T,
RVREAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKF




and truncation
LSQDELVYTFGETVALGASGIVIWGTLSITRTKES




after P471
CQAIKEYMDTTLNPYIINVTLAAKMCSQVLCQEQG




at the
VCIRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGK




C-terminus
PTLEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAV





DVCIADGVCIDAFLKP





HM17
75
Substitution of


FRGPLLPNR
PFLWAWNAPSEFCLGKFDEPLDMSLF





L36~V47 with
SFIGSPRINATGQGVTIFYVDRLGYYPYIDSITGV




FRGPLLPNR, and
TVNGGIPQKISLQDHLDKAKKDITFYMPVDNLGMA




amino acids
VIDWEEWRPTWARNWKPKDVYKNRSIELVQQQNVQ




substitution of
LSLTEATEKAKQEFEKAGKDFLVETIKLGKLLRPN




M345T, S347T,
HLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKRNDD




M348K, K349E,
LSWLWNESTALYPSIYLNTQQSPVAATLYVRNRVR




L352Q, L353A,
EAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKFLSQ




L354I, D355K,
DELVYTFGETVALGASGIVIWGILSITRTKESCQA




N356E, E359D


IKE
YMDTTLNPYIINVTLAAKMCSQVLCQEQGVCI





and I361T
RKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGKPTL





EDLEQFSEKFYCSCYSTLSCKEKADVKDTDAVDVC





IADGVCIDAFLKPPMETEEPQIFYNASPSTLS





HM18
76
Substitution of


FRGPLLPNRPFTTV
WNAPSEFCLGKFDEPLDMSLF





L36~A52 with
SFIGSPRINATGQGVTIFYVDRLGYYPYIDSITGV




FRGPLLPNRPFTTV,
TVNGGIPQKISLQDHLDKAKKDITFYMPVDNLGMA




and amino acids
VIDWEEWRPTWARNWKPKDVYKNRSIELVQQQNVQ




substitution of
LSLTEATEKAKQEFEKAGKDFLVETIKLGKLLRPN




M345T, S347T,
HLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKRNDD




M348K, K349E,
LSWLWNESTALYPSIYLNTQQSPVAATLYVRNRVR




L352Q, L353A,
EAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKFLSQ




L354I, D355K,
DELVYTFGETVALGASGIVIWGILSITRTKESCQA




N356E, E359D


IKE
YMDTTLNPYIINVTLAAKMCSQVLCQEQGVCI





and I361T
RKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGKPTL





EDLEQFSEKFYCSCYSTLSCKEKADVKDTDAVDVC





IADGVCIDAFLKPPMETEEPQIFYNASPSTLS





HM19
77
14 amino acids
FRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDMSL




substitution of
FSFIGSPRINATGQGVTIFYVDRLGYYPYIDSITG




L342W, S343E,
VTVNGGIPQKISLQDHLDKAKKDITFYMPVDNLGM




I344N, M345T,
AVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQNV




S347T, M348K,
QLSLTEATEKAKQEFEKAGKDFLVETIKLGKLLRP




K349E, L352Q,
NHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKRND




L353A, L354I,
DLSWLWNESTALYPSIYLNTQQSPVAATLYVRNRV




D355K, N356E,
REAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKFLS




E359D and
QDELVYTFGETVALGASGIVIWGTWENTRTKESCQ




I361T,


AIKE
YMDTTLNPYIINVTLAAKMCSQVLCQEQGVC





truncation
IRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGKPT




before F38 at
LEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAVDV




the N-terminus,
CIADGVCIDAFLK




and truncation





after K470 at





the C-terminus






HM20
78
14 amino acids
FRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDMSL




substitution of
FSFIGSPRINATGQGVTIFYVDRLGYYPYIDSITG




L342W, S343E,
VTVNGGIPQKISLQDHLDKAKKDITFYMPVDNLGM




I344N, M345T,
AVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQNV




S347T, M348K,
QLSLTEATEKAKQEFEKAGKDFLVETIKLGKLLRP




K349E, L352Q,
NHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKRND




L353A, L354I,
DLSWLWNESTALYPSIYLNTQQSPVAATLYVRNRV




D355K, N356E,
REAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKFLS




E359D and
QDELVYTFGETVALGASGIVIWGTWENTRTKESCQ




I361T,


AIKE
YMDTTLNPYIINVTLAAKMCSQVLCQEQGVC





truncation
IRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGKPT




before F38 at
LEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAVDV




the N-terminus,
CIADGVCIDAF




and truncation





after F468 at





the C-terminus






HM21
79
15 amino acids
LNFRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDM




substitution of
SLFSFIGSPRINATGQGVTIFYVDRLGYYPYIDSI




T341S, L342W,
TGVTVNGGIPQKISLQDHLDKAKKDITFYMPVDNL




S343E, I344N,
GMAVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQ




M345T, S347T,
NVQLSLTEATEKAKQEFEKAGKDFLVETIKLGKLL




M348K, K349E,
RPNHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKR




L352Q, L353A,
NDDLSWLWNESTALYPSIYLNTQQSPVAATLYVRN




L354I, D355K,
RVREAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKF




N356E, E359D
LSQDELVYTFGETVALGASGIVIWGSWENTRTKES




and I361T
CQAIKEYMDTTLNPYIINVTLAAKMCSQVLCQEQG





VCIRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGK





PTLEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAV





DVCIADGVCIDAFLKPPMETEEPQIFYNASPSTLS





HM24
80
11 amino acids
APPVIPNVPFLWAWNAPSEFCLGKFDEPLDMSLFS




substitution of
FIGSPRINATGQGVTIFYVDRLGYYPYIDSITGVT




M345T, S347T,
VNGGIPQKISLQDHLDKAKKDITFYMPVDNLGMAV




M348K, K349E,
IDWEEWRPTWARNWKPKDVYKNRSIELVQQQNVQL




L352Q, L353A,
SLTEATEKAKQEFEKAGKDFLVETIKLGKLLRPNH




L354I, D355K,
LWGYYLFPDCYNHHYKKPGYNGSCFNVEIKRNDDL




N356E, E359D
SWLWNESTALYPSIYLNTQQSPVAATLYVRNRVRE




and I361T,
AIRVSKIPDAKSPLPVFAYTRIVFTDQVLKFLSQD




and truncation
ELVYTEGETVALGASGIVIWGILSITRTKESCQAI




before A40 at


KE
YMDTTLNPYIINVTLAAKMCSQVLCQEQGVCIR





the N-terminus
KNWNSSDYLHLNPDNFAIQLEKGGKFTVRGKPTLE





DLEQFSEKFYCSCYSTLSCKEKADVKDTDAVDVCI





ADGVCIDAFLKPPMETEEPQIFYNASPSTLS





HM25
81
11 amino acids
PVIPNVPFLWAWNAPSEFCLGKFDEPLDMSLFSFI




substitution of
GSPRINATGQGVTIFYVDRLGYYPYIDSITGVIVN




M345T, S347T,
GGIPQKISLQDHLDKAKKDITFYMPVDNLGMAVID




M348K, K349E,
WEEWRPTWARNWKPKDVYKNRSIELVQQQNVQLSL




L352Q, L353A,
TEATEKAKQEFEKAGKDFLVETIKLGKLLRPNHLW




L354I, D355K,
GYYLFPDCYNHHYKKPGYNGSCFNVEIKRNDDLSW




N356E, E359D
LWNESTALYPSIYLNTQQSPVAATLYVRNRVREAI




and I361T,
RVSKIPDAKSPLPVFAYTRIVFTDQVLKFLSQDEL




truncation
VYTEGETVALGASGIVIWGILSITRTKESCQAIKE




before P42
YMDTTLNPYIINVTLAAKMCSQVLCQEQGVCIRKN




at the
WNSSDYLHLNPDNFAIQLEKGGKFTVRGKPTLEDL




N-terminus
EQFSEKFYCSCYSTLSCKEKADVKDTDAVDVCIAD





GVCIDAFLKPPMETEEPQIFYNASPSTLS





HM29
82
14 amino acids
LNFRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDM




substitution of
SLFSFIGSPRINATGQGVTIFYVDRLGYYPYIDSI




L342W, S343E,
TGVIVNGGIPQKISLQDHLDKAKKDITFYMPVDNL




I344N, M345T,
GMAVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQ




S347T, M348K,
NVQLSLTEATEKAKQEFEKAGKDFLVETIKLGKLL




K349E, L352Q,
RPNHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKR




L353A, L354I,
NDDLSWLWNESTALYPSIYLNTQQSPVAATLYVRN




D355K, N356E,
RVREAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKF




E359D and
LSQDELVYTEGETVALGASGIVIWGTWENTRTKES




I361T,
CQAIKEYMDTTLNPYIINVTLAAKMCSQVLCQEQG




truncation
VCIRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGK




before L36
PTLEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAV




at the
DVCIADGVCIDA




N-terminus,





and





truncation





after A467





at the





C-terminus






HM30
83
14 amino acids
LNFRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDM




substitution of
SLFSFIGSPRINATGQGVTIFYVDRLGYYPYIDSI




L342W, S343E,
TGVIVNGGIPQKISLQDHLDKAKKDITFYMPVDNL




I344N, M345T,
GMAVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQ




S347T, M348K,
NVQLSLTEATEKAKQEFEKAGKDFLVETIKLGKLL




K349E, L352Q,
RPNHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKR




L353A, L354I,
NDDLSWLWNESTALYPSIYLNTQQSPVAATLYVRN




D355K, N356E,
RVREAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKF




E359D and
LSQDELVYTEGETVALGASGIVIWGTWENTRTKES




I361T,
CQAIKEYMDTTLNPYIINVTLAAKMCSQVLCQEQG




truncation
VCIRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGK




before L36
PTLEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAV




at the
DVCIADGVC




N-terminus,





and





truncation





after C464





at the





C-terminus






HM31
84
14 amino acids
LNFRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDM




substitution of
SLFSFIGSPRINATGQGVTIFYVDRLGYYPYIDSI




L342W, S343E,
TGVIVNGGIPQKISLQDHLDKAKKDITFYMPVDNL




I344N, M345T,
GMAVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQ




S347T, M348K,
NVQLSLTEATEKAKQEFEKAGKDFLVETIKLGKLL




K349E, L352Q,
RPNHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKR




L353A, L354I,
NDDLSWLWNESTALYPSIYLNTQQSPVAATLYVRN




D355K, N356E,
RVREAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKF




E359D and
LSQDELVYTEGETVALGASGIVIWGTWENTRTKES




I361T,
CQAIKEYMDTTLNPYIINVTLAAKMCSQVLCQEQG




truncation
VCIRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGK




before L36
PTLEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAV




at the
DVCIAD




N-terminus,





and truncation





after D461





at the





C-terminus






HM32
85
14 amino acids
LNFRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDM




substitution of
SLFSFIGSPRINATGQGVTIFYVDRLGYYPYIDSI




L342W, S343E,
TGVIVNGGIPQKISLQDHLDKAKKDITFYMPVDNL




I344N, M345T,
GMAVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQ




S347T, M348K,
NVQLSLTEATEKAKQEFEKAGKDFLVETIKLGKLL




K349E, L352Q,
RPNHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKR




L353A, L354I,
NDDLSWLWNESTALYPSIYLNTQQSPVAATLYVRN




D355K, N356E,
RVREAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKF




E359D and
LSQDELVYTEGETVALGASGIVIWGTWENTRTKES




I361T,
CQAIKEYMDTTLNPYIINVTLAAKMCSQVLCQEQG




truncation
VCIRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGK




before L36
PTLEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAV




at the
DVC




N-terminus,





and





truncation





after C458





at the





C-terminus






HM33
86
14 amino acids
LNFRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDM




substitution of
SLFSFIGSPRINATGQGVTIFYVDRLGYYPYIDSI




L342W, S343E,
TGVIVNGGIPQKISLQDHLDKAKKDITFYMPVDNL




I344N, M345T,
GMAVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQ




S347T, M348K,
NVQLSLTEATEKAKQEFEKAGKDFLVETIKLGKLL




K349E, L352Q,
RPNHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKR




L353A, L354I,
NDDLSWLWNESTALYPSIYLNTQQSPVAATLYVRN




D355K, N356E,
RVREAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKF




E359D and
LSQDELVYTEGETVALGASGIVIWGTWENTRTKES




I361T,
CQAIKEYMDTTLNPYIINVTLAAKMCSQVLCQEQG




truncation
VCIRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGK




before L36
PTLEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAV




at the





N-terminus,





and





truncation





after V455





at the





C-terminus






HP34
87
15 amino acids
FRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDMSL




substitution of
FSFIGSPRINATGQGVTIFYVDRLGYYPYIDSITG




T341S, L342W,
VTVNGGIPQKISLQDHLDKAKKDITFYMPVDNLGM




S343E, I344N,
AVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQNV




M345T, S347T,
QLSLTEATEKAKQEFEKAGKDFLVETIKLGKLLRP




M348K, K349E,
NHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKRND




L352Q, L353A,
DLSWLWNESTALYPSIYLNTQQSPVAATLYVRNRV




L354I, D355K,
REAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKFLS




N356E, E359D
QDELVYTEGETVALGASGIVIWGSWENTRTKESCQ




and I361T,


AIKE
YMDTTLNPYIINVTLAAKMCSQVLCQEQGVC





truncation
IRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGKPT




before F38 at
LEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAVDV




the N-terminus,
CIADGVCIDAFLK




and





truncation





after K470





at the





C-terminus






HM35
88
14 amino acids
FRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDMSL




substitution of
FSFIGSPRINATGQGVTIFYVDRLGYYPYIDSITG




L342W, S343E,
VTVNGGIPQKISLQDHLDKAKKDITFYMPVDNLGM




I344N, M345T,
AVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQNV




S347T, M348K,
QLSLTEATEKAKQEFEKAGKDFLVETIKLGKLLRP




K349E, L352Q,
NHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKRND




L353A, L354I,
DLSWLWNESTALYPSIYLNTQQSPVAATLYVRNRV




D355K, N356E,
REAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKFLS




E359D and
QDELVYTEGETVALGASGIVIWGTWENTRTKESCQ




I361T,


AIKE
YMDTTLNPYIINVTLAAKMCSQVLCQEQGVC





truncation
IRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGKPT




before F38
LEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAVDV




at the
CIADGVCIDAFLKPP




N-terminus,





and





truncation





after P472





at the





C-terminus






HM36
89
14 amino acids
FRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDMSL




substitution of
FSFIGSPRINATGQGVTIFYVDRLGYYPYIDSITG




L342W, S343E,
VTVNGGIPQKISLQDHLDKAKKDITFYMPVDNLGM




I344N, M345T,
AVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQNV




S347T, M348K,
QLSLTEATEKAKQEFEKAGKDFLVETIKLGKLLRP




K349E, L352Q,
NHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKRND




L353A, L354I,
DLSWLWNESTALYPSIYLNTQQSPVAATLYVRNRV




D355K, N356E,
REAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKFLS




E359D and
QDELVYTEGETVALGASGIVIWGTWENTRTKESCQ




I361T,


AIKE
YMDTTLNPYIINVTLAAKMCSQVLCQEQGVC





truncation
IRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGKPT




before F38
LEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAVDV




at the
CIADGVCIDAFLKPPM




N-terminus,





and





truncation





after M473





at the





C-terminus






HM37
90
14 amino acids
FRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDMSL




substitution of
FSFIGSPRINATGQGVTIFYVDRLGYYPYIDSITG




L342W, S343E,
VTVNGGIPQKISLQDHLDKAKKDITFYMPVDNLGM




I344N, M345T,
AVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQNV




S347T, M348K,
QLSLTEATEKAKQEFEKAGKDFLVETIKLGKLLRP




K349E, L352Q,
NHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKRND




L353A, L354I,
DLSWLWNESTALYPSIYLNTQQSPVAATLYVRNRV




D355K, N356E,
REAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKFLS




E359D and
QDELVYTEGETVALGASGIVIWGTWENTRTKESCQ




I361T,


AIKE
YMDTTLNPYIINVTLAAKMCSQVLCQEQGVC





truncation
IRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGKPT




before
LEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAVDV




F38 at the
CIADGVCIDAFLKPPME




N-terminus,





and





truncation





after





E474 at the





C-terminus






HM38
91
14 amino acids
FRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDMSL




substitution of
FSFIGSPRINATGQGVTIFYVDRLGYYPYIDSITG




L342W, S343E,
VTVNGGIPQKISLQDHLDKAKKDITFYMPVDNLGM




I344N, M345T,
AVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQNV




S347T, M348K,
QLSLTEATEKAKQEFEKAGKDFLVETIKLGKLLRP




K349E, L352Q,
NHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKRND




L353A, L354I,
DLSWLWNESTALYPSIYLNTQQSPVAATLYVRNRV




D355K, N356E,
REAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKFLS




E359D and
QDELVYTEGETVALGASGIVIWGTWENTRTKESCQ




I361T,


AIKE
YMDTTLNPYIINVTLAAKMCSQVLCQEQGVC





truncation
IRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGKPT




before
LEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAVDV




F38 at
CIADGVCIDAFLKPPMET




the N-terminus,





and





truncation





after T475





at the





C-terminus






HM39
92
14 amino acids
FRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDMSL




substitution of
FSFIGSPRINATGQGVTIFYVDRLGYYPYIDSITG




L342W, S343E,
VTVNGGIPQKISLQDHLDKAKKDITFYMPVDNLGM




I344N, M345T,
AVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQNV




S347T, M348K,
QLSLTEATEKAKQEFEKAGKDFLVETIKLGKLLRP




K349E, L352Q,
NHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKRND




L353A, L354I,
DLSWLWNESTALYPSIYLNTQQSPVAATLYVRNRV




D355K, N356E,
REAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKFLS




E359D and
QDELVYTEGETVALGASGIVIWGTWENTRTKESCQ




I361T,


AIKE
YMDTTLNPYIINVTLAAKMCSQVLCQEQGVC





truncation
IRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGKPT




before
LEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAVDV




F38 at the
CIADGVCIDAFLKPPMETE




N-terminus,





and





truncation





after E476





at the





C-terminus






HM40
93
11 amino acids
NFRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDMS




substitution of
LFSFIGSPRINATGQGVTIFYVDRLGYYPYIDSIT




M345T, S347T,
GVTVNGGIPQKISLQDHLDKAKKDITFYMPVDNLG




M348K, K349E,
MAVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQN




L352Q, L353A,
VQLSLTEATEKAKQEFEKAGKDFLVETIKLGKLLR




L354I, D355K,
PNHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKRN




N356E, E359D
DDLSWLWNESTALYPSIYLNTQQSPVAATLYVRNR




and I361T,
VREAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKFL




and
SQDELVYTEGETVALGASGIVIWGILSITRTKESC




truncation


QAIKE
YMDTTLNPYIINVTLAAKMCSQVLCQEQGV





before
CIRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGKP




N37 at the
TLEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAVD




N-terminus
VCIADGVCIDAFLKPPMETEEPQIFYNASPSTLS





HM41
94
11 amino acids
RAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDMSLF




substitution of
SFIGSPRINATGQGVTIFYVDRLGYYPYIDSITGV




M345T, S347T,
TVNGGIPQKISLQDHLDKAKKDITFYMPVDNLGMA




M348K, K349E,
VIDWEEWRPTWARNWKPKDVYKNRSIELVQQQNVQ




L352Q, L353A,
LSLTEATEKAKQEFEKAGKDFLVETIKLGKLLRPN




L354I, D355K,
HLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKRNDD




N356E, E359D
LSWLWNESTALYPSIYLNTQQSPVAATLYVRNRVR




and I361T,
EAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKFLSQ




and
DELVYTEGETVALGASGIVIWGILSITRTKESCQA




truncation


IKE
YMDTTLNPYIINVTLAAKMCSQVLCQEQGVCI





before R39
RKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGKPIL




at the
EDLEQFSEKFYCSCYSTLSCKEKADVKDTDAVDVC




N-terminus
IADGVCIDAFLKPPMETEEPQIFYNASPSTLS





HM42
95
11 amino acids
PPVIPNVPFLWAWNAPSEFCLGKFDEPLDMSLFSF




substitution of
IGSPRINATGQGVTIFYVDRLGYYPYIDSITGVTV




M345T, S347T,
NGGIPQKISLQDHLDKAKKDITFYMPVDNLGMAVI




M348K, K349E,
DWEEWRPTWARNWKPKDVYKNRSIELVQQQNVQLS




L352Q, L353A,
LTEATEKAKQEFEKAGKDFLVETIKLGKLLRPNHL




L354I, D355K,
WGYYLFPDCYNHHYKKPGYNGSCFNVEIKRNDDLS




N356E, E359D
WLWNESTALYPSIYLNTQQSPVAATLYVRNRVREA




and I361T,
IRVSKIPDAKSPLPVFAYTRIVFTDQVLKFLSQDE




and
LVYTEGETVALGASGIVIWGILSITRTKESCQAIK




truncation


E
YMDTTLNPYIINVTLAAKMCSQVLCQEQGVCIRK





before P41
NWNSSDYLHLNPDNFAIQLEKGGKFTVRGKPTLED




at the
LEQFSEKFYCSCYSTLSCKEKADVKDTDAVDVCIA




N-terminus
DGVCIDAFLKPPMETEEPQIFYNASPSTLS





HM43
96
14 amino acids
FRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDMSL




substitution of
FSFIGSPRINATGQGVTIFYVDRLGYYPYIDSITG




L342W, S343E,
VTVNGGIPQKISLQDHLDKAKKDITFYMPVDNLGM




I344N, M345T,
AVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQNV




S347T, M348K,
QLSLTEATEKAKQEFEKAGKDFLVETIKLGKLLRP




K349E, L352Q,
NHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKRND




L353A, L354I,
DLSWLWNESTALYPSIYLNTQQSPVAATLYVRNRV




D355K, N356E,
REAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKFLS




E359D and
QDELVYTEGETVALGASGIVIWGSWENTRTKESCQ




I361T,


AIKE
YMDTTLNPYIINVTLAAKMCSQVLCQEQGVC





truncation
IRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGKPT




before
LEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAVDV




F38 at the
CIADGVCI




N-terminus,





and





truncation





after I465





at the





C-terminus






HM44
97
14 amino acids
FRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDMSL




substitution of
FSFIGSPRINATGQGVTIFYVDRLGYYPYIDSITG




L342W, S343E,
VTVNGGIPQKISLQDHLDKAKKDITFYMPVDNLGM




I344N, M345T,
AVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQNV




S347T, M348K,
QLSLTEATEKAKQEFEKAGKDFLVETIKLGKLLRP




K349E, L352Q,
NHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKRND




L353A, L354I,
DLSWLWNESTALYPSIYLNTQQSPVAATLYVRNRV




D355K, N356E,
REAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKFLS




E359D and
QDELVYTEGETVALGASGIVIWGSWENTRTKESCQ




I361T,


AIKE
YMDTTLNPYIINVTLAAKMCSQVLCQEQGVC





truncation
IRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGKPT




before
LEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAVDV




F38 at the
CIADGVCID




N-terminus,





and





truncation





after D466





at the





C-terminus






HM45
98
14 amino acids
FRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDMSL




substitution of
FSFIGSPRINATGQGVTIFYVDRLGYYPYIDSITG




L342W, S343E,
VTVNGGIPQKISLQDHLDKAKKDITFYMPVDNLGM




I344N, M345T,
AVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQNV




S347T, M348K,
QLSLTEATEKAKQEFEKAGKDFLVETIKLGKLLRP




K349E, L352Q,
NHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKRND




L353A, L354I,
DLSWLWNESTALYPSIYLNTQQSPVAATLYVRNRV




D355K, N356E,
REAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKFLS




E359D and
QDELVYTEGETVALGASGIVIWGSWENTRTKESCQ




I361T,


AIKE
YMDTTLNPYIINVTLAAKMCSQVLCQEQGVC





truncation
IRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGKPT




before
LEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAVDV




F38 at the
CIADGVCIDA




N-terminus,





and





truncation





after A467





at the





C-terminus






HP46
99
15 amino acids
FRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDMSL




substitution of
FSFIGSPRINATGQGVTIFYVDRLGYYPYIDSITG




T341S, L342W,
VTVNGGIPQKISLQDHLDKAKKDITFYMPVDNLGM




S343E, I344N,
AVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQNV




M345T, S347T,
QLSLTEATEKAKQEFEKAGKDFLVETIKLGKLLRP




M348K, K349E,
NHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKRND




L352Q, L353A,
DLSWLWNESTALYPSIYLNTQQSPVAATLYVRNRV




L354I, D355K,
REAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKFLS




N356E, E359D
QDELVYTEGETVALGASGIVIWGSWENTRTKESCQ




and I361T,


AIKE
YMDTTLNPYIINVTLAAKMCSQVLCQEQGVC





truncation
IRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGKPT




before F38
LEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAVDV




at the
CIADGVCIDAF




N-terminus,





and





truncation





after F468





at the





C-terminus






HM47
100
14 amino acids
FRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDMSL




substitution of
FSFIGSPRINATGQGVTIFYVDRLGYYPYIDSITG




L342W, S343E,
VTVNGGIPQKISLQDHLDKAKKDITFYMPVDNLGM




I344N, M345T,
AVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQNV




S347T, M348K,
QLSLTEATEKAKQEFEKAGKDFLVETIKLGKLLRP




K349E, L352Q,
NHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKRND




L353A, L354I,
DLSWLWNESTALYPSIYLNTQQSPVAATLYVRNRV




D355K, N356E,
REAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKFLS




E359D and
QDELVYTEGETVALGASGIVIWGTWENTRTKESCQ




I361T,


AIKE
YMDTTLNPYIINVTLAAKMCSQVLCQEQGVC





truncation
IRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGKPT




before F38
LEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAVDV




at the
CIADGVCIDAFLKPPMETEEP




N-terminus,





and





truncation





after P478





at the





C-terminus






HM48
101
14 amino acids
FRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDMSL




substitution of
FSFIGSPRINATGQGVTIFYVDRLGYYPYIDSITG




L342W, S343E,
VTVNGGIPQKISLQDHLDKAKKDITFYMPVDNLGM




I344N, M345T,
AVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQNV




S347T, M348K,
QLSLTEATEKAKQEFEKAGKDFLVETIKLGKLLRP




K349E, L352Q,
NHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKRND




L353A, L354I,
DLSWLWNESTALYPSIYLNTQQSPVAATLYVRNRV




D355K, N356E,
REAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKFLS




E359D and
QDELVYTEGETVALGASGIVIWGTWENTRTKESCQ




I361T,


AIKE
YMDTTLNPYIINVTLAAKMCSQVLCQEQGVC





truncation
IRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGKPT




before
LEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAVDV




F38 at the
CIADGVCIDAFLKPPMETEEPQI




N-terminus,





and





truncation





after I480





at the





C-terminus






HM49
102
14 amino acids
FRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDMSL




substitution of
FSFIGSPRINATGQGVTIFYVDRLGYYPYIDSITG




L342W, S343E,
VTVNGGIPQKISLQDHLDKAKKDITFYMPVDNLGM




I344N, M345T,
AVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQNV




S347T, M348K,
QLSLTEATEKAKQEFEKAGKDFLVETIKLGKLLRP




K349E, L352Q,
NHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKRND




L353A, L354I,
DLSWLWNESTALYPSIYLNTQQSPVAATLYVRNRV




D355K, N356E,
REAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKFLS




E359D and
QDELVYTEGETVALGASGIVIWGTWENTRTKESCQ




I361T,


AIKE
YMDTTLNPYIINVTLAAKMCSQVLCQEQGVC





truncation
IRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGKPT




before
LEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAVDV




F38 at the
CIADGVCIDAFLKPPMETEEPQIFY




N-terminus,





and





truncation





after Y482





at the





C-terminus






HM50
103
14 amino acids
FRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDMSL




substitution of
FSFIGSPRINATGQGVTIFYVDRLGYYPYIDSITG




L342W, S343E,
VTVNGGIPQKISLQDHLDKAKKDITFYMPVDNLGM




I344N, M345T,
AVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQNV




S347T, M348K,
QLSLTEATEKAKQEFEKAGKDFLVETIKLGKLLRP




K349E, L352Q,
NHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKRND




L353A, L354I,
DLSWLWNESTALYPSIYLNTQQSPVAATLYVRNRV




D355K, N356E,
REAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKFLS




E359D and
QDELVYTEGETVALGASGIVIWGTWENTRTKESCQ




I361T,


AIKE
YMDTTLNPYIINVTLAAKMCSQVLCQEQGVC





truncation
IRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGKPT




before F38
LEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAVDV




at the
CIADGVCIDAFLKPPMETEEPQIFYNA




N-terminus,





and truncation





after A484





at the





C-terminus






HM51
104
14 amino acids
FRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDMSL




substitution of
FSFIGSPRINATGQGVTIFYVDRLGYYPYIDSITG




L342W, S343E,
VTVNGGIPQKISLQDHLDKAKKDITFYMPVDNLGM




I344N, M345T,
AVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQNV




S347T, M348K,
QLSLTEATEKAKQEFEKAGKDFLVETIKLGKLLRP




K349E, L352Q,
NHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKRND




L353A, L354I,
DLSWLWNESTALYPSIYLNTQQSPVAATLYVRNRV




D355K, N356E,
REAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKFLS




E359D and
QDELVYTFGETVALGASGIVIWGTWENTRTKESCQ




I361T,


AIKE
YMDTTLNPYIINVTLAAKMCSQVLCQEQGVC





truncation
IRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGKPT




before
LEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAVDV




F38 at the
CIADGVCIDAFLKPPMETEEPQIFYNASP




N-terminus,





and





truncation





after P486





at the





C-terminus






HM52
105
14 amino acids
FRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDMSL




substitution of
FSFIGSPRINATGQGVTIFYVDRLGYYPYIDSITG




L342W, S343E,
VTVNGGIPQKISLQDHLDKAKKDITFYMPVDNLGM




I344N, M345T,
AVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQNV




S347T, M348K,
QLSLTEATEKAKQEFEKAGKDFLVETIKLGKLLRP




K349E, L352Q,
NHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKRND




L353A, L354I,
DLSWLWNESTALYPSIYLNTQQSPVAATLYVRNRV




D355K, N356E,
REAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKFLS




E359D and
QDELVYTFGETVALGASGIVIWGTWENTRTKESCQ




I361T,


AIKE
YMDTTLNPYIINVTLAAKMCSQVLCQEQGVC





truncation
IRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGKPT




before
LEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAVDV




F38 at the
CIADGVCIDAFLKPPMETEEPQIFYNASPST




N-terminus,





and





truncation





after T488





at the





C-terminus






HM53
106
15 amino acids
LNFRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDM




substitution of
SLFSFIGSPRINATGQGVTIFYVDRLGYYPYIDSI




T341G, L342W,
TGVTVNGGIPQKISLQDHLDKAKKDITFYMPVDNL




S343E, I344N,
GMAVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQ




M345T, S347T,
NVQLSLTEATEKAKQEFEKAGKDFLVETIKLGKLL




M348K, K349E,
RPNHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKR




L352Q, L353A,
NDDLSWLWNESTALYPSIYLNTQQSPVAATLYVRN




L354I, D355K,
RVREAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKF




N356E, E359D
LSQDELVYTFGETVALGASGIVIWGGWENTRTKES




and I361T,
CQAIKEYMDTTLNPYIINVTLAAKMCSQVLCQEQG




truncation
VCIRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGK




before L36
PTLEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAV




at the
DVCIADGVCIDAFLKPPMETEEPQIFYNASPSTLS




N-terminus,





and





truncation 





after S490





at the





C-terminus






HM54
107
15 amino acids
LNFRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDM




substitution of
SLFSFIGSPRINATGQGVTIFYVDRLGYYPYIDSI




T341A, L342W,
TGVTVNGGIPQKISLQDHLDKAKKDITFYMPVDNL




S343E, I344N,
GMAVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQ




M345T, S347T,
NVQLSLTEATEKAKQEFEKAGKDFLVETIKLGKLL




M348K, K349E,
RPNHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKR




L352Q, L353A,
NDDLSWLWNESTALYPSIYLNTQQSPVAATLYVRN




L354I, D355K,
RVREAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKF




N356E, E359D
LSQDELVYTEGETVALGASGIVIWGAWENTRTKES




and I361T,
CQAIKEYMDTTLNPYIINVTLAAKMCSQVLCQEQG




truncation
VCIRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGK




before L36
PTLEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAV




at the
DVCIADGVCIDAFLKPPMETEEPQIFYNASPSTLS




N-terminus,





and





truncation





after S490





at the





C-terminus






HM55
108
15 amino acids
LNFRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDM




substitution of
SLFSFIGSPRINATGQGVTIFYVDRLGYYPYIDSI




T341C, L342W,
TGVIVNGGIPQKISLQDHLDKAKKDITFYMPVDNL




S343E, I344N,
GMAVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQ




M345T, S347T,
NVQLSLTEATEKAKQEFEKAGKDFLVETIKLGKLL




M348K, K349E,
RPNHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKR




L352Q, L353A,
NDDLSWLWNESTALYPSIYLNTQQSPVAATLYVRN




L354I, D355K,
RVREAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKF




N356E, E359D
LSQDELVYTEGETVALGASGIVIWGCWENTRTKES




and I361T,
CQAIKEYMDTTLNPYIINVTLAAKMCSQVLCQEQG




truncation
VCIRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGK




before L36
PTLEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAV




at the
DVCIADGVCIDAFLKPPMETEEPQ109IFYNASPS




N-terminus,
TLS




and





truncation





after S490





at the





C-terminus






HM56
109
15 amino acids
LNFRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDM




substitution of
SLFSFIGSPRINATGQGVTIFYVDRLGYYPYIDSI




T341D, L342W,
TGVIVNGGIPQKISLQDHLDKAKKDITFYMPVDNL




S343E, I344N,
GMAVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQ




M345T, S347T,
NVQLSLTEATEKAKQEFEKAGKDFLVETIKLGKLL




M348K, K349E,
RPNHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKR




L352Q, L353A,
NDDLSWLWNESTALYPSIYLNTQQSPVAATLYVRN




L354I, D355K,
RVREAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKF




N356E, E359D
LSQDELVYTEGETVALGASGIVIWGDWENTRTKES




and I361T,
CQAIKEYMDTTLNPYIINVTLAAKMCSQVLCQEQG




truncation
VCIRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGK




before L36
PTLEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAV




at the
DVCIADGVCIDAFLKPPMETEEPQIFYNASPSTLS




N-terminus,





and





truncation





after S490





at the





C-terminus






HP57
110
12 amino acids
FRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDMSL




substitution of
FSFIGSPRINATGQGVTIFYVDRLGYYPYIDSITG




I344N, M345T,
VTVNGGIPQKISLQDHLDKAKKDITFYMPVDNLGM




S347T, M348K,
AVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQNV




K349E, L352Q,
QLSLTEATEKAKQEFEKAGKDFLVETIKLGKLLRP




L353A, L354I,
NHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKRND




D355K, N356E,
DLSWLWNESTALYPSIYLNTQQSPVAATLYVRNRV




E359D and
REAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKFLS




I361T,
QDELVYTFGETVALGASGIVIWGILSNTRTKESCQ




truncation


AIKE
YMDTTLNPYIINVTLAAKMCSQVLCQEQGVC





before F38
IRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGKPT




at the
LEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAVDV




N-terminus,
CIADGVCIDAF




and truncation





after





F468 at the





C-terminus






HP58
111
13 amino acids
FRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDMSL




substitution of
FSFIGSPRINATGQGVTIFYVDRLGYYPYIDSITG




S343E, I344N,
VTVNGGIPQKISLQDHLDKAKKDITFYMPVDNLGM




M345T, S347T,
AVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQNV




M348K, K349E,
QLSLTEATEKAKQEFEKAGKDFLVETIKLGKLLRP




L352Q, L353A,
NHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKRND




L354I, D355K,
DLSWLWNESTALYPSIYLNTQQSPVAATLYVRNRV




N356E, E359D
REAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKFLS




and I361T,
QDELVYTFGETVALGASGIVIWGTLENTRTKESCQ




truncation


AIKE
YMDTTLNPYIINVTLAAKMCSQVLCQEQGVC





before F38
IRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGKPT




at the
LEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAVDV




N-terminus,
CIADGVCIDAF




and





truncation





after F468





at the





C-terminus






HP59
112
15 amino acids
FRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDMSL




substitution of
FSFIGSPRINATGQGVTIFYVDRLGYYPYIDSITG




T341A, L342W,
VTVNGGIPQKISLQDHLDKAKKDITFYMPVDNLGM




S343E, I344N,
AVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQNV




M345T, S347T,
QLSLTEATEKAKQEFEKAGKDFLVETIKLGKLLRP




M348K, K349E,
NHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKRND




L352Q, L353A,
DLSWLWNESTALYPSIYLNTQQSPVAATLYVRNRV




L354I, D355K,
REAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKFLS




N356E, E359D
QDELVYTFGETVALGASGIVIWGAWENTRTKESCQ




and I361T


AIKE
YMDTTLNPYIINVTLAAKMCSQVLCQEQGVC





truncation
IRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGKPT




before F38
LEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAVDV




at the
CIADGVCIDAF




N-terminus,





and





truncation





after F468





at the





C-terminus






HP60
113
15 amino acids
FRAPPVIPNVPFLWAWNAPSEFCLGKFDEPLDMSL




substitution of
FSFIGSPRINATGQGVTIFYVDRLGYYPYIDSITG




T341G, L342W,
VTVNGGIPQKISLQDHLDKAKKDITFYMPVDNLGM




S343E, I344N,
AVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQNV




M345T, S347T,
QLSLTEATEKAKQEFEKAGKDFLVETIKLGKLLRP




M348K, K349E,
NHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKRND




L352Q, L353A,
DLSWLWNESTALYPSIYLNTQQSPVAATLYVRNRV




L354I, D355K,
REAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKFLS




N356E, E359D
QDELVYTFGETVALGASGIVIWGGWENTRTKESCQ




and I361T,


AIKE
YMDTTLNPYIINVTLAAKMCSQVLCQEQGVC





truncation
IRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGKPT




before F38
LEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAVDV




at the
CIADGVCIDAF




N-terminus,





and





truncation





after F468





at the





C-terminus






HP61
114
16 amino acids
FRGPPVIPNVPFLWAWNAPSEFCLGKFDEPLDMSL




substitution of
FSFIGSPRINATGQGVTIFYVDRLGYYPYIDSITG




A40G, T341S,
VTVNGGIPQKISLQDHLDKAKKDITFYMPVDNLGM




L342W, S343E,
AVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQNV




I344N, M345T,
QLSLTEATEKAKQEFEKAGKDFLVETIKLGKLLRP




S347T, M348K,
NHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKRND




K349E, L352Q,
DLSWLWNESTALYPSIYLNTQQSPVAATLYVRNRV




L353A, L354I,
REAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKFLS




D355K, N35 6E,
QDELVYTEGETVALGASGIVIWGSWENTRTKESCQ




E359D and


AIKE
YMDTTLNPYIINVTLAAKMCSQVLCQEQGVC





I361T,
IRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGKPT




truncation
LEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAVDV




before F38
CIADGVCIDAF




at the





N-terminus,





and





truncation





after F468





at the





C-terminus






HP62
115
Removal of P42,
FRGPPVIPNVPFLWAWNAPSEFCLGKFDEPLDMSL




15 amino acids
FSFIGSPRINATGQGVTIFYVDRLGYYPYIDSITG




substitution of
VTVNGGIPQKISLQDHLDKAKKDITFYMPVDNLGM




T341S, L342W,
AVIDWEEWRPTWARNWKPKDVYKNRSIELVQQQNV




S343E, I344N,
QLSLTEATEKAKQEFEKAGKDFLVETIKLGKLLRP




M345T, S347T,
NHLWGYYLFPDCYNHHYKKPGYNGSCFNVEIKRND




M348K, K349E,
DLSWLWNESTALYPSIYLNTQQSPVAATLYVRNRV




L352Q, L353A,
REAIRVSKIPDAKSPLPVFAYTRIVFTDQVLKFLS




L354I, D355K,
QDELVYTEGETVALGASGIVIWGSWENTRTKESCQ




N356E, E359D


AIKE
YMDTTLNPYIINVTLAAKMCSQVLCQEQGVC





and I361T,
IRKNWNSSDYLHLNPDNFAIQLEKGGKFTVRGKPT




truncation
LEDLEQFSEKFYCSCYSTLSCKEKADVKDTDAVDV




before F38
CIADGVCIDAF




at the





N-terminus,





and





truncation





after F468





at the





C-terminus









Example 2. Construction of PH20 Variants HM1 and HM6

As shown in FIG. 1B, amino acid residues M345 to N363 of PH20 correspond to the alpha-helix 8 region and the linker region between alpha-helix 7 and alpha-helix 8 in the protein tertiary structure model. Among the amino acids of alpha-helix 8, C351 forms a disulfide bond with C60 of alpha-helix 1; Y357 forms hydrophobic interaction with F315 of alpha-helix 7; and N363 forms a hydrogen bond with D69 of alpha-helix 1, thereby stabilizing secondary structures adjacent to alpha-helix 8 (FIG. 1C).


In order to construct variants having higher enzymatic activity and thermal stability than WT by substituting amino acids located in the alpha-helix 8 region of PH20 and the linker region between alpha-helix 7 and alpha-helix 8, the following variants were constructed: the variant HM1 in which amino acids in the M345 to N363 region were substituted; the variant HM2 in which 7 amino acids in the Y365 to V379 region were substituted; and the variant HM3 in which 19 amino acids in the M345 to V379 region were substituted. Among the amino acids located in alpha-helix 8 of PH20, C351, which is involved in disulfide bonding, and Y357 which is involved in hydrophobic interaction, were not substituted. The substituted amino acid sequences in the variants HM1, HM2 and HM3 are shown in Table 11 above. When ExpiCHO cells were transfected with the pcDNA3.4-TOPO plasmid comprising the gene of the variant HM1, HM2 or HM3, the variant HM1 was expressed in the ExpiCHO cells (FIG. 3A), and the variants HM2 and HM3 were not expressed in the cells. Whether the protein would be expressed was confirmed not only by measurement of the enzymatic activity, but also by Western blot analysis using an antibody (Abcam, ab193009) against human PH20. The epitope of the antibody is Q173 to P222 region. These experimental results suggest that amino acid substitution of Y365 to V379 region of the amino acid sequence of alpha-helix 8, used in construction of the variants HM2 and HM3, causes a serious effect on the protein structure; thus the proteins of the variants HM2 and HM3 were not expressed. The expression level of the variant HM1 was 3.4-fold higher than that of wild-type WT (FIG. 2). The variant HM1 is one in which the hydrogen bond between N363 of alpha-helix 8 and D69 of alpha-helix 1 was removed by substituting N363 with glycine (FIG. 1C). To restore the hydrogen bond between N363 and D69, G363 in HM1 was substituted with asparagine, thereby constructing the variant HM6. The substituted amino acids in the variant HM6 are shown in Table 11 above.


The variant HM6 was expressed in ExpiCHO cells, and the expression level thereof was similar to that of the variant HM1 (the expression level was 3.4-fold higher than that of WT) (FIG. 2). When the enzymatic activity was measured by turbidimetric assay, it was shown that the enzymatic activity of the variant HM6 was 1.3-fold higher than that of WT (FIG. 3B). In the substrate-gel assay, generally, SDS is removed with 2.5% Triton X-100 (w/v) after SDS-PAGE, and enzymatic reaction is performed at 37° C. for 1 to 4 h, during which the extent to which hyaluronidase hydrolyzes hyaluronic acid is measured using Alcian blue dye. It is known that when SDS is removed from the substrate gel, protein folding immediately occurs, and the substrate does not affect the protein folding. When the enzymatic activities of WT and the variants HM1 and HM6 were measured by the substrate-gel assay at 37° C. for 1 to 4 h, HM6 exhibited higher enzymatic activity than those of WT and the variant HM1 (FIG. 3C). This result suggests that protein folding of the variants HM1 and HM6 and the resulting renaturation were faster than those of WT and that the constructed variants exhibit higher enzymatic activity than WT. When the signal peptide of PH20 itself was used, the expression level of the protein in ExpiCHO cells was low, and to solve this problem, the signal peptide sequence of human serum albumin or human Hyal1 was used. As shown in FIG. 3C, when each of the signal peptides of human serum albumin and human Hyal1 was used as the signal peptide of the variant HM1, the expression of the protein increased, and there was no significant difference between the two signal peptides. When the enzymatic activity was measured by the substrate-gel assay, WT having the signal peptide of human serum albumin and the variant HM1 having the signal peptide of human serum albumin or Hyal1 exhibited enzymatic activity at the pH ranging from 5 to 8 (FIG. 3D). In the present invention, the signal peptide of human serum albumin was used as the signal peptide of the variant constructed after HM1.


When the thermal stabilities of WT and the variants HM1 and HM6 were compared based on the aggregation temperatures, the aggregation temperatures were 46.5° C., 53.0° C. and 50.5° C., respectively, indicating that the aggregation temperatures of the variants HM1 and HM6 were 6.5° C. and 4.0° C. higher, than that of WT, respectively (FIG. 10). The results of measuring the aggregation temperatures were consistent with the protein refolding results shown in the substrate-gel assay. These results suggest for the first time that the hydrogen bond formed by the N363 residue of alpha-helix 8 plays an important role in the thermal stability and enzymatic activity of PH20.


In addition, when the hydrophilic/hydrophobic natures of WT and the variants HM1 and HM6 were compared using a phenyl column chromatography, HM1 and HM6 were all eluted earlier than WT. This suggests that the variants HM1 and HM6 have a more hydrophilic nature than WT due to the substitution of the amino acids. However, the variants HM1 and HM6 eluted from the phenyl column showed two peaks, unlike WT, and exhibited the same molecular weight when treated with PNGase F. This appears to be a difference caused by N-glycosylation (FIG. 3E).


Example 3. Construction of PH20 Variants HM4, HM7, HM8, HM9, HM10, HM11 and HM12

It is believed that the amino acid substitution of the M345 to N363 and M345 to I361 regions by construction of the variants HM1 and HM6 in Example 2 resulted in an increase in both the enzymatic activity and thermal stability of PH20, has made great advances in terms of protein engineering. Thus, based on the variants HM1 and HM6, other amino acids in the N-terminal and C-terminal directions were additionally substituted.


First, the variants HM4 and HM7 were constructed, which comprise the substituted amino acids in the variants HM1 and HM6 and in which the amino acids between G340 and I344 were additionally substituted with G340V, T341S, L342W, S343E and I344N. The sequences of the substituted amino acid sequences in the variants HM4 and HM7 are shown in Table 11 above. The variants HM4 and HM7 were expressed in ExpiCHO cells. The results of protein purification for HM7 are shown in FIG. 5A. HM4 and HM7 showed an increase in protein expression of 6.3-fold compared to WT, and showed increases in aggregation temperature of 10° C. and 11.5° C., respectively, compared to WT. However, the enzymatic activities of HM4 and HM7, measured by turbidimetric assay, were about 15% of that of WT (FIG. 5B). Enzymatic activity and thermal stability generally have a trade-off relationship, but in the present invention, it appeared that the substitution introduced in the variants HM1 and HM6 increased the thermal stability of the variants while maintaining the enzymatic activity, and the enzymatic activities of the variants HM4 and HM7 decreased due to an excessive increase in the thermal stability. An increase in aggregation temperature of 11.5° C., which appeared in the variants HM4 and HM7 is a very significant result in terms of protein engineering. When the structural flexibilities of the proteins were analyzed by a Stern-Volmer plot, it was shown that the structural flexibilities of HM1, HM6, HM4 and HM7, obtained by substituting alpha-helix 8 and its linker region, were all higher than that of WT (FIG. 11A). This result suggests that the increase in local thermal stability resulted in an increase in the flexibility of the entire protein structure.


The difference between the variants HM6 and HM7 is amino acids between G340 and I344. In order to identify amino acids which are involved in the increased thermal stability of the variant HM7, the following variants were constructed based on the variant HM6: HM8 in which I344N was substituted; HM9 in which S343 and I344N were substituted; HM10 in which L342W, S343E and I344N were substituted; and HM21 in which T341S, L342W, S343E and I344N were substituted. The sequences of the substituted amino acids in the variants HM8, HM9, HM10 and HM21 are shown in Table 11 above. The variants HM8, HM9, HM10 and HM21 were expressed in ExpiCHO cells (FIG. 5A). As I344N, S343E and L342W were introduced in the N-terminal direction loop of alpha-helix 8 based on the variant HM6, the aggregation temperatures of the variants HM8, HM9 and HM10 increased to 52.5° C., 53° C. and 55.5° C., respectively (FIG. 10). However, the variants HM8, HM9 and HM10 maintained enzymatic activities similar to that of WT (FIG. 5B). This result suggests that the amino acid substitution introduced in the variants HM8, HM9 and HM10 had a local effect on the thermal stabilities of the enzymes, but had no significant effect on the enzymatic activities. However, HM21 showed reduced thermal stability compared to HM10, but showed approximately 2-fold higher enzymatic activity compared to WT at pH 7.0. When WT and each variant were allowed to react with the substrate for h in the substrate-gel assay, the enzymatic activity was reduced in the order of HM21>HM10>HM9>HM8>HM6>WT (FIG. 5C).


When the physical properties of the variants HM7, HM8, HM9, HM10 and HM21 were examined using a phenyl column chromatography, these PH20 variants were eluted earlier than WT, suggesting that these variants all have a hydrophilic nature. However, the pattern of the main peak at the amino acid substitution position appeared differently (FIG. 5D).


The variant HM7 also showed two peaks in the phenyl column chromatography, like the variants HM1 and HM6, suggesting that two different types were present.


In order to examine the patterns of migration of WT and the variants depending on their isoelectric points, isoelectric focusing (hereinafter referred to as IEF) analysis was performed (FIG. 5E). On IEF gel, WT and the variant HM6 and HM8 showed similar migration patterns, and the variants HM9, HM10, HM21 and HM7 comprising the S343E mutation migrated to a more acidic region. This result suggests that a change in the isoelectric point of the protein occurs due to the introduction of glutamic acid by S343E substitution of the amino acid between G340 and I344.


In addition, based on the variant HM6, other amino acids in the C-terminal regions of alpha-helix 8 were substituted, thereby constructing the variants HM11 and HM12. The sequences of the substituted amino acids in the variants HM11 and HM12 are shown in Table 11 above. The variant HM11 was expressed in ExpiCHO cells, but the expression level thereof was lower than that of WT (FIG. 2), and the variant HM12 was not expressed in ExpiCHO cells. The variant HM11 exhibited an activity corresponding to 32% of that of WT (FIG. 6B).


Example 4. Construction of N-Terminal Amino Acid-Truncated PH20 Variants Based on HM6

The C-terminal region of PH20 is already well known to play an important role in the expression and enzymatic activity of PH20, but the role of the N-terminal region of PH20 is not well known. In order to examine the effect of cleavage of the amino acid at the N-terminal region of PH20 on the enzymatic activity, variants HM40, HM13, HM41, HM24, HM42 and HM25 having the N-terminus cleaved at N37, F38, R39, A40, P41 or P42 were constructed based on the variant HM6 (Table 11). Furthermore, HP61 and HP62 with modifications at the N-terminal amino acids were additionally constructed.


The variants HM40, HM13, HM41, HM24, HM42, HP61 and HP62 were expressed in ExpiCHO cells, but HM25 was not expressed (FIGS. 7A and 7B). The N-terminal truncated PH20 variants showed a difference in enzyme activity depending on the position where the N-terminus started. The variants HM40, HM13 and HM41, in which one to three amino acids were cleaved, showed enzymatic activity similar to that of the template HM6, but HM24 and HM42, in which four to five amino acids were cleaved, showed slightly lower activity than HM6 (FIG. 7C). However, HM25 in which six amino acids were cleaved were little expressed in ExpiCHO cells, and the enzymatic activity was also significantly low (3.5 U/μg). It appears that changes in the enzymatic activities of HP61 and HP62 with modifications at N-terminal amino acids are not significant.


Regarding the enzymatic activities of the N-terminal cleaved PH20 variants, measured by the substrate-gel assay (1 h of reaction), the enzymatic activities of HM40, HM13 and HM41 were similar to that of the HM6 variant, but the enzymatic activities of HM24 and HM42 were lower than that of HM6 (FIG. 7D). HM25 in which six amino acids were cleaved could not be analyzed, because the amount of the protein produced was small.


When the physical properties of the variants HM40, HM13, HM41, HM24 and HM42 were analyzed using a phenyl column chromatography, these variants were eluted from the column earlier than WT, suggesting that these variants have a hydrophilic nature (FIG. 7E). This result suggests that the variants HM40, HM13, HM41, HM24 and HM42 constructed based on the variant HM6 maintained the hydrophilic nature of HM6 when considering the characteristics of L36 to A40 residues.


The aggregation temperatures of the N-terminal cleaved PH20 variants, measured by DLS, were different among the variants depending on the position where the amino acid started (FIG. 7F). Although the N-terminal amino acid residues of the variants HM40, HM13, HM41 and HM42 were cleaved, they showed an aggregation temperature of 50° C. or higher, indicating that the characteristics of the template HM6 were remained intact. Among these variants, the variants HM40 and HM42 exhibited an aggregation temperature which was 3 to 4° C. higher than that of HM6, indicating that the thermal stability of these variants increased. In addition, in order to examine the effect of substitution of the N-terminal amino acids of PH20 on protein expression and enzymatic activity, the following variants were constructed: the variant HM17 in which N-terminal amino acid residues 36 to 47 (LNFRAPPVIPNV) of PH20 were substituted with FRGPLLPNR; and the variant HM18 in which N-terminal amino acid residues 36 to 52 (LNFRAPPVIPNVPFLWA) of PH20 were substituted with FRGPLLPNRPFTTV. The sequences of the substituted amino acids in the variants HM17 and HM18 are shown in Table 11 above. The variants HM17 and HM18 were not expressed in ExpiCHO cells. This suggests that, even when up to five amino acids located at the N-terminus were cleaved, the variants showed protein expression and enzymatic activity; however, substitution of more amino acid residues, such as 36 to 47 residues or 36 to 52 residues, had an effect on protein folding.


Example 5. Construction of the C-Terminal Amino Acid-Truncated HM6-Based Variants HM14, HM15 and HM16 of PH20

The C-terminal region of PH20 is known to play an important role in protein expression and enzymatic activity. In the present invention, the variants HM14, HM15 and HM16, in which the C-terminal amino acids were cleaved at I465, F468 and K471, respectively, were constructed based on the variant HM6. The sequences of the substituted amino acids in these variants HM14, HM15 and HM16 are shown in Table 11 above. These variants HM14, HM15 and HM16 were expressed in ExpiCHO cells (FIG. 8A), and the protein expression levels thereof were in the order of HM16>HM15>HM14, indicating that the protein expression levels decreased as the number of C-terminal amino acids cleaved increased (FIG. 8A). However, the enzymatic activities of the variants HM14, HM15 and HM16 were in the order of HM16>HM14 (≈WT)>HM15 (FIG. 8B). According to Frost et al., the C-terminal 477-483 region of PH20 is necessary for soluble expression, and when the C-terminus is cleaved at 467, the enzymatic activity of the variant is only 10% of a PH20 variant whose C-terminus was cleaved at residues 477 to 483, and when the C-terminus is cleaved before residue 467, the variant has no enzymatic activity. However, the C-terminal cleaved variants HM14, HM15 and HM16 constructed based on the variant HM6 in the present invention showed increased protein folding due to amino acid substitution of the M345 to I361 region, and thus the thermal stability thereof increased. For this reason, even when the C-terminus was cleaved after I465, F468 or P471, the variant showed enzymatic activity similar to that of WT, and the enzymatic activity thereof did not significantly decrease.


The structural flexibilities of WT and the variants HM14, HM15 and HM16 were examined by fluorescence quenching using acrylamide (FIG. 11B). The variants HM14, HM15 and HM16 were all structurally more flexible than WT. This result suggests that the C-terminal cleaved variants constructed using the variant HM6 also maintained their structural flexibility.


The enzymatic activities of the variants HM14, HM15 and HM16, measured by turbidimetric assay, were also confirmed in substrate-gel assay (FIG. 8C).


When the physical properties of the C-terminal cleaved variants were analyzed using a phenyl column chromatography, the variants HM14, HM15 and HM16 were all eluted earlier than WT, indicating that they had a hydrophilic nature. In addition, the hydrophobicities of these variants were in the order of HM16>HM14>HM15 (FIG. 8D).


Example 6. Construction of the HM10-Based Variants HM19 and HM20 Comprising N-Terminal and C-Terminal Amino Acid Cleavage

The PH20 variants constructed in the present invention were based on HM6, and HM8, HM9 and HM10, in which amino acid residues G340 to I344 were additionally substituted, exhibited better performance than WT in terms of protein expression levels, enzymatic activities and thermal stabilities. The variants HM19 and HM20 were constructed which had an N-terminus cleaved at F38 and a C-terminus cleaved at F468 based on HM10 having high enzymatic activity and thermal stability among variants HM8, HM9 and HM10. HM19 and HM20 were all expressed in ExpiCHO cells and purified using a HisTrap column chromatography (FIG. 9A). When the enzymatic activities of these variants were measured by turbidimetric assay, HM19 and HM20 exhibited an enzymatic activity which was 10% higher than that of WT (FIG. 9B). In substrate-gel assay, HM19 and HM20 also exhibited higher enzymatic activity than WT (FIG. 9C).


Example 7. Characterization of HM10-Based PH20 Variants

The expression levels of HM10-based C-terminal truncated variants in ExpiCHO cells showed a tendency to decrease as the length of the C-terminal region became shorter, and these variants were not expressed when the C-terminus was truncated at C464 or shorter (FIG. 12). C464 is necessary because it forms a disulfide bond with C437 and is important for maintaining the protein structure.


In order to examine whether the variant is not expressed in ExpiCHO cells when the C-terminus is cleaved at residue 464 or shorter, Western blot analysis was performed. As shown in FIG. 13, the HM30, HM31, HM32 and HM33 variants were not detected in Western blots.


The enzymatic activities of the HM10-based C-terminal truncated variants, measured by turbidimetric assay, are shown in FIGS. 14A and 14B. The C-terminal truncated PH20 variants exhibited an enzymatic activity of ±20% compared to WT. When the C-terminus was cleaved after I480, the enzyme activity increased overall. In addition, the variants HP19 and HP20, obtained by removing the 6×His-tag from HM19 and HM20, showed decreases in enzymatic activity of 23% and 9.6%, respectively, compared to when the 6×His-tag was present. This suggests that the 6×His-tag has an effect on the enzymatic activity.


When the enzymatic activities of the HM10-based C-terminal truncated variants were measured by substrate-gel assay, these variants exhibited higher enzymatic activity than WT, and showed enzymatic activity similar to that of the template HM10, indicating that the difference in enzymatic activity depending on the length of the C-terminal region was not significant (FIG. 14C).


Example 8. Characterization of HM21-Based PH20 Variants

The variant HP34 was purified by four-step column chromatography (FIG. 15A), and the variant HP46 was purified by three-step column chromatography (FIG. 15B). The amounts of HP34 and HP46 produced were 1.73 mg/L and 25.6 mg/L, respectively. HP34 and HP46 are 6×His-tag-free variants, and the process of purifying these variants differs from the process of purifying variants having the 6×His-tag, and thus it is difficult to compare the expression levels of the proteins.


In turbidimetric assay, the activities of HP34 and HP46 were 45.6 U/μg and 47.2 U/μg, respectively, which were about 2-fold higher than that of WT and were about 10% higher than that of the template HM21 (FIG. 16A).


The kinetics of each variant was measured by Morgan-Elson assay, and the results of the measurement are shown in FIG. 16B. The catalytic efficiencies (kcat/Km) of HP34 and HP46 were 1.7 to 2 times higher than that of wild-type HW2. This result is consistent with the result that the specific activity was higher than that of WT. The Michaelis constant (Km) was lower in these variants than in HW2, indicating the substrate affinities of these variants increased. From these results, it can be concluded that the HM21, HP34 and HP46 variants bind strongly to the substrate and have the property of converting the substrate into a product with high efficiency. This property is attributable to the effect of substitution of T341 with serine. When threonine located at position 341 is substituted with an amino acid, such as alanine, glycine, aspartic acid or the like, the effect of the substitution on the enzymatic activity can be predicted.


The aggregation temperatures of HP34 and HP46, measured by DLS, were 51.5° C. and 51.0° C., respectively, which were similar to that of the template HM21 and were about 5° C. higher than that of HW2, indicating that these variants were thermally stable (FIG. 17A). The enzymatic activity of HP20, measured by substrate-gel assay, was similar to that of HP20, whereas HP46 exhibited higher enzymatic activity than HP20, indicating that the protein folding of HM21 as a template was better than that of HM10 (FIG. 17B).


Wild-type HW2 and the variant HP46 were left to stand overnight at pH 7.0 and pH 3.0, and then the enzymatic activities thereof were compared by substrate-gel assay. As a result, it was shown that HP46 exhibited high activity not only at pH 7.0 but also at pH 3.0, indicating that it had excellent stability (FIG. 17C).


HM53, HM54, HM55, HM56, HP59 and HP60 are variants having a mutation at the amino acid at position 341, among HM21-based variants. It was confirmed that the mutation of the amino acid at position 341 had various effects on the expression level and activity of the variants (FIGS. 17D and 17E).


Example 9. In Vitro Immunogenicity Assay of PH20 Variant

Biopharmaceuticals with higher molecular weights than those of low-molecular-weight synthetic chemicals have a risk of triggering unintended immune responses when they enter the human body. An outside contact surface, created by folding or interaction with adjacent domains in the secondary structure or tertiary structure of a large-molecular-weight biomaterial, can promote immune response to the biomaterial by providing an epitope to the immune system in the human body. This immune response can produce an anti-drug antibody (ADA), and this response can inhibit the drug's effectiveness, or cause hypersensitivity to the drug, or promote clearance of the drug in the human body. The immune response to the drug may therefore affect the results in clinical trials, and may cause serious abnormal reactions upon long-term use. This immune response can be influenced by various factors, and triggered by the specific response to the drug itself or disease, or by factors that depend on the method of drug administration or individual patients. Factors caused by the drug itself include the similarity or dissimilarity of the biopharmaceuticals to human peptides, posttranslational modification, impurities, aggregate formation, and the characteristics of the formulation. Factors that varies among individual patients include sex, reactivity with other drugs being taken, and genetic factors depending on the type of human leukocyte antigen (HLA).


This immunogenic response is triggered by CD4+ T cells or CD8+ T cells that recognize epitopes regardless of the cause of the immune response. Because of HLA class II gene diversity in individuals, epitopes of CD4+ T cells are different among individuals, and therefore the responsiveness to biopharmaceuticals in CD4+ T cells in each blood provided by the healthy donor can be a very important criterion for evaluating immune responses that may arise in clinical processes. CD4+ T cells are activated by antigen presenting cells (APCs) recognizing the antigens presented through their type II MHC (major histocompatibility complex). Activated CD4+ T cells release cytokines that activate macrophages, cytotoxic T cells, and B cells, resulting in high levels of antibody production. On the contrary, CD8+ T cells have direct cytotoxicity and directly remove cells infected with antigens, damaged cells, or cells that lost their function. CD8+ T cells have T cell receptors that can recognize specific antigen peptides bound to type I MHC molecules located on the surface of every cell. CD8+ T cells can also be activated by recognizing antigens presented by antigen presenting cells, and this activation can be further enhanced by cytokines of CD4+ T cells. Therefore, when the activation level of CD4+ T cells and CD8+ T cells by new biomaterials is measured in vitro, it is possible to predict immunogenic responses that can be induced in clinical processes. In this example, in order to predict the immunogenicity of the PH20 variant in comparison with a control, CD4+ T cells and CD8+ T cells were isolated from PBMCs, and then treated with 1.5 ng/mL and 15 ng/mL of the control PH20 and the PH20 variant (HP46), and then the distributions of activated CD4+ T cells and CD8+ T cells were measured. The activation level of each type of T cells was measured using Stimulating Index, and Stimulating Index (SI) is defined as follows:





Stimulating Index (SI)=(T-cell activation level after treatment with test sample)/(T-cell activation level after treatment with vehicle)


If the SI value of cells is 2 or more, it can be determined that the cells were activated at the significant level.


Immunogenic responses may vary depending on the HLA type. Therefore, experiments for measuring responses in more various HLA types were performed using T cells isolated from PBMCs provided from 10 healthy donors. The HLA types of the 10 PBMCs used are shown in Table 12 below.









TABLE 12







HLA types of tested PBMCs













No.
HLAA
HLAB
HLAC1
DRB1
DQB1
DPB1






















1
0206
3303
3501
5801
0302
0801
1405
1501
0303
0602
0201
0301


2
0201
0207
4601
5101
0102
1502
0803
1101
0301
0601
0201
0202


3
0206
3101
1501
3501
0304
0401
0803
0901
0303
0601
0201
0501


4
0101
3101
1501
1517
0303
0701
1302
1501
0602
0604
0401
0501


5
0201
3101
5101
6701
0702
1402
0401
0403
0301
0302
0201
0401


6
0201
1101
1501
3901
0401
0702
0102
0901
0402
0501
0101
1401


7
0201
0206
4002
5502
0102
0304
0802
1454
0302
0502
0402
0501


8
0201
0201
1501
4001
0303
0304
0802
1406
0301
0302
0501
1301


9
1101
2402
4006
5101
0801
1502
0701
0901
0202
0303
0201
1301


10
2602
3101
4002
5101
0304
1502
0405
1501
0401
0602
0201
0501









The results of measuring the activation levels of CD4+ and CD8+ T cells treated with each of PH20 and the PH20 variant are summarized in Table 13 below. When looking at the results of measuring the activation levels of CD4+ and CD8+ T cells, it appears that PH20 and the PH20 variant all show relatively low activation levels. In the case of PH20, the activation level of CD4+ T cells was measured to be 2 or more in two experiments, and in the case of the PH20 variant, the activation of CD4+ T cells was not detected. In the case of PH20, the activation of CD8+ T cells was detected in one experiment, and in the case of the PH20 variant, the activation of CD8+ T cells was also detected in one experiment. However, in the case of PH20, the SI value was measured to be 2 or more at both 1.5 ng/mL and 15 ng/mL, but in the case of the PH20 variant, an SI value of 2 or less at 1.5 ng/mL and an SI value of 2 or more at 15 ng/mL were measured (see FIGS. 18 and 19).


Therefore, at the lower concentration, the activation level of CD8+ T cells was observed to be low in the case of the PH20 variant, and it is determined that the activation level of CD8+ T cells by PH20 is higher than the activation level of CD8+ T cells by the PH20 variant. The conclusions drawn from the above results are as follows:


1) the activation levels of CD4+ T cells and CD8+ T cells by PH20 and the PH20 variant are relatively low; and


2) the possibility of activation of CD4+ T cells and CD8+ T cells by the PH20 variant is lower than that by PH20.


From these results, it is expected that the PH20 variant will have a lower possibility of triggering an immunogenic response in clinical processes than PH20.









TABLE 13







Stimulating Index (SI) measured from in vitro immunogenicity assay results










CD4
CD8











PBMC
PH20 (ng/mL)
PH20 variant (ng/mL)
PH20 (ng/mL)
PH20 variant (ng/mL)















No.
1.5
15
1.5
15
1.5
15
1.5
15


















1
1.03
0.859
0.728
1.176
1.38
1.42
1.22
1.60


2
1.05
0.09
0.596
0.922
1.20
1.11
1.00
1.07


3
1.90
1.44
1.56
1.12
1.43
1.31
1.59
1.35


4
2.51
2.79
0.788
1.82
1.28
1.90
0.579
0.974


5
1.27
1.47
1.09
1.14
0.987
0.932
0.972
1.02


6
0.825
0.834
0.998
0.946
0.904
1.13
1.09
0.986


7
2.07
2.07
1.43
1.90
2.08
2.34
1.30
2.46


8
0.898
1.079
0.967
0.822
0.926
1.06
0.896
0.813


9
0.882
0.957
0.895
0.941
0.853
0.993
0.950
1.09


10
0.983
0.970
1.12
1.13
1.15
1.08
1.15
1.24









Advantageous Effects

The PH20 variants or fragments thereof according to the present invention have increased protein expression levels and show an increase in protein aggregation temperature of 4-11.5° C. or so when expressed in CHO (ExpiCHO) cells so that they can be efficiently produced while having high thermal stability, compared to the mature wild-type PH20.


Further, as the result of substrate-gel assay, one of tests to measure the activity of hyaluronidase, the PH20 variants or fragments thereof according to the present invention have improved protein refolding so that they are re-natured faster than that of the mature wild-type PH20, and the original enzymatic activity is maintained regardless of the C-terminal cleavage position.


Furthermore, the PH20 variants or fragments thereof according to the present invention have low immunogenicity so that they can be repeatedly administered to the human body.


REFERENCE



  • Arming, S., Strobl, B., Wechselberger, C., and Kreil, G. (1997). In vitro mutagenesis of PH-20 hyaluronidase from human sperm. Eur J Biochem 247, 810-814.

  • Bookbinder, L. H., Hofer, A., Haller, M. F., Zepeda, M. L., Keller, G. A., Lim, J. E., Edgington, T. S., Shepard, H. M., Patton, J. S., and Frost, G. I. (2006). A recombinant human enzyme for enhanced interstitial transport of therapeutics. J Control Release 114, 230-241.

  • Chao, K. L., Muthukumar, L., and Herzberg, O. (2007). Structure of human hyaluronidase-1, a hyaluronan hydrolyzing enzyme involved in tumor growth and angiogenesis. Biochemistry 46, 6911-6920.

  • Frost, G. I. (2007). Recombinant human hyaluronidase (rHuPH20): an enabling platform for subcutaneous drug and fluid administration. Expert Opin Drug Deliv 4, 427-440.



Sequence List (Free Text)

The electronic file is attached.

Claims
  • 1. An PH20 variant or fragment thereof, which comprises one or more amino acid residue substitutions selected from the group consisting of T341A, T341C, T341G, S343E, M345T, K349E, L353A, L354I, N356E and I361T in wild-type PH20 having the amino acid sequence of SEQ ID NO: 1.
  • 2. The PH20 variant or fragment thereof of claim 1, which comprises one or more amino acid residue substitutions selected from the group consisting of T341A, T341C, L354I and N356E.
  • 3. The PH20 variant or fragment thereof of claim 1, which further comprises one or more amino acid residue substitutions in the region corresponding to an alpha-helix region and/or its linker region in wild-type PH20 having the amino acid sequence of SEQ ID NO: 1.
  • 4. The PH20 variant or fragment thereof of claim 3, wherein the alpha-helix region is an alpha-helix 8 region (S347 to C381), and its linker region is a linker region (A333 to R346) between alpha-helix 7 and alpha-helix 8.
  • 5. The PH20 variant or fragment thereof of claim 4, wherein the region corresponding to the alpha-helix region and the its linker region is T341 to N363, T341 to I361, L342 to I361, S343 to I361, I344 to I361, M345 to I361, or M345 to N363.
  • 6. The PH20 variant or fragment thereof of claim 4, wherein the alpha-helix 8 region (S347 to C381) and/or the linker region (A333 to R346) between alpha-helix 7 and alpha-helix 8 are/is substituted with one or more amino acid residues of the corresponding region of Hyal1.
  • 7. The PH20 variant or fragment thereof of claim 1, which comprises amino acid residue substitutions of L354I and/or N356E, and further comprises an amino acid residue substitution at one or more positions selected from the group consisting of T341, L342, S343, I344, M345, S347, M348, K349, L352, L353, D355, E359, I361 and N363.
  • 8. The PH20 variant or fragment thereof of claim 7, which comprises amino acid residue substitutions of L354I and/or N356E, and further comprises one or more amino acid residue substitutions selected from the group consisting of T341A, T341C, T341D, T341G, T341S, L342W, S343E, I344N, M345T, S347T, M348K, K349E, L352Q, L353A, D355K, E359D, I361T and N363G.
  • 9. The PH20 variant or fragment thereof of claim 7, which comprises M345T, S347T, M348K, K349E, L352Q, L353A, L354I, D355K, N356E, E359D and I361T.
  • 10. The PH20 variant or fragment thereof of claim 9, which further comprises one or more amino acid residue substitutions selected from the group consisting of T341A, T341C, T341D, T341G, T341S, L342W, S343E, I344N and N363G.
  • 11. The PH20 variant or fragment thereof of claim 10, which comprises any one amino acid residue substitution selected from the following amino acid residue substitution groups: (a) T341S, L342W, S343E, I344N, M345T, S347T, M348K, K349E, L352Q, L353A, L354I, D355K, N356E, E359D and I361T;(b) L342W, S343E, I344N, M345T, S347T, M348K, K349E, L352Q, L353A, L354I, D355K, N356E, E359D and I361T;(c) M345T, S347T, M348K, K349E, L352Q, L353A, L354I, D355K, N356E, E359D, I361T and N363G;(d) T341G, L342W, S343E, I344N, M345T, S347T, M348K, K349E, L352Q, L353A, L354I, D355K, N356E, E359D and I361T;(e) T341A, L342W, S343E, I344N, M345T, S347T, M348K, K349E, L352Q, L353A, L354I, D355K, N356E, E359D and I361T;(f) T341C, L342W, S343E, I344N, M345T, S347T, M348K, K349E, L352Q, L353A, L354I, D355K, N356E, E359D and I361T;(g) T341D, L342W, S343E, I344N, M345T, S347T, M348K, K349E, L352Q, L353A, L354I, D355K, N356E, E359D and I361T;(h) I344N, M345T, S347T, M348K, K349E, L352Q, L353A, L354I, D355K, N356E, E359D and I361T; and(i) S343E, I344N, M345T, S347T, M348K, K349E, L352Q, L353A, L354I, D355K, N356E, E359D and I361T.
  • 12. The PH20 variant or fragment thereof of claim 1, wherein further one or more of the N-terminal or C-terminal amino acid residues are deleted.
  • 13. The PH20 variant or fragment thereof of claim 12, wherein cleavage occurs before an amino acid residue selected from the group consisting of M1 to P42 at the N-terminus, so that one or more amino acid residues at the N-terminus are deleted.
  • 14. The PH20 variant or fragment thereof of claim 13, wherein cleavage occurs before an amino acid residue L36, N37, F38, R39, A40, P41, or P42 at the N-terminus, so that one or more amino acid residues at the N-terminus are deleted.
  • 15. The PH20 variant or fragment thereof of claim 12, wherein cleavage occurs after an amino acid residue selected from the group consisting of V455 to L509 at the C-terminus, so that one or more amino acid residues at the C-terminus are deleted.
  • 16. The PH20 variant or fragment thereof of claim 15, wherein cleavage occurs after an amino acid residue selected from the group consisting of V455 to S490 at the C-terminus, so that one or more amino acid residues at the C-terminus are deleted.
  • 17. The PH20 variant or fragment thereof of claim 16, wherein cleavage occurs after an amino acid residue V455, C458, D461, C464, I465, D466, A467, F468, K470, P471, P472, M473, E474, T475, E476, P478, I480, Y482, A484, P486, T488, or S490 at the C-terminus, so that one or more amino acid residues at the C-terminus are deleted.
  • 18. The PH20 variant or fragment thereof of claim 1, wherein the N-terminus comprises a human growth hormone-derived signal peptide having an amino acid sequence MATGSRTSLLLAFGLLCLPWLQEGSA of SEQ ID NO: 3, a human serum albumin-derived signal peptide having an amino acid sequence MKWVTFISLLFLFSSAYS of SEQ ID NO: 4, or a human Hyal1-derived signal peptide having an amino acid sequence MAAHLLPICALFLTLLDMAQG of SEQ ID NO: 5.
  • 19. The PH20 variant or fragment thereof of claim 1, wherein the PH20 variant or fragment thereof is selected from the group consisting of amino acid sequences of SEQ ID NOs: 60 to 115.
  • 20. The PH20 variant or fragment thereof of claim 19, which has an amino acid sequence of SEQ ID NO: 99.
  • 21. A composition for treating cancer, which comprises the PH20 variant or fragment thereof of claim 1.
  • 22. The composition for treating cancer of claim 21, wherein the composition is used for combined treatment with other anticancer drugs.
  • 23. The composition for treating cancer of claim 22, wherein the other anticancer drugs are immuno-oncologic agents.
  • 24. The composition for treating cancer of claim 23, wherein the immuno-oncologic agents are immune checkpoint inhibitors.
  • 25. A nucleic acid encoding the PH20 variant or fragment thereof of claim 1.
  • 26. A recombinant expression vector comprising the nucleic acid of claim 25.
  • 27. A host cell transformed with the recombinant expression vector of claim 26.
  • 28.-29. (canceled)
Priority Claims (2)
Number Date Country Kind
10-2018-0086308 Jul 2018 KR national
10-2019-0029758 Mar 2019 KR national
PCT Information
Filing Document Filing Date Country Kind
PCT/KR2019/009215 7/25/2019 WO 00