US20170137482A1 - Cell-permeable (icp)-socs3 recombinant protein and uses thereof - Google Patents

Cell-permeable (icp)-socs3 recombinant protein and uses thereof Download PDF

Info

Publication number
US20170137482A1
US20170137482A1 US15/361,701 US201615361701A US2017137482A1 US 20170137482 A1 US20170137482 A1 US 20170137482A1 US 201615361701 A US201615361701 A US 201615361701A US 2017137482 A1 US2017137482 A1 US 2017137482A1
Authority
US
United States
Prior art keywords
aliphatic
socs3
icp
recombinant protein
cell
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/361,701
Inventor
Daewoong Jo
Youngsil CHOI
Kuysook LEE
Seulmee SHIN
Jihye Kim
Jeongmin Kim
Chohyun KIM
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Cellivery Therapeutics Inc
Original Assignee
Cellivery Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Cellivery Therapeutics Inc filed Critical Cellivery Therapeutics Inc
Priority to US15/361,701 priority Critical patent/US20170137482A1/en
Assigned to CELLIVERY THERAPEUTICS, INC., JO, DAEWOONG reassignment CELLIVERY THERAPEUTICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CHOI, Youngsil, JO, DAEWOONG, KIM, Chohyun, KIM, JEONGMIN, KIM, JIHYE, LEE, Kuysook, SHIN, Seulmee
Assigned to CELLIVERY THERAPEUTICS, INC. reassignment CELLIVERY THERAPEUTICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: JO, DAEWOONG
Assigned to CELLIVERY THERAPEUTICS, INC. reassignment CELLIVERY THERAPEUTICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: JO, DAEWOONG
Publication of US20170137482A1 publication Critical patent/US20170137482A1/en
Priority to US16/426,751 priority patent/US10961292B2/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/475Growth factors; Growth regulators
    • C07K14/51Bone morphogenetic factor; Osteogenins; Osteogenic factor; Bone-inducing factor
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • A61K38/1761Apoptosis related proteins, e.g. Apoptotic protease-activating factor-1 (APAF-1), Bax, Bax-inhibitory protein(s)(BI; bax-I), Myeloid cell leukemia associated protein (MCL-1), Inhibitor of apoptosis [IAP] or Bcl-2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • C07K14/4703Inhibitors; Suppressors
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/08Linear peptides containing only normal peptide links having 12 to 20 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/08Peptides having 5 to 11 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/10Peptides having 12 to 20 amino acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/1703Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • A61K38/1709Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4702Regulators; Modulating activity
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/01Fusion polypeptide containing a localisation/targetting motif
    • C07K2319/10Fusion polypeptide containing a localisation/targetting motif containing a tag for extracellular membrane crossing, e.g. TAT or VP22
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/20Fusion polypeptide containing a tag with affinity for a non-protein ligand
    • C07K2319/21Fusion polypeptide containing a tag with affinity for a non-protein ligand containing a His-tag
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/40Fusion polypeptide containing a tag for immunodetection, or an epitope for immunisation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/04Linear peptides containing only normal peptide links
    • C07K7/06Linear peptides containing only normal peptide links having 5 to 11 amino acids

Definitions

  • the present invention relates to providing improved cell-permeable (iCP)-SOCS3 recombinant protein and uses thereof.
  • the iCP-SOCS3 recombinant protein may be used as protein-based anti-solid tumor agent by utilizing the platform technology for macromolecule intracellular transduction.
  • the Janus kinase signal transducers and activators of transcription signaling plays important roles in immune responses, including oncogenesis. So many investigations demonstrated that STAT-3, an important member of STAT proteins, was considered as a proto-oncogene in various types of disorder. STAT-3 is phosphorylated and dimerizes by the Janus kinase (JAK), and its overexpression and constitutive activation can significantly induce cell proliferation, tumor angiogenesis, invasion. Meanwhile, inhibition of JAK-STAT signaling led to suppress the cancer cell growth and induce apoptosis.
  • SOCS3 cytokine signaling-3
  • Gastric cancer remains the second leading cause of cancer-related death in the world. Advances in early detection and decreased chronic Helicobacter pylori infection rates have led to a substantial reduction in gastric cancer rates worldwide. However, effective treatment regimens for gastric cancers, especially advanced gastric cancer, are still lacking; therefore, the prognosis of patients with this disease remains poor.
  • SOCS3 mRNA levels are higher in adjacent normal mucosal tissues, however, gastric cancer patients with high simultaneous expression of SOCS3 have a better overall survival than those with low simultaneous expression. Based on this, SOCS3 may represent new therapeutic target to treat gastric cancer.
  • Colorectal cancer is one of the most fatal neoplastic diseases worldwide and a serious global health problem, with over one million new cases and half million mortalities worldwide each year. It has been reported as being relevant to some inflammatory bowel diseases, such as Crohn's disease and ulcerative colitis.
  • the pathogenesis of colorectal carcinoma is complex, with the involvement of multiple cellular transduction pathways including IL-6/STAT3 signaling.
  • Reduced or silenced SOCS3 has been found in many human types of cancer including colorectal cancer, and restoring SOCS3 expression in the cancer cells inhibits IL-6-mediated STAT3 activation, induces tumor cell apoptosis and decreases cell proliferation. Therefore, suppression of the IL-6/STAT3 pathway via modulation of SOCS3 has been a promising strategy for anti-colon/colorectal cancer therapy.
  • Glioblastoma the most common neoplasm among diffuse infiltrating astrocytomas, is notorious for its ability to evade immune-surveillance as well as for its invasive and angiogenic properties. Gliomas are the most common type of primary brain tumors are highly malignant and are associated with a very poor prognosis. Glioblastoma is a very aggressive subtype of glioma with very short life expectancy and limited treatment options. A hallmark of this lethal disorder is the presence of activated STAT3. Because SOCS3 is a negative regulator of STAT-3 activation, it hypothesized that SOCS3 may function as a tumor suppressor in glioblastoma tissues.
  • Breast cancer is a disease that arises from the accumulation of alterations in the genome of cells that make up the mammary gland.
  • Breast cancer is the most common type of cancer among women, with an estimated 1.38 million new cases of cancer diagnosed in 2008 (23% of all cancers), and the second most common type of cancer overall (10.9% of all cancers).
  • Expression of SOCS3 protein is significantly down-regulated in breast cancer specimens and replacing of SOCS3 protein may directly influence the treatment of breast cancer.
  • Cytokine signaling is strictly regulated by the SOCS family proteins induced by different classes of agonists, including cytokines, hormones and infectious agents.
  • SOCS1 and SOCS3 are relatively specific to STAT1 and STAT3, respectively.
  • SOCS1 inhibits JAK activation through its N-terminal kinase inhibitory region (KIR) by the direct binding to the activation loop of JAKs, while SOCS3 binds to janus kinases (JAKs)-proximal sites on the receptor through its SH2 domain and inhibits JAK activity that blocks recruitment of STAT3. Both promote anti-inflammatory effects due to the suppression of inflammation-inducing cytokine signaling.
  • KIR N-terminal kinase inhibitory region
  • JAKs janus kinases
  • SOCS box another domain in SOCS proteins, interacts with E3 ubiquitin ligases and/or couples the SH2 domain-binding proteins to the ubiquitin-proteasome pathway. Therefore, SOCSs inhibit cytokine signaling by suppressing JAK kinase activity and degrading the activated cytokine receptor complex.
  • the SOCS1 gene has been implicated as an anti-oncogene in the tumor development.
  • Previous studies have reported that aberrant methylation in the CpG island of SOCS1 induces its transcriptional silencing in cancer cell lines, and SOCS1 heterozygous mice are hypersensitive to various cancers.
  • abnormalities of SOCS3 are also associated with the solid tumors. Hypermethylation of CpG islands in the SOCS3 promoter is correlated with its transcriptional silencing in tumors cell lines.
  • SOCS3 overexpression down-regulates active STAT3, induces apoptosis, and suppresses growth in cancer cells.
  • aMTD Advanced macromolecule transduction domain
  • SDs solubilization domains
  • iCP-SOCS3 aMTD/SD-fused iCP-SOCS3 recombinant proteins
  • iCP-SOCS3 iCP-SOCS3 recombinant proteins
  • the newly developed iCP-SOCS3 proteins have now been demonstrated to have therapeutic application in treating solid tumor, exploiting the ability of SOCS3 to suppress JAK/STAT signaling.
  • the present application represents that macromolecule intracellular transduction technology (MITT) enabled by the new hydrophobic CPPs that are aMTD may provide novel protein therapy through SOCS3-intracellular protein replacement against the solid tumor.
  • MITT macromolecule intracellular transduction technology
  • One aspect disclosed in the present application provides an improved Cell-Permeable (iCP)-SOCS3 recombinant protein, which comprises a SOCS3 protein; and an advanced macromolecule transduction domain (aMTD) being composed of 9-13 amino acid sequences and having improved cell and/or tissue permeability, wherein the aMTD is fused to one end or both ends of the SOCS3 protein and has the following features of:
  • one or more solubilization domain (SD)(s) are further fused to the end(s) of one or more of the SOCS3 protein and the aMTD.
  • the aMTD may form ⁇ -Helix structure.
  • the aMTD may be composed of 12 amino acid sequences and represented by the following general formula:
  • X(s) independently refer to Alanine (A), Valine (V), Leucine (L) or Isoleucine (I); and Proline (P) can be positioned in one of U(s) (either 5′, 6′, 7′ or 8′). The remaining U(s) are independently composed of A, V, L or I, P at the 12′ is Proline.
  • iCP-SOCS3 recombinant protein which is represented by any one of the following structural formulae:
  • A is an advanced macromolecule transduction domain (aMTD) having improved cell and/or tissue permeability
  • B is a SOCS3 protein
  • C is a solubilization domain (SD);
  • the aMTD is composed of 9-13 amino acid sequences and has the following features of:
  • the SOCS3 protein may have an amino acid sequence of SEQ ID NO: 814.
  • the SOCS3 protein may be encoded by a polynucleotide sequence of SEQ ID NO: 815.
  • the SOCS3 protein may further include a ligand selectively binding to a receptor of a cell, a tissue, or an organ.
  • the aMTD may have an amino acid sequence selected from the group consisting of SEQ ID NOs: 1-240 and 822.
  • the aMTD may be encoded by a polynucleotide sequence selected from the group consisting of SEQ ID NOs: 241-480 and 823.
  • the SD(s) may have an amino acid sequence independently selected from the group consisting of SEQ ID NOs: 798, 799, 800, 801, 802, 803, and 804.
  • the SD(s) may be encoded by a polynucleotide sequence independently selected from the group consisting of SEQ ID NOs: 805, 806, 807, 808, 809, 810, and 811.
  • the iCP-SOCS3 recombinant protein may have a histidine-tag affinity domain additionally fused to one end thereof.
  • the histidine-tag affinity domain may have an amino acid sequence of SEQ ID NO: 812.
  • the histidine-tag affinity domain may be encoded by a polynucleotide sequence of SEQ ID NO: 813.
  • the fusion may be formed via a peptide bond or a chemical bond.
  • the iCP-SOCS3 recombinant protein may be used for the treating, preventing, or delaying the onset of, solid tumor.
  • Still another aspect disclosed in the present application provides a polynucleotide sequence encoding the iCP-SOCS3 recombinant protein.
  • the polynucleotide sequence may be a polynucleotide sequence represented by SEQ ID NO: 825.
  • Still another aspect disclosed in the present application provides a recombinant expression vector including the polynucleotide sequence.
  • Still another aspect disclosed in the present application provides a transformant transformed with the recombinant expression vector.
  • Still another aspect disclosed in the present application provides a preparing method of the iCP-SOCS3 recombinant protein including preparing the recombinant expression vector; preparing the transformant using the recombinant expression vector; culturing the transformant; and recovering the recombinant protein expressed by the culturing.
  • compositions including the iCP-SOCS3 recombinant protein as an active ingredient.
  • Still another aspect disclosed in the present application provides a pharmaceutical composition for the treating, preventing, or delaying the onset of, solid tumor including the iCP-SOCS3 recombinant protein as an active ingredient; and a pharmaceutically acceptable carrier.
  • Still another aspect disclosed in the present application provides use of the iCP-SOCS3 recombinant protein as a medicament for the treating, preventing, or delaying the onset of, solid tumor.
  • Still another aspect disclosed in the present application provides a medicament including the iCP-SOCS3 recombinant protein.
  • Still another aspect disclosed in the present application provides use of the iCP-SOCS3 recombinant protein in the preparation of a medicament for the treating, preventing, or delaying the onset of, solid tumor.
  • Still another aspect disclosed in the present application provides a method of treating, preventing, or delaying the onset of, solid tumor in a subject, the method including identifying a subject in need of the treating, preventing, or delaying the onset of, solid tumor; and administering to the subject a therapeutically effective amount of the iCP-SOCS3 recombinant protein.
  • the subject may be a mammal.
  • a “peptide,” as used herein, refers to a chain-type polymer formed by amino acid residues which are linked to each other via peptide bonds, and used interchangeably with “polypeptide.” Further, a “polypeptide” includes a peptide and a protein.
  • peptide includes amino acid sequences that are conservative variations of those peptides specifically exemplified herein.
  • conservative variation denotes the replacement of an amino acid residue by another, biologically similar residue. Examples of conservative variations include substitution of one hydrophobic residue, such as isoleucine, valine, leucine, alanine, cysteine, glycine, phenylalanine, proline, tryptophan, tyrosine, norleucine, or methionine for another, or substitution of one polar residue for another, for example, substitution of arginine for lysine, glutamic acid for aspartic acid, or glutamine for asparagine, and the like.
  • Neutral hydrophilic amino acids which may be substituted for one another include asparagine, glutamine, serine, and threonine.
  • conservative variation also includes use of a substituted amino acid in place of an unsubstituted parent amino acid, provided that antibodies raised to the substituted polypeptide also immunoreact with the unsubstituted polypeptide. Such conservative substitutions are within the definition of the classes of the peptides disclosed in the present application.
  • one aspect disclosed in the present application provides peptides corresponding to amino acid sequences (e.g. SEQ ID NOs: 1 to 240 and 822) provided herein, as well as analogues, homologs, isomers, derivatives, amidated variations, and conservative variations thereof, as long as the cell permeability of the peptide remains.
  • amino acid sequences e.g. SEQ ID NOs: 1 to 240 and 822
  • All peptides may be synthesized using L-amino acids, but D forms of all of the peptides may be synthetically produced.
  • C-terminal derivatives such as C-terminal methyl esters and C-terminal amidates, may be produced in order to increase the cell permeability of the peptide according to one embodiment disclosed in the present application.
  • deletion of one or more amino acids may also result in a modification to the structure of the resultant molecule without any significant change in its cell permeability. This may lead to the development of a smaller active molecule which may also have utility. For example, amino- or carboxyl-terminal amino acids which may not be required for the cell permeability of a particular peptide may be removed.
  • the term “gene” refers to an arbitrary nucleic acid sequence or a part thereof having a functional role in protein coding or transcription, or regulation of other gene expression.
  • the gene may be composed of all nucleic acids encoding a functional protein or a part of the nucleic acid encoding or expressing the protein.
  • the nucleic acid sequence may include a gene mutation in exon, intron, initiation or termination region, promoter sequence, other regulatory sequence, or a unique sequence adjacent to the gene.
  • primer refers to an oligonucleotide sequence that hybridizes to a complementary RNA or DNA target polynucleotide and serves as the starting points for the stepwise synthesis of a polynucleotide from mononucleotides by the action of a nucleotidyltransferase as occurs, for example, in a polymerase chain reaction.
  • coding region refers to a nucleic acid sequence, a complement thereof, or a part thereof which encodes a particular gene product or a fragment thereof for which expression is desired, according to the normal base pairing and codon usage relationships. Coding sequences include exons in genomic DNA or immature primary RNA transcripts, which are joined together by the cellular biochemical machinery to provide a mature mRNA. The anti-sense strand is the complement of the nucleic acid, and the coding sequence may be deduced therefrom.
  • an iCP-SOCS3 recombinant protein which comprises a SOCS3 protein and an advanced macromolecule transduction domain (aMTD) being composed of 9-13 amino acid sequences, preferably 10-12 amino acid sequences, and having improved cell and/or tissue permeability, wherein the aMTD is fused to one end or both ends of the SOCS3 protein and has the following features of:
  • (a) being preferably composed of 3 or more amino acid sequences selected from the group consisting of Ala, Val, Ile, Leu, and Pro;
  • GRAVY grand average of hydropathy
  • aMTD sequences were analyzed, identified, and determined according to one embodiment disclosed in the present application. These aMTD sequences are artificially assembled based on the critical factors (CFs) determined from in-depth analysis of previously published hydrophobic CPPs.
  • CFs critical factors
  • the aMTD sequences according to one aspect disclosed in the present application are the first artificially developed cell permeable polypeptides capable of mediating the transduction of biologically active macromolecules—including peptides, polypeptides, protein domains, or full-length proteins—through the plasma membrane of cells.
  • one or more solubilization domain (SD)(s) are further fused to one or more of the SOCS3 protein and the aMTD, preferably one end or both ends of the SOCS3 protein, and more preferably the C-terminus of the SOCS3 protein.
  • the aMTD may form a-Helix structure.
  • the aMTD may be preferably composed of 12 amino acid sequences and represented by the following general formula:
  • X(s) independently refer to Alanine (A), Valine (V), Leucine (L) or Isoleucine (I); and Proline (P) can be positioned in one of U(s) (either 5′, 6′, 7′ or 8′). The remaining U(s) are independently composed of A, V, L or I, P at the 12′ is Proline.
  • Still another aspect disclosed in the present application provides an iCP-SOCS3 recombinant protein which is represented by any one of structural formulae A-B-C, A-C-B, B-A-C, B-C-A, C-A-B, C-B-A and A-C-B-C, and preferably by A-B-C or C-B-A:
  • A is an advanced macromolecule transduction domain (aMTD) having improved cell and/or tissue permeability
  • B is a SOCS3 protein
  • C is a solubilization domain (SD);
  • the aMTD is composed of 9-13, preferably 10-12 amino acid sequences and has the following features of:
  • the SOCS3 protein may have an amino acid sequence of SEQ ID NO: 814.
  • the SOCS3 protein may be encoded by a polynucleotide sequence of SEQ ID NO: 815.
  • the SOCS3 protein may form a fusion product, together with an extracellular domain of a ligand capable of selectively binding to a receptor which is specifically expressed on the particular cell, tissue, or organ, or monoclonal antibody (mAb) capable of specifically binding to the receptor or the ligand and a modified form thereof.
  • a ligand capable of selectively binding to a receptor which is specifically expressed on the particular cell, tissue, or organ
  • mAb monoclonal antibody
  • the binding of the peptide and a biologically active substance may be formed either by indirect linkage by a cloning technique using an expression vector at a nucleotide level or by direct linkage via chemical or physical covalent or non-covalent bond of the peptide and the biologically active substance.
  • the SOCS3 protein may preferably further include a ligand selectively binding to a receptor of a cell, a tissue, or an organ.
  • the aMTD may have an amino acid sequence selected from the group consisting of SEQ ID NOs: 1-240 and 822, preferably SEQ ID NOs: 2, 16, 22, 32, 40, 43, 63, 65, 77, 84, 85, 86, 110, 131, 142, 143, 177, 228, 229, 233, 237, 239 and 822, more preferably SEQ ID NO: 43.
  • the aMTD may be encoded by a polynucleotide sequence selected from the group consisting of SEQ ID NOs: 241-480 and 823, preferably SEQ ID NOs: 242, 256, 262, 272, 280, 283, 303, 305, 317, 324, 325, 326, 350, 371, 382, 383, 417, 468, 469 473, 477, 479 and 823, more preferably SEQ ID NO: 283.
  • the SD(s) may have an amino acid sequence independently selected from the group consisting of SEQ ID NOs: 798, 799, 800, 801, 802, 803, and 804.
  • the SD may be preferably SDA of SEQ ID NO: 798, SDB of SEQ ID NO: 799, or SDB′ of SEQ ID NO: 804, and more preferably, SDB of SEQ ID NO: 799 which has superior structural stability, or SDB′ of SEQ ID NO: 804 which has a modified amino acid sequence of SDB to avoid immune responses upon in vivo application.
  • the modification of the amino acid sequence in SDB may be replacement of an amino acid residue, Valine, corresponding to position 28 of the amino acid sequence of SDB (SEQ ID NO: 799) by Leucine.
  • the SDs may be encoded by a polynucleotide sequence independently selected from the group consisting of SEQ ID NOs: 805, 806, 807, 808, 809, 810, and 811.
  • the SD may be preferably SDA encoded by a polynucleotide sequence of SEQ ID NO: 805, SDB encoded by a polynucleotide sequence of SEQ ID NO: 806, or SDB′ for deimmunization (or humanization) encoded by a polynucleotide sequence of SEQ ID NO: 811, and more preferably, SDB having superior structural stability, which is encoded by a polynucleotide sequence of SEQ ID NO: 806, or SDB′ having a modified polynucleotide sequence of SDB to avoid immune responses upon in vivo application, which is encoded by a polynucleotide sequence of SEQ ID NO: 811.
  • the iCP-SOCS3 recombinant protein may be preferably selected from the group consisting of:
  • a recombinant protein in which SOCS3 having an amino acid sequence of SEQ ID NO: 814 is fused to the N-terminus or the C-terminus of aMTD having any one amino acid sequence selected from the group consisting of SEQ ID NOs: 1 to 240 and 822, 2, 16, 22, 32, 40, 43, 63, 65, 77, 84, 85, 86, 110, 131, 142, 143, 177, 228, 229, 233, 237, 239 and 822, more preferably SEQ ID NO: 43;
  • recombinant protein in which a Histidine tag having an amino acid sequence of 812 is further fused to the N-terminus of the recombinant protein of 1) or 2).
  • the iCP-SOCS3 recombinant protein may be composed of an amino acid sequence selected from the group consisting of SEQ ID NO: 825.
  • the SOCS3 protein may exhibit a physiological phenomenon-related activity or a therapeutic purpose-related activity by intracellular or in-vivo delivery.
  • the recombinant expression vector may include a tag sequence which makes it easy to purify the recombinant protein, for example, consecutive histidine codon, maltose binding protein codon, Myc codon, etc., and further include a fusion partner to enhance solubility of the recombinant protein, etc.
  • the recombinant expression vector may further include one or more glycine, proline, and spacer amino acid or polynucleotide sequences including AAY amino acids.
  • the recombinant expression vector may include a sequence specifically digested by an enzyme in order to remove an unnecessary region of the recombinant protein, an expression regulatory sequence, and a marker or reporter gene sequence to verify intracellular delivery, but is not limited thereto.
  • the iCP-SOCS3 recombinant protein may preferably have a histidine-tag affinity domain additionally fused to one end thereof.
  • the histidine-tag affinity domain may have an amino acid sequence of SEQ ID NO: 812.
  • the histidine-tag affinity domain may be encoded by a polynucleotide sequence of SEQ ID NO: 813.
  • the fusion may be formed via a peptide bond or a chemical bond.
  • the chemical bond may be preferably selected from the group consisting of disulfide bonds, diamine bonds, sulfide-amine bonds, carboxyl-amine bonds, ester bonds, and covalent bonds.
  • the iCP-SOCS3 recombinant protein may be used for the treating, preventing, or delaying the onset of, solid tumor.
  • Still another aspect disclosed in the present application provides a polynucleotide sequence encoding the iCP-SOCS3.
  • the polynucleotide sequence may be fused with a histidine-tag affinity domain.
  • Still another aspect disclosed in the present application provides a recombinant expression vector including the polynucleotide sequence.
  • the vector may be inserted in a host cell and recombined with the host cell genome, or refers to any nucleic acid including a nucleotide sequence competent to replicate spontaneously as an episome.
  • a vector may include a linear nucleic acid, a plasmid, a phagemid, a cosmid, an RNA vector, a viral vector, etc.
  • the vector may be genetically engineered to incorporate the nucleic acid sequence encoding the recombinant protein in an orientation either N-terminal and/or C-terminal to a nucleic acid sequence encoding a peptide, a polypeptide, a protein domain, or a full-length protein of interest, and in the correct reading frame so that the recombinant protein consisting of aMTD, SOCS3 protein, and preferably SD may be expressed.
  • Expression vectors may be selected from those readily available for use in prokaryotic or eukaryotic expression systems.
  • Standard recombinant nucleic acid methods may be used to express a genetically engineered recombinant protein.
  • the nucleic acid sequence encoding the recombinant protein according to one embodiment disclosed in the present application may be cloned into a nucleic acid expression vector, e.g., with appropriate signal and processing sequences and regulatory sequences for transcription and translation, and the protein may be synthesized using automated organic synthetic methods. Synthetic methods of producing proteins are described in, for example, the literature [Methods in Enzymology, Volume 289: Solid-Phase Peptide Synthesis by Gregg B. Fields (Editor), Sidney P. Colowick, Melvin I. Simon (Editor), Academic Press (1997)].
  • the recombinant protein sequence may be typically subcloned into an expression vector that includes a strong promoter for directing transcription, a transcription/translation terminator, and in the case of a nucleic acid encoding a protein, a ribosome binding site for translational initiation.
  • a strong promoter for directing transcription e.g., a transcription/translation terminator
  • a nucleic acid encoding a protein e.g., a ribosome binding site for translational initiation.
  • Suitable bacterial promoters are well known in the art and are described, e.g., in the literatures [Sambrook & Russell, Molecular Cloning: A Laboratory Manual, 3d Edition, Cold Spring Harbor Laboratory, N.Y.
  • Bacterial expression systems for expression of the recombinant protein are available in, e.g., E. coli, Bacillus sp., and Salmonella (Palva et al., Gene 22: 229-235 (1983); Mosbach et al., Nature 302: 543-545 (1983)). Kits for such expression systems are commercially available. Eukaryotic expression systems for mammalian cells, yeast, and insect cells are well known in the art and are also commercially available.
  • the eukaryotic expression vector may be preferably an adenoviral vector, an adeno-associated vector, or a retroviral vector.
  • the expression vector for expressing the cell permeable recombinant protein in which the cargo protein, i.e. ⁇ SOCS3 protein, is attached to the N-terminus, C-terminus, or both termini of aMTD may include regulatory sequences including, for example, a promoter, operably attached to a sequence encoding the advanced macromolecule transduction domain.
  • Non-limiting examples of inducible promoters include steroid-hormone responsive promoters (e.g., ecdysone-responsive, estrogen-responsive, and glutacorticoid-responsive promoters), tetracycline “Tet-On” and “Tet-Off” systems, and metal-responsive promoters.
  • steroid-hormone responsive promoters e.g., ecdysone-responsive, estrogen-responsive, and glutacorticoid-responsive promoters
  • Tet-On tetracycline “Tet-On” and “Tet-Off” systems
  • metal-responsive promoters metal-responsive promoters.
  • the polynucleotide sequence according to one embodiment disclosed in the present application may be present in a vector in which the polynucleotide sequence is operably linked to regulatory sequences capable of providing for the expression of the polynucleotide sequence by a suitable host cell.
  • polynucleotide sequence may be selected from the following groups:
  • a polynucleotide sequence in which any one polynucleotide sequence selected from the group consisting of SEQ ID NOs: 241-480 and 823, preferably SEQ ID NOs: 242, 252, 274, 279, 322, 331, 338, 345, 347, 361, 365, 370, 371, 383, 387, 417, 462, 468, 469, 473, 477, 479 and 823, more preferably SEQ ID NO: 283, is operably linked with a polynucleotide sequence of SEQ ID NO: 815; and
  • any one polynucleotide sequence selected from the group consisting of SEQ ID NOs: 805 to 811 is further operably linked to the polynucleotide sequence of 1), or further operably linked to between: any one polynucleotide sequence selected from the group consisting of SEQ ID NOs: 241-480 and 823, preferably SEQ ID NOs: 242, 256, 262, 272, 280, 283, 303, 305, 317, 324, 325, 326, 350, 371, 382, 383, 417, 468, 469, 473, 477, 479 and 823, more preferably SEQ ID NO: 283; and a polynucleotide sequence of SEQ ID NO: 815.
  • operably linked is intended to mean that the polynucleotide sequence of interest is linked to the regulatory sequence(s) in a manner which allows for expression of the polynucleotide sequence.
  • regulatory sequence is intended to include promoters, enhancers, and other expression control elements.
  • the expression vectors may contain a signal sequence or a leader sequence for membrane targeting or secretion, as well as regulatory sequences such as a promoter, an operator, an initiation codon, a termination codon, a polyadenylation signal, an enhancer and the like.
  • the promoter may be a constitutive or an inducible promoter.
  • the expression vector may include one or more selectable marker genes for selecting the host cell containing the expression vector, and may further include a polynucleotide sequence that enables the vector to replicate in the host cell in question.
  • the expression vector constructed according to one embodiment disclosed in the present application may be the vector where the polynucleotide encoding the iCP-SOCS3 recombinant protein (where an aMTD is fused to the N-terminus or C-terminus of a SOCS3 protein) is inserted within the multiple cloning sites (MCS), preferably within the Nde1/Sal1 site or BamH1/Sal1 site of a pET-28a(+)(Novagen, Darmstadt, Germany) or pET-26b(+) vector(Novagen, Darmstadt, Germany).
  • MCS multiple cloning sites
  • the polynucleotide encoding the SD being additionally fused to the N-terminus or C-terminus of a SOCS3 protein or an aMTD may be inserted into a cleavage site of restriction enzyme (Ndel, BamH1 and Sal1, etc.) within the multiple cloning sites (MCS) of a pET-28a(+)(Novagen, Darmstadt, Germany) or pET-26b(+) vector(Novagen, Darmstadt, Germany).
  • a cleavage site of restriction enzyme Ndel, BamH1 and Sal1, etc.
  • the polynucleotide encoding the iCP-SOCS3 recombinant protein may be cloned into a pET-28a(+) vector bearing a His-tag sequence so as to fuse six histidine residues to the N-terminus of the iCP-SOCS3 recombinant protein to allow easy purification.
  • the polynucleotide sequence may be a polynucleotide sequence selected from the group consisting of SEQ ID NOS: 824, 826, 828 and 830.
  • the recombinant protein may be introduced into an appropriate host cell, e.g., a bacterial cell, a yeast cell, an insect cell, or a tissue culture cell.
  • the recombinant protein may also be introduced into embryonic stem cells in order to generate a transgenic organism. Large numbers of suitable vectors and promoters are known to those skilled in the art and are commercially available for generating the recombinant protein.
  • vectors including the polynucleotide sequence according to one embodiment disclosed in the present application and appropriate transcriptional/translational control signals.
  • These methods include in vitro recombinant DNA techniques, synthetic techniques, and in vivo recombination/genetic recombination.
  • these techniques are described in the literatures [Sambrook & Russell, Molecular Cloning: A Laboratory Manual, 3d Edition, Cold Spring Harbor Laboratory, N. Y. (2001); and Ausubel et al., Current Protocols in Molecular Biology Greene Publishing Associates and Wiley Interscience, N.Y. (1989)].
  • Still another aspect disclosed in the present application provides a transformant transformed with the recombinant expression vector.
  • the transformation includes transfection, and refers to a process whereby a foreign (extracellular) DNA, with or without an accompanying material, enters into a host cell.
  • the “transfected cell” refers to a cell into which the foreign DNA is introduced into the cell, and thus the cell harbors the foreign DNA.
  • the DNA may be introduced into the cell so that a nucleic acid thereof may be integrated into the chromosome or replicable as an extrachromosomal element.
  • the cell introduced with the foreign DNA, etc. is called a transformant.
  • introducing’ of a protein, a peptide, an organic compound into a cell may be used interchangeably with the expression of ‘carrying,’ ‘penetrating,’‘transporting,’‘delivering,’ ‘permeating’ or ‘passing.’
  • the host cell refers to a eukaryotic or prokaryotic cell into which one or more DNAs or vectors are introduced, and refers not only to the particular subject cell but also to the progeny or potential progeny thereof. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein.
  • the host cells may be preferably bacterial cells, and as the bacterial cells, there are, in principle, no limitations. They may be eubacteria (gram-positive or gram-negative) or archaebacteria, as long as they allow genetic manipulation for insertion of a gene of interest, preferably for site-specific integration, and they may be cultured on a manufacturing scale. Preferably, the host cells may have the property to allow cultivation to high cell densities.
  • Examples of bacterial host cells that may be used in the preparation of the recombinant protein are E. coli (Lee, 1996; Hannig and Makrides, 1998), Bacillus subtilis, Pseudomonas fluorescens (Squires et al., 2004; Retallack et al., 2006) as well as various Corynebacterium (US 2006/0003404 A1) and Lactococcus lactis (Mierau et al., 2005) strains.
  • the host cells are Escherichia coli cells.
  • host cells with a foreign strong RNA polymerase may be used.
  • Escherichia coli strains engineered to carry a foreign RNA polymerase e.g. like in the case of using a T7 promoter a T7-like RNA polymerase in the so-called “T7 strains” integrated in their genome may be used.
  • T7 strains e.g. BL21(DE3), HMS174(DE3), and their derivatives or relatives (see Novagen, pET System manual, 11 th edition
  • BL21-CodonPlus (DE3)-RIL or BL21-CodonPlus (DE3)-RIPL Algilent Technologies
  • strains are DE3 lysogens containing the T7 RNA polymerase gene under control of the lacUV5 promoter. Induction with IPTG allows production of T7 RNA polymerase which then directs the expression of the gene of interest under the control of the T7 promoter.
  • the host cell strains E. coli BL21(DE3) or HMS174(DE3), which have received their genome-based T7 RNA polymerase via the phage DE3, are lysogenic. It is preferred that the T7 RNA polymerase contained in the host cell has been integrated by a method which avoids, or preferably excludes, the insertion of residual phage sequences in the host cell genome since lysogenic strains have the disadvantage to potentially exhibit lytic properties, leading to undesirable phage release and cell lysis.
  • Still another aspect disclosed in the present application provides a preparing method of the iCP-SOCS3 recombinant protein including preparing the recombinant expression vector; preparing the transformant using the recombinant expression vector; culturing the transformant; and recovering the recombinant protein expressed by culturing.
  • Culturing may be preferably in a mode that employs the addition of a feed medium, this mode being selected from the fed-batch mode, semi-continuous mode, or continuous mode, and the bacterial expression host cells may include a DNA construct, integrated in their genome, carrying the DNA sequence encoding the protein of interest under the control of a promoter that enables expression of said protein.
  • the culture medium may be semi-defined, i.e. containing complex media compounds (e.g. yeast extract, soy peptone, casamino acids), or it may be chemically defined, without any complex compounds.
  • a defined medium may be used.
  • the defined media also called minimal or synthetic media
  • glucose may be used as a carbon source.
  • the carbon source of the feed medium serves as the growth-limiting component which controls the specific growth rate.
  • Host cells may be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption, or the use of cell lysing agents.
  • the literature describes a number of general methods for purifying recombinant (and non-recombinant) proteins.
  • the methods may include, e.g., ion-exchange chromatography, size-exclusion chromatography, affinity chromatography, selective precipitation, dialysis, and hydrophobic interaction chromatography. These methods may be adapted to devise a purification strategy for the cell permeable recombinant protein. If the cell permeable recombinant protein includes a purification handle, such as an epitope tag or a metal chelating sequence, affinity chromatography may be used to easily purify the protein.
  • a purification handle such as an epitope tag or a metal chelating sequence
  • the amount of the protein produced may be evaluated by detecting the advanced macromolecule transduction domain directly (e.g., using Western analysis) or indirectly (e.g., by assaying materials derived from the cells for specific DNA binding activity, such as by electrophoretic mobility shift assay). Proteins may be detected prior to purification, during any stage of purification, or after purification. In some implementations, purification or complete purification may not be necessary.
  • the iCP-SOCS3 recombinant proteins may be preferably used for the treating, preventing, or delaying the onset of, solid tumor.
  • the cell permeable recombinant proteins may be delivered to the interior of the cell, eliminating the need to transfect or transform the cell with a recombinant vector.
  • the cell permeable recombinant proteins may be used in vitro to investigate protein function or may be used to maintain cells in a desired state.
  • compositions including the iCP-SOCS3 Recombinant Protein as an active ingredient.
  • Still another aspect disclosed in the present application provides a pharmaceutical composition for treating, preventing, or delaying the onset of, solid tumor including the iCP-SOCS3 Recombinant Protein as an active ingredient; and a pharmaceutically acceptable carrier.
  • the iCP-SOCS3 Recombinant Protein may be used in a single agent, or in combination with one or more other anti-cancer agents.
  • Solid tumor described herein include, but are not limited to, described herein include, but are not limited to, prostate cancer, renal cancer, hepatic cancer, colon cancer, rectal cancer, colorectal cancer, brain cancer, renal cancer, colorectal cancer, pancreatic cancer, gastric cancer, breast cancer, lung cancer, cancers of the head and neck, thyroid cancer, glioblastoma, Kaposi's sarcoma, melanoma, oesophageal cancer, gastro-oesophageal cancer, cervical cancer, hepatocellular carcinoma, endometrial cancer, urothelial cancer, or ovarian cancer.
  • the tumor may be early stage tumor, metastatic tumor, non-metastatic tumor, primary tumor, resected tumor, advanced tumor, locally advanced tumor, unresectable tumor, tumor in remission, or recurrent tumor.
  • the composition may be for injectable (e.g. intraperitoneal, intravenous, and intra-arterial, etc.) and may include the active ingredient in an amount of 0.001 mg/kg to 1000 mg/kg, preferably 0.01 mg/kg to 100 mg/kg, more preferably 0.1 mg/kg to 50 mg/kg for human.
  • injectable e.g. intraperitoneal, intravenous, and intra-arterial, etc.
  • the active ingredient in an amount of 0.001 mg/kg to 1000 mg/kg, preferably 0.01 mg/kg to 100 mg/kg, more preferably 0.1 mg/kg to 50 mg/kg for human.
  • dosages per day normally fall within the range of about 0.001 to about 1000 mg/kg of body weight. In the treatment of adult humans, the range of about 0.1 to about 50 mg/kg/day, in single or divided dose, is especially preferred.
  • concentration of the iCP-SOCS3 recombinant protein actually administered will be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the age, weight, and response of the individual patient, and the severity of the patient's symptoms, and therefore the above dosage ranges are not intended to limit the scope of the invention in any way. In some instances dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed without causing any harmful side effect, provided that such larger doses are first divided into several smaller doses for administration throughout the day.
  • Still another aspect disclosed in the present application provides use of the iCP-SOCS3 recombinant protein as a medicament for treating, preventing, or delaying the onset of, solid tumor.
  • Still another aspect disclosed in the present application provides a medicament including the iCP-SOCS3 recombinant protein.
  • Still another aspect disclosed in the present application provides a method of treating, preventing, or delaying the onset of, solid tumor in a subject including identifying a subject in need of treating, preventing, or delaying the onset of, solid tumor; and administering to the subject a therapeutically effective amount of the iCP-SOCS3 recombinant protein.
  • the subject may be preferably a mammal.
  • the subject in need of treating, preventing, or delaying the onset of, solid tumor may be identified by any conventional diagnostic methods known in the art including ultrasound, CT scan, MRI, alpha-fetoprotein testing, and biopsy, etc.
  • the pharmaceutical composition according to one embodiment disloced in the present application may be prepared by using pharmaceutically suitable and physiologically acceptable additives, in addition to the active ingredient, and the additives may include excipients, disintegrants, sweeteners, binders, coating agents, blowing agents, lubricants, glidants, flavoring agents, etc.
  • the pharmaceutical composition may be preferably formulated by further including one or more pharmaceutically acceptable carriers in addition to the above-described active ingredient.
  • Dosage forms of the pharmaceutical composition may include granules, powders, tablets, coated tablets, capsules, suppositories, liquid formulations, syrups, juice, suspensions, emulsions, drops, injectable liquid formulations, etc.
  • the active ingredient may be combined with any oral, non-toxic pharmaceutically acceptable inert carrier, such as ethanol, glycerol, water, etc.
  • suitable binders, lubricants, disintegrants, and colorants may be additionally included as a mixture.
  • Suitable binder may include, but are not limited to, starch, gelatin, natural sugars such as glucose or beta-lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth, or sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride, etc.
  • examples of the disintegrant may include, but are not limited to, starch, methyl cellulose, agar, bentonite, xanthan gum, etc.
  • a pharmaceutically acceptable carrier which is sterile and biocompatible may be used, such as saline, sterile water, a Ringer's solution, buffered saline, an albumin infusion solution, a dextrose solution, a maltodextrin solution, glycerol, and ethanol, and these materials may be used alone or in any combination thereof. If necessary, other common additives, such as antioxidants, buffers, bacteriostatic agents, etc., may be added.
  • compositions may be additionally added to prepare injectable formulations such as aqueous solutions, suspensions, and emulsions, or pills, capsules, granules, or tablets.
  • injectable formulations such as aqueous solutions, suspensions, and emulsions, or pills, capsules, granules, or tablets.
  • the composition may be preferably formulated, depending upon diseases and ingredients, using any appropriate method known in the art, as disclosed in Remington's Pharmaceutical Science, Mack Publishing Company, Easton Pa.
  • the treatment or treating mean improving or stabilizing the subject's condition or disease; or preventing or relieving the development or worsening of symptoms associated with the subject's condition or disease.
  • the prevention, prophylaxis and preventive treatment are used herein as synonyms.
  • the treating, preventing, or delaying the onset of, solid tumor may be any one or more of the following: alleviating one or more symptoms of tumor, delaying progressing of tumor, shrinking tumor size in patient, inhibiting tumor growth, prolonging overall survival, prolonging disease-free survival, prolonging time to disease progression, preventing or delaying tumor metastasis, reducing or eradiating preexisting tumor metastasis, reducing incidence or burden of preexisting tumor metastasis, preventing recurrence of tumor.
  • the subject and patient are used herein interchangeably. They refer to a human or another mammal (e.g., mouse, rat, rabbit, dog, cat, cattle, swine, sheep, horse or primate) that can be afflicted with or is susceptible to a disease or disorder but may or may not have the disease or disorder.
  • the subject is a human being.
  • the amount effective or effective amount is the amount of an active ingredient or a pharmaceutical composition disclosed herein that when administered to a subject for treating a disease, is sufficient to effect such treatment of the disease. Any improvement in the patient is considered sufficient to achieve treatment.
  • An effective amount of an active ingredient or a pharmaceutical composition disclosed herein, used for the preventing, or delaying the onset of, solid tumor may vary depending upon the manner of administration, the age, body weight, and general health of the patient. Ultimately, the prescribers or researchers will decide the appropriate amount and dosage regimen.
  • the composition including the iCP-SOCS3 recombinant protein as an active ingredient may be administered in a common manner via oral, buccal, rectal, intravenous, intra-arterial, intraperitoneal, intramuscular, intrasternal, percutaneous, topical, intraocular or subcutaneous route, more preferably via intraperitoneal, intravenous, or intra-arterial injection route.
  • FIG. 1 shows Structure of aMTD- or rPeptide-Fused Recombinant Proteins.
  • a schematic diagram of the His-tagged CRA recombinant proteins is illustrated and constructed according to the present invention.
  • the his-tag for affinity purification (white), aMTD or rPeptide (gray) and cargo A (CRA, black) are shown.
  • FIG. 2 a shows Construction of Expression Vectors for aMTDs- or rPeptide-Fused Recombinant Proteins.
  • FIGS. 2 b and 2 c show the agarose gel electrophoresis analysis showing plasmid DNA fragments at 645 bp insert encoding aMTDs or rPeptide-fused CRA cloned into the pET28a(+) vector according to the present invention.
  • FIGS. 3 a to 3 d show Inducible Expression of aMTD- or rPeptide-Fused Recombinant Proteins.
  • Expressed recombinant aMTD- or random peptide-fused CRA recombinant proteins were transformed in E. coli BL21 (DE3) strain.
  • Expression of recombinant proteins in E. coli before ( ⁇ ) and after (+) induction with IPTG was monitored by SDS-PAGE, and stained with Coomassie blue.
  • FIGS. 4 a and 4 b show Purification of aMTD- or rPeptide-Fused Recombinant Proteins. Expressed recombinant proteins were purified by Ni 2 + affinity chromatography under the natural condition. Purification of recombinant proteins displayed through SDS-PAGE analysis.
  • FIGS. 5 a to 5 u show Determination of aMTD-Mediated Cell-Permeability.
  • Cell-permeability of a negative control (A: rP38) and reference hydrophobic CPPs (MTM12 and MTD85) are shown.
  • the cell-permeability of each aMTD and/or rPeptide is visually compared to that of the cargo protein lacking peptide sequence (HCA).
  • Gray shaded area represents untreated RAW 264.7 cells (vehicle); thin light gray line represents the cells treated with equal molar concentration of FITC (FITC only); dark thick line indicates the cells treated with FITC-his-tagged CRA protein (HCA); and the cells treated with the FITC-proteins (HMCA) fused to negative control (rP38), reference CPP (MTM12 or MTD85) or new hydrophobic CPP (aMTD) are shown with light thick line and indicated by arrows.
  • FIGS. 6 a to 6 c show Determination of rPeptide-Mediated Cell-Permeability.
  • the cell-permeability of each aMTD and/or rPeptide was visually compared to that of the cargo protein lacking peptide sequence (HCA).
  • Gray shaded area represents untreated RAW 264.7 cells (vehicle); thin light gray line represents the cells treated with equal molar concentration of FITC (FITC only); dark thick line indicates the cells treated with FITC-his-tagged CRA protein (HCA); and the cells treated with the FITC-proteins fused to rPeptides are shown with light thick line and indicated by arrows.
  • FIGS. 7 a to 7 k shows Visualized Cell-Permeability of aMTD-Fused Recombinant Proteins.
  • NIH3T3 clls were treated with FITC-labeled protein (10 ⁇ M) fused to aMTD for 1 hour at 37.
  • Cell-permeability of the proteins was visualized by laser scanning confocal microscopy (LSM700 version).
  • FIG. 8 shows Visualized Cell-Permeability of rPeptide-Fused Recombinant Proteins.
  • Cell-permeability of rPeptide-fused recombinant proteins was visualized by laser scanning confocal microscopy (LSM700 version).
  • FIGS. 9 a to 9 c show Relative Cell-Permeability of aMTD-Fused Recombinant Proteins Compared to Negative Control (rP38).
  • the FIG. shows graphs comparing the cell-permeability of the recombinant proteins fused to aMTDs and a negative control (A: rP38).
  • FIGS. 11 a to 11 c show Relative Cell-Permeability of aMTD-Fused Recombinant Proteins Compared to Reference CPP (MTD85).
  • the FIG. shows graphs comparing the cell-permeability of the recombinant proteins fused to aMTDs and a reference CPP (MTD85).
  • FIG. 12 shows Relative Cell-Permeability of rPeptide-Mediated Recombinant Proteins Compared to Average that of aMTDs.
  • the FIG. shows graphs comparing the cell-permeability of the recombinant proteins fused to rPeptides and that (average value: aMTD AVE) of aMTDs.
  • FIGS. 14 a and 14 b show Association of Cell-Permeability with Critical Factors in aMTDs. These graphs show the association of cell-permeability with critical factors [bending potential: proline position (PP), rigidity/flexibility: instability index (II), structural feature: aliphatic index (AI) and hydropathy: grand average of hydropathy (GRAVY)].
  • PP proline position
  • II instability index
  • AI structural feature
  • GRAVY grand average of hydropathy
  • FIGS. 15 a and 15 b show Relative Relevance of aMTD-Mediated Cell-Permeability with Critical Factors.
  • Cell-permeability of 10 high and 10 low ranked aMTDs in their delivery potential were examined for their association with the critical factors [bending potential: proline position (PP), rigidity/flexibility: instability index (II), structural feature: aliphatic index (AI) and hydropathy: grand average of hydropathy (GRAVY)].
  • PP proline position
  • II instability index
  • AI structural feature
  • GRAVY grand average of hydropathy
  • FIG. 16 shows Relative Relevance of rPeptide-Mediated Cell-Permeability with Hydropathy Range (GRAVY). This graph and a chart illustrate relative relevance of rPeptide-mediated cell-permeability with its hydropathy range (GRAVY).
  • FIG. 17 shows a structure of iCP-SOCS3 recombinant protein designed according to example 6-1.
  • FIG. 20 shows aMTD-Mediated cell-permeability of SOCS3 recombinant proteins in RAW 264.7 cells according to example 7-1.
  • FIG. 21 shows aMTD-Mediated intracellular delivery and localization of SOCS3 Recombinant Proteins in NIH3T3 cells cells according to example 7-1.
  • FIG. 22 shows systemic delivery of aMTD/SD-fused SOCS3 recombinant proteins in vivo according to example 7-2.
  • FIG. 23 shows inhibition of IFN- ⁇ -induced STAT phosphorylation by iCP-SOCS3 recombinant protein according to example 8-1.
  • FIG. 24 shows inhibition of LPS-induced cytokines secretion by iCP-SOCS3 recombinant protein according to example 8-2.
  • FIG. 25 shows the structures of SOCS3 recombinant protein lacking aMTD prepared as a negative control according to example 9.
  • FIG. 26 shows expression, purification, and solubility/yield of HS3 (lacking aMTD and SD) and HS3B (lacking aMTD) determined according to example 6-3.
  • FIG. 27 shows the agarose gel electrophoresis analysis showing plasmid DNA fragments insert encoding His-aMTD#-SOCS3-SDB (HM # S3B) and His-rP#-SOCS3-SDB cloned into the pET28a (+) vector according to example 10.
  • FIGS. 28 a and 28 b show expression, purification, and solubility/yield of His-aMTD#-SOCS3-SDB (HM # S3B) determined according to example 10.
  • FIG. 29 shows expression, purification, and solubility/yield of His-rP#-SOCS3-SDB (HrP # S3B) determined according to example 10.
  • FIG. 30 shows solubility/yield of His-aMTD#-SOCS3-SDB (HM # S3B) determined according to example 10.
  • FIG. 31 show aMTD-mediated cell-permeability.
  • the cell-permeability of each SOCS3 recombinant protein fused with SD and various aMTD is visually compared to that of the cargo protein lacking CPP (HS3B) or lacking CPP and SD (HS3). Gray shaded area represents untreated E.
  • coli cells (diluent); green line represents the cells treated with equal molar concentration of FITC (FITC only); black line indicates the cells treated with FITC-his-SOCS protein (FITC-HS3); blue line indicates the cells treated with FITC-his-SOCS-SDB protein (FITC-HS3B) purple line indicates the cells treated with FITC-his-aMTD # -SOCS-SDB protein (FITC-HM # S3B).
  • FIG. 32 shows relative cell-permeability of His-aMTD # -SOCS3-SDB-Fused recombinant proteins Compared to control (Vehicle, FITC only, HS3 and HS3B).
  • FIG. 33 shows random Peptide-Mediated cell-permeability.
  • the cell-permeability of each SOCS3 recombinant protein fused with SDB and aMTD 165 or various rP is visually compared to that of the cargo protein lacking CPP (HS3B) or lacking CPP and SD (HS3). Gray shaded area represents untreated E.
  • coli cells (diluent); green line represents the cells treated with equal molar concentration of FITC (FITC only); black line indicates the cells treated with FITC-his-SOCS protein (FITC-HS3); blue line indicates the cells treated with FITC-his-SOCS-SDB protein (FITC-HS3B) and purple line indicates the cells treated with FITC-his-rPeptide # -SOCS-SDB protein (FITC-HrP # S3B).
  • FIG. 34 shows relative cell-permeability of His-rP # -SOCS3-SDB-Fused recombinant proteins Compared to control (Vehicle, FITC only, HS3 and HS3B).
  • FIG. 35 shows apoptotic cells analysis according to example 11-1.
  • FIG. 36 shows induction of apoptosis by iCP-SOCS3 recombinant proteins according to example 11-2.
  • FIG. 37 shows cell migration inhibition by iCP-SOCS3 recombinant protein according to example 11-3.
  • FIG. 38 shows solubility/yield, permeability and biological activity of His-aMTD#-SOCS3-SDB (HM # S3B) determined according to example 10 to 11-3.
  • FIG. 39 shows expression, purification, and solubility/yield of M 165 S3SB (lacking his-tag) determined according to example 12-1.
  • FIG. 40 shows cell-permeability of SOCS3 recombinant proteins (lacking his-tag) in RAW 264.7 cells according to example 12-2.
  • FIG. 41 shows Annexin V analysis according to example 12-3.
  • FIG. 42 shows cell migration inhibition (bottom) by iCP-SOCS3 recombinant protein according to example 12-3.
  • FIG. 43 shows a structure of iCP-SOCS3 recombinant protein (His-aMTD 165 -50C53-SDB′) constructed according to example 12-4.
  • FIG. 44 shows expression, purification, and solubility/yield of HM 165 S3SB and HM 165 S3SB′ determined according to example 12-4.
  • FIG. 45 shows aMTD-Mediated cell-permeability of iCP-SOCS3 recombinant proteins (HM 165 S3B and HM 165 S3B′(V28L)) in RAW 264.7 cells according to example 12-5.
  • FIG. 46 shows antiproliferative activity of iCP-SOCS3 recombinant proteins (HM 165 S3B and HM 165 S3B′(V28L)) according to example 12-6.
  • FIG. 47 shows induction of apoptosis by iCP-SOCS3 recombinant proteins (HM 165 S3B and HM 165 S3B′(V28L)) according to example 12-6.
  • FIG. 49 a shows a structure of iCP-SOCS3 recombinant proteins (top) and agarose gel electrophoresis analysis (bottom) according to example 12-7 and
  • FIG. 49 b shows inducible expressions and purifications of iCP-SOCS3 recombinant protein in E. coli (bottom) according to example 12-7.
  • FIG. 50 shows inhibition of IFN- ⁇ -induced STAT phosphorylation by iCP-SOCS3 recombinant protein according to example 13.
  • FIG. 52 shows effect of treating taxol ( FIG. 52 A) and antimycin ( FIG. 52 B) on aMTD-mediated SOCS3 protein uptake into cells according to example 14.
  • FIG. 53 shows effect of temperature on aMTD-mediated SOCS3 protein uptake into cells according to example 14.
  • FIG. 54 shows aMTD-mediated cell-to-cell delivery assessed according to example 14.
  • FIG. 55 shows bioavailability of iCP-SOCS3 recombinant protein in PBMC, splenocytes and hepatocytes analyzed by fluorescence microscopy according to example 15.
  • FIG. 58 shows systemic delivery of aMTD/SD-fused SOCS3 recombinant proteins into Various Tissues according to example 16.
  • FIG. 59 shows expression level of endogenous SOCS3 mRNA in gastric cell line analyzed by the agarose gel electrophoresis according to example 17-1-2.
  • FIG. 62 shows expression level of SOCS3 gene, and phosphorylation of JAK1 and JAK2 in colon cancer cell line analyzed by western blot analysis according to example 17-2-3.
  • FIG. 63 shows methylation and unmethylation level of endogenous SOCS3 in glioblastoma cell line analyzed by the agarose gel electrophoresis according to example 17-3-1.
  • FIG. 64 shows expression level of SOCS3 gene, and phosphorylation of JAK1 and JAK2 in glioblastoma cancer cell line analyzed by western blot analysis according to example 17-3-2.
  • FIG. 65 shows expression level of endogenous SOCS3 mRNA in breast cancer cell line analyzed by the agarose gel electrophoresis according to example 17-4-1.
  • FIG. 66 shows methylation and unmethylation level of endogenous SOCS3 in breast cancer cell line analyzed by the agarose gel electrophoresis according to example 17-4-2.
  • FIG. 67 shows expression level of SOCS3 gene, and phosphorylation of JAK1 and JAK2 in breast cancer cell line analyzed by western blot analysis according to example 17-4-3.
  • FIG. 68 shows antiproliferative activity of iCP-SOCS3 recombinant protein in gastric cancer cell line according to example 18-1.
  • FIG. 69 shows antiproliferative activity of iCP-SOCS3 recombinant protein in colorectal cancer cell line according to example 18-1.
  • FIG. 70 shows antiproliferative activity of iCP-SOCS3 recombinant protein in glioblastoma cell line according to example 18-1.
  • FIG. 71 shows antiproliferative activity of iCP-SOCS3 recombinant protein in breast cancer cell line according to example 18-1.
  • FIG. 73 shows cell migration inhibition activity by iCP-SOCS3 recombinant protein in colorectal cancer cell line (HCT116) according to example 18-2.
  • FIG. 74 shows cell migration inhibition activity by iCP-SOCS3 recombinant protein in glioblastoma cell line (U-87 MG) according to example 18-2.
  • FIG. 75 shows cell migration inhibition activity by iCP-SOCS3 recombinant protein in breast cancer cell line (MDA-MB-231) according to example 18-2.
  • FIG. 76 show transwell migration inhibition activity by iCP-SOCS3 recombinant protein in gastric cancer cell line (AGS) ( FIG. 76 , left), and decrease in invasion by iCP-SOCS3 recombinant protein in gastric cancer cell line (AGS) ( FIG. 76 , right) analyzed according to example 18-2.
  • FIG. 77 show transwell migration inhibition activity by iCP-SOCS3 recombinant protein in glioblstoma cell line (U-87 MG) analyzed according to example 18-2.
  • FIGS. 78 to 82 show induction of apoptosis in normal cells ( FIG. 78 ), in gastric cancer cells ( FIG. 79 ), in colorectal cancer cells (HCT116, FIG. 80 ), in glioblastoma cells (U-87 MG, FIG. 81 ), and in breast cancer cells (MDA-MB-231, FIG. 82 ) by iCP-SOCS3 recombinant proteins assessed by Annexin V staining according to example 18-3.
  • FIG. 83 show induction of apoptosis in colon cancer cells (HCT116) by iCP-SOCS3 recombinant proteins analyzed by TUNEL assay according to example 18-4.
  • FIGS. 84 to 88 show inhibition of cell cycle progression in normal cells ( FIG. 84 ), in gastric cancer cells (AGS, FIG. 85 ), in colon cancer cells (HCT116, FIG. 86 ), in glioblastoma cells (U-87 MG, FIG. 87 ), and in breast cancer cells (MDA-MB-231, FIG. 88 ) by iCP-SOCS3 recombinant proteins assessed by flow cytometric analysis according to example 18-5.
  • FIG. 89 shows down regulation of expression of apoptosis by iCP-SOCS3 recombinant proteins in breast cancer cells (MDA-MB-231) determined by immunoblotting according to example 18-6.
  • FIG. 90 shows suppression of the tumor growth by iCP-SOCS3 recombinant proteins in gastric cancer cells (NCI-N87) assessed according to example 19-1.
  • FIG. 91 shows suppression of the tumor growth by iCP-SOCS3 recombinant proteins in colorectal cancer cells (HCT116) assessed according to example 19-2.
  • FIG. 92 shows suppression of the tumor growth by iCP-SOCS3 recombinant proteins in glioblastoma cells (U-87 MG) assessed according to example 19-3.
  • FIG. 93 shows suppression of the tumor growth by iCP-SOCS3 recombinant proteins in breast cancer cells (MDA-MB-231) assessed according to example 19-4.
  • FIG. 94 shows humanized SDB domain according to example 12-4.
  • FIG. 95 shows methylation and unmethylation level of endogenous SOCS3 in gastric cancer cell line analyzed by the agarose gel electrophoresis according to example 17-1-1.
  • FIG. 96 shows methylation and unmethylation level of endogenous SOCS3 in colon cancer cell line analyzed by the agarose gel electrophoresis according to example 17-2-1.
  • FIG. 97 shows sequences of amino acid and nucleotide of basic CPP, and primers used in example 6-4.
  • FIG. 98 shows structure, expression, purification and solubility/yield of aMTD/SD-fused SOCS3 recombinant protein and basic CPP/SD-fused SOCS3 recombinant protein according to example 6-4.
  • FIG. 99 shows comparison of cell-permeability between aMTD/SD fused SOCS3 recombinant proteins and basic/SD-fused in RAW 264.7 cells according to example 7-1-2.
  • FIG. 100 shows comparison of tissue-permeability between aMTD/SD fused SOCS3 recombinant proteins and basic/SD-fused in various tissues of ICR mice according to example 7-2-2.
  • FIG. 101 shows effect of treating protein K ( FIG. 101 A) and Taxol ( FIG. 101 B) on aMTD (or basic CPP)-mediated SOCS3 protein uptake into cells according to example 14-2.
  • FIGS. 102 and 103 show aMTD (or basic CPP)-mediated cell-to-cell delivery ( FIG. 102 ) and cell-to-cell function assessed according to example 14-2.
  • MTDs were selected from a screen of more than 1,500 signal peptide sequences. Although the MTDs that have been developed did not have a common sequence or sequence motif, they were all derived from the hydrophobic (H) regions of signal sequences (HRSSs) that also lack common sequences or motifs except their hydrophobicity and the tendency to adopt alpha-helical conformations.
  • HRSSs hydrophobic regions of signal sequences
  • the wide variation in H-region sequences may reflect prior evolution for proteins with membrane translocating activity and subsequent adaptation to the SRP/Sec61 machinery, which utilizes a methionine-rich signal peptide binding pocket in SRP to accommodate a wide-variety of signal peptide sequences.
  • Previously described hydrophobic CPPs were derived from the hydrophobic regions present in the signal peptides of secreted and cell surface proteins.
  • the prior art consists first, of ad hoc use of H-region sequences (MTS/MTM), and second, of H-region sequences (with and without modification) with highest CPP activity selected from a screen of 1,500 signal sequences (MTM).
  • MTS/MTM H-region sequences
  • MTM 1,500 signal sequences
  • the modified H-region derived hydrophobic CPP sequences had advanced in diversity with multiple number of available sequences apart from MTS/MTM derived from fibroblast growth factor (FGF) 4.
  • FGF fibroblast growth factor
  • the hydrophobic CPPs like the signal peptides from which they originated, did not conform to a consensus sequence, and they had adverse effects on protein solubility when incorporated into protein cargo.
  • CFs critical factors
  • CPPs critical factors
  • aMTDs advanced macromolecule transduction domains
  • cell-permeability could be predicted by their character analysis before conducting any in vitro and/or in vivo experiments.
  • Table 1 shows the summary of published hydrophobic Cell-Penetrating Peptides which were chosen.
  • Table 3 shows characteristics of published hydrophobic Cell-Penetrating Peptides (A) which were analyzed.
  • Table 4 summarizes Critical Factors (CFs) of published hydrophobic Cell-Penetrating Peptides (A) which were analyzed.
  • Bending potential was determined based on the fact whether proline (P) exists and/or where the amino acid(s) providing bending potential to the peptide in recombinant protein is/are located.
  • Proline differs from the other common amino acids in that its side chain is bonded to the backbone nitrogen atom as well as the alpha-carbon atom. The resulting cyclic structure markedly influences protein architecture which is often found in the bends of folded peptide/protein chain.
  • Instability Index (II) Since one of the crucial structural features of any peptide is based on the fact whether the motif is rigid or flexible, which is an intact physicochemical characteristic of the peptide sequence, instability index (II) of the sequence was determined. The index value representing rigidity/flexibility of the peptide was extremely varied (8.9-79.1), but average value was 40.1 ⁇ 21.9 which suggested that the peptide should be somehow flexible, but not too much rigid or flexible (Table 3).
  • Alanine (V), valine (V), leucine (L) and isoleucine (I) contain aliphatic side chain and are hydrophobic—that is, they have an aversion to water and like to cluster. These amino acids having hydrophobicity and aliphatic residue enable them to pack together to form compact structure with few holes.
  • Analyzed peptide sequence showed that all composing amino acids were hydrophobic (A, V, L and I) except glycine (G) in only one out of 17 (MTD10—Table 3) and aliphatic (A, V, L, I, and P).
  • Their hydropathic index (Grand Average of Hydropathy: GRAVY) and aliphatic index (AI) were 2.5 ⁇ 0.4 and 217.9 ⁇ 43.6, respectively.
  • Their amino acid composition is also indicated in the Table 3.
  • the CPP sequences may be supposed to penetrate the plasma membrane directly after inserting into the membranes in a “bent” configuration with hydrophobic sequences having ⁇ -helical conformation.
  • our analysis strongly indicated that bending potential was crucial for membrane penetration. Therefore, structural analysis of the peptides was conducted to determine whether the sequences were to form helix or not.
  • Nine peptides were helix and eight were not (Table 3). It seems to suggest that helix structure may not be required.
  • Critical Factors for the development of new hydrophobic CPPs—advanced MTDs: amino acid length, bending potential (proline presence and location), rigidity/flexibility (instability index: II), structural feature (aliphatic index: AI), hydropathy (GRAVY) and amino acid composition/residue structure (hydrophobic and aliphatic A/a) (Table 3 and Table 4).
  • Table 5 shows characteristics of published hydrophobic Cell-Penetrating Peptides (B): selected CPPs that were used to each cargo in vivo.
  • Table 6 shows summarized Critical Factors of published hydrophobic Cell-Penetrating Peptides (B).
  • Hydrophobic CPPs which did not have a bending potential, rigid or too much flexible sequences (too much low or too much high Instability Index), or too low or too high hydrophobic CPPs were unselected, but secondary structure was not considered because helix structure of sequence was not required.
  • Table 7 shows characteristics of published hydrophobic Cell-Penetrating Peptides (C): selected CPPs that provided bending potential and higher flexibility.
  • Table 8 shows summarized Critical Factors of published hydrophobic Cell-Penetrating Peptides (C)
  • H-regions of signal sequence (HRSS)-derived CPPs do not have a common sequence, sequence motif, and/or common-structural homologous feature.
  • the aim is to develop improved hydrophobic CPPs formatted in the common sequence- and structural-motif which satisfy newly determined ‘Critical Factors’ to have ‘Common Function,’ namely, to facilitate protein translocation across the membrane with similar mechanism to the analyzed reference CPPs.
  • the common homologous features have been analyzed to determine the critical factors that influence the cell-permeability.
  • the range value of each critical factor has been determined to include the analyzed index of each critical factor from analysis A, B and C to design novel aMTDs (Table 9).
  • Table 9 shows comparison the range/feature of each Critical Factor between the value of analyzed CPPs and the value determined for new design of novel aMTDs sequences
  • Amino Acid Composition Hydrophobic and Aliphatic amino acids—A, V, L, I and P
  • aMTDs advanced macromolecule transduction domains
  • aMTD sequences have been newly designed, numbered from 1 to 240, as shown in Table 10-15.
  • sequence ID Number is a sequence listings for reference, and aMTD numbers refer to amino acid listing numbers that actually have been used at the experiments. For further experiments, aMTD numbers have been used.
  • polynucleotide sequences shown in the sequence lists have been numbered from SEQ ID NO: 241 to SEQ ID NO: 480.
  • Tables 10 to 15 shows 240 new hydrophobic aMTD sequences that were developed to satisfy all critical factors.
  • rPs rPeptides
  • Table 16 shows the peptides that do not have any proline in the middle (at 5′, 6′, 7′ or 8′) and at the end of the sequences. In addition, Table 16 describes the peptides that do not have proline in the middle of the sequences. All these peptides are supposed to have no-bending potential.
  • New hydrophobic CPPs—aMTDs are consisted with only hydrophobic and aliphatic amino acids (A, V, L, I and P) with average ranges of the indexes—AI: 180-220 and GRAVY: 2.1-2.6 (Table 9).
  • the peptides which contain an aromatic residue (W, F or Y) are shown in Table 19 and the peptides which are hydrophobic with non-aromatic sequences but have amino acids residue other than A, V, L, I, P or additional proline residues are designed (Table 20).
  • hydrophilic and/or bending peptides which are consisted with non-aliphatic amino acids are shown in Table 21.
  • aMTD hydrophobic, flexible, bending, aliphatic and 12-A/a length peptides
  • Designed rPeptides that do not satisfy at least one of the critical factors are 141 that no bending peptide sequences are 26; rigid peptide (II ⁇ 40) sequences are 23; too much flexible peptides are 24; aromatic peptides (aromatic ring presences) are 27; hydrophobic, but non-aromatic peptides are 23; and hydrophilic, but non-aliphatic peptides are 18.
  • aMTD-fused cargo materials would be biologically active molecules that could be one of the following: enzymes, transcription factors, toxic, antigenic peptides, antibodies and antibody fragments.
  • biologically active molecules could be one of these following macromolecules: enzymes, hormones, carriers, immunoglobulin, membrane-bound proteins, transmembrane proteins, internal proteins, external proteins, secreted proteins, virus proteins, native proteins, glycoproteins, fragmented proteins, disulfide bonded proteins, recombinant proteins, chemically modified proteins and prions.
  • these biologically active molecules could be one of the following: nucleic acid, coding nucleic acid sequence, mRNAs, antisense RNA molecule, carbohydrate, lipid and glycolipid.
  • the recombinant proteins were forcedly expressed in E. coli BL21 (DE3) cells grown to an OD 600 of 0.6 and induced for 2 hours with 0.7 mM isopropyl- ⁇ -D-thiogalactopyranoside (IPTG).
  • the proteins were purified by Ni 2+ affinity chromatography as directed by the supplier (Qiagen, Hilden, Germany) in natural condition. After the purification, purified proteins were dissolved in a physiological buffer such as DMEM medium.
  • 316 aMTD sequences have been designed.
  • 141 rPeptides are also developed that lack one of these critical factors: no bending peptides: i) absence of proline both in the middle and at the end of sequence or ii) absence of proline either in the middle or at the end of sequence, rigid peptides, too much flexible peptides, aromatic peptides (aromatic ring presence), hydrophobic but non-aromatic peptides, and hydrophilic but non-aliphatic peptides (Table 22).
  • Table 23 shows Comparison Analysis of Cell-Permeability of aMTDs with a Negative Control (A: rP38).
  • Relative cell-permeability (relative fold) of aMTDs to the reference CPPs [B: MTM12 (AAVLLPVLLAAP), C: MTD85 (AVALLILAV)] was also analyzed (Tables 40 and 41)
  • Table 25 shows Comparison Analysis of Cell-Permeability of aMTDs with a Reference CPP (C: MTD85).
  • Geometric means of negative control histidine-tagged rP38-fused CRA recombinant protein subtracted by that of naked protein (histidine-tagged CRA protein) lacking any peptide (rP38 or aMTD) was standardized as relative fold of 1.
  • Relative cell-permeability of 240 aMTDs to the negative control was significantly increased by up to 164 fold, with average increase of 19.6 ⁇ 1.6 (Table 26-31).
  • novel 240 aMTDs averaged of 13 ⁇ 1.1 (maximum 109.9) and 6.6 ⁇ 0.5 (maximum 55.5) fold higher cell-permeability, respectively (Tables 26-31).
  • cell-permeability of 31 rPeptides has been compared with that of 240 aMTDs (0.3 ⁇ 0.04; Tables 32 and 33).
  • relative cell-permeability of aMTDs has shown maximum of 164.0, 109.9 and 55.5 fold higher to rP38, MTM12 and MTD85, respectively.
  • rP38 negative control
  • Relative cell-permeability of negative control (rP38) to the 240 aMTDs is only 0.3 ⁇ 0.04 fold.
  • Recombinant proteins fused to the aMTDs were tested to determine their intracellular delivery and localization by laser scanning confocal microscopy with a negative control (rP38) and previous published CPPs (MTM12 and MTD85) as the positive control references.
  • NIH3T3 cells were exposed to 10 ⁇ M of FITC-labeled protein for 1 hour at 37, and nuclei were counterstained with DAPI. Then, cells were examined by confocal laser scanning microscopy ( FIG. 7 ). Recombinant proteins fused to aMTDs clearly display intracellular delivery and cytoplasmic localization ( FIG. 7 ) that are typically higher than the reference CPPs (MTM12 and MTD85).
  • rP38-fused recombinant protein did not show internalized fluorescence signal ( FIG. 7 a ).
  • rPeptides his-tagged CRA recombinant proteins fused to each rPeptide display lower- or non-cell-permeability.
  • Histidine-tagged aMTD-fused cargo recombinant proteins have been greatly enhanced in their solubility and yield.
  • FITC-conjugated recombinant proteins have also been tested to quantitate and visualize intracellular localization of the proteins and demonstrated higher cell-permeability compared to the reference CPPs.
  • aMTD-fused recombinant proteins have promoted the ability of protein transduction into the cells compared to the recombinant proteins containing rPeptides and/or reference hydrophobic CPPs (MTM12 and MTD85). According to the results, it has been demonstrated that critical factors of cell-penetrating peptide sequences play a major role to determine peptide-mediated intracellular delivery by penetrating plasma membrane. In addition, cell-permeability can considerably be improved by following the rational that all satisfy the critical factors.
  • SAR structure/sequence activity relationship
  • aMTDs with its proline at 7′ or 8′ amino acid in their sequences have much higher cell-permeability compared to the sequences in which their proline position is at 5′ or 6′ ( FIGS. 14 a and 15 a ).
  • rPeptides have shown similar SAR correlations in the cell-permeability, pertaining to their proline position (PP) and hydropathy (GRAVY). These results confirms that rPeptides with high GRAVY (2.4 ⁇ 2.6) have better cell-permeability ( FIG. 16 ).
  • each aMTD-fused recombinant protein has equally two proline sequences in the composition.
  • Alanine In the composition of amino acids, the result does not show a significant difference by the number of alanine in terms of the aMTD's delivery potential because all of the aMTDs have three to five alanines. In the sequences, however, four alanine compositions show the most effective delivery potential (geometric mean) ( FIG. 13 a ).
  • Leucine and Isoleucine show inverse relationship between the number of amino acid (I and L) and delivery potential of aMTDs. Lower number of isoleucine and leucine in the sequences tends to have higher delivery potential (geometric mean) ( FIGS. 13 a and 13 b ).
  • Valine Conversely, the composition of valine of aMTD sequences shows positive correlation with their cell-permeability. When the number of valine in the sequence is low, the delivery potential of aMTD is also relatively low ( FIG. 13 b ).
  • the range and feature of five out of six critical factors have been empirically and experimentally determined that are also included in the index range and feature of the critical factors initially proposed before conducting the experiments and SAR analysis.
  • index range and feature of critical factors of newly developed 240 aMTDs the bending potential (proline position: PP), rigidity/flexibility (Instability Index: II), structural feature (Aliphatic Index: AI), hydropathy (GRAVY), amino acid length and composition are all within the characteristics of the critical factors derived from analysis of reference hydrophobic CPPs.
  • the expression vectors were designed for histidine-tagged CRA proteins fused with aMTDs or rPeptides.
  • PCR polymerase chain reaction
  • plasmids were mixed with competent cells of E. coli DH5-alpha strain on the ice for 10 minutes. This mixture was placed on the ice for 2 minutes after it was heat shocked in the water bath at 42° C. for 90 seconds. Then, the mixture added with LB broth media was recovered in 37° C. shaking incubator for 1 hour. Transformant was plated on LB broth agar plate with kanamycin (50 ⁇ g/mL) (Biopure, Johnson City, Tenn., USA) before incubating at 37° C. overnight.
  • kanamycin 50 ⁇ g/mL
  • PCR primers for the CRA recombinant proteins fused to aMTD and random peptides are summarized in Tables 23 to 30. Amino acid sequences of aMTD and rPeptide primers are shown in Tables 31 to 38.
  • pET-28a(+) vectors for the expression of CRA proteins fused to a negative control [rPeptide 38 (rP38)], reference hydrophobic CPPs (MTM 12 and MTD 85 ) and aMTDs were transformed in E. coli BL21 (DE3) strains.
  • Induced recombinant proteins were loaded on 15% SDS-PAGE gel and stained with Coomassie Brilliant Blue (InstantBlue, Expedeon, Novexin, UK) ( FIG. 3 ).
  • the E. coli cultures were harvested by centrifugation at 5,000 ⁇ rpm for 10 minutes, and the supernatant was discarded. The pellet was re-suspended in the lysis buffer (50 mM NaH 2 PO 4 , 10 mM Imidazol, 300 mM NaCl, pH 8.0). The cell lysates were sonicated on ice using a sonicator (Sonics and Materials, Inc., Newtown, Conn., USA) equipped with a probe.
  • a sonicator Sonics and Materials, Inc., Newtown, Conn., USA
  • Recombinant proteins purified under natural condition were analyzed on 15% SDS-PAGE gel and stained with Coomassie Brilliant Blue ( FIG. 4 ). All of the recombinant proteins were dialyzed for 8 hours and overnight against physiological buffer, a 1:1 mixture of cell culture medium (Dulbecco's Modified Eagle's Medium: DMEM, Hyclone, Logan, Utah, USA) and Dulbecco's phosphate buffered saline (DPBS, Gibco, Grand Island, N.Y., USA). From 316 aMTDs and 141 rPeptides cloned, 240 aMTD- and 31 rPeptide-fused recombinant proteins were induced, purified, prepared and analyzed for their cell-permeability.
  • DMEM Dulbecco's Modified Eagle's Medium
  • DPBS Dulbecco's phosphate buffered saline
  • FITC fluorescein isothiocyanate
  • RAW 264.7 cells were treated with 10 ⁇ M FITC-labeled recombinant proteins for 1 hour at 37° C., washed three times with cold PBS, treated with 0.25% tripsin/EDTA (Sigma-Aldrich, St. Louis, Mo.) for 20 minutes at 37° C. to remove cell-surface bound proteins.
  • NIH3T3 cells were cultured for 24 hours on coverslip in 24-wells chamber slides, treated with 10 ⁇ M FITC-conjugated recombinant proteins for 1 hour at 37° C., and washed three times with cold PBS.
  • Treated cells were fixed in 4% paraformaldehyde (PFA, Junsei, Tokyo, Japan) for 10 minutes at room temperature, washed three times with PBS, and mounted with VECTASHIELD Mounting Medium (Vector laboratories, Burlingame, Calif., USA), and counter stained with DAPI (4′,6-diamidino-2-phenylindole).
  • PFA paraformaldehyde
  • VECTASHIELD Mounting Medium Vector laboratories, Burlingame, Calif., USA
  • DAPI 4,6-diamidino-2-phenylindole
  • Full-length cDNA for human SOCS3 (SEQ ID NO: 815) was purchased from Origene (USA). Histidine-tagged human SOCS3 proteins were constructed by amplifying the SOCS3 cDNA (225 amino acids) using primers for aMTD fused to SOCS3 cargo.
  • the PCR reactions 100 ng genomic DNA, 10 pmol each primer, each 0.2 mM dNTP mixture, 1 ⁇ reaction buffer and 2.5 U Pfu(+) DNA polymerase (Doctor protein, Korea)) were digested on the restriction enzyme site between Nde I (5′) and Sal I (3′) involving 35 cycles of denaturing (95° C.), annealing (62° C.), and extending (72° C.) for 45 sec each.
  • PCR reactions remained for 10 min at 72° C.
  • the PCR products were subcloned into 6 ⁇ His expression vector, pET-28a(+) (Novagen, Darmstadt, Germany). Coding sequence for SDA or SDB fused to C terminus of his-tagged aMTD-SOCS3 was cloned at BamHI (5′) and SalI (3′) in pET-28a(+) from PCR-amplified DNA segments and confirmed by DNA sequence analysis of the resulting plasmids.
  • Each of the recombinant expression vectors, HS3, HMS3, HMS3A, HMS3B, HMS3C, HMS3D, and HMS3E prepared in example 6-1 was transformed into E. coli BL21 CodonPlus(DE3)-RIL by a heat shock method, and then 600 ul of each was incubated in an LB medium (Biopure, Johnson City, Tenn., USA) containing 50 ⁇ g/ml of kanamycin at 37° C. for 1 hour. Thereafter, the recombinant protein gene-introduced E. coli was inoculated in 7 ml of LB medium, and then incubated at 37° C. overnight. The E.
  • coli was inoculated in 700 ml of LB medium and incubated at 37° C. until OD 600 reached 0.6.
  • 0.6 mM of isopropyl- ⁇ -D-thiogalactoside IPTG, Gen Depot, USA
  • IPTG isopropyl- ⁇ -D-thiogalactoside
  • the cell pellet thus recovered was suspended in a lysis buffer (100 mM NaH 2 PO 4 , 10 mM Tris-HCl, 8 M Urea, pH 8.0), and cells were disrupted by sonication (on/off time: 30 sec/30 sec, on time 2 hours, amplify 40%), and centrifuged at 15,000 rpm for 30 min to obtain a soluble fraction and an insoluble fraction.
  • a lysis buffer 100 mM NaH 2 PO 4 , 10 mM Tris-HCl, 8 M Urea, pH 8.0
  • This insoluble fraction was suspended in a denature lysis buffer (8 M Urea, 10 mM Tris, 100 mM Sodium phosphate) and purified by Ni 2+ affinity chromatography as directed by the supplier(Qiagen, Hilden, Germany) and refolded by dialyzing with a refolding buffer (0.55 M guanidine HCl, 0.44 M L-arginine, 50 mM Tris-HCl, 150 mM NaCl, 1 mM EDTA, 100 mM NDSB, 2 mM reduced glutathione, and 0.2 mM oxidized glutathione).
  • the proteins were put in a SnakeSkin Dialysis Tubing bag (pore size: 10000 mw, Thermo scientific, USA) and then they were dialyzed by physiological buffer (DMEM). The strain lysate where protein expression was not induced, the strain lysate where protein expression was induced by addition of IPTG, and purified proteins were loaded on SDS-PAGE to analyze protein expression characteristics and expression levels ( FIGS. 19 and 20 ).
  • SOCS3 recombinant proteins showed high expression levels in the BL21CodonPlus(DE3)-RIL strain.
  • SOCS3 recombinant proteins containing aMTD 165 and solubilization domain had little tendency to precipitate whereas recombinant SOCS3 proteins lacking a solubilization domain (HM 165 S3) or lacking an aMTD and a SD (HS3) were largely insoluble. Solubility of aMTD/SD-fused SOCS3 proteins was scored on a 5 point scale compared with that of SOCS3 proteins lacking the solubilization domain.
  • Example 6-3 Determination of Solubility/Yield of aMTD/SD-Fused SOCS3 Recombinant Proteins According to SD Type
  • solubilization domains were replaced in the same manner as in Example 6-2 to prepare 5 kinds of aMTD/SD-fused SOCS3 recombinant proteins, and their solubility/yield were measured ( FIG. 19 ).
  • Example 6-4 Comparison Between aMTD/SD-Fused SOCS3 Recombinant Protein and Basic CPP/SD-Fused SOCS3 Recombinant Protein
  • SOCS3 recombinant proteins were conjugated to 5/6-fluorescein isothiocyanate (FITC).
  • FITC 5/6-fluorescein isothiocyanate
  • FIG. 20 RAW 264.7 (KCLB, Seoul, South Korea) ( FIG. 20 ) or NIH3T3 cells (KCLB, Seoul, South Korea) ( FIG. 21 ) were treated with 10 ⁇ M FITC-labeled SOCS3 recombinant proteins and cultivated for 1 hr at 37° C.
  • NIH3T3 cells were washed three times with ice-cold PBS (Phosphate-buffered saline, Hyclone, USA) and treated with proteinase K (10 ⁇ g/mL, SIGMA, USA) to remove surface-bound proteins, and internalized proteins were measured by flow cytometry (FlowJo cytometric analysis software, Guava, Millipore, Darmstadt, Germany). Untreated cells (gray) and equimolar concentration of unconjugated FITC (FITC only, green)-treated cells were served as control ( FIG. 20 ). Each of NIH3T3 cells was incubated for 1 hour at 37° C. with 10 ⁇ M FITC-labeled SOCS3 protein.
  • VECTASHIELD Mounting Medium Vector Laboratories, Burlingame, Calif.
  • DAPI 4,6-diamidino-2-phenylindole
  • SOCS3 recombinant proteins containing aMTD 165 (HM 165 S3, HM 165 S3A and HM 165 S3B) efficiently entered the cells ( FIGS. 20 and 21 ) and were localized to various extents in cytoplasm ( FIG. 21 ).
  • SOCS3 protein containing non-aMTD (HS3) did not appear to enter cells. While all SOCS3 proteins containing aMTD 165 transduced into the cells, HM 165 S3B displayed more uniform cellular distribution, and protein uptake of HM 165 S3B was also very efficient.
  • the cell-permeability of basic CPP/SD-fused SOCS3 recombinant proteins was assessed by the same method as used in Example 7-1 except for a known basic CPP (TAT or PolyR) being used instead of aMTD. The results of the assessment were shown in FIG. 99 .
  • ICR mice Doo-Yeol Biotech Co. Ltd., Seoul, Korea
  • IP intraperitoneal
  • FITC Fluorescein isothiocyanate, SIGMA, USA
  • SIGMA Fluorescein isothiocyanate
  • the liver, kidney, spleen, lung, heart, and brain were removed and washed with PBS, and then placed on a dry ice, and embedded with an O.C.T. compound (Sakura). After cryosectioning at 20 ⁇ m, tissue distributions of fluorescence-labeled-SOCS3 proteins in different organs was analyzed by fluorescence microscopy (Carl Zeiss, Gottingen, Germany)( FIG. 22 ).
  • SOCS3 recombinant proteins fused to aMTD 165 were distributed to a variety of tissues (liver, kidney, spleen, lung, heart and, to a lesser extent, brain). Liver showed highest levels of fluorescent cell-permeable SOCS3 since intraperitoneal administration favors the delivery of proteins to this organ via the portal circulation.
  • SOCS3 containing aMTD 165 was detectable to a lesser degree in lung, spleen and heart.
  • Example 7-2-2 Comparison Between aMTD/SD-Fused SOCS3 Recombinant Protein and Basic CPP/SD-Fused SOCS3 Recombinant Protein
  • tissue-permeability of basic CPP/SD-fused SOCS3 recombinant proteins was assessed by the same method as used in Example 7-2 except for a known basic CPP (TAT or PolyR) being used instead of aMTD. The results of the assessment were shown in FIG. 100 .
  • Example 8-1 Biological Activity Test of iCP-SOCS3—Inhibition Activity of IFN- ⁇ -Induced STAT Phosphorylation
  • PANC-1 Cells (KCLB, Seoul, South Korea) were treated with 10 ng/ml IFN- ⁇ (R&D systems, Abingdon, UK) for 15 min and treated with either DMEM (vehicle) or 10 ⁇ M aMTD/SD-fused SOCS3 recombinant proteins for 2 hrs.
  • the cells were lysed in RIPA lysis buffer (Biosesang, Seongnam, Korea) containing proteinase inhibitor cocktail (Roche, Indianapolis, Ind., USA), incubated for 15 min at 4° C., and centrifuged at 13,000 rpm for 10 min at 4° C. Equal amounts of lysates were separated on 10% SDS-PAGE gels and transferred to a nitrocellulose membrane.
  • the well-known cytokine signaling inhibitory actions of SOCS3 are inflammation inhibition through i) inhibition of IFN- ⁇ -mediated JAK/STAT and ii) inhibition of LPS-mediated cytokine secretion.
  • the ultimate test of cell-penetrating efficiency is a determination of intracellular activity of SOCS3 proteins transported by aMTD. Since endogenous SOCS3 are known to block phosphorylation of STAT1 and STAT3 by IFN- ⁇ -mediated Janus kinases (JAK) 1 and 2 activation, we demonstrated whether cell-permeable SOCS3 inhibits the phosphorylation of STATs. As shown in FIG.
  • Example 8-2 Biological Activity Test of iCP-SOCS3
  • SOCS3 recombinant protein containing aMTD and SDB (HM 165 S3B) is a prototype of a new generation of improved cell-permeable SOCS3 (iCP-SOCS3), and will be selected for further evaluation as a potential anti-tumor agent.
  • SOCS3 recombinant proteins lacking SD (HM 165 S3) or both aMTD and SD (HS3) were found to be less soluble, produced lower yields, and showed tendency to precipitate when they were expressed and purified in E. coli . Therefore, we additionally designed and constructed SOCS3 recombinant protein containing only SDB (without aMTD 165 : HS3B) as a negative control ( FIG. 25 ). Preparation, expression and purification, and measurement of solubility/yield of the recombinant proteins were performed in the same manner as in Examples 6-2 and 6-3.
  • aMTDs were selected for development of iCP-SOCS3 recombinant protein (Tables 38 and 39), based on the critical Factors, in order to examine which aMTD provides the highest cell-/tissue-permeability.
  • Example 11-1 Investigation of Biological Activity for Determination of Optimal aMTD-Fused SOCS3 Recombinant Protein—1
  • PANC-1 cells pancreatic carcinoma cell line
  • aMTD/SD-fused SOCS3 serum-free medium
  • Apoptotic cells were analyzed using terminal dUTP nick-end labeling (TUNEL) assay with In Situ Cell Death Detection kit TMR red (Roche, 4056 Basel, Switzerland).
  • TUNEL terminal dUTP nick-end labeling
  • TMR red In Situ Cell Death Detection kit
  • AGS cells gastric carcinoma cell line
  • ATCC American Type Culture Collection
  • 10 uM of aMTD/SD-fused SOCS3 for 14 hours.
  • Cancer cell death was analyzed by Annexin V analysis.
  • Annexin V/7-Aminoactinomycin D (7-AAD) staining was performed using flow cytometry according to the manufacturer's guidelines (BD Pharmingen, San Diego, Calif., USA). Briefly, cells were washed three times with ice-cold PBS.
  • the cells were then resuspended in 100 ⁇ l of binding buffer and incubated with 1 ⁇ l of 7-AAD and 1 ⁇ l of annexin V-PE for 30 min in the dark at 37° C.
  • Flow cytometric analysis was immediately performed using a guava easyCyteTM 8 Instrument (Merck Millipore, Darmstadt, Germany).
  • AGS cells gastric cancer cell line
  • Confluent AGS cells were incubated with 10 ⁇ M HM#S3B in serum-free medium for 2 hrs prior to changing the growth medium (DMEM/F12, Hyclone, Logan, Utah, USA) and washed twice with PBS, and the monolayer at the center of the well was “wounded” by scraping with a sterilized white pipette tip.
  • Cells were cultured for an additional 24 hrs and cell migration was observed by phase contrast microscopy (Nikon, ECLIPSE Ts2). The migration was quantified by counting the number of cells that migrated from the wound edge into the clear area.
  • Histidine-tag free human SOCS3 proteins were constructed by amplifying the SOCS3 cDNA (225 amino acids) for aMTD fused to SOCS3 cargo.
  • the PCR reactions (100 ng genomic DNA, 10 pmol each primer, each 0.2 mM dNTP mixture, 1 ⁇ reaction buffer and 2.5 U Pfu(+) DNA polymerase (Doctor protein, Korea)) were digested on the restriction enzyme site between Nde I (5′) and Sal I (3′) involving 35 cycles of denaturing (95° C.), annealing (62° C.), and extending (72° C.) for 45 sec each. For the last extension cycle, the PCR reactions remained for 10 min at 72° C.
  • PCR products were subcloned into pET-26b(+) (Novagen, Darmstadt, Germany). Coding sequence for SDB fused to C terminus of aMTD-SOCS3 was cloned at BamHI (5′) and SalI (3′) in pET-26b(+) from PCR-amplified DNA segments and confirmed by DNA sequence analysis of the resulting plasmids.
  • Example 7-1 RAW264.7 cells were treated with FITC-labeled HS3B, HM 165 S3B, and M 165 S3B proteins, and cell permeability was evaluated.
  • both HM 165 S3B and M 165 S3B were found to have high cell permeability.
  • iCP-SOCS3 was prepared in the same manner as in Example 6-1, except that SDB′ having a substitution of valine with leucine at an amino acid position 28 was used ( FIG. 94 ). Further, protein purification was performed in the same manner as in Example 6-2 using the primer as below (Table 43).
  • both HM 165 S3B and HM 165 S3B′(V28L) were found to have high solubility/yield.
  • both HM 165 S3B and HM 165 S3B′(V28L) were found to have high cell permeability.
  • HM 165 S3B and HM 165 S3B′(V28L) recombinant proteins anti-proliferative activity was examined and induction of apoptosis of gastric carcinoma cell line (AGS) was analyzed by Annexin V staining in the same manner as in Example 11-2, and inhibition of migration was analyzed in the same manner as in Example 11-3.
  • AGS gastric carcinoma cell line
  • Antiproliferative activity were evaluated with the CellTiter-Glo Cell Viability Assay.
  • AGS cells (3 ⁇ 10 3 /well) were seeded in 96 well plates. The next day, cells were treated with DMEM (vehicle) or 10 ⁇ M HM 165 S3B, HM 165 S3B′(V28L) for 96 hrs in the presence of serum (2%). Proteins were replaced daily. Cell growth and survival were evaluated with the CellTiter-Glo Cell Viability Assay (Promega, Madison, Wis.). Measurements using a Luminometer (Turner Designs, Sunnyvale, Calif.) were conducted following the manufacturer's protocol.
  • HM 165 S3B and HM 165 S3B′(V28L) showed high anti-proliferative effects on gastric carcinoma cells ( FIG. 46 ), and also effects of inducing apoptosis ( FIG. 47 ) and of inhibiting migration of gastric carcinoma cells ( FIG. 48 ), and in particular, HM 165 S3B′(V28L) exhibited efficacy equivalent to or higher than HM 165 S3B.
  • iCP-SOCS3 of BS3M 165 structure was prepared in the same manner as in Example 6-1, and B′S3M 165 iCP-SOCS3 was also prepared by humanized SDB domain ( FIG. 49 a ).
  • iCP-SOCS3 recombinant protein (BS3M 165 , B′S3M 165 ) in E. coli (bottom) were analyzed in the same manner as in Examples 6-2 and 6-3, respectively, and shown in FIG. 49 b . Further, E. coli codon-optimized iCP-SOCS3 was prepared.
  • iCP-SOCS3 (HM 165 S3B) recombinant protein inhibits activation of the JAK/STAT-signaling pathway was examined by the method of Example 8-1.
  • PANC-1 Cells (KCLB, Seoul, South Korea) were treated with 10 ng/ml IFN- ⁇ (R&D systems, Abingdon, UK) for 15 min and treated with either DMEM (vehicle) or 1, 5, 10 ⁇ M aMTD/SD-fused SOCS3 recombinant proteins for 2 hrs.
  • the cells were lysed in RIPA lysis buffer (Biosesang, Seongnam, Korea) containing proteinase inhibitor cocktail (Roche, Indianapolis, Ind., USA), incubated for 15 min at 4° C., and centrifuged at 13,000 rpm for 10 min at 4° C. Equal amounts of lysates were separated on 10% SDS-PAGE gels and transferred to a nitrocellulose membrane.
  • the membranes were blocked using 5% skim milk in TBST and for western blot analysis incubated with the following antibodies: anti-phospho-STAT3 (Cell Signaling Technology, USA), then HRP conjugated anti-rabbit secondary antibody (Santacruz).
  • the well-known cytokine signaling inhibitory actions of SOCS3 are inflammation inhibition through i) inhibition of IFN- ⁇ -mediated JAK/STAT and ii) inhibition of LPS-mediated cytokine secretion.
  • the ultimate test of cell-penetrating efficiency is a determination of intracellular activity of SOCS3 proteins transported by aMTD. Since endogenous SOCS3 are known to block phosphorylation of STAT3 by IFN- ⁇ -mediated Janus kinases (JAK) 1 and 2 activation, we demonstrated whether cell-permeable SOCS3 inhibits the phosphorylation of STATs. As shown in FIG.
  • iCP-SOCS3 (HM 165 S3B) suppressed IFN- ⁇ -induced phosphorylation of STAT3 in dose dependent manner.
  • STAT phosphorylation was readily detected in cells exposed to HS3B, which lacks the aMTD motif required for membrane penetration, indicating that HS3B, which lacks an MTD sequence did not enter the cells, has no biological activity.
  • the mechanism of aMTD 165 -mediated intracellular delivery was investigated.
  • RAW 264.7 cells were pretreated with 100 mM EDTA for 3 hours, and then treated with 10 ⁇ M of iCP-SOCS3 (HM 165 S3B) recombinant protein for 1 hour, followed by flow cytometry in the same manner as in Example 7-1 ( FIG. 51A ).
  • RAW 264.7 cells were pretreated with 5 ⁇ g/ml of proteinase K for 10 minutes, and then treated with 10 ⁇ M of iCP-SOCS3 (HM 165 S3B) recombinant protein for 1 hour, followed by flow cytometry ( FIG. 51B ).
  • RAW 264.7 cells were pretreated with 20 ⁇ M taxol for 30 minutes, and then treated with 10 ⁇ M of iCP-SOCS3 (HM 165 S3B) recombinant protein for 1 hour, followed by flow cytometry ( FIG. 52A ).
  • RAW 264.7 cells were pretreated with 1 mM ATP and 10 ⁇ M antimycin singly or in combination for 2 hours, and then treated with 10 ⁇ M of iCP-SOCS3 (HM 165 S3B) recombinant protein for 1 hour, followed by flow cytometry ( FIG. 52B ).
  • iCP-SOCS3 HM 165 S3B
  • RAW 264.7 cells were left at 4° C. and 37° C. for 1 hour, respectively, and then treated with 10 ⁇ M of iCP-SOCS3 (HM 165 S3B) recombinant protein for 1 hour, followed by flow cytometry ( FIG. 53 ).
  • iCP-SOCS3 HM 165 S3B
  • the aMTD-mediated intracellular delivery of SOCS3 protein did not require protease-sensitive protein domains displayed on the cell surface ( FIG. 51B ), microtubule function ( FIG. 52A ), or ATP utilization ( FIG. 52B ), since aMTD 165 -dependent uptake, compare to HS3 and HS3B, was essentially unaffected by treating cells with proteinase K, taxol, or the ATP depleting agent, antimycin.
  • iCP-SOCS3 (HM 165 S3B) proteins uptake was blocked by treatment with EDTA and low temperature ( FIGS. 51A and 53 ), indicating the importance of membrane integrity and fluidity for aMTD-mediated protein transduction.
  • RAW 264.7 cells were treated with 10 ⁇ M of FITC-labeled iCP-SOCS3 (HM 165 S3B) recombinant protein for 1 hour. Thereafter, these cells were co-cultured with PerCP-Cy5.5-CD14-stained RAW 264.7 cells for 2 hours. Cell-to-cell protein transfer was assessed by flow cytometry, scoring for CD14/FITC double-positive cells. Efficient cell-to-cell transfer of HM 165 S3B, but not HS3 or HS3B ( FIG. 54 ), suggests that SOCS3 recombinant proteins containing aMTD 165 are capable of bidirectional passage across the plasma membrane.
  • FIG. 101 it was confirmed that aMTD165/SD-fused SOCS3 recombinant proteins are independent to cell surface receptor ( FIG. 101A ) and the cell-permeability of aMTD165/SD-fused SOCS3 recombinant proteins is not due to endocytosis ( FIG. 101B ).
  • RAW 264.7 cells were treated with 5 ⁇ M of FITC-labeled HM 165 S3B, HTS3B for 2 hour and washed with PBS two times. Thereafter, they were seeded on PANC-1 cell, incubated for 2 hours and treated with 20 ng/ml of IFN- ⁇ for 15 minutes, followed by Western blotting in the same manner as in Example 8-1. And Cell-to-cell protein transfer was assessed by flow cytometry.
  • phospho-STAT3 was only reduced in cells treated with HM 165 S3B.
  • mice ICR mice (Doo-Yeol Biotech Co. Ltd., Seoul, Korea) were intravenous (IV) injected with 600 ⁇ g/head of 10 ⁇ M FITC (Fluorescein isothiocyanate, SIGMA, USA)-labeled SOCS3 recombinant proteins (HS3B, HM 165 S3B) and after 15 min, 30 min, 1H, 2H, 4H, 8H, 12H, 16H, 24H, 36H, 48H, mice of each group were sacrificed. From the mice, peripheral blood mononuclear cells (PBMCs), splenocytes, and hepatocytes were separated.
  • PBMCs peripheral blood mononuclear cells
  • the spleen was removed and washed with PBS, and then placed on a dry ice and embedded in an O.C.T. compound (Sakura). After cryosectioning at 20 ⁇ m, tissue distributions of fluorescence-labeled-SOCS3 proteins in different organs was analyzed by fluorescence microscopy (Carl Zeiss, Gottingen, Germany).
  • mice were laparotomized and the spleen or liver were removed.
  • the spleen or liver thus removed was separated into single cells using a Cell Strainer (SPL, Korea). These cells were collected in a microtube, followed by centrifugation at 4,000 rpm and 4° C. for 5 minutes.
  • 0.5 ml of RBC lysis buffer was added thereto, followed by vortexing.
  • the microtube was left at room temperature for 5 minutes, and then centrifuged at 4,000 rpm and 4° C. for 5 minutes.
  • 0.5 ml of PBS was added to a pellet, followed by pipetting and flow cytometry (FlowJo cytometric analysis software, Guava, Millipore, Darmstadt, Germany).
  • iCP-SOCS3 As shown in FIG. 55 , in PBMCs, the maximum permeability of iCP-SOCS3 was observed at 30 minutes, and in splenocytes, the maximum permeability of iCP-SOCS3 was observed at 2 hours and maintained up to 16 hours. In hepatocytes, the maximum permeability of iCP-SOCS3 was observed at 15 minutes and maintained up to 16 hours. As shown in FIG. 56 , in the pancreas tissue, very high distribution of iCP-SOCS3 was observed at 2 hours, and maintained up to 8 hours. Therefore, it can be seen that iCP-SOCS3 is rapidly delivered from blood to various tissues within 2 hours, and maintained up to 8-16 hours depending on the tissues.
  • iCP-SOCS3 recombinant protein As a therapeutic agent for solid tumors, its permeability to solid tumors (gastric cancer, colorectal cancer, breast cancer, glioblastoma cell line) and various tissues was investigated.
  • AGS gastric cancer cell line
  • HCT116 colonal cancer cell line
  • MDA-MB-231 breast cancer cell line
  • U-87 MG glioblastoma cell line
  • Cells were treated with 10 ⁇ M FITC-labeled iCP-SOCS3 recombinant proteins and cultivated for 1 hr at 37° C.
  • the cells were treated with proteinase K (10 ⁇ g/mL, SIGMA, USA) and washed three times with ice-cold PBS (Phosphate-buffered saline, Hyclone, USA) to remove surface-bound proteins, and internalized proteins were measured by flow cytometry (FlowJo cytometric analysis software, Guava, Millipore, Darmstadt, Germany). Untreated cells (gray) and equimolar concentration of unconjugated FITC (FITC only, green)-treated cells were served as control ( FIG. 57 ).
  • proteinase K 10 ⁇ g/mL, SIGMA, USA
  • PBS Phosphate-buffered saline, Hyclone, USA
  • iCP-SOCS3 recombinant proteins (FITC-HM 165 S3SB) enhanced the systemic delivery to various tissues after intraperitoneal injection ( FIG. 58 ). Therefore, these data indicate that iCP-SOCS3 protein could be intracellularly delivered and distributed to the various cancer cells and various tissues, contributing for beneficial biotherapeutic effects.
  • Example 17-1 Investigation of Anti-Cancer Efficacy of iCP-SOCS3 Recombinant Protein in Gastric Cancer Cells
  • Example 17-1-1 Analysis of Hypermethylation Level in Gastric Cancer Cells
  • SOCS3 expression is suppressed due to methylation in cancer cells, and therefore, inflammation or cancer development is increased. Further, to investigate the effect of cell permeable SOCS3, a cancer cell line where endogenous SOCS3 expression is suppressed should be selected or applied to a model, and therefore, the present experiments were performed.
  • Unmethyl-F was 5′-tag tgt gta agt tgt agg aga gtg g-3′(SEQ ID NO: 816), Unmethyl-R was 5′-cta aac ata aaa aaa taa cac taa tcc aaa-3′ (SEQ ID NO: 817), Methyl-F was 5′-gta gtg cgt aag ttg tag gag agc-3′ (SEQ ID NO: 818), Methyl-R was 5′-gta aaaaa taa cgc taa tcc gaa-3′ (SEQ ID NO: 819), PCR was performed for 30 cycles consisting of pre-denaturation at 95° C. for 5 minutes, denaturation at 95° C. for 30 seconds, annealing at 60° C. for 45 seconds, extension at 72° C. for 1 minute
  • SOCS3 mRNA expression levels in cancer cells were analyzed by RT-PCR. mRNAs were isolated from normal cell lines and cancer cell lines according to a method provided in a manufacturer's sheet of Hybrid-R (Geneall, Korea), and PCR was performed using SOCS3 primer F 5′-cct act gaa ccc tcc tcc ga-3′ (SEQ ID NO: 820) and SOCS3 primer R 5′-gca get ggg tga ctt tct ca-3′ (SEQ ID NO: 821) for 30 cycles consisting of denaturing (95° C.), annealing (60° C.), and extending (72° C.) for 45 seconds each.
  • Example 17-1-3 Analysis of Endogenous SOCS3 and JAK/STAT Signaling Activation Status in Gastric Cell Line
  • SOCS3 levels endogenously expressed and activation of JAK/STAT-signaling pathway were investigated in different colorectal cell lines, normal cells, and normal hepatic cells.
  • SOCS3 expression is suppressed due to methylation in cancer cells, and therefore, inflammation or cancer development is increased. Further, to investigate the effect of cell permeable SOCS3, a cancer cell line where endogenous SOCS3 expression is suppressed should be selected or applied to a model, and therefore, the present experiments were performed.
  • Unmethyl-F was 5′-tag tgt gta agt tgt agg aga gtg g-3′(SEQ ID NO: 816), Unmethyl-R was 5′-cta aac ata aaa aaa taa cac taa tcc aaa-3′ (SEQ ID NO: 817), Methyl-F was 5′-gta gtg cgt aag ttg tag gag agc-3′ (SEQ ID NO: 818), Methyl-R was 5′-gta aaaaa taa cgc taa tcc gaa-3′ (SEQ ID NO: 819), PCR was performed for 30 cycles consisting of pre-denaturation at 95° C. for 5 minutes, denaturation at 95° C. for 30 seconds, annealing at 60° C. for 45 seconds, extension at 72° C. for 1 minute
  • Example 17-2-2 Analysis of Expression Level of Endogenous SOCS3 mRNA in Colorectal Cancer Cell Line
  • SOCS3 mRNA expression levels in cancer cells were analyzed by RT-PCR. mRNAs were isolated from normal cell lines and cancer cell lines according to a method provided in a manufacturer's sheet of Hybrid-R (Geneall, Korea), and PCR was performed using SOCS3 primer F 5′-cct act gaa ccc tcc tcc ga-3′ (SEQ ID NO: 820) and SOCS3 primer R 5′-gca get ggg tga ctt tct ca-3′ (SEQ ID NO: 821) for 30 cycles consisting of denaturing (95° C.), annealing (60° C.), and extending (72° C.) for 45 seconds each.
  • Example 17-2-3 Analysis of Expression of Endogenous SOCS3 and JAK/STAT Signaling Activation Status in Colorectal Cancer Cell Line
  • JAK/STAT3 activation in cancer cells was analyzed by Western blot analysis.
  • the normal HEK293 cells, and colorectal cancer cell lines, HCT116, SW480, RKO, and HT29 were washed with PBS, and then the cells were lysed in RIPA lysis buffer (Biosesang, Seongnam, Korea) containing proteinase inhibitor cocktail (Roche, Indianapolis, Ind., USA), incubated for 15 min at 4° C., and centrifuged at 13,000 rpm for 10 min at 4° C. to isolate proteins, followed by western blotting in the same manner as in Example 8-1.
  • SOCS3 iCP-SOCS3 recombinant protein as a mechanism-specific therapeutic agent for solid tumors
  • SOCS3 levels endogenously expressed and activation of JAK/STAT-signaling pathway were investigated in different glioblastoma cells, normal cells, and normal hepatic cells.
  • Example 17-3-1 Analysis of Hypermethylation Level in Glioblastoma Cell Line
  • SOCS3 expression is suppressed due to methylation in cancer cells, and therefore, inflammation or cancer development is increased. Further, to investigate the effect of cell permeable SOCS3, a cancer cell line where endogenous SOCS3 expression is suppressed should be selected or applied to a model, and therefore, the present experiments were performed.
  • PCR was performed for 30 cycles consisting of pre-denaturation at 95° C. for 5 minutes, denaturation at 95° C. for 30 seconds, annealing at 60° C. for 45 seconds, and extension at 72° C. for 1 minute, and then final extension at 72° C. for 8 minutes.
  • Example 17-3-2 Analysis of Expression of Endogenous SOCS3 and JAK/STAT Signaling Activation Status in Glioblastoma Cells
  • SOCS3 levels endogenously expressed and activation of JAK/STAT-signaling pathway were investigated in different breast cancer cell lines, normal cells, and normal hepatic cells.
  • Example 17-4-1 Analysis of Expression Level of Endogenous SOCS3 mRNA in Breast Cancer Cell Line
  • SOCS3 mRNA expression levels in cancer cells were analyzed by RT-PCR. mRNAs were isolated from normal cell lines and cancer cell lines according to a method provided in a manufacturer's sheet of Hybrid-R (Geneall, Korea), and PCR was performed using SOCS3 primer F 5′-cct act gaa ccc tcc tcc ga-3′ (SEQ ID NO: 820) and SOCS3 primer R 5′-gca get ggg tga ctt tct ca-3′ (SEQ ID NO: 821) for 30 cycles consisting of denaturing (95° C.), annealing (60° C.), and extending (72° C.) for 45 seconds each.
  • Example 17-4-2 Analysis of Hypermethylation Level in Breast Cancer Cell Line
  • SOCS3 expression is suppressed due to methylation in cancer cells, and therefore, inflammation or cancer development is increased. Further, to investigate the effect of cell permeable SOCS3, a cancer cell line where endogenous SOCS3 expression is suppressed should be selected or applied to a model, and therefore, the present experiments were performed.
  • Unmethyl-F was 5′-tag tgt gta agt tgt agg aga gtg g-3′(SEQ ID NO: 816)
  • Unmethyl-R was 5′-cta aac ata aaa aaa taa cac taa tcc aaa-3′ (SEQ ID NO: 817)
  • Methyl-F was 5′-gta gtg cgt aag ttg tag gag agc-3′ (SEQ ID NO: 818)
  • Methyl-R was 5′-gta aaaaa taa cgc taa tcc gaa-3′ (SEQ ID NO: 819).
  • PCR was performed for 30 cycles consisting of pre-denaturation at 95° C. for 5 minutes, denaturation at 95° C. for 30 seconds, annealing at 60° C. for 45 seconds, and extension at 72° C. for 1 minute, and then final extension at 72° C. for 8 minutes.
  • JAK/STAT3 activation in cancer cells was analyzed by Western blot analysis.
  • the normal HEK293 cells, and breast cancer cell lines, MDA-MB-231, MCF7, SK-BR3, and T47D were washed with PBS, and then the cells were lysed in RIPA lysis buffer (Biosesang, Seongnam, Korea) containing protease inhibitor cocktail (Roche, Indianapolis, Ind., USA), incubated for 15 min at 4° C., and centrifuged at 13,000 rpm for 10 min at 4° C. to isolate proteins, followed by western blotting in the same manner as in Example 8-1.
  • RIPA lysis buffer Biosesang, Seongnam, Korea
  • protease inhibitor cocktail Roche, Indianapolis, Ind., USA
  • Cells originated from human gastric cancer cells (AGS, MKN45, MKN75, NCI-N87), colorectal cancer cells (HCT116), glioblastoma (U-87 MG), breast cancer cells (MDA-MB-231), and mouse fibroblast (NIH3T3) were purchased from ATCC (Manassas, Va., USA) and KCLB (Seoul, Korea). All the cells were maintained as recommended by the supplier. These cells (3 ⁇ 10 3 /well) were seeded in 96 well plates. The next day, cells were treated with DMEM (vehicle) or 10 ⁇ M recombinant SOCS3 proteins for 96 hrs in the presence of serum (2%). Proteins were replaced daily. Cell growth and survival were evaluated with the CellTiter-Glo Cell Viability Assay (Promega, Madison, Wis.). Measurements using a Luminometer (Turner Designs, Sunnyvale, Calif.) were conducted following the manufacturer's protocol.
  • HM 165 S3B iCP-SOCS3 protein was the most cytotoxic to colorectal cancer cells—over 60% in 10 ⁇ M treatment (p ⁇ 0.01)—especially compared to vehicle alone (i.e. exposure of cells to culture media without recombinant proteins; left).
  • HM 165 S3B iCP-SOCS3 protein was the most cytotoxic to glioblastoma cells—over 80% in 10 ⁇ M treatment (p ⁇ 0.01)—especially compared to vehicle alone (i.e. exposure of cells to culture media without recombinant proteins; left).
  • HM 165 S3B iCP-SOCS3 protein was the most cytotoxic to breast cancer cells—over 40% in 10 ⁇ M treatment (p ⁇ 0.01)—especially compared to vehicle alone (i.e. exposure of cells to culture media without recombinant proteins; left).
  • Non-CP-SOCS3 HS3B
  • iCP-SOCS3 HM 165 S3B
  • serum-free medium serum-free medium
  • the cells were washed twice with PBS, and the monolayer at the center of the well was “wounded” by scraping with a pipette tip.
  • Cells were cultured for an additional 24-48 hrs and cell migration was observed by phase contrast microscopy (Nikon, ECLIPSE Ts2). The migration was quantified by counting the number of cells that migrated from the wound edge into the clear area.
  • iCP-SOCS3 (HM 165 S3B) suppressed the repopulation of wounded monolayer although SOCS3 protein lacking aMTD 165 (HS3B) had no effect on the cell migration, in AGS ( FIG. 72 ), HCT116 ( FIG. 73 ), U-87 MG ( FIG. 74 ), and MDA-MB-231 ( FIG. 75 ), respectly.
  • iCP-SOCS3 (HM 165 S3B) had no effect on the cell migration.
  • Transwell inserts (Costar) was coated with 0.1% gelatin, and the membranes were allowed to dry for 1 hr at room temperature.
  • the Transwell inserts were assembled into a 24-well plate, and the lower chamber was filled with growth media containing 10% FBS and FGF2 (40 ng/ml). Cells were added to each upper chamber at a density of 5 ⁇ 10 5 , and the plate was incubated at 37° C. in a 5% CO 2 incubator for 24 hrs. Migrated cells were stained with 0.6% hematoxylin and 0.5% eosin and counted.
  • AGS ( FIG. 76 ) and U-87 MG ( FIG. 77 ) cells treated with iCP-SOCS3 (HM 165 S3B) protein also showed significant inhibitory effect on their Transwell migration compared with untreated cells (Vehicle) and non-permeable SOCS3 protein-treated cells.
  • Transwell inserts The lower surface of Transwell inserts (Costar) was coated with 0.1% gelatin, the upper surface of Transwell inserts was coated with matrigel (40 ⁇ g per well; BD Pharmingen, San Diego, Calif., USA), and the membranes were allowed to dry for 1 hr at room temperature.
  • the Transwell inserts were assembled into a 24-well plate, and the lower chamber was filled with growth media containing 10% FBS and FGF2 (40 ng/ml). Cells (5 ⁇ 10 5 ) were added to each upper chamber, and the plate was incubated at 37° C. in a 5% CO 2 incubator for 24 hrs. Migrated cells were stained with 0.6% hematoxylin and 0.5% eosin and counted.
  • iCP-SOCS3 As shown in FIG. 76 (right), AGS treated with iCP-SOCS3 (HM 165 S3B) protein caused remarkable decrease in invasion compared with the control proteins. Taken together, these data indicate that iCP-SOCS3 contributes to inhibit tumorigenic activities of solid tumors.
  • iCP-SOCS3 regulates apoptosis in various cancer cells, such as gastric cancer cells (AGS and MKN45), colorectal cancer cells (HCT116), glioblastoma cells (U-87 MG), and breast cancer cells (MDA-MB-231).
  • Annexin V/7-Aminoactinomycin D (7-AAD) staining was performed using flow cytometry according to the manufacturer's guidelines (BD Pharmingen, San Diego, Calif., USA). Briefly, 1 ⁇ 10 6 cells were washed three times with ice-cold PBS. The cells were then resuspended in 100 ⁇ l of binding buffer and incubated with 1 ⁇ l of 7-AAD and 1 ⁇ l of Annexin V-7-AAD for 30 min in the dark at 4° C. Flow cytometric analysis was immediately performed using a guava easyCyteTM 8 Instrument (Merck Millipore, Darmstadt, Germany). In this regard, gemcitabine (0.1 ⁇ 10 ⁇ M) was used as a reference drug.
  • iCP-SOCS3 HM 165 S3B protein did not induce apoptosis in normal cell lines (NIH3T3 and HEK293).
  • 10 uM iCP-SOCS3 HM 165 S3B was efficient inducer of apoptosis in cancer cells, as assessed by Annexin V staining. Consistently, no changes in Annexin V staining were observed in cancer cells treated with HS3B compared to untreated cell (Vehicle).
  • Example 18-4 Investigation of Anti-Cancer Efficacy (Induction of Apoptosis) of iCP-SOCS3
  • iCP-SOCS3 (HM 165 S3B) protein was considerably efficient inducer of apoptosis in HCT116 cells ( FIG. 83 ), as assessed by a fluorescent terminal dUTP nick-end labeling (TUNEL) assay. Consistently, no changes in TUNEL was observed in HCT116 cells treated with HS3B compared to untreated cell (Vehicle).
  • iCP-SOCS3 regulates cell cycle progression in various cancer cells, such as gastric cancer cells (AGS), colorectal cancer cells (HCT116), glioblastoma cells (U-87 MG, and breast cancer cells (MDA-MB-231).
  • AGS gastric cancer cells
  • HCT116 colorectal cancer cells
  • glioblastoma cells U-87 MG
  • MDA-MB-2331 breast cancer cells
  • Cancer cells and normal cells were treated with 10 uM protein (Non-CP-SOCS3 (HS3B) and iCP-SOSC3 (HM 165 S3B)) for 8 hrs. After the treatment, cells were washed twice with cold PBS and re-suspended in 1 ml cold PBS, fixed in cold 70% ethanol, washed with cold PBS twice and re-suspended in PI master mix (PI 10 ug/ml (sigma), 50 ug/ml RNase A (invitrogen) in staining buffer) at a final cell density of 2 ⁇ 10 5 cell/ml. The cell mixtures were incubated 30 min in the dark at 4° C. Flow cytometric analysis was immediately performed using a guava easyCyteTM 8 Instrument (Merck Millipore, Darmstadt, Germany).
  • iCP-SOCS3 (HM 165 S3B) protein efficiently inhibited cell cycle progression in various cancer cells, such as AGS, HCT116, U-87 MG, and MDA-MB-231 ( FIGS. 85 to 88 ), while iCP-SOCS3 (HM 165 S3B) protein did not inhibit cell cycle progression in normal cells ( FIG. 84 ).
  • Cells were treated for 24 hr with 10 ⁇ M HS3B or HM 165 S3B proteins, lysed in RIPA lysis buffer containing proteinase inhibitor cocktail, incubated for 15 min at 4° C., and centrifuged at 13,000 rpm for 10 min at 4° C. Equal amounts of lysates were separated on 15% SDS-PAGE gels and transferred to a nitrocellulose membrane. The membranes were blocked using 5% skim milk or 5% Albumin in TBST and incubated with the following antibodies: anti-Bcl-2 (Santa Cruz biotechnology), anti-Cleaved Caspase 3 (Cell Signaling Technology), then HRP conjugated anti-mouse or anti-rabbit secondary antibody. The expression of each protein was determined by immunoblotting with indicated antibodies. An antibody against ⁇ -actin was used as a loading control.
  • MDA-MB-231 cells treated with iCP-SOCS3 (HM 165 S3B) protein dramatically reduced the expression of anti-apoptotic protein such as B-cell lymphoma 2 (Bcl-2) and increased the level of cleaved cysteine-aspartic acid protease (caspase-3) at 24 hr ( FIG. 89 ).
  • Bcl-2 B-cell lymphoma 2
  • caspase-3 cleaved cysteine-aspartic acid protease
  • Example 19-1 Investigation of Anti-Cancer Efficacy of iCP-SOCS3 in Gastric Cancer Cell (NCI-N87)-Derived Xenograft (CDX) Model
  • Tumor tissues were finely minced using a homogenizer according to the manufacturer's protocol of Hybrid-R (geneall, Korea), and then mRNA was isolated therefrom. 1 ug of mRNA thus separated was used to synthesize cDNA using an Accupower RT Premix (Bioneer, Korea). PCR was performed using 2 ul of cDNA and the primers of Table 45. PCR was performed using an Accupower PCR Premix (Bioneer, Korea) for 30 cycles consisting of denaturing (95° C.), annealing (60° C.), and extending (72° C.) for 45 seconds each.
  • Tissue samples were fixed in 4% Paraformaldehyde (Duksan, South Korea) for 3 days, dehydrated, cleared with xylene and embedded in Paraplast. Sections (6 ⁇ m thick) of tumor were placed onto poly-L-lysine coated slides. To block endogenous peroxidase activity, sections were incubated for 15 min with 3% H 2 O 2 in methanol. After washing three times with PBS, slides were incubated for 30 min with blocking solution (5% fetal bovine serum in PBS).
  • blocking solution 5% fetal bovine serum in PBS
  • Mouse anti-Bax antibody (sc-7480, Santa Cruz Biotechnology, SantaCruz, Calif., USA) and rabbit anti-VEGF (ab46154, Abcam, Cambridge, UK) were diluted 1:1000 (to protein concentration 0.1 ⁇ g/ml) in blocking solution, applied to sections, and incubated at 4° C. for 24 hrs.
  • mice Female Balb/c nu/nu mice (5-week-old, Doo-Yeol Biotech Co. Ltd. Korea) were subcutaneously implanted with Hep3B2.1-7 tumor block (1 mm 3 ) into the left back side of the mouse. Tumor-bearing mice were intravenously administered with 600 ⁇ g/head of iCP-SOCS3 (HM 165 S3B) or the control proteins (HS3B) for 21 days and observed for 2 weeks following the termination of the treatment.
  • HM 165 S3B the control proteins
  • mice After protein treatment, mice were killed, and six organs (brain, heart, lung, liver, kidney, and spleen) from each were collected and kept in a suitable fixation solution until the next step. Tumor size was monitored by measuring the longest (length) and shortest dimensions (width) once a day with a dial caliper, and tumor volume was calculated as width2 ⁇ length ⁇ 0.5.
  • mRNA levels were analyzed using the tumor tissues removed at 2 weeks (Day 35) after termination of drug administration to analyze changes in expressions of major biomarker genes by iCP-SOCS3.
  • cyclinA1 cell cycle (cyclinA1) and angiogenesis (VEGF)-related genes were significantly regulated by treatment of iCP-SOCS3.
  • iCP-SOCS3 is able to inhibit development of gastric cancer through expressions of various factors regulating cancer activity such as cell cycle, angiogenesis, etc.
  • tumor tissues removed at 2 weeks (Day 35) after termination of drug administration were used to perform Western blotting and IHC, and increase or decrease in the expressions of cancer activity regulating factors by iCP-SOCS3 was observed.
  • Example 19-2 Investigation of Anti-Cancer Efficacy of iCP-SOCS3 in Colorectal Cancer Cell (HCT116)-Derived Xenograft (CDX) Model
  • HM 165 S3B protein significantly suppressed the tumor growth (p ⁇ 0.05) during the treatment and the effect persisted for at least 2 weeks after the treatment was terminated (79% at day 35, respectively).
  • the growth of HS3B-treated tumors increased, matching the rates observed in control mice (Vehicle).
  • apoptosis (Bax) and angiogenesis (VEGF)-related genes were significantly regulated by treatment of iCP-SOCS3.
  • tumor tissues removed at 2 weeks (Day 35) after termination of drug administration were used to perform Western blotting and IHC, and increase or decrease in the expressions of cancer activity regulating factors by iCP-SOCS3 was observed.
  • iCP-SOCS3-treated tumor tissues expressions of a cell cycle regulator, p21 and an apoptosis inducer, Bax were increased. Further, expression of an angiogenesis inducer VEGF was also remarkably decreased ( FIG. 91 ). Accordingly, it was demonstrated that cell cycle, apoptosis, and angiogenesis are also regulated by iCP-SOCS3 in-vivo.
  • HM 165 S3B protein significantly suppressed the tumor growth (p ⁇ 0.05) during the treatment and the effect persisted for at least 2 weeks after the treatment was terminated (76% at day 42, respectively).
  • the growth of HS3B-treated tumors increased, matching the rates observed in control mice (Vehicle).
  • apoptosis (Bax), cell cycle (CDK4, CDK2, cyclinE, cyclinAl), angiogenesis (HIF1a, VEGF), and migration (FAK)-related genes were significantly regulated by treatment of iCP-SOCS3.
  • VEGF and angiopoietin I which are biomarker genes of angiogenesis were significantly decreased by treatment of iCP-SOCS3.
  • iCP-SOCS3 is able to inhibit development of glioblastoma through expressions of various factors regulating cancer activity such as apoptosis, cell cycle, migration, angiogenesis, etc.
  • HM 165 S3B protein significantly suppressed the tumor growth (p ⁇ 0.05) during the treatment and the effect persisted for at least 2 weeks after the treatment was terminated (63% at day 35, respectively).
  • the growth of HS3B-treated tumors increased, matching the rates observed in control mice (Vehicle).
  • iCP-SOCS3 is able to inhibit development of breast cancer through expressions of various factors regulating cancer activity such as cell cycle, angiogenesis, etc., indicating characteristics of iCP-SOCS3 as a mechanism-specific therapeutic agent targeting solid tumors.
  • tumor tissues removed at 2 weeks (Day 35) after termination of drug administration were used to perform Western blotting and IHC, and increase or decrease in the expressions of cancer activity regulating factors by iCP-SOCS3 was observed.

Abstract

The present invention relates to providing improved cell-permeable (iCP)-SOCS3 recombinant protein and uses thereof. Preferably, the iCP-SOCS3 recombinant protein may be used as protein-based anti-solid tumor agent by utilizing the platform technology for macromolecule intracellular transduction.

Description

    TECHNICAL FIELD
  • The present invention relates to providing improved cell-permeable (iCP)-SOCS3 recombinant protein and uses thereof. Preferably, the iCP-SOCS3 recombinant protein may be used as protein-based anti-solid tumor agent by utilizing the platform technology for macromolecule intracellular transduction.
  • BACKGROUND ART
  • The Janus kinase signal transducers and activators of transcription signaling (JAK/STAT) plays important roles in immune responses, including oncogenesis. So many investigations demonstrated that STAT-3, an important member of STAT proteins, was considered as a proto-oncogene in various types of disorder. STAT-3 is phosphorylated and dimerizes by the Janus kinase (JAK), and its overexpression and constitutive activation can significantly induce cell proliferation, tumor angiogenesis, invasion. Meanwhile, inhibition of JAK-STAT signaling led to suppress the cancer cell growth and induce apoptosis. Suppressor of cytokine signaling-3 (SOCS3), a kind of endogenous protein inhibitor of JAK/STAT pathway, was identified to be inversely associated with the STATS expression and phosphorylation in vivo and in vitro and aberrant expression of SOCS3 protein was observed in human solid tumors including gastric, colorectal and breast cancer, and glioblastoma.
  • Gastric cancer remains the second leading cause of cancer-related death in the world. Advances in early detection and decreased chronic Helicobacter pylori infection rates have led to a substantial reduction in gastric cancer rates worldwide. However, effective treatment regimens for gastric cancers, especially advanced gastric cancer, are still lacking; therefore, the prognosis of patients with this disease remains poor. SOCS3 mRNA levels are higher in adjacent normal mucosal tissues, however, gastric cancer patients with high simultaneous expression of SOCS3 have a better overall survival than those with low simultaneous expression. Based on this, SOCS3 may represent new therapeutic target to treat gastric cancer.
  • Colorectal cancer is one of the most fatal neoplastic diseases worldwide and a serious global health problem, with over one million new cases and half million mortalities worldwide each year. It has been reported as being relevant to some inflammatory bowel diseases, such as Crohn's disease and ulcerative colitis. The pathogenesis of colorectal carcinoma is complex, with the involvement of multiple cellular transduction pathways including IL-6/STAT3 signaling. Reduced or silenced SOCS3 has been found in many human types of cancer including colorectal cancer, and restoring SOCS3 expression in the cancer cells inhibits IL-6-mediated STAT3 activation, induces tumor cell apoptosis and decreases cell proliferation. Therefore, suppression of the IL-6/STAT3 pathway via modulation of SOCS3 has been a promising strategy for anti-colon/colorectal cancer therapy.
  • Glioblastoma, the most common neoplasm among diffuse infiltrating astrocytomas, is notorious for its ability to evade immune-surveillance as well as for its invasive and angiogenic properties. Gliomas are the most common type of primary brain tumors are highly malignant and are associated with a very poor prognosis. Glioblastoma is a very aggressive subtype of glioma with very short life expectancy and limited treatment options. A hallmark of this lethal disorder is the presence of activated STAT3. Because SOCS3 is a negative regulator of STAT-3 activation, it hypothesized that SOCS3 may function as a tumor suppressor in glioblastoma tissues.
  • Breast cancer is a disease that arises from the accumulation of alterations in the genome of cells that make up the mammary gland. Breast cancer is the most common type of cancer among women, with an estimated 1.38 million new cases of cancer diagnosed in 2008 (23% of all cancers), and the second most common type of cancer overall (10.9% of all cancers). Expression of SOCS3 protein is significantly down-regulated in breast cancer specimens and replacing of SOCS3 protein may directly influence the treatment of breast cancer.
  • Cytokine signaling is strictly regulated by the SOCS family proteins induced by different classes of agonists, including cytokines, hormones and infectious agents. Among them, SOCS1 and SOCS3 are relatively specific to STAT1 and STAT3, respectively. SOCS1 inhibits JAK activation through its N-terminal kinase inhibitory region (KIR) by the direct binding to the activation loop of JAKs, while SOCS3 binds to janus kinases (JAKs)-proximal sites on the receptor through its SH2 domain and inhibits JAK activity that blocks recruitment of STAT3. Both promote anti-inflammatory effects due to the suppression of inflammation-inducing cytokine signaling. Furthermore, the SOCS box, another domain in SOCS proteins, interacts with E3 ubiquitin ligases and/or couples the SH2 domain-binding proteins to the ubiquitin-proteasome pathway. Therefore, SOCSs inhibit cytokine signaling by suppressing JAK kinase activity and degrading the activated cytokine receptor complex.
  • In connection with SOCSs and various solid tumors including gastric, colorectal and breast cancer, and glioblastoma, the SOCS1 gene has been implicated as an anti-oncogene in the tumor development. Previous studies have reported that aberrant methylation in the CpG island of SOCS1 induces its transcriptional silencing in cancer cell lines, and SOCS1 heterozygous mice are hypersensitive to various cancers. In addition, abnormalities of SOCS3 are also associated with the solid tumors. Hypermethylation of CpG islands in the SOCS3 promoter is correlated with its transcriptional silencing in tumors cell lines. SOCS3 overexpression down-regulates active STAT3, induces apoptosis, and suppresses growth in cancer cells. The importance of STAT3 to inflammation-associated carcinogenesis is underlined by the previous study that cancer-specific deletion of SOCS3 in a mouse carcinoma model results in larger and more numerous tumors. This means that SOCS3 plays a major role in the negative regulation of the JAK/STAT pathway in carcinogenesis and contributes to the suppression of tumor development by protecting the tissue cells.
  • REFERENCES
    • 1. Fischer P M., Cellular uptake mechanisms and potential therapeutic utility of peptidic cell delivery vectors: progress 2001-2006, Med Res Rev. 2007; 27:755-95.
    • 2. Heitz F, Morris M C, Divita G., Twenty years of cell-penetrating peptides: from molecular mechanisms to therapeutics, Br J Pharmacol. 2009; 157:195-206.
    • 3. Lapenna S, Giordano A., Cell cycle kinases as therapeutic targets for cancer, Nat Rev Drug Discov. 2009; 8:547-66.
    • 4. Lim J, Kim J, Duong T, Lee G, Kim J, Yoon J. et al., Antitumor activity of cell-permeable p18(INK4c) with enhanced membrane and tissue penetration, Mol Ther. 2012; 20:1540-9.
    • 5. Jo D, Liu D, Yao S, Collins R D, Hawiger J., Intracellular protein therapy with SOCS3 inhibits inflammation and apoptosis, Nat Med. 2005; 11:892-8.
    • 6. Jo D, Nashabi A, Doxsee C, Lin Q, Unutmaz D, Chen J. et al., Epigenetic regulation of gene structure and function with a cell-permeable Cre recombinase, Nat Biotechnol. 2001; 19:929-33.
    • 7. Liu D, Li C, Chen Y, Burnett C, Liu X Y, Downs S. et al., Nuclear import of proinflammatory transcription factors is required for massive liver apoptosis induced by bacterial lipopolysaccharide, J Biol Chem. 2004; 279:48434-42.
    • 8. Liu D, Liu X Y, Robinson D, Burnett C, Jackson C, Seele L. et al., Suppression of Staphylococcal Enterotoxin B-induced Toxicity by a Nuclear Import Inhibitor, J Biol Chem. 2004; 279:19239-46.
    • 9. Liu D, Zienkiewicz J, DiGiandomenico A, Hawiger J., Suppression of acute lung inflammation by intracellular peptide delivery of a nuclear import inhibitor, Mol Ther. 2009; 17:796-802.
    • 10. Moore D J, Zienkiewicz J, Kendall P L, Liu D, Liu X, Veach R A. et al., In vivo islet protection by a nuclear import inhibitor in a mouse model of type 1 diabetes, PLoS One. 2010; 5:e13235.
    • 11. Lim J, Jang G, Kang S, Lee G, Nga do T T, Phuong do T L. et al., Cell-permeable NM23 blocks the maintenance and progression of established pulmonary metastasis, Cancer Res. 2011; 71:7216-25.
    • 12. Duong T, Kim J, Ruley H E, Jo D., Cell-permeable parkin proteins suppress Parkinson disease-associated phenotypes in cultured cells and animals, PLoS One. 2014; 9:e102517.
    • 13. Lim J, Duong T, Do N, Do P, Kim J, Kim H. et al., Antitumor activity of cell-permeable RUNX3 protein in gastric cancer cells. Clin Cancer Res. 2013; 19:680-90.
    • 14. Lim J, Duong T, Lee G, Seong B L, El-Rifai W, Ruley H E et al. The effect of intracellular protein delivery on the anti-tumor activity of recombinant human endostatin, Biomaterials. 2013; 34:6261-71.
    • 15. Lim J, Kim J, Kang J, Jo D., Partial somatic to stem cell transformations induced by cell-permeable reprogramming factors, Scientific Reports. 2014; 4:4361.
    DISCLOSURE Technical Problem
  • To negatively control JAK/STAT signaling, recombinant SOCS3 proteins that contain a cell-penetrating peptide (CPP)—membrane-translocating motif (MTM) from fibroblast growth factor (FGF)-4 has been reported. These recombinant SOCS3 proteins inhibited STAT phosphorylation, inflammatory cytokines production and MHC-II expression in cultured and primary macrophages. In addition, SOCS3 fused to MTM protected mice challenged with a lethal dose of the SEB super-antigen, by suppressing apoptosis and hemorrhagic necrosis in multiple organs. However, the SOCS3 proteins fused to FGF4-derived MTM displayed extremely low solubility, poor yields and relatively low cell- and tissue-permeability. Therefore, the MTM-fused SOCS3 proteins were not suitable for further clinical development as therapeutic agents.
  • Technical Solution
  • For MITT, six critical factors (length, bending potential, instability index, aliphatic index, GRAVY, amino acid composition) have been determined through analysis of baseline hydrophobic CPPs. Advanced macromolecule transduction domain (aMTD), newly designed based on these six critical factors, could optimize cell-/tissue-permeability of SOCS3 proteins that have a therapeutic effects and develop them as protein-based drugs. Further, in order to increase solubility and yield of recombinant protein, solubilization domains (SDs) additionally fused to the aMTD-SOCS3 recombinant protein, thereby notably increased the solubility and manufacturing yield of the recombinant protein.
  • In this application, aMTD/SD-fused iCP-SOCS3 recombinant proteins (iCP-SOCS3), much improved physicochemical characteristics (solubility and yield) and functional activity (cell-/tissue-permeability) compared with the protein fused only to FGF-4-derived MTM. In addition, the newly developed iCP-SOCS3 proteins have now been demonstrated to have therapeutic application in treating solid tumor, exploiting the ability of SOCS3 to suppress JAK/STAT signaling. The present application represents that macromolecule intracellular transduction technology (MITT) enabled by the new hydrophobic CPPs that are aMTD may provide novel protein therapy through SOCS3-intracellular protein replacement against the solid tumor. These findings suggest that restoration of SOCS3 by replenishing the intracellular SOCS3 with iCP-SOCS3 protein creates a new paradigm for anti-cancer therapy, and the intracellular protein replacement therapy with the SOCS3 recombinant protein fused to the combination of aMTD and SD pair may be useful to treat the solid tumor.
  • One aspect disclosed in the present application provides an improved Cell-Permeable (iCP)-SOCS3 recombinant protein, which comprises a SOCS3 protein; and an advanced macromolecule transduction domain (aMTD) being composed of 9-13 amino acid sequences and having improved cell and/or tissue permeability, wherein the aMTD is fused to one end or both ends of the SOCS3 protein and has the following features of:
  • (a) being composed of 3 or more amino acid sequences selected from the group consisting of Ala, Val, Ile, Leu, and Pro;
  • (b) having proline as amino acid sequences corresponding to any one or more of positions 5 to 8, and 12 of its amino acid sequence; and
  • (c) having an instability index of 40-60; an aliphatic index of 180-220; and a grand average of hydropathy (GRAVY) of 2.1-2.6, as measured by Protparam.
  • According to one embodiment, one or more solubilization domain (SD)(s) are further fused to the end(s) of one or more of the SOCS3 protein and the aMTD.
  • According to another embodiment, the aMTD may form α-Helix structure. According to still another embodiment, the aMTD may be composed of 12 amino acid sequences and represented by the following general formula:
  • wherein X(s) independently refer to Alanine (A), Valine (V), Leucine (L) or Isoleucine (I); and Proline (P) can be positioned in one of U(s) (either 5′, 6′, 7′ or 8′). The remaining U(s) are independently composed of A, V, L or I, P at the 12′ is Proline.
  • Another aspect disclosed in the present application provides an iCP-SOCS3 recombinant protein which is represented by any one of the following structural formulae:

  • A-B-C, A-C-B, B-A-C, B-C-A, C-A-B, C-B-A and A-C-B-C
  • wherein A is an advanced macromolecule transduction domain (aMTD) having improved cell and/or tissue permeability, B is a SOCS3 protein, and C is a solubilization domain (SD); and
  • the aMTD is composed of 9-13 amino acid sequences and has the following features of:
  • (a) being composed of 3 or more amino acids selected from the group consisting of Ala, Val, Ile, Leu, and Pro;
  • (b) having proline as amino acid sequences corresponding to any one or more of positions 5 to 8, and 12 of its amino acid sequence;
  • (c) having an instability index of 40-60; an aliphatic index of 180-220; and a grand average of hydropathy (GRAVY) of 2.1-2.6, as measured by Protparam; and
  • (d) forming α-Helix structure.
  • According to one embodiment disclosed in the present application, the SOCS3 protein may have an amino acid sequence of SEQ ID NO: 814.
  • According to another embodiment disclosed in the present application, the SOCS3 protein may be encoded by a polynucleotide sequence of SEQ ID NO: 815.
  • According to still another embodiment disclosed in the present application, the SOCS3 protein may further include a ligand selectively binding to a receptor of a cell, a tissue, or an organ. According to still another embodiment disclosed in the present application, the aMTD may have an amino acid sequence selected from the group consisting of SEQ ID NOs: 1-240 and 822.
  • According to still another embodiment disclosed in the present application, the aMTD may be encoded by a polynucleotide sequence selected from the group consisting of SEQ ID NOs: 241-480 and 823.
  • According to still another embodiment disclosed in the present application, the SD(s) may have an amino acid sequence independently selected from the group consisting of SEQ ID NOs: 798, 799, 800, 801, 802, 803, and 804.
  • According to still another embodiment disclosed in the present application, the SD(s) may be encoded by a polynucleotide sequence independently selected from the group consisting of SEQ ID NOs: 805, 806, 807, 808, 809, 810, and 811.
  • According to still another embodiment disclosed in the present application, the iCP-SOCS3 recombinant protein may have a histidine-tag affinity domain additionally fused to one end thereof.
  • According to still another embodiment disclosed in the present application, the histidine-tag affinity domain may have an amino acid sequence of SEQ ID NO: 812.
  • According to still another embodiment disclosed in the present application, the histidine-tag affinity domain may be encoded by a polynucleotide sequence of SEQ ID NO: 813.
  • According to still another embodiment disclosed in the present application, the fusion may be formed via a peptide bond or a chemical bond.
  • According to still another embodiment disclosed in the present application, the iCP-SOCS3 recombinant protein may be used for the treating, preventing, or delaying the onset of, solid tumor.
  • Still another aspect disclosed in the present application provides a polynucleotide sequence encoding the iCP-SOCS3 recombinant protein.
  • According to one embodiment disclosed in the present application, the polynucleotide sequence may be a polynucleotide sequence represented by SEQ ID NO: 825.
  • Still another aspect disclosed in the present application provides a recombinant expression vector including the polynucleotide sequence.
  • Still another aspect disclosed in the present application provides a transformant transformed with the recombinant expression vector.
  • Still another aspect disclosed in the present application provides a preparing method of the iCP-SOCS3 recombinant protein including preparing the recombinant expression vector; preparing the transformant using the recombinant expression vector; culturing the transformant; and recovering the recombinant protein expressed by the culturing.
  • Still another aspect disclosed in the present application provides a composition including the iCP-SOCS3 recombinant protein as an active ingredient.
  • Still another aspect disclosed in the present application provides a pharmaceutical composition for the treating, preventing, or delaying the onset of, solid tumor including the iCP-SOCS3 recombinant protein as an active ingredient; and a pharmaceutically acceptable carrier.
  • Still another aspect disclosed in the present application provides use of the iCP-SOCS3 recombinant protein as a medicament for the treating, preventing, or delaying the onset of, solid tumor.
  • Still another aspect disclosed in the present application provides a medicament including the iCP-SOCS3 recombinant protein.
  • Still another aspect disclosed in the present application provides use of the iCP-SOCS3 recombinant protein in the preparation of a medicament for the treating, preventing, or delaying the onset of, solid tumor.
  • Still another aspect disclosed in the present application provides a method of treating, preventing, or delaying the onset of, solid tumor in a subject, the method including identifying a subject in need of the treating, preventing, or delaying the onset of, solid tumor; and administering to the subject a therapeutically effective amount of the iCP-SOCS3 recombinant protein.
  • According to one embodiment disclosed in the present application, the subject may be a mammal.
  • Unless defined otherwise, all terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the present invention belongs. Although a certain method and a material is described herein, it should not be construed as being limited thereto, any similar or equivalent method and material to those may also be used in the practice or testing of the present invention. All publications mentioned herein are incorporated herein by reference in their entirety to disclose and describe the methods and/or materials in connection with which the publications are cited. It must be noted that as used herein and in the appended claims, the singular forms “a,” “an,” and “the” include plural referents unless the context clearly dictates otherwise.
  • A “peptide,” as used herein, refers to a chain-type polymer formed by amino acid residues which are linked to each other via peptide bonds, and used interchangeably with “polypeptide.” Further, a “polypeptide” includes a peptide and a protein.
  • Further, the term “peptide” includes amino acid sequences that are conservative variations of those peptides specifically exemplified herein. The term “conservative variation,” as used herein, denotes the replacement of an amino acid residue by another, biologically similar residue. Examples of conservative variations include substitution of one hydrophobic residue, such as isoleucine, valine, leucine, alanine, cysteine, glycine, phenylalanine, proline, tryptophan, tyrosine, norleucine, or methionine for another, or substitution of one polar residue for another, for example, substitution of arginine for lysine, glutamic acid for aspartic acid, or glutamine for asparagine, and the like. Neutral hydrophilic amino acids which may be substituted for one another include asparagine, glutamine, serine, and threonine.
  • The term “conservative variation” also includes use of a substituted amino acid in place of an unsubstituted parent amino acid, provided that antibodies raised to the substituted polypeptide also immunoreact with the unsubstituted polypeptide. Such conservative substitutions are within the definition of the classes of the peptides disclosed in the present application.
  • A person having ordinary skill in the art may make similar substitutions to obtain peptides having higher cell permeability and a broader host range. For example, one aspect disclosed in the present application provides peptides corresponding to amino acid sequences (e.g. SEQ ID NOs: 1 to 240 and 822) provided herein, as well as analogues, homologs, isomers, derivatives, amidated variations, and conservative variations thereof, as long as the cell permeability of the peptide remains.
  • Minor modifications to primary amino acid sequence disclosed in the present application may result in peptides which have substantially equivalent or enhanced cell permeability, as compared to the specific peptides described herein. Such modifications may be deliberate, as by site-directed mutagenesis, or may be spontaneous.
  • All peptides may be synthesized using L-amino acids, but D forms of all of the peptides may be synthetically produced. In addition, C-terminal derivatives, such as C-terminal methyl esters and C-terminal amidates, may be produced in order to increase the cell permeability of the peptide according to one embodiment disclosed in the present application.
  • All of the peptides produced by these modifications are included herein, as long as in the case of amidated versions of the peptide, the cell permeability of the original peptide is altered or enhanced such that the amidated peptide is therapeutically useful. It is envisioned that such modifications are useful for altering or enhancing cell permeability of a particular peptide.
  • Furthermore, deletion of one or more amino acids may also result in a modification to the structure of the resultant molecule without any significant change in its cell permeability. This may lead to the development of a smaller active molecule which may also have utility. For example, amino- or carboxyl-terminal amino acids which may not be required for the cell permeability of a particular peptide may be removed.
  • The term “gene” refers to an arbitrary nucleic acid sequence or a part thereof having a functional role in protein coding or transcription, or regulation of other gene expression. The gene may be composed of all nucleic acids encoding a functional protein or a part of the nucleic acid encoding or expressing the protein. The nucleic acid sequence may include a gene mutation in exon, intron, initiation or termination region, promoter sequence, other regulatory sequence, or a unique sequence adjacent to the gene.
  • The term “primer” refers to an oligonucleotide sequence that hybridizes to a complementary RNA or DNA target polynucleotide and serves as the starting points for the stepwise synthesis of a polynucleotide from mononucleotides by the action of a nucleotidyltransferase as occurs, for example, in a polymerase chain reaction.
  • The term “coding region” or “coding sequence” refers to a nucleic acid sequence, a complement thereof, or a part thereof which encodes a particular gene product or a fragment thereof for which expression is desired, according to the normal base pairing and codon usage relationships. Coding sequences include exons in genomic DNA or immature primary RNA transcripts, which are joined together by the cellular biochemical machinery to provide a mature mRNA. The anti-sense strand is the complement of the nucleic acid, and the coding sequence may be deduced therefrom.
  • One aspect disclosed in the present application provides an iCP-SOCS3 recombinant protein, which comprises a SOCS3 protein and an advanced macromolecule transduction domain (aMTD) being composed of 9-13 amino acid sequences, preferably 10-12 amino acid sequences, and having improved cell and/or tissue permeability, wherein the aMTD is fused to one end or both ends of the SOCS3 protein and has the following features of:
  • (a) being preferably composed of 3 or more amino acid sequences selected from the group consisting of Ala, Val, Ile, Leu, and Pro;
  • (b) having proline as amino acid sequences corresponding to any one or more of positions 5 to 8, and 12 of its amino acid sequence, and preferably one or more of positions 5 to 8 and position 12 of its amino acid sequence; and
  • (c) having an instability index of preferably 40-60 and more preferably 41-58; an aliphatic index of preferably 180-220 and more preferably 185-225; and a grand average of hydropathy (GRAVY) of preferably 2.1-2.6 and more preferably 2.2-2.6 as measured by Protparam (see http://web.expasy.org/protparam/).
  • These critical factors that facilitate the cell permeable ability of aMTD sequences were analyzed, identified, and determined according to one embodiment disclosed in the present application. These aMTD sequences are artificially assembled based on the critical factors (CFs) determined from in-depth analysis of previously published hydrophobic CPPs.
  • The aMTD sequences according to one aspect disclosed in the present application are the first artificially developed cell permeable polypeptides capable of mediating the transduction of biologically active macromolecules—including peptides, polypeptides, protein domains, or full-length proteins—through the plasma membrane of cells.
  • According to one embodiment, one or more solubilization domain (SD)(s) are further fused to one or more of the SOCS3 protein and the aMTD, preferably one end or both ends of the SOCS3 protein, and more preferably the C-terminus of the SOCS3 protein.
  • According to another embodiment, the aMTD may form a-Helix structure.
  • According to still another embodiment, the aMTD may be preferably composed of 12 amino acid sequences and represented by the following general formula:
  • Here, X(s) independently refer to Alanine (A), Valine (V), Leucine (L) or Isoleucine (I); and Proline (P) can be positioned in one of U(s) (either 5′, 6′, 7′ or 8′). The remaining U(s) are independently composed of A, V, L or I, P at the 12′ is Proline.
  • Still another aspect disclosed in the present application provides an iCP-SOCS3 recombinant protein which is represented by any one of structural formulae A-B-C, A-C-B, B-A-C, B-C-A, C-A-B, C-B-A and A-C-B-C, and preferably by A-B-C or C-B-A:
  • wherein A is an advanced macromolecule transduction domain (aMTD) having improved cell and/or tissue permeability, B is a SOCS3 protein, and C is a solubilization domain (SD); and
  • the aMTD is composed of 9-13, preferably 10-12 amino acid sequences and has the following features of:
  • (a) being composed of 3 or more amino acid sequences selected from the group consisting of Ala, Val, Ile, Leu, and Pro;
  • (b) having proline as amino acid sequences corresponding to any one or more of positions 5 to 8, and 12 of its amino acid sequence, and preferably, one or more of positions 5 to 8 and position 12 of its amino acid sequence;
  • (c) having an instability index of 40-60, preferably 41-58 and more preferably 50-58; an aliphatic index of 180-220. preferably 185-225 and more preferably 195-205; and a grand average of hydropathy (GRAVY) of 2.1-2.6 and preferably 2.2-2.6, as measured by Protparam (see http://web.expasy.org/protparam/); and
  • (d) preferably forming α-Helix structure.
  • In one embodiment disclosed in the present application, the SOCS3 protein may have an amino acid sequence of SEQ ID NO: 814.
  • In another embodiment disclosed in the present application, the SOCS3 protein may be encoded by a polynucleotide sequence of SEQ ID NO: 815.
  • When the iCP-SOCS3 recombinant protein is intended to be delivered to a particular cell, tissue, or organ, the SOCS3 protein may form a fusion product, together with an extracellular domain of a ligand capable of selectively binding to a receptor which is specifically expressed on the particular cell, tissue, or organ, or monoclonal antibody (mAb) capable of specifically binding to the receptor or the ligand and a modified form thereof.
  • The binding of the peptide and a biologically active substance may be formed either by indirect linkage by a cloning technique using an expression vector at a nucleotide level or by direct linkage via chemical or physical covalent or non-covalent bond of the peptide and the biologically active substance.
  • In still another embodiment disclosed in the present application, the SOCS3 protein may preferably further include a ligand selectively binding to a receptor of a cell, a tissue, or an organ.
  • In one embodiment disclosed in the present application, the aMTD may have an amino acid sequence selected from the group consisting of SEQ ID NOs: 1-240 and 822, preferably SEQ ID NOs: 2, 16, 22, 32, 40, 43, 63, 65, 77, 84, 85, 86, 110, 131, 142, 143, 177, 228, 229, 233, 237, 239 and 822, more preferably SEQ ID NO: 43.
  • In still another embodiment disclosed in the present application, the aMTD may be encoded by a polynucleotide sequence selected from the group consisting of SEQ ID NOs: 241-480 and 823, preferably SEQ ID NOs: 242, 256, 262, 272, 280, 283, 303, 305, 317, 324, 325, 326, 350, 371, 382, 383, 417, 468, 469 473, 477, 479 and 823, more preferably SEQ ID NO: 283. In still another embodiment disclosed in the present application, the SD(s) may have an amino acid sequence independently selected from the group consisting of SEQ ID NOs: 798, 799, 800, 801, 802, 803, and 804. The SD may be preferably SDA of SEQ ID NO: 798, SDB of SEQ ID NO: 799, or SDB′ of SEQ ID NO: 804, and more preferably, SDB of SEQ ID NO: 799 which has superior structural stability, or SDB′ of SEQ ID NO: 804 which has a modified amino acid sequence of SDB to avoid immune responses upon in vivo application. The modification of the amino acid sequence in SDB may be replacement of an amino acid residue, Valine, corresponding to position 28 of the amino acid sequence of SDB (SEQ ID NO: 799) by Leucine.
  • In still another embodiment disclosed in the present application, the SDs may be encoded by a polynucleotide sequence independently selected from the group consisting of SEQ ID NOs: 805, 806, 807, 808, 809, 810, and 811. The SD may be preferably SDA encoded by a polynucleotide sequence of SEQ ID NO: 805, SDB encoded by a polynucleotide sequence of SEQ ID NO: 806, or SDB′ for deimmunization (or humanization) encoded by a polynucleotide sequence of SEQ ID NO: 811, and more preferably, SDB having superior structural stability, which is encoded by a polynucleotide sequence of SEQ ID NO: 806, or SDB′ having a modified polynucleotide sequence of SDB to avoid immune responses upon in vivo application, which is encoded by a polynucleotide sequence of SEQ ID NO: 811.
  • In still another embodiment disclosed in the present application, the iCP-SOCS3 recombinant protein may be preferably selected from the group consisting of:
  • 1) a recombinant protein, in which SOCS3 having an amino acid sequence of SEQ ID NO: 814 is fused to the N-terminus or the C-terminus of aMTD having any one amino acid sequence selected from the group consisting of SEQ ID NOs: 1 to 240 and 822, 2, 16, 22, 32, 40, 43, 63, 65, 77, 84, 85, 86, 110, 131, 142, 143, 177, 228, 229, 233, 237, 239 and 822, more preferably SEQ ID NO: 43;
  • 2) a recombinant protein, in which SD having any one amino acid sequence selected from the group consisting of SEQ ID NOs: 798 to 804 is further fused to one or more of the N-terminus or the C-terminus of the SOCS3 and aMTD in the recombinant protein of 1); and
  • 3) a recombinant protein, in which a Histidine tag having an amino acid sequence of 812 is further fused to the N-terminus of the recombinant protein of 1) or 2).
  • According to one embodiment, the iCP-SOCS3 recombinant protein may be composed of an amino acid sequence selected from the group consisting of SEQ ID NO: 825.
  • The SOCS3 protein may exhibit a physiological phenomenon-related activity or a therapeutic purpose-related activity by intracellular or in-vivo delivery. The recombinant expression vector may include a tag sequence which makes it easy to purify the recombinant protein, for example, consecutive histidine codon, maltose binding protein codon, Myc codon, etc., and further include a fusion partner to enhance solubility of the recombinant protein, etc. Further, for the overall structural and functional stability of the recombinant protein or flexibility of the proteins encoded by respective genes, the recombinant expression vector may further include one or more glycine, proline, and spacer amino acid or polynucleotide sequences including AAY amino acids. Furthermore, the recombinant expression vector may include a sequence specifically digested by an enzyme in order to remove an unnecessary region of the recombinant protein, an expression regulatory sequence, and a marker or reporter gene sequence to verify intracellular delivery, but is not limited thereto.
  • In still another embodiment disclosed in the present application, the iCP-SOCS3 recombinant protein may preferably have a histidine-tag affinity domain additionally fused to one end thereof.
  • In still another embodiment disclosed in the present application, the histidine-tag affinity domain may have an amino acid sequence of SEQ ID NO: 812.
  • In still another embodiment disclosed in the present application, the histidine-tag affinity domain may be encoded by a polynucleotide sequence of SEQ ID NO: 813.
  • In still another embodiment disclosed in the present application, the fusion may be formed via a peptide bond or a chemical bond.
  • The chemical bond may be preferably selected from the group consisting of disulfide bonds, diamine bonds, sulfide-amine bonds, carboxyl-amine bonds, ester bonds, and covalent bonds.
  • In still another embodiment disclosed in the present application, the iCP-SOCS3 recombinant protein may be used for the treating, preventing, or delaying the onset of, solid tumor.
  • Still another aspect disclosed in the present application provides a polynucleotide sequence encoding the iCP-SOCS3.
  • According to still another embodiment disclosed in the present application, the polynucleotide sequence may be fused with a histidine-tag affinity domain.
  • Still another aspect disclosed in the present application provides a recombinant expression vector including the polynucleotide sequence.
  • Preferably, the vector may be inserted in a host cell and recombined with the host cell genome, or refers to any nucleic acid including a nucleotide sequence competent to replicate spontaneously as an episome. Such a vector may include a linear nucleic acid, a plasmid, a phagemid, a cosmid, an RNA vector, a viral vector, etc.
  • Preferably, the vector may be genetically engineered to incorporate the nucleic acid sequence encoding the recombinant protein in an orientation either N-terminal and/or C-terminal to a nucleic acid sequence encoding a peptide, a polypeptide, a protein domain, or a full-length protein of interest, and in the correct reading frame so that the recombinant protein consisting of aMTD, SOCS3 protein, and preferably SD may be expressed. Expression vectors may be selected from those readily available for use in prokaryotic or eukaryotic expression systems.
  • Standard recombinant nucleic acid methods may be used to express a genetically engineered recombinant protein. The nucleic acid sequence encoding the recombinant protein according to one embodiment disclosed in the present application may be cloned into a nucleic acid expression vector, e.g., with appropriate signal and processing sequences and regulatory sequences for transcription and translation, and the protein may be synthesized using automated organic synthetic methods. Synthetic methods of producing proteins are described in, for example, the literature [Methods in Enzymology, Volume 289: Solid-Phase Peptide Synthesis by Gregg B. Fields (Editor), Sidney P. Colowick, Melvin I. Simon (Editor), Academic Press (1997)].
  • In order to obtain high level expression of a cloned gene or nucleic acid, for example, a cDNA encoding the recombinant protein according to one embodiment disclosed in the present application, the recombinant protein sequence may be typically subcloned into an expression vector that includes a strong promoter for directing transcription, a transcription/translation terminator, and in the case of a nucleic acid encoding a protein, a ribosome binding site for translational initiation. Suitable bacterial promoters are well known in the art and are described, e.g., in the literatures [Sambrook & Russell, Molecular Cloning: A Laboratory Manual, 3d Edition, Cold Spring Harbor Laboratory, N.Y. (2001); and Ausubel, et al., Current Protocols in Molecular Biology, Greene Publishing Associates and Wiley Interscience, N. Y. (1989)]. Bacterial expression systems for expression of the recombinant protein are available in, e.g., E. coli, Bacillus sp., and Salmonella (Palva et al., Gene 22: 229-235 (1983); Mosbach et al., Nature 302: 543-545 (1983)). Kits for such expression systems are commercially available. Eukaryotic expression systems for mammalian cells, yeast, and insect cells are well known in the art and are also commercially available. The eukaryotic expression vector may be preferably an adenoviral vector, an adeno-associated vector, or a retroviral vector.
  • Generally, the expression vector for expressing the cell permeable recombinant protein according to one embodiment disclosed in the present application in which the cargo protein, i.e. ΔSOCS3 protein, is attached to the N-terminus, C-terminus, or both termini of aMTD may include regulatory sequences including, for example, a promoter, operably attached to a sequence encoding the advanced macromolecule transduction domain. Non-limiting examples of inducible promoters that may be used include steroid-hormone responsive promoters (e.g., ecdysone-responsive, estrogen-responsive, and glutacorticoid-responsive promoters), tetracycline “Tet-On” and “Tet-Off” systems, and metal-responsive promoters.
  • The polynucleotide sequence according to one embodiment disclosed in the present application may be present in a vector in which the polynucleotide sequence is operably linked to regulatory sequences capable of providing for the expression of the polynucleotide sequence by a suitable host cell.
  • According to one embodiment disclosed in the present application, the polynucleotide sequence may be selected from the following groups:
  • 1) a polynucleotide sequence, in which any one polynucleotide sequence selected from the group consisting of SEQ ID NOs: 241-480 and 823, preferably SEQ ID NOs: 242, 252, 274, 279, 322, 331, 338, 345, 347, 361, 365, 370, 371, 383, 387, 417, 462, 468, 469, 473, 477, 479 and 823, more preferably SEQ ID NO: 283, is operably linked with a polynucleotide sequence of SEQ ID NO: 815; and
  • 2) a polynucleotide sequence, in which any one polynucleotide sequence selected from the group consisting of SEQ ID NOs: 805 to 811 is further operably linked to the polynucleotide sequence of 1), or further operably linked to between: any one polynucleotide sequence selected from the group consisting of SEQ ID NOs: 241-480 and 823, preferably SEQ ID NOs: 242, 256, 262, 272, 280, 283, 303, 305, 317, 324, 325, 326, 350, 371, 382, 383, 417, 468, 469, 473, 477, 479 and 823, more preferably SEQ ID NO: 283; and a polynucleotide sequence of SEQ ID NO: 815.
  • Within an expression vector, the term “operably linked” is intended to mean that the polynucleotide sequence of interest is linked to the regulatory sequence(s) in a manner which allows for expression of the polynucleotide sequence. The term “regulatory sequence” is intended to include promoters, enhancers, and other expression control elements. Such operable linkage with the expression vector can be achieved by conventional gene recombination techniques known in the art, while site-directed DNA cleavage and linkage are carried out by using conventional enzymes known in the art.
  • The expression vectors may contain a signal sequence or a leader sequence for membrane targeting or secretion, as well as regulatory sequences such as a promoter, an operator, an initiation codon, a termination codon, a polyadenylation signal, an enhancer and the like. The promoter may be a constitutive or an inducible promoter. Further, the expression vector may include one or more selectable marker genes for selecting the host cell containing the expression vector, and may further include a polynucleotide sequence that enables the vector to replicate in the host cell in question.
  • The expression vector constructed according to one embodiment disclosed in the present application may be the vector where the polynucleotide encoding the iCP-SOCS3 recombinant protein (where an aMTD is fused to the N-terminus or C-terminus of a SOCS3 protein) is inserted within the multiple cloning sites (MCS), preferably within the Nde1/Sal1 site or BamH1/Sal1 site of a pET-28a(+)(Novagen, Darmstadt, Germany) or pET-26b(+) vector(Novagen, Darmstadt, Germany).
  • In still another embodiment disclosed in the present application, the polynucleotide encoding the SD being additionally fused to the N-terminus or C-terminus of a SOCS3 protein or an aMTD may be inserted into a cleavage site of restriction enzyme (Ndel, BamH1 and Sal1, etc.) within the multiple cloning sites (MCS) of a pET-28a(+)(Novagen, Darmstadt, Germany) or pET-26b(+) vector(Novagen, Darmstadt, Germany).
  • In still another embodiment disclosed in the present application, the polynucleotide encoding the iCP-SOCS3 recombinant protein may be cloned into a pET-28a(+) vector bearing a His-tag sequence so as to fuse six histidine residues to the N-terminus of the iCP-SOCS3 recombinant protein to allow easy purification.
  • According to one embodiment disclosed in the present application, the polynucleotide sequence may be a polynucleotide sequence selected from the group consisting of SEQ ID NOS: 824, 826, 828 and 830.
  • The recombinant protein may be introduced into an appropriate host cell, e.g., a bacterial cell, a yeast cell, an insect cell, or a tissue culture cell. The recombinant protein may also be introduced into embryonic stem cells in order to generate a transgenic organism. Large numbers of suitable vectors and promoters are known to those skilled in the art and are commercially available for generating the recombinant protein.
  • Known methods may be used to construct vectors including the polynucleotide sequence according to one embodiment disclosed in the present application and appropriate transcriptional/translational control signals. These methods include in vitro recombinant DNA techniques, synthetic techniques, and in vivo recombination/genetic recombination. For example, these techniques are described in the literatures [Sambrook & Russell, Molecular Cloning: A Laboratory Manual, 3d Edition, Cold Spring Harbor Laboratory, N. Y. (2001); and Ausubel et al., Current Protocols in Molecular Biology Greene Publishing Associates and Wiley Interscience, N.Y. (1989)].
  • Still another aspect disclosed in the present application provides a transformant transformed with the recombinant expression vector.
  • The transformation includes transfection, and refers to a process whereby a foreign (extracellular) DNA, with or without an accompanying material, enters into a host cell. The “transfected cell” refers to a cell into which the foreign DNA is introduced into the cell, and thus the cell harbors the foreign DNA. The DNA may be introduced into the cell so that a nucleic acid thereof may be integrated into the chromosome or replicable as an extrachromosomal element. The cell introduced with the foreign DNA, etc. is called a transformant.
  • As used herein, ‘introducing’ of a protein, a peptide, an organic compound into a cell may be used interchangeably with the expression of ‘carrying,’ ‘penetrating,’‘transporting,’‘delivering,’ ‘permeating’ or ‘passing.’
  • It is understood that the host cell refers to a eukaryotic or prokaryotic cell into which one or more DNAs or vectors are introduced, and refers not only to the particular subject cell but also to the progeny or potential progeny thereof. Because certain modifications may occur in succeeding generations due to either mutation or environmental influences, such progeny may not, in fact, be identical to the parent cell, but are still included within the scope of the term as used herein.
  • The host cells may be preferably bacterial cells, and as the bacterial cells, there are, in principle, no limitations. They may be eubacteria (gram-positive or gram-negative) or archaebacteria, as long as they allow genetic manipulation for insertion of a gene of interest, preferably for site-specific integration, and they may be cultured on a manufacturing scale. Preferably, the host cells may have the property to allow cultivation to high cell densities.
  • Examples of bacterial host cells that may be used in the preparation of the recombinant protein are E. coli (Lee, 1996; Hannig and Makrides, 1998), Bacillus subtilis, Pseudomonas fluorescens (Squires et al., 2004; Retallack et al., 2006) as well as various Corynebacterium (US 2006/0003404 A1) and Lactococcus lactis (Mierau et al., 2005) strains. Preferably, the host cells are Escherichia coli cells.
  • More preferably, the host cell may include an RNA polymerase capable of binding to a promoter regulating the gene of interest. The RNA polymerase may be endogenous or exogenous to the host cell.
  • Preferably, host cells with a foreign strong RNA polymerase may be used. For example, Escherichia coli strains engineered to carry a foreign RNA polymerase (e.g. like in the case of using a T7 promoter a T7-like RNA polymerase in the so-called “T7 strains”) integrated in their genome may be used. Examples of T7 strains, e.g. BL21(DE3), HMS174(DE3), and their derivatives or relatives (see Novagen, pET System manual, 11th edition), may be widely used and commercially available. Preferably, BL21-CodonPlus (DE3)-RIL or BL21-CodonPlus (DE3)-RIPL (Agilent Technologies) may be used. These strains are DE3 lysogens containing the T7 RNA polymerase gene under control of the lacUV5 promoter. Induction with IPTG allows production of T7 RNA polymerase which then directs the expression of the gene of interest under the control of the T7 promoter.
  • The host cell strains, E. coli BL21(DE3) or HMS174(DE3), which have received their genome-based T7 RNA polymerase via the phage DE3, are lysogenic. It is preferred that the T7 RNA polymerase contained in the host cell has been integrated by a method which avoids, or preferably excludes, the insertion of residual phage sequences in the host cell genome since lysogenic strains have the disadvantage to potentially exhibit lytic properties, leading to undesirable phage release and cell lysis.
  • Still another aspect disclosed in the present application provides a preparing method of the iCP-SOCS3 recombinant protein including preparing the recombinant expression vector; preparing the transformant using the recombinant expression vector; culturing the transformant; and recovering the recombinant protein expressed by culturing.
  • Culturing may be preferably in a mode that employs the addition of a feed medium, this mode being selected from the fed-batch mode, semi-continuous mode, or continuous mode, and the bacterial expression host cells may include a DNA construct, integrated in their genome, carrying the DNA sequence encoding the protein of interest under the control of a promoter that enables expression of said protein.
  • There are no limitations in the type of the culture medium. The culture medium may be semi-defined, i.e. containing complex media compounds (e.g. yeast extract, soy peptone, casamino acids), or it may be chemically defined, without any complex compounds. Preferably, a defined medium may be used. The defined media (also called minimal or synthetic media) are exclusively composed of chemically defined substances, i.e. carbon sources such as glucose or glycerol, salts, vitamins, and, in view of a possible strain auxotrophy, specific amino acids or other substances such as thiamine. Most preferably, glucose may be used as a carbon source. Usually, the carbon source of the feed medium serves as the growth-limiting component which controls the specific growth rate.
  • Host cells may be disrupted by any convenient method, including freeze-thaw cycling, sonication, mechanical disruption, or the use of cell lysing agents. The literature [Scopes, Protein Purification: Principles and Practice, New York: Springer-Verlag (1994)] describes a number of general methods for purifying recombinant (and non-recombinant) proteins. The methods may include, e.g., ion-exchange chromatography, size-exclusion chromatography, affinity chromatography, selective precipitation, dialysis, and hydrophobic interaction chromatography. These methods may be adapted to devise a purification strategy for the cell permeable recombinant protein. If the cell permeable recombinant protein includes a purification handle, such as an epitope tag or a metal chelating sequence, affinity chromatography may be used to easily purify the protein.
  • The amount of the protein produced may be evaluated by detecting the advanced macromolecule transduction domain directly (e.g., using Western analysis) or indirectly (e.g., by assaying materials derived from the cells for specific DNA binding activity, such as by electrophoretic mobility shift assay). Proteins may be detected prior to purification, during any stage of purification, or after purification. In some implementations, purification or complete purification may not be necessary.
  • The iCP-SOCS3 recombinant proteins according to one embodiment disclosed in the present application are cell permeable proteins, and may be used as protein-based vaccines, particularly in the case where killed or attenuated whole organism vaccines are impractical.
  • The iCP-SOCS3 recombinant proteins according to one embodiment disclosed in the present application may be preferably used for the treating, preventing, or delaying the onset of, solid tumor. The cell permeable recombinant proteins may be delivered to the interior of the cell, eliminating the need to transfect or transform the cell with a recombinant vector. The cell permeable recombinant proteins may be used in vitro to investigate protein function or may be used to maintain cells in a desired state.
  • Still another aspect disclosed in the present application provides a composition including the iCP-SOCS3 Recombinant Protein as an active ingredient.
  • Still another aspect disclosed in the present application provides a pharmaceutical composition for treating, preventing, or delaying the onset of, solid tumor including the iCP-SOCS3 Recombinant Protein as an active ingredient; and a pharmaceutically acceptable carrier.
  • According to one embodiment disclosed in the present application, the iCP-SOCS3 Recombinant Protein may be used in a single agent, or in combination with one or more other anti-cancer agents.
  • Solid tumor described herein include, but are not limited to, described herein include, but are not limited to, prostate cancer, renal cancer, hepatic cancer, colon cancer, rectal cancer, colorectal cancer, brain cancer, renal cancer, colorectal cancer, pancreatic cancer, gastric cancer, breast cancer, lung cancer, cancers of the head and neck, thyroid cancer, glioblastoma, Kaposi's sarcoma, melanoma, oesophageal cancer, gastro-oesophageal cancer, cervical cancer, hepatocellular carcinoma, endometrial cancer, urothelial cancer, or ovarian cancer.
  • According to one embodiment disclosed in the present invention, the tumor may be early stage tumor, metastatic tumor, non-metastatic tumor, primary tumor, resected tumor, advanced tumor, locally advanced tumor, unresectable tumor, tumor in remission, or recurrent tumor.
  • Preferably, the composition may be for injectable (e.g. intraperitoneal, intravenous, and intra-arterial, etc.) and may include the active ingredient in an amount of 0.001 mg/kg to 1000 mg/kg, preferably 0.01 mg/kg to 100 mg/kg, more preferably 0.1 mg/kg to 50 mg/kg for human.
  • For examples, dosages per day normally fall within the range of about 0.001 to about 1000 mg/kg of body weight. In the treatment of adult humans, the range of about 0.1 to about 50 mg/kg/day, in single or divided dose, is especially preferred. However, it will be understood that the concentration of the iCP-SOCS3 recombinant protein actually administered will be determined by a physician, in the light of the relevant circumstances, including the condition to be treated, the chosen route of administration, the age, weight, and response of the individual patient, and the severity of the patient's symptoms, and therefore the above dosage ranges are not intended to limit the scope of the invention in any way. In some instances dosage levels below the lower limit of the aforesaid range may be more than adequate, while in other cases still larger doses may be employed without causing any harmful side effect, provided that such larger doses are first divided into several smaller doses for administration throughout the day.
  • Still another aspect disclosed in the present application provides use of the iCP-SOCS3 recombinant protein as a medicament for treating, preventing, or delaying the onset of, solid tumor.
  • Still another aspect disclosed in the present application provides a medicament including the iCP-SOCS3 recombinant protein.
  • Still another aspect disclosed in the present application provides use of the iCP-SOCS3 recombinant protein for the preparation of a medicament for treating, preventing, or delaying the onset of, solid tumor.
  • Still another aspect disclosed in the present application provides a method of treating, preventing, or delaying the onset of, solid tumor in a subject including identifying a subject in need of treating, preventing, or delaying the onset of, solid tumor; and administering to the subject a therapeutically effective amount of the iCP-SOCS3 recombinant protein.
  • In one embodiment disclosed in the present application, the subject may be preferably a mammal.
  • Preferably, the subject in need of treating, preventing, or delaying the onset of, solid tumor may be identified by any conventional diagnostic methods known in the art including ultrasound, CT scan, MRI, alpha-fetoprotein testing, and biopsy, etc.
  • The pharmaceutical composition according to one embodiment disloced in the present application may be prepared by using pharmaceutically suitable and physiologically acceptable additives, in addition to the active ingredient, and the additives may include excipients, disintegrants, sweeteners, binders, coating agents, blowing agents, lubricants, glidants, flavoring agents, etc.
  • For administration, the pharmaceutical composition may be preferably formulated by further including one or more pharmaceutically acceptable carriers in addition to the above-described active ingredient.
  • Dosage forms of the pharmaceutical composition may include granules, powders, tablets, coated tablets, capsules, suppositories, liquid formulations, syrups, juice, suspensions, emulsions, drops, injectable liquid formulations, etc. For formulation of the composition into a tablet or capsule, for example, the active ingredient may be combined with any oral, non-toxic pharmaceutically acceptable inert carrier, such as ethanol, glycerol, water, etc. If desired or necessary, suitable binders, lubricants, disintegrants, and colorants may be additionally included as a mixture.
  • Examples of the suitable binder may include, but are not limited to, starch, gelatin, natural sugars such as glucose or beta-lactose, corn sweeteners, natural and synthetic gums such as acacia, tragacanth, or sodium oleate, sodium stearate, magnesium stearate, sodium benzoate, sodium acetate, sodium chloride, etc. Examples of the disintegrant may include, but are not limited to, starch, methyl cellulose, agar, bentonite, xanthan gum, etc. For formulation of the composition into a liquid preparation, a pharmaceutically acceptable carrier which is sterile and biocompatible may be used, such as saline, sterile water, a Ringer's solution, buffered saline, an albumin infusion solution, a dextrose solution, a maltodextrin solution, glycerol, and ethanol, and these materials may be used alone or in any combination thereof. If necessary, other common additives, such as antioxidants, buffers, bacteriostatic agents, etc., may be added. Further, diluents, dispersants, surfactants, binders, and lubricants may be additionally added to prepare injectable formulations such as aqueous solutions, suspensions, and emulsions, or pills, capsules, granules, or tablets. Furthermore, the composition may be preferably formulated, depending upon diseases and ingredients, using any appropriate method known in the art, as disclosed in Remington's Pharmaceutical Science, Mack Publishing Company, Easton Pa.
  • Preferably, the treatment or treating mean improving or stabilizing the subject's condition or disease; or preventing or relieving the development or worsening of symptoms associated with the subject's condition or disease. The prevention, prophylaxis and preventive treatment are used herein as synonyms.
  • Preferably, the treating, preventing, or delaying the onset of, solid tumor may be any one or more of the following: alleviating one or more symptoms of tumor, delaying progressing of tumor, shrinking tumor size in patient, inhibiting tumor growth, prolonging overall survival, prolonging disease-free survival, prolonging time to disease progression, preventing or delaying tumor metastasis, reducing or eradiating preexisting tumor metastasis, reducing incidence or burden of preexisting tumor metastasis, preventing recurrence of tumor.
  • The subject and patient are used herein interchangeably. They refer to a human or another mammal (e.g., mouse, rat, rabbit, dog, cat, cattle, swine, sheep, horse or primate) that can be afflicted with or is susceptible to a disease or disorder but may or may not have the disease or disorder. In certain embodiments, the subject is a human being.
  • Preferably, the amount effective or effective amount is the amount of an active ingredient or a pharmaceutical composition disclosed herein that when administered to a subject for treating a disease, is sufficient to effect such treatment of the disease. Any improvement in the patient is considered sufficient to achieve treatment. An effective amount of an active ingredient or a pharmaceutical composition disclosed herein, used for the preventing, or delaying the onset of, solid tumor may vary depending upon the manner of administration, the age, body weight, and general health of the patient. Ultimately, the prescribers or researchers will decide the appropriate amount and dosage regimen.
  • In the treatment or prevention method according to one embodiment disclosed in the present application, the composition including the iCP-SOCS3 recombinant protein as an active ingredient may be administered in a common manner via oral, buccal, rectal, intravenous, intra-arterial, intraperitoneal, intramuscular, intrasternal, percutaneous, topical, intraocular or subcutaneous route, more preferably via intraperitoneal, intravenous, or intra-arterial injection route.
  • Advantageous Effects
  • According to one aspect disclosed in the present application, development and establishment of improved cell-permeable SOCS3 recombinant protein, as therapeutics of solid tumor are provided. Because iCP-SOCS3 was designed based on endogenous proteins, it would be a safety anti-solid tumor drug without side-effect.
  • However, the effects of the disclosures in the present application are not limited to the above-mentioned effects, and another effects not mentioned will be clearly understood by those skilled in the art from the following description.
  • DESCRIPTION OF DRAWINGS
  • FIG. 1 shows Structure of aMTD- or rPeptide-Fused Recombinant Proteins. A schematic diagram of the His-tagged CRA recombinant proteins is illustrated and constructed according to the present invention. The his-tag for affinity purification (white), aMTD or rPeptide (gray) and cargo A (CRA, black) are shown.
  • FIG. 2a shows Construction of Expression Vectors for aMTDs- or rPeptide-Fused Recombinant Proteins. FIGS. 2b and 2c show the agarose gel electrophoresis analysis showing plasmid DNA fragments at 645 bp insert encoding aMTDs or rPeptide-fused CRA cloned into the pET28a(+) vector according to the present invention.
  • FIGS. 3a to 3d show Inducible Expression of aMTD- or rPeptide-Fused Recombinant Proteins. Expressed recombinant aMTD- or random peptide-fused CRA recombinant proteins were transformed in E. coli BL21 (DE3) strain. Expression of recombinant proteins in E. coli before (−) and after (+) induction with IPTG was monitored by SDS-PAGE, and stained with Coomassie blue.
  • FIGS. 4a and 4b show Purification of aMTD- or rPeptide-Fused Recombinant Proteins. Expressed recombinant proteins were purified by Ni2+ affinity chromatography under the natural condition. Purification of recombinant proteins displayed through SDS-PAGE analysis.
  • FIGS. 5a to 5u show Determination of aMTD-Mediated Cell-Permeability. Cell-permeability of a negative control (A: rP38) and reference hydrophobic CPPs (MTM12 and MTD85) are shown. The cell-permeability of each aMTD and/or rPeptide is visually compared to that of the cargo protein lacking peptide sequence (HCA). Gray shaded area represents untreated RAW 264.7 cells (vehicle); thin light gray line represents the cells treated with equal molar concentration of FITC (FITC only); dark thick line indicates the cells treated with FITC-his-tagged CRA protein (HCA); and the cells treated with the FITC-proteins (HMCA) fused to negative control (rP38), reference CPP (MTM12 or MTD85) or new hydrophobic CPP (aMTD) are shown with light thick line and indicated by arrows.
  • FIGS. 6a to 6c show Determination of rPeptide-Mediated Cell-Permeability. The cell-permeability of each aMTD and/or rPeptide was visually compared to that of the cargo protein lacking peptide sequence (HCA). Gray shaded area represents untreated RAW 264.7 cells (vehicle); thin light gray line represents the cells treated with equal molar concentration of FITC (FITC only); dark thick line indicates the cells treated with FITC-his-tagged CRA protein (HCA); and the cells treated with the FITC-proteins fused to rPeptides are shown with light thick line and indicated by arrows.
  • FIGS. 7a to 7k shows Visualized Cell-Permeability of aMTD-Fused Recombinant Proteins. NIH3T3 clls were treated with FITC-labeled protein (10 μM) fused to aMTD for 1 hour at 37. Cell-permeability of the proteins was visualized by laser scanning confocal microscopy (LSM700 version).
  • FIG. 8 shows Visualized Cell-Permeability of rPeptide-Fused Recombinant Proteins. Cell-permeability of rPeptide-fused recombinant proteins was visualized by laser scanning confocal microscopy (LSM700 version).
  • FIGS. 9a to 9c show Relative Cell-Permeability of aMTD-Fused Recombinant Proteins Compared to Negative Control (rP38). The FIG. shows graphs comparing the cell-permeability of the recombinant proteins fused to aMTDs and a negative control (A: rP38).
  • FIGS. 10a to 10c show Relative Cell-Permeability of aMTD-Fused Recombinant Proteins Compared to Reference CPP (MTM12). The FIG. shows graphs comparing the cell-permeability of the recombinant proteins fused to aMTDs and a reference CPP (MTM12).
  • FIGS. 11a to 11c show Relative Cell-Permeability of aMTD-Fused Recombinant Proteins Compared to Reference CPP (MTD85). The FIG. shows graphs comparing the cell-permeability of the recombinant proteins fused to aMTDs and a reference CPP (MTD85).
  • FIG. 12 shows Relative Cell-Permeability of rPeptide-Mediated Recombinant Proteins Compared to Average that of aMTDs. The FIG. shows graphs comparing the cell-permeability of the recombinant proteins fused to rPeptides and that (average value: aMTD AVE) of aMTDs.
  • FIGS. 13a and 13b show Association of Cell-Permeability with Amino Acid Composition in aMTD Sequences. These graphs display delivery potential (Geometric Mean) of aMTDs influenced with amino acid composition (A, I, V and L).
  • FIGS. 14a and 14b show Association of Cell-Permeability with Critical Factors in aMTDs. These graphs show the association of cell-permeability with critical factors [bending potential: proline position (PP), rigidity/flexibility: instability index (II), structural feature: aliphatic index (AI) and hydropathy: grand average of hydropathy (GRAVY)].
  • FIGS. 15a and 15b show Relative Relevance of aMTD-Mediated Cell-Permeability with Critical Factors. Cell-permeability of 10 high and 10 low ranked aMTDs in their delivery potential were examined for their association with the critical factors [bending potential: proline position (PP), rigidity/flexibility: instability index (II), structural feature: aliphatic index (AI) and hydropathy: grand average of hydropathy (GRAVY)].
  • FIG. 16 shows Relative Relevance of rPeptide-Mediated Cell-Permeability with Hydropathy Range (GRAVY). This graph and a chart illustrate relative relevance of rPeptide-mediated cell-permeability with its hydropathy range (GRAVY).
  • FIG. 17 shows a structure of iCP-SOCS3 recombinant protein designed according to example 6-1.
  • FIG. 18 shows the agarose gel electrophoresis analysis showing plasmid DNA fragments insert encoding His-SOCS3-SDB (HS3B), His-aMTD165-SOC53-SDB (HM165S3B), His-aMTD165-SOCS3-SDC (HM165S3C), His-aMTD165-SOC53-SDD (HM165S3D), His-aMTD165-SOC53-SDE (HM165S3E) cloned into the pET28a (+) vector according to example 6-1.
  • FIG. 19 shows inducible expression and purification of iCP-SOCS3 recombinant protein in E. coli according to example 6-2 and improvement of solubility/yield of iCP-SOCS3 recombinant protein by fusing aMTD/SD according to example 6-3.
  • FIG. 20 shows aMTD-Mediated cell-permeability of SOCS3 recombinant proteins in RAW 264.7 cells according to example 7-1.
  • FIG. 21 shows aMTD-Mediated intracellular delivery and localization of SOCS3 Recombinant Proteins in NIH3T3 cells cells according to example 7-1.
  • FIG. 22 shows systemic delivery of aMTD/SD-fused SOCS3 recombinant proteins in vivo according to example 7-2.
  • FIG. 23 shows inhibition of IFN-γ-induced STAT phosphorylation by iCP-SOCS3 recombinant protein according to example 8-1.
  • FIG. 24 shows inhibition of LPS-induced cytokines secretion by iCP-SOCS3 recombinant protein according to example 8-2.
  • FIG. 25 shows the structures of SOCS3 recombinant protein lacking aMTD prepared as a negative control according to example 9.
  • FIG. 26 shows expression, purification, and solubility/yield of HS3 (lacking aMTD and SD) and HS3B (lacking aMTD) determined according to example 6-3.
  • FIG. 27 shows the agarose gel electrophoresis analysis showing plasmid DNA fragments insert encoding His-aMTD#-SOCS3-SDB (HM#S3B) and His-rP#-SOCS3-SDB cloned into the pET28a (+) vector according to example 10.
  • FIGS. 28a and 28b show expression, purification, and solubility/yield of His-aMTD#-SOCS3-SDB (HM#S3B) determined according to example 10.
  • FIG. 29 shows expression, purification, and solubility/yield of His-rP#-SOCS3-SDB (HrP#S3B) determined according to example 10.
  • FIG. 30 shows solubility/yield of His-aMTD#-SOCS3-SDB (HM#S3B) determined according to example 10.
  • FIG. 31 show aMTD-mediated cell-permeability. The cell-permeability of each SOCS3 recombinant protein fused with SD and various aMTD is visually compared to that of the cargo protein lacking CPP (HS3B) or lacking CPP and SD (HS3). Gray shaded area represents untreated E. coli cells (diluent); green line represents the cells treated with equal molar concentration of FITC (FITC only); black line indicates the cells treated with FITC-his-SOCS protein (FITC-HS3); blue line indicates the cells treated with FITC-his-SOCS-SDB protein (FITC-HS3B) purple line indicates the cells treated with FITC-his-aMTD#-SOCS-SDB protein (FITC-HM#S3B).
  • FIG. 32 shows relative cell-permeability of His-aMTD#-SOCS3-SDB-Fused recombinant proteins Compared to control (Vehicle, FITC only, HS3 and HS3B).
  • FIG. 33 shows random Peptide-Mediated cell-permeability. The cell-permeability of each SOCS3 recombinant protein fused with SDB and aMTD165 or various rP is visually compared to that of the cargo protein lacking CPP (HS3B) or lacking CPP and SD (HS3). Gray shaded area represents untreated E. coli cells (diluent); green line represents the cells treated with equal molar concentration of FITC (FITC only); black line indicates the cells treated with FITC-his-SOCS protein (FITC-HS3); blue line indicates the cells treated with FITC-his-SOCS-SDB protein (FITC-HS3B) and purple line indicates the cells treated with FITC-his-rPeptide#-SOCS-SDB protein (FITC-HrP#S3B).
  • FIG. 34 shows relative cell-permeability of His-rP#-SOCS3-SDB-Fused recombinant proteins Compared to control (Vehicle, FITC only, HS3 and HS3B).
  • FIG. 35 shows apoptotic cells analysis according to example 11-1.
  • FIG. 36 shows induction of apoptosis by iCP-SOCS3 recombinant proteins according to example 11-2.
  • FIG. 37 shows cell migration inhibition by iCP-SOCS3 recombinant protein according to example 11-3.
  • FIG. 38 shows solubility/yield, permeability and biological activity of His-aMTD#-SOCS3-SDB (HM#S3B) determined according to example 10 to 11-3.
  • FIG. 39 shows expression, purification, and solubility/yield of M165S3SB (lacking his-tag) determined according to example 12-1.
  • FIG. 40 shows cell-permeability of SOCS3 recombinant proteins (lacking his-tag) in RAW 264.7 cells according to example 12-2.
  • FIG. 41 shows Annexin V analysis according to example 12-3.
  • FIG. 42 shows cell migration inhibition (bottom) by iCP-SOCS3 recombinant protein according to example 12-3.
  • FIG. 43 shows a structure of iCP-SOCS3 recombinant protein (His-aMTD165-50C53-SDB′) constructed according to example 12-4.
  • FIG. 44 shows expression, purification, and solubility/yield of HM165S3SB and HM165S3SB′ determined according to example 12-4.
  • FIG. 45 shows aMTD-Mediated cell-permeability of iCP-SOCS3 recombinant proteins (HM165S3B and HM165S3B′(V28L)) in RAW 264.7 cells according to example 12-5.
  • FIG. 46 shows antiproliferative activity of iCP-SOCS3 recombinant proteins (HM165S3B and HM165S3B′(V28L)) according to example 12-6.
  • FIG. 47 shows induction of apoptosis by iCP-SOCS3 recombinant proteins (HM165S3B and HM165S3B′(V28L)) according to example 12-6.
  • FIG. 48 shows cell migration inhibition by SOCS3 recombinant proteins (HM165S3B and HM165S3B′(V28L)) according to example 12-6.
  • FIG. 49a shows a structure of iCP-SOCS3 recombinant proteins (top) and agarose gel electrophoresis analysis (bottom) according to example 12-7 and FIG. 49b shows inducible expressions and purifications of iCP-SOCS3 recombinant protein in E. coli (bottom) according to example 12-7.
  • FIG. 50 shows inhibition of IFN-γ-induced STAT phosphorylation by iCP-SOCS3 recombinant protein according to example 13.
  • FIG. 51 shows effect of treating EDTA (FIG. 51A) and proteinase K (FIG. 51B) on aMTD-mediated SOCS3 protein uptake into cells according to example 14.
  • FIG. 52 shows effect of treating taxol (FIG. 52 A) and antimycin (FIG. 52 B) on aMTD-mediated SOCS3 protein uptake into cells according to example 14.
  • FIG. 53 shows effect of temperature on aMTD-mediated SOCS3 protein uptake into cells according to example 14.
  • FIG. 54 shows aMTD-mediated cell-to-cell delivery assessed according to example 14.
  • FIG. 55 shows bioavailability of iCP-SOCS3 recombinant protein in PBMC, splenocytes and hepatocytes analyzed by fluorescence microscopy according to example 15.
  • FIG. 56 shows biodistribution of iCP-SOCS3 recombinant protein in pancreas tissues analyzed by confocal microscope according to example 15.
  • FIG. 57 shows aMTD-Mediated cell-permeability of SOCS3 recombinant proteins in various cancer cells according to example 16.
  • FIG. 58 shows systemic delivery of aMTD/SD-fused SOCS3 recombinant proteins into Various Tissues according to example 16.
  • FIG. 59 shows expression level of endogenous SOCS3 mRNA in gastric cell line analyzed by the agarose gel electrophoresis according to example 17-1-2.
  • FIG. 60 shows expression level of SOCS3 gene, and phosphorylation of JAK1 and JAK2 in gastric cancer cell line analyzed by western blot analysis according to example 17-1-3.
  • FIG. 61 shows expression level of endogenous SOCS3 mRNA in colon cancer cell line analyzed by the agarose gel electrophoresis according to example 17-2-2.
  • FIG. 62 shows expression level of SOCS3 gene, and phosphorylation of JAK1 and JAK2 in colon cancer cell line analyzed by western blot analysis according to example 17-2-3.
  • FIG. 63 shows methylation and unmethylation level of endogenous SOCS3 in glioblastoma cell line analyzed by the agarose gel electrophoresis according to example 17-3-1.
  • FIG. 64 shows expression level of SOCS3 gene, and phosphorylation of JAK1 and JAK2 in glioblastoma cancer cell line analyzed by western blot analysis according to example 17-3-2.
  • FIG. 65 shows expression level of endogenous SOCS3 mRNA in breast cancer cell line analyzed by the agarose gel electrophoresis according to example 17-4-1.
  • FIG. 66 shows methylation and unmethylation level of endogenous SOCS3 in breast cancer cell line analyzed by the agarose gel electrophoresis according to example 17-4-2.
  • FIG. 67 shows expression level of SOCS3 gene, and phosphorylation of JAK1 and JAK2 in breast cancer cell line analyzed by western blot analysis according to example 17-4-3.
  • FIG. 68 shows antiproliferative activity of iCP-SOCS3 recombinant protein in gastric cancer cell line according to example 18-1.
  • FIG. 69 shows antiproliferative activity of iCP-SOCS3 recombinant protein in colorectal cancer cell line according to example 18-1.
  • FIG. 70 shows antiproliferative activity of iCP-SOCS3 recombinant protein in glioblastoma cell line according to example 18-1.
  • FIG. 71 shows antiproliferative activity of iCP-SOCS3 recombinant protein in breast cancer cell line according to example 18-1.
  • FIG. 72 shows cell migration inhibition activity by iCP-SOCS3 recombinant protein in gastric cancer cell line (AGS) according to example 18-2.
  • FIG. 73 shows cell migration inhibition activity by iCP-SOCS3 recombinant protein in colorectal cancer cell line (HCT116) according to example 18-2.
  • FIG. 74 shows cell migration inhibition activity by iCP-SOCS3 recombinant protein in glioblastoma cell line (U-87 MG) according to example 18-2.
  • FIG. 75 shows cell migration inhibition activity by iCP-SOCS3 recombinant protein in breast cancer cell line (MDA-MB-231) according to example 18-2.
  • FIG. 76 show transwell migration inhibition activity by iCP-SOCS3 recombinant protein in gastric cancer cell line (AGS) (FIG. 76, left), and decrease in invasion by iCP-SOCS3 recombinant protein in gastric cancer cell line (AGS) (FIG. 76, right) analyzed according to example 18-2.
  • FIG. 77 show transwell migration inhibition activity by iCP-SOCS3 recombinant protein in glioblstoma cell line (U-87 MG) analyzed according to example 18-2.
  • FIGS. 78 to 82 show induction of apoptosis in normal cells (FIG. 78), in gastric cancer cells (FIG. 79), in colorectal cancer cells (HCT116, FIG. 80), in glioblastoma cells (U-87 MG, FIG. 81), and in breast cancer cells (MDA-MB-231, FIG. 82) by iCP-SOCS3 recombinant proteins assessed by Annexin V staining according to example 18-3.
  • FIG. 83 show induction of apoptosis in colon cancer cells (HCT116) by iCP-SOCS3 recombinant proteins analyzed by TUNEL assay according to example 18-4.
  • FIGS. 84 to 88 show inhibition of cell cycle progression in normal cells (FIG. 84), in gastric cancer cells (AGS, FIG. 85), in colon cancer cells (HCT116, FIG. 86), in glioblastoma cells (U-87 MG, FIG. 87), and in breast cancer cells (MDA-MB-231, FIG. 88) by iCP-SOCS3 recombinant proteins assessed by flow cytometric analysis according to example 18-5.
  • FIG. 89 shows down regulation of expression of apoptosis by iCP-SOCS3 recombinant proteins in breast cancer cells (MDA-MB-231) determined by immunoblotting according to example 18-6.
  • FIG. 90 shows suppression of the tumor growth by iCP-SOCS3 recombinant proteins in gastric cancer cells (NCI-N87) assessed according to example 19-1.
  • FIG. 91 shows suppression of the tumor growth by iCP-SOCS3 recombinant proteins in colorectal cancer cells (HCT116) assessed according to example 19-2.
  • FIG. 92 shows suppression of the tumor growth by iCP-SOCS3 recombinant proteins in glioblastoma cells (U-87 MG) assessed according to example 19-3.
  • FIG. 93 shows suppression of the tumor growth by iCP-SOCS3 recombinant proteins in breast cancer cells (MDA-MB-231) assessed according to example 19-4.
  • FIG. 94 shows humanized SDB domain according to example 12-4.
  • FIG. 95 shows methylation and unmethylation level of endogenous SOCS3 in gastric cancer cell line analyzed by the agarose gel electrophoresis according to example 17-1-1.
  • FIG. 96 shows methylation and unmethylation level of endogenous SOCS3 in colon cancer cell line analyzed by the agarose gel electrophoresis according to example 17-2-1.
  • FIG. 97 shows sequences of amino acid and nucleotide of basic CPP, and primers used in example 6-4.
  • FIG. 98 shows structure, expression, purification and solubility/yield of aMTD/SD-fused SOCS3 recombinant protein and basic CPP/SD-fused SOCS3 recombinant protein according to example 6-4.
  • FIG. 99 shows comparison of cell-permeability between aMTD/SD fused SOCS3 recombinant proteins and basic/SD-fused in RAW 264.7 cells according to example 7-1-2.
  • FIG. 100 shows comparison of tissue-permeability between aMTD/SD fused SOCS3 recombinant proteins and basic/SD-fused in various tissues of ICR mice according to example 7-2-2.
  • FIG. 101 shows effect of treating protein K (FIG. 101 A) and Taxol (FIG. 101 B) on aMTD (or basic CPP)-mediated SOCS3 protein uptake into cells according to example 14-2.
  • FIGS. 102 and 103 show aMTD (or basic CPP)-mediated cell-to-cell delivery (FIG. 102) and cell-to-cell function assessed according to example 14-2.
  • MODE FOR INVENTION 1. Analysis of Reference Hydrophobic CPPs to Identify ‘Critical Factors’ for Development of Advanced MTDs
  • Previously reported MTDs were selected from a screen of more than 1,500 signal peptide sequences. Although the MTDs that have been developed did not have a common sequence or sequence motif, they were all derived from the hydrophobic (H) regions of signal sequences (HRSSs) that also lack common sequences or motifs except their hydrophobicity and the tendency to adopt alpha-helical conformations. The wide variation in H-region sequences may reflect prior evolution for proteins with membrane translocating activity and subsequent adaptation to the SRP/Sec61 machinery, which utilizes a methionine-rich signal peptide binding pocket in SRP to accommodate a wide-variety of signal peptide sequences.
  • Previously described hydrophobic CPPs (e.g. MTS/MTM and MTD) were derived from the hydrophobic regions present in the signal peptides of secreted and cell surface proteins. The prior art consists first, of ad hoc use of H-region sequences (MTS/MTM), and second, of H-region sequences (with and without modification) with highest CPP activity selected from a screen of 1,500 signal sequences (MTM). Second prior art, the modified H-region derived hydrophobic CPP sequences had advanced in diversity with multiple number of available sequences apart from MTS/MTM derived from fibroblast growth factor (FGF) 4. However, the number of MTDs that could be modified from naturally occurring secreted proteins are somewhat limited. Because there is no set of rules in determining their cell-permeability, no prediction for the cell-permeability of modified MTD sequences can be made before testing them.
  • The hydrophobic CPPs, like the signal peptides from which they originated, did not conform to a consensus sequence, and they had adverse effects on protein solubility when incorporated into protein cargo. We therefore set out to identify optimal sequence and structural determinants, namely critical factors (CFs), to design new hydrophobic CPPs with enhanced ability to deliver macromolecule cargoes including proteins into the cells and tissues while maintaining protein solubility. These newly developed CPPs, advanced macromolecule transduction domains (aMTDs) allowed almost infinite number of possible designs that could be designed and developed based on the critical factors. Also, their cell-permeability could be predicted by their character analysis before conducting any in vitro and/or in vivo experiments. These critical factors below have been developed by analyzing all published reference hydrophobic CPPs.
  • 1-1. Analysis of Hydrophobic CPPs
  • Seventeen different hydrophobic CPPs (Table 1) published from 1995 to 2014 (Table 2) were selected. After physiological and chemical properties of selected hydrophobic CPPs were analyzed, 11 different characteristics that may be associated with cell-permeability have been chosen for further analysis. These 11 characteristics are as follows: sequence, amino acid length, molecular weight, pI value, bending potential, rigidity/flexibility, structural feature, hydropathy, residue structure, amino acid composition and secondary structure of the sequences (Table 3).
  • Table 1 shows the summary of published hydrophobic Cell-Penetrating Peptides which were chosen.
  • TABLE 1
    # Pepides Origin Protein Ref.
    1 MTM Homo sapiens NP_001998 Kaposi fibroblast growth factor (K-FGF) 1
    2 MTS Homo sapiens NP_001998 Kaposi fibroblast growth factor (K-FGF) 2
    3 MTD10 Streptomyces coelicolor NP_625021 Glycosyl hydrolase 8
    4 MTD13 Streptomyces coelicolor NP_639877 Putative secreted protein 3
    5 MTD47 Streptomyces coelicolor NP_627512 Secreted protein 4
    6 MTD56 Homo sapiens P23274 Peptidyl-prolyl cis-trans isomerase B precursor 5
    7 MTD73 Drosophila melanogaster AAA17887 Spatzle (spz) protein 5
    8 MTD77 Homo sapiens NP_003231 Kaposi fibroblast growth factor (K-FGF) 6
    9 MTD84 Phytophthora cactorum AAK63068 Phytotoxic protein PcF precusor 4
    10 MTD85 Streptomyces coelicolor NP_629842 Peptide transport system peptide binding 7
    protein
    11 MTD86 Streptomyces coelicolor NP_629842 Peptide transport system secreted peptide 7
    binding protein
    12 MTD103 Homo sapiens TMBV19 domain Family member B 8
    13 MTD132 Streptomyces coelicolor NP_628377 P60-family secreted protein 4
    14 MTD151 Streptomyces coelicolor NP_630126 Secreted chitinase 8
    15 MTD173 Streptomyces coelicolor NP_624384 Secreted protein 4
    16 MTD174 Streptomyces coelicolor NP_733505 Large, multifunctional secreted protein 8
    17 MTD181 Neisseria meningitidis Z2491 CAB84257.1 Putative secreted protein 4
  • Table 2 summarizes reference information
  • TABLE 2
    References
    # Title Journal Year Vol Issue Page
    1 Inhibition of Nuclear Translocation of Transcription Factor JOURNAL OF 1995 270 24 14255
    NF-kB by a Synthetic peptide Containing a Cell Membrane- BIOLOGICAL
    permeable Motif and Nuclear Localization Sequence CHEMISTRY
    2 Epigenetic Regulation of Gene Structure and Function with NATURE 2001 19 10 929
    a Cell-Permeable Cre Recombinase BIOTECHNOLOGY
    3 Cell-Permeable NM23 Blocks the Maintenance and CANCER 2011 71 23 7216
    Progression of Established Pulmonary Metastasis RESEARCH
    4 Antitumor Activity of Cell-Permeable p18INK4c With MOLECULAR 2012 20 8 1540
    Enhanced Membrane and Tissue Penetration THERAPY
    5 Antitumor Activity of Cell-Permeable RUNX3 Protein in CLINICAL 2012 19 3 680
    Gastric Cancer Cells CANCER
    RESEARCH
    6 The Effect of Intracellular Protein Delivery on the Anti- BIOMATERIALS 2013 34 26 6261
    Tumor Activity of Recombinant Human Endostatin
    7 Partial Somatic to Stem Cell Transformations Induced By SCIENTIFIC 2014 4 10 4361
    Cell-Permeable Reprogramming Factors REPORTS
    8 Cell-Permeable Parkin Proteins Suppress Parkinson PLOS ONE 2014 9 7 17
    Disease-Associated Phenotypes in Cultured Cells and
    Animals
  • Table 3 shows characteristics of published hydrophobic Cell-Penetrating Peptides (A) which were analyzed.
  • TABLE 3
    Rigidity/ Structural
    Flexibility Feature
    Molecular Bending (Instability (Aliphatic
    # Peptides Sequence Length Weight pI Potential Index: II) Index: AI)
    1 MTM AAVALLPAVLLALLAP 16 1,515.9 5.6 Bending 45.5 220.0
    2 MTS AAVLLPVLLAAP 12 1,147.4 5.6 Bending 57.3 211.7
    3 MTD10 LGGAVVAAPVAAAVAP 16 1,333.5 5.5 Bending 47.9 140.6
    4 MTD13 LAAAALAVLPL 11 1,022.3 5.5 Bending 26.6 213.6
    5 MTD47 AAAVPVLVAA 10 881.0 5.6 Bending 47.5 176.0
    6 MTD56 VLLAAALIA 9 854.1 5.5 No- 8.9 250.0
    Bending
    7 MTD73 PVLLLLA 7 737.9 6.0 No- 36.1 278.6
    Bending
    8 MTD77 AVALLILAV 9 882.1 5.6 No- 30.3 271.1
    Bending
    9 MTD84 AVALVAVVAVA 11 982.2 5.6 No- 9.1 212.7
    Bending
    10 MTD85 LLAAAAALLLA 11 1,0102 5.5 No- 9.1 231.8
    Bending
    11 MTD86 LLAAAAALLLA 11 1,010.2 5.5 No- 9.1 231.8
    Bending
    12 MTD103 LALPVLLLA 9 922.2 5.5 Bending 51.7 271.1
    13 MTD132 AVVVPAIVLAAP 12 1,119.4 5.6 Bending 50.3 195.0
    14 MTD151 AAAPVAAVP 9 1,031.4 5.5 Bending 73.1 120.0
    15 MTD173 AVIPILAVP 9 892.1 5.6 Bending 48.5 216.7
    16 MTD174 LILLLPAVALP 12 1,011.8 5.5 Bending 79.1 257.3
    17 MTD181 AVLLLPAAA 9 838.0 5.6 Bending 51.7 206.7
    AVE 10.8 ± 2.4 1,011 ± 189.6 5.6 ± 0.1 Proline 40.1 ± 21.9 217.9 ± 43.6
    Presence
    Hydro- A/a
    pathy Residue Composition Secondary
    # Peptides Sequence (GRAVY) Structure A V L I P G Structure Cargo Ref.
    1 MTM AAVALLPAVLLALLAP 2.4 Aliphatic 6 2 6 0 2 0 Helix p50 1
    Ring
    2 MTS AAVLLPVLLAAP 2.3 Aliphatic 4 2 4 0 2 0 No-Helix CRE 2
    Ring
    3 MTD10 LGGAVVAAPVAAAVAP 1.8 Aliphatic 7 4 1 0 2 2 Helix Parkin 8
    Ring
    4 MTD13 LAAAALAVLPL 2.4 Aliphatic 5 1 4 0 1 0 No-Helix RUNX3 3
    Ring
    5 MTD47 AAAVPVLVAA 2.4 Aliphatic 5 3 1 0 1 0 No-Helix CMYC 4
    Ring
    6 MTD56 VLLAAALIA 3.0 Aliphatic 4 1 3 1 0 0 Helix ES 5
    Ring
    7 MTD73 PVLLLLA 2.8 Aliphatic 1 1 4 0 1 0 Helix ES 5
    Ring
    8 MTD77 AVALLILAV 3.3 Aliphatic 3 2 3 1 0 0 Helix NM23 6
    Ring
    9 MTD84 AVALVAVVAVA 3.1 Aliphatic 5 5 1 0 0 0 Helix OCT4 4
    Ring
    10 MTD85 LLAAAAALLLA 2.7 Aliphatic 6 0 5 0 0 0 No-Helix RUNX3 7
    Ring
    11 MTD86 LLAAAAALLLA 2.7 Aliphatic 6 0 5 0 0 0 No-Helix SOX2 7
    Ring
    12 MTD103 LALPVLLLA 2.8 Aliphatic 2 1 5 0 1 0 Helix p18 8
    Ring
    13 MTD132 AVVVPAIVLAAP 2.4 Aliphatic 4 4 1 1 2 0 No-Helix LIN28 4
    Ring
    14 MTD151 AAAPVAAVP 1.6 Aliphatic No-Helix Parkin 8
    Ring
    15 MTD173 AVIPILAVP 2.4 Aliphatic 2 2 1 2 2 0 Helix KLF4 4
    Ring
    16 MTD174 LILLLPAVALP 2.6 Aliphatic Helix Parkin 8
    Ring
    17 MTD181 AVLLLPAAA 2.4 Aliphatic 4 1 3 0 1 0 No-Helix SOX2 4
    Ring
    AVE 2.5 ± 0.4
  • Two peptide/protein analysis programs were used (ExPasy: SoSui: http://harrier.nagahama-i-bio.ac.jp/sosui/sosui_submit.html) to determine various indexes and structural features of the peptide sequences and to design new sequence. Followings are important factors analyzed.
  • 1-2. Characteristics of Analyzed Peptides: Length, Molecular Weight and Pl Value
  • Average length, molecular weight and pl value of the peptides analyzed were 10.8±2.4, 1,011±189.6 and 5.6±0.1, respectively (Table 4)
  • Table 4 summarizes Critical Factors (CFs) of published hydrophobic Cell-Penetrating Peptides (A) which were analyzed.
  • TABLE 4
    Length: 10.8 ± 2.4
    Molecular Weight: 1,011 ± 189.6
    pI: 5.6 ± 0.1
    Bending Potential (BP): Proline presences in the middle and/or the end of
    peptides, or No Proline.
    Instability Index (II): 40.1 ± 21.9
    Residue Structure & Aliphatic Index (AI): 217.9 ± 43.6
    Hydropathy (GRAVY): 2.5 ± 0.4
    Aliphatic Ring: Non-polar hydrophobic & aliphatic amino acid
    (A, V, L, I).
    Secondary Structure: α-Helix is favored but not required.
  • 1-3. Characteristics of Analyzed Peptides: Bending Potential—Proline Position (PP)
  • Bending potential (bending or no-bending) was determined based on the fact whether proline (P) exists and/or where the amino acid(s) providing bending potential to the peptide in recombinant protein is/are located. Proline differs from the other common amino acids in that its side chain is bonded to the backbone nitrogen atom as well as the alpha-carbon atom. The resulting cyclic structure markedly influences protein architecture which is often found in the bends of folded peptide/protein chain.
  • Eleven out of 17 were determined as ‘Bending’ peptide which means that proline is present in the middle of sequence for peptide bending and/or located at the end of the peptide for protein bending. As indicated above, peptide sequences could penetrate the plasma membrane in a “bent” configuration. Therefore, bending or no-bending potential is considered as one of the critical factors for the improvement of current hydrophobic CPPs.
  • 1-4. Characteristics of Analyzed Peptides: Rigidity/Flexibility
  • Instability Index (II) Since one of the crucial structural features of any peptide is based on the fact whether the motif is rigid or flexible, which is an intact physicochemical characteristic of the peptide sequence, instability index (II) of the sequence was determined. The index value representing rigidity/flexibility of the peptide was extremely varied (8.9-79.1), but average value was 40.1±21.9 which suggested that the peptide should be somehow flexible, but not too much rigid or flexible (Table 3).
  • 1-5. Characteristics of Analyzed Peptides: Structural Features
  • structural feature (Aliphatic Index: AI) and hydropathy (Grand Average of Hydropathy: GRAVY)
  • Alanine (V), valine (V), leucine (L) and isoleucine (I) contain aliphatic side chain and are hydrophobic—that is, they have an aversion to water and like to cluster. These amino acids having hydrophobicity and aliphatic residue enable them to pack together to form compact structure with few holes. Analyzed peptide sequence showed that all composing amino acids were hydrophobic (A, V, L and I) except glycine (G) in only one out of 17 (MTD10—Table 3) and aliphatic (A, V, L, I, and P). Their hydropathic index (Grand Average of Hydropathy: GRAVY) and aliphatic index (AI) were 2.5±0.4 and 217.9±43.6, respectively. Their amino acid composition is also indicated in the Table 3.
  • 1-6. Characteristics of analyzed peptides: secondary structure (Helicity)
  • As explained above, the CPP sequences may be supposed to penetrate the plasma membrane directly after inserting into the membranes in a “bent” configuration with hydrophobic sequences having α-helical conformation. In addition, our analysis strongly indicated that bending potential was crucial for membrane penetration. Therefore, structural analysis of the peptides was conducted to determine whether the sequences were to form helix or not. Nine peptides were helix and eight were not (Table 3). It seems to suggest that helix structure may not be required.
  • 1-7. Determination of Critical Factors (CFs)
  • In the 11 characteristics analyzed, the following 6 are selected namely “Critical Factors” for the development of new hydrophobic CPPs—advanced MTDs: amino acid length, bending potential (proline presence and location), rigidity/flexibility (instability index: II), structural feature (aliphatic index: AI), hydropathy (GRAVY) and amino acid composition/residue structure (hydrophobic and aliphatic A/a) (Table 3 and Table 4).
  • 2. Analysis of Selected Hydrophobic CPPs to Optimize ‘Critical Factors’
  • Since the analyzed data of the 17 different hydrophobic CPPs (analysis A, Table 3 and 4) previously developed during the past 2 decades showed high variation and were hard to make common- or consensus-features, analysis B (Table 5 and 6) and C (Table 7 and 8) were also conducted to optimize the critical factors for better design of improved CPPs—aMTDs. Therefore, 17 hydrophobic CPPs have been grouped into two groups and analyzed the groups for their characteristics in relation to the cell permeable property. The critical factors have been optimized by comparing and contrasting the analytical data of the groups and determining the common homologous features that may be critical for the cell permeable property.
  • 2-1. Selective analysis (B) of peptides used to biologically active cargo protein for in vivo
  • In analysis B, eight CPPs were used with each biologically active cargo in vivo. Length was 11±3.2, but 3 out of 8 CPPs possessed little bending potential. Rigidity/Flexibility (instability index: II) was 41±15, but removing one [MTD85: rigid, with minimal II (9.1)] of the peptides increased the overall instability index to 45.6±9.3. This suggested that higher flexibility (40 or higher II) is potentially be better. All other characteristics of the 8 CPPs were similar to the analysis A, including structural feature and hydropathy (Table 5 and 6)
  • Table 5 shows characteristics of published hydrophobic Cell-Penetrating Peptides (B): selected CPPs that were used to each cargo in vivo.
  • TABLE 5
    Structural
    Rigidity/Flexibility Feature
    Molecular Bending (Instability (Aliphatic
    # Peptides Sequence Length Weight pI Potential Index: II) Index: AI)
    1 MTM AAVALLPAVLLALLAP 16 1,515.9 5.6 Bending 45.5 220.0
    2 MTS AAVLLPVLLAAP 12 1,147.4 5.6 Bending 57.3 211.7
    3 MTD10 LGGAVVAAPVAAAVAP 16 1,333.5 5.5 Bending 47.9 140.6
    4 MTD73 PVLLLLA 7 737.9 6.0 No- 36.1 278.6
    Bending
    5 MTD77 AVALLILAV 9 882.1 5.6 No- 30.3 271.1
    Bending
    6 MTD85 LLAAAAALLLA 11 1,010.2 5.5 No- 9.1* 231.8
    Bending
    7 MTD103 LALPVLLLA 9 922.2 5.5 Bending 51.7 271.1
    8 MTD132 AVVVPAIVLAAP 12 1,119.4 5.6 Bending 50.3 195.0
    AVE 11 ± 3.2 1,083 ± 252 5.6 ± 0.1 Proline 41 ± 15 227 ± 47
    Presence
    A/a
    Hydropathy Residue Composition Secondary
    # Peptides Sequence (GRAVY) Structure A V L I P G Structure Cargo Ref.
    1 MTM AAVALLPAVLLALLAP 2.4 Aliphatic 6 2 6 0 2 0 Helix p50 1
    Ring
    2 MTS AAVLLPVLLAAP 2.3 Aliphatic 4 2 4 0 2 0 No-Helix CRE 2
    Ring
    3 MTD10 LGGAVVAAPVAAAVAP 1.8 Aliphatic 7 4 1 0 2 2 Helix Parkin 8
    Ring
    4 MTD73 PVLLLLA 2.8 Aliphatic 1 1 4 0 1 0 Helix ES 6
    Ring
    5 MTD77 AVALLILAV 3.3 Aliphatic 3 2 3 1 0 0 Helix NM23 3
    Ring
    6 MTD85 LLAAAAALLLA 2.7 Aliphatic 6 0 5 0 0 0 No-Helix RUNX3 5
    Ring
    7 MTD103 LALPVLLLA 2.8 Aliphatic 2 1 5 0 1 0 Helix p18 4
    Ring
    8 MTD132 AVVVPAIVLAAP 2.4 Aliphatic 4 4 1 1 2 0 No-Helix LIN28 7
    Ring
    AVE 2.5 ± 0.4
    *Removing the MTD85 increases II to 45.6 ± 9.3.
  • Table 6 shows summarized Critical Factors of published hydrophobic Cell-Penetrating Peptides (B).
  • TABLE 6
    Length: 11 ± 3.2
    Molecular Weight: 1,083 ± 252
    pI: 5.6 ± 0.1
    Bending Potential (BP): Proline presences in the middle and/or the end of
    peptides, or No Proline.
    Instability Index (II): 41.0 ± 15 (′Removing the MTD85 increases
    II to 45.6 ± 9.3)
    Residue Structure & Aliphatic Index (AI): 227 ± 47
    Hydropathy (GRAVY): 2.5 ± 0.4
    Aliphatic Ring: Non-polar hydrophobic & aliphatic amino acid
    (A, V, L, I).
    Secondary Structure: α-Helix is favored but not required.
  • 2-2. Selective Analysis (C) of Peptides that Provided Bending Potential and Higher Flexibility
  • To optimize the ‘Common Range and/or Consensus Feature of Critical Factor’ for the practical design of aMTDs and the random peptides (rPs or rPeptides), which were to prove that the ‘Critical Factors’ determined in the analysis A, B and C were correct to improve the current problems of hydrophobic CPPs—protein aggregation, low solubility/yield, and poor cell-/tis sue-permeability of the recombinant proteins fused to the MTS/MTM or MTD, and non-common sequence and non-homologous structure of the peptides, empirically selected peptides were analyzed for their structural features and physicochemical factor indexes.
  • Hydrophobic CPPs which did not have a bending potential, rigid or too much flexible sequences (too much low or too much high Instability Index), or too low or too high hydrophobic CPPs were unselected, but secondary structure was not considered because helix structure of sequence was not required.
  • In analysis C, eight selected CPP sequences that could provide a bending potential and higher flexibility were finally analyzed (Table 7 and 8). Common amino acid length is 12 (11.6±3.0). Proline is presence in the middle of and/or the end of sequence. Rigidity/Flexibility (II) is 45.5-57.3 (Avg: 50.1±3.6). AI and GRAVY representing structural feature and hydrophobicity of the peptide are 204.7±37.5 and 2.4±0.3, respectively. All peptides are consisted with hydrophobic and aliphatic amino acids (A, V, L, I, and P). Therefore, analysis C was chosen as a standard for the new design of new hydrophobic CPPs—aMTDs.
  • Table 7 shows characteristics of published hydrophobic Cell-Penetrating Peptides (C): selected CPPs that provided bending potential and higher flexibility.
  • TABLE 7
    Structural
    Rigidity/Flexibility Feature
    Molecular Bending (Instability (Aliphatic
    # Peptides Sequence Length Weight pI Potential Index: II) Index: AI)
    1 MTM AAVALLPAVLLALLAP 16 1515.9 5.6 Bending 45.5 220.0
    2 MTS AAVLLPVLLAAP 12 1147.4 5.6 Bending 57.3 211.7
    3 MTD10 LGGAVVAAPVAAAVAP 16 1333.5 5.5 Bending 47.9 140.6
    4 MTD47 AAAVPVLVAA 10 881.0 5.6 Bending 47.5 176.0
    5 MTD103 LALPVLLLA 9 922.2 5.5 Bending 51.7 271.1
    6 MTD132 AVVVPAIVLAAP 12 1119.4 5.6 Bending 50.3 195.0
    7 MTD173 AVIPILAVP 9 892.1 5.6 Bending 48.5 216.7
    8 MTD181 AVLLLPAAA 9 838.0 5.6 Bending 51.7 206.7
    AVE 11.6 ± 3.0 1081.2 ± 244.6 5.6 ± 0.1 Proline 50.1 ± 3.6 204.7 ± 37.5
    Presence
    A/a
    Hydropathy Residue Composition Secondary
    # Peptides Sequence (GRAVY) Structure A V L I P G Structure Cargo Ref.
    1 MTM AAVALLPAVLLALLAP 2.4 Aliphatic 6 2 6 0 2 0 Helix p50 1
    Ring
    2 MTS AAVLLPVLLAAP 2.3 Aliphatic 4 2 4 0 2 0 No-Helix CRE 2
    Ring
    3 MTD10 LGGAVVAAPVAAAVAP 1.8 Aliphatic 7 4 1 0 2 2 Helix Parkin 8
    Ring
    4 MTD47 AAAVPVLVAA 2.4 Aliphatic 5 3 1 0 1 0 No-Helix CMYC 4
    Ring
    5 MTD103 LALPVLLLA 2.8 Aliphatic 2 1 5 0 1 0 Helix p18 8
    Ring
    6 MTD132 AVVVPAIVLAAP 2.4 Aliphatic 4 4 1 1 2 0 No-Helix LIN28 4
    Ring
    7 MTD173 AVIPILAVP 2.4 Aliphatic 2 2 1 2 2 0 Helix KLF4 4
    Ring
    8 MTD181 AVLLLPAAA 2.4 Aliphatic 4 1 3 0 1 0 No-Helix SOX2 4
    Ring
    AVE 2.4 ± 0.3
  • Table 8 shows summarized Critical Factors of published hydrophobic Cell-Penetrating Peptides (C)
  • TABLE 8
    Length: 11.6 ± 3.0
    Molecular Weight: 1,081.2 ± 224.6
    pI: 5.6 ± 0.1
    Bending Potential (BP): Proline presences in the middle and/or the end of
    peptides.
    Instability Index (II): 50.1 ± 3.6
    Residue Structure & Aliphatic Index (AI): 204.7 ± 37.5
    Hydropathy (GRAVY): 2.4 ± 0.3
    Aliphatic Ring: Non-polar hydrophobic & aliphatic amino acid
    (A, V, L, I).
    Secondary Structure: α-Helix is favored but not required.

    3. New design of improved hydrophobic CPPs—aMTDs based on the optimized Critical Factors
  • 3-1. Determination of common sequence and/or common homologous structure
  • As mentioned above, H-regions of signal sequence (HRSS)-derived CPPs (MTS/MTM and MTD) do not have a common sequence, sequence motif, and/or common-structural homologous feature. In this invention, the aim is to develop improved hydrophobic CPPs formatted in the common sequence- and structural-motif which satisfy newly determined ‘Critical Factors’ to have ‘Common Function,’ namely, to facilitate protein translocation across the membrane with similar mechanism to the analyzed reference CPPs. Based on the analysis A, B and C, the common homologous features have been analyzed to determine the critical factors that influence the cell-permeability. The range value of each critical factor has been determined to include the analyzed index of each critical factor from analysis A, B and C to design novel aMTDs (Table 9). These features have been confirmed experimentally with newly designed aMTDs in their cell-permeability.
  • Table 9 shows comparison the range/feature of each Critical Factor between the value of analyzed CPPs and the value determined for new design of novel aMTDs sequences
  • TABLE 9
    Summarized Critical Factors of aMTD
    Selected CPPs Newly Designed CPPs
    Critical Factor Range Range
    Bending Potential Proline presences in Proline presences in
    (Proline Position: PP) the middle and/or at the middle (5′, 6′, 7′
    the end of peptides or 8′) and at the
    end of peptides
    Rigidity/Flexibility 45.5-57.3 40-60
    (Instability Index: II) (50.1 ± 3.6)
    Structural Feature 140.6-220.0 180-220
    (Aliphatic Index: AI) (204.7 ± 37.5)
    Hydropathy 1.8-2.8 2.1-2.6
    (Grand Average of (2.4 ± 0.3)
    Hydropathy GRAVY)
    Length 11.6 ± 3.0  9-13
    (Number of Amino Acid)
    Amino acid Composition A, V, I, L, P A, V, I, L, P
  • In Table 9, universal common features and sequence/structural motif are provided. Length is 9-13 amino acids, and bending potential is provided with the presence of proline in the middle of sequence (at 5′, 6′, 7′ or 8′ amino acid) for peptide bending and at the end of peptide for recombinant protein bending and Rigidity/Flexibility of aMTDs is II>40 are described in Table 9.
  • 3-2. Critical Factors for development of advanced MTDs
  • Recombinant cell-permeable proteins fused to the hydrophobic CPPs to deliver therapeutically active cargo molecules including proteins into live cells had previously been reported, but the fusion proteins expressed in bacteria system were hard to be purified as a soluble form due to their low solubility and yield. To address the crucial weakness for further clinical development of the cell-permeable proteins as protein-based biotherapeutics, greatly improved form of the hydrophobic CPP, named as advanced MTD (aMTD) has newly been developed through critical factors-based peptide analysis. The critical factors used for the current invention of the aMTDs are herein (Table 9).
  • 1. Amino Acid Length: 9-13
  • 2. Bending Potential (Proline Position: PP)
  • : Proline presences in the middle (from 5′ to 8′ amino acid) and at the end of sequence
  • 3. Rigidity/Flexibility (Instability Index: II): 40-60
  • 4. Structural Feature (Aliphatic Index: AI): 180-220
  • 5. Hydropathy (GRAVY): 2.1-2.6
  • 6. Amino Acid Composition: Hydrophobic and Aliphatic amino acids—A, V, L, I and P
  • 3-3. Design of potentially best aMTDs that all Critical Factors are considered and satisfied
  • After careful consideration of six critical factors derived from analysis of unique features of hydrophobic CPPs, advanced macromolecule transduction domains (aMTDs) have been designed and developed based on the common 12 amino acid platform which satisfies the critical factors including amino acid length (9-13) determined from the analysis.
  • Unlike previously published hydrophobic CPPs that require numerous experiments to determine their cell-permeability, newly developed aMTD sequences could be designed by performing just few steps as follows using above mentioned platform to follow the determined range value/feature of each critical factor.
  • First, prepare the 12 amino acid sequence platform for aMTD. Second, place proline (P) in the end (12′) of sequence and determine where to place proline in one of four U(s) in 5′, 6′, 7′, and 8. Third, alanine (A), valine (V), leucine (L) or isoleucine (I) is placed in either X(s) and/or U(s), where proline is not placed. Lastly, determine whether the amino acid sequences designed based on the platform, satisfy the value or feature of six critical factors to assure the cell permeable property of aMTD sequences. Through these processes, numerous novel aMTD sequences have been constructed. The expression vectors for preparing non-functional cargo recombinant proteins fused to each aMTD, expression vectors have been constructed and forcedly expressed in bacterial cells. These aMTD-fused recombinant proteins have been purified in soluble form and determined their cell-permeability quantitatively. aMTD sequences have been newly designed, numbered from 1 to 240, as shown in Table 10-15. In Table 10-15, sequence ID Number is a sequence listings for reference, and aMTD numbers refer to amino acid listing numbers that actually have been used at the experiments. For further experiments, aMTD numbers have been used. In addition, polynucleotide sequences shown in the sequence lists have been numbered from SEQ ID NO: 241 to SEQ ID NO: 480.
  • Tables 10 to 15 shows 240 new hydrophobic aMTD sequences that were developed to satisfy all critical factors.
  • TABLE 10
    Rigidity/ Sturctural
    Sequence Flexibility Feature Hydropathy Residue
    ID Number aMTD Sequences Length (II) (AI) (GRAVY) Structure
    1 1 AAALAPVVLALP 12 57.3 187.5 2.1 Aliphatic
    2 2 AAAVPLLAVVVP 12 41.3 195.0 2.4 Aliphatic
    3 3 AALLVPAAVLAP 12 57.3 187.5 2.1 Aliphatic
    4 4 ALALLPVAALAP 12 57.3 195.8 2.1 Aliphatic
    5 5 AAALLPVALVAP 12 57.3 187.5 2.1 Aliphatic
    6 11 VVALAPALAALP 12 57.3 187.5 2.1 Aliphatic
    7 12 LLAAVPAVLLAP 12 57.3 211.7 2.3 Aliphatic
    8 13 AAALVPVVALLP 12 57.3 203.3 2.3 Aliphatic
    9 21 AVALLPALLAVP 12 57.3 211.7 2.3 Aliphatic
    10 22 AVVLVPVLAAAP 12 57.3 195.0 2.4 Aliphatic
    11 23 VVLVLPAAAAVP 12 57.3 195.0 2.4 Aliphatic
    12 24 IALAAPALIVAP 12 50.2 195.8 2.2 Aliphatic
    13 25 IVAVAPALVALP 12 50.2 203.3 2.4 Aliphatic
    14 42 VAALPVVAVVAP 12 57.3 186.7 2.4 Aliphatic
    15 43 LLAAPLVVAAVP 12 41.3 187.5 2.1 Aliphatic
    16 44 ALAVPVALLVAP 12 57.3 203.3 2.3 Aliphatic
    17 61 VAALPVLLAALP 12 57.3 211.7 2.3 Aliphatic
    18 62 VALLAPVALAVP 12 57.3 203.3 2.3 Aliphatic
    19 63 AALLVPALVAVP 12 57.3 203.3 2.3 Aliphatic
  • TABLE 11
    Rigidity/ Sturctural
    Sequence Flexibility Feature Hydropathy Residue
    ID Number aMTD Sequences Length (II) (AI) (GRAVY) Structure
    20 64 AIVALPVAVLAP 12 50.2 203.3 2.4 Aliphatic
    21 65 IAIVAPVVALAP 12 50.2 203.3 2.4 Aliphatic
    22 81 AALLPALAALLP 12 57.3 204.2 2.1 Aliphatic
    23 82 AVVLAPVAAVLP 12 57.3 195.0 2.4 Aliphatic
    24 83 LAVAAPLALALP 12 41.3 195.8 2.1 Aliphatic
    25 84 AAVAAPLLLALP 12 41.3 195.8 2.1 Aliphatic
    26 85 LLVLPAAALAAP 12 57.3 195.8 2.1 Aliphatic
    27 101 LVALAPVAAVLP 12 57.3 203.3 2.3 Aliphatic
    28 102 LALAPAALALLP 12 57.3 204.2 2.1 Aliphatic
    29 103 ALIAAPILALAP 12 57.3 204.2 2.2 Aliphatic
    30 104 AVVAAPLVLALP 12 41.3 203.3 2.3 Aliphatic
    31 105 LLALAPAALLAP 12 57.3 204.1 2.1 Aliphatic
    32 121 AIVALPALALAP 12 50.2 195.8 2.2 Aliphatic
    33 123 AAIIVPAALLAP 12 50.2 195.8 2.2 Aliphatic
    34 124 IAVALPALIAAP 12 50.3 195.8 2.2 Aliphatic
    35 141 AVIVLPALAVAP 12 50.2 203.3 2.4 Aliphatic
    36 143 AVLAVPAVLVAP 12 57.3 195.0 2.4 Aliphatic
    37 144 VLAIVPAVALAP 12 50.2 203.3 2.4 Aliphatic
    38 145 LLAVVPAVALAP 12 57.3 203.3 2.3 Aliphatic
    39 161 AVIALPALIAAP 12 57.3 195.8 2.2 Aliphatic
    40 162 AVVALPAALIVP 12 50.2 203.3 2.4 Aliphatic
    41 163 LALVLPAALAAP 12 57.3 195.8 2.1 Aliphatic
    42 164 LAAVLPALLAAP 12 57.3 195.8 2.1 Aliphatic
    43 165 ALAVPVALAIVP 12 50.2 203.3 2.4 Aliphatic
    44 182 ALIAPVVALVAP 12 57.3 203.3 2.4 Aliphatic
    45 183 LLAAPVVIALAP 12 57.3 211.6 2.4 Aliphatic
    46 184 LAAIVPAIIAVP 12 50.2 211.6 2.4 Aliphatic
    47 185 AALVLPLIIAAP 12 41.3 220.0 2.4 Aliphatic
    48 201 LALAVPALAALP 12 57.3 195.8 2.1 Aliphatic
    49 204 LIAALPAVAALP 12 57.3 195.8 2.2 Aliphatic
    50 205 ALALVPAIAALP 12 57.3 195.8 2.2 Aliphatic
    51 221 AAILAPIVALAP 12 50.2 195.8 2.2 Aliphatic
    52 222 ALLIAPAAVIAP 12 57.3 195.8 2.2 Aliphatic
    53 223 AILAVPIAVVAP 12 57.3 203.3 2.4 Aliphatic
    54 224 ILAAVPIALAAP 12 57.3 195.8 2.2 Aliphatic
    55 225 VAALLPAAAVLP 12 57.3 187.5 2.1 Aliphatic
    56 241 AAAVVPVLLVAP 12 57.3 195.0 2.4 Aliphatic
    57 242 AALLVPALVAAP 12 57.3 187.5 2.1 Aliphatic
    58 243 AAVLLPVALAAP 12 57.3 187.5 2.1 Aliphatic
    59 245 AAALAPVLALVP 12 57.3 187.5 2.1 Aliphatic
    60 261 LVLVPLLAAAAP 12 41.3 211.6 2.3 Aliphatic
    61 262 ALIAVPAIIVAP 12 50.2 211.6 2.4 Aliphatic
    62 263 ALAVIPAAAILP 12 54.9 195.8 2.2 Aliphatic
    63 264 LAAAPVVIVIAP 12 50.2 203.3 2.4 Aliphatic
    64 265 VLAIAPLLAAVP 12 41.3 211.6 2.3 Aliphatic
    65 281 ALIVLPAAVAVP 12 50.2 203.3 2.4 Aliphatic
    66 282 VLAVAPALIVAP 12 50.2 203.3 2.4 Aliphatic
    67 283 AALLAPALIVAP 12 50.2 195.8 2.2 Aliphatic
    68 284 ALIAPAVALIVP 12 50.2 211.7 2.4 Aliphatic
    69 285 AIVLLPAAVVAP 12 50.2 203.3 2.4 Aliphatic
  • TABLE 12
    Rigidity/ Sturctural
    Sequence Flexibility Feature Hydropathy Residue
    ID Number aMTD Sequences Length (II) (AI) (GRAVY) Structure
    70 301 VIAAPVLAVLAP 12 57.3 203.3 2.4 Aliphatic
    71 302 LALAPALALLAP 12 57.3 204.2 2.1 Aliphatic
    72 304 AIILAPIAAIAP 12 57.3 204.2 2.3 Aliphatic
    73 305 IALAAPILLAAP 12 57.3 204.2 2.2 Aliphatic
    74 321 IVAVALPALAVP 12 50.2 203.3 2.3 Aliphatic
    75 322 VVAIVLPALAAP 12 50.2 203.3 2.3 Aliphatic
    76 323 IVAVALPVALAP 12 50.2 203.3 2.3 Aliphatic
    77 324 IVAVALPAALVP 12 50.2 203.3 2.3 Aliphatic
    78 325 IVAVALPAVALP 12 50.2 203.3 2.3 Aliphatic
    79 341 IVAVALPAVLAP 12 50.2 203.3 2.3 Aliphatic
    80 342 VIVALAPAVLAP 12 50.2 203.3 2.3 Aliphatic
    81 343 IVAVALPALVAP 12 50.2 203.3 2.3 Aliphatic
    82 345 ALLIVAPVAVAP 12 50.2 203.3 2.3 Aliphatic
    83 361 AVVIVAPAVIAP 12 50.2 195.0 24 Aliphatic
    84 363 AVLAVAPALIVP 12 50.2 203.3 2.3 Aliphatic
    85 364 LVAAVAPALIVP 12 50.2 203.3 2.3 Aliphatic
    86 365 AVIVVAPALLAP 12 50.2 203.3 2.3 Aliphatic
    87 381 VVAIVLPAVAAP 12 50.2 195.0 24 Aliphatic
    88 382 AAALVIPAILAP 12 54.9 195.8 2.2 Aliphatic
    89 383 VIVALAPALLAP 12 50.2 211.6 2.3 Aliphatic
    90 384 VIVAIAPALLAP 12 50.2 211.6 2.4 Aliphatic
    91 385 IVAIAVPALVAP 12 50.2 203.3 2.4 Aliphatic
    92 401 AALAVIPAAILP 12 54.9 195.8 2.2 Aliphatic
    93 402 ALAAVIPAAILP 12 54.9 195.8 2.2 Aliphatic
    94 403 AAALVIPAAILP 12 54.9 195.8 2.2 Aliphatic
    95 404 LAAAVIPAAILP 12 54.9 195.8 2.2 Aliphatic
    96 405 LAAAVIPVAILP 12 54.9 211.7 2.4 Aliphatic
    97 421 AAILAAPLIAVP 12 57.3 195.8 2.2 Aliphatic
    98 422 VVAILAPLLAAP 12 57.3 211.7 2.4 Aliphatic
    99 424 AVVVAAPVLALP 12 57.3 195.0 2.4 Aliphatic
    100 425 AVVAIAPVLALP 12 57.3 203.3 2.4 Aliphatic
    101 442 ALAALVPAVLVP 12 57.3 203.3 2.3 Aliphatic
    102 443 ALAALVPVALVP 12 57.3 203.3 2.3 Aliphatic
    103 444 LAAALVPVALVP 12 57.3 203.3 2.3 Aliphatic
    104 445 ALAALVPALVVP 12 57.3 203.3 2.3 Aliphatic
    105 461 IAAVIVPAVALP 12 50.2 203.3 2.4 Aliphatic
    106 462 IAAVLVPAVALP 12 57.3 203.3 2.4 Aliphatic
    107 463 AVAILVPLLAAP 12 57.3 211.7 2.4 Aliphatic
    108 464 AVVILVPLAAAP 12 57.3 203.3 2.4 Aliphatic
    109 465 IAAVIVPVAALP 12 50.2 203.3 2.4 Aliphatic
    110 481 AIAIAIVPVALP 12 50.2 211.6 2.4 Aliphatic
    111 482 ILAVAAIPVAVP 12 54.9 203.3 2.4 Aliphatic
    112 483 ILAAAIIPAALP 12 54.9 204.1 2.2 Aliphatic
    113 484 LAVVLAAPAIVP 12 50.2 203.3 2.4 Aliphatic
    114 485 AILAAIVPLAVP 12 50.2 211.6 2.4 Aliphatic
    115 501 VIVALAVPALAP 12 50.2 203.3 2.4 Aliphatic
    116 502 AIVALAVPVLAP 12 50.2 203.3 2.4 Aliphatic
    117 503 AAIIIVLPAALP 12 50.2 220.0 2.4 Aliphatic
    118 504 LIVALAVPALAP 12 50.2 211.7 2.4 Aliphatic
    119 505 AIIIVIAPAAAP 12 50.2 195.8 2.3 Aliphatic
  • TABLE 13
    Rigidity/ Sturctural
    Sequence Flexibility Feature Hydropathy Residue
    ID Number aMTD Sequences Length (II) (AI) (GRAVY) Structure
    120 521 LAALIVVPAVAP 12 50.2 203.3 2.4 Aliphatic
    121 522 ALLVIAVPAVAP 12 57.3 203.3 2.4 Aliphatic
    122 524 AVALIVVPALAP 12 50.2 203.3 2.4 Aliphatic
    123 525 ALAIVVAPVAVP 12 50.2 195.0 2.4 Aliphatic
    124 541 LLALIIAPAAAP 12 57.3 204.1 2.1 Aliphatic
    125 542 ALALIIVPAVAP 12 50.2 211.6 2.4 Aliphatic
    126 543 LLAALIAPAALP 12 57.3 204.1 2.1 Aliphatic
    127 544 IVALIVAPAAVP 12 43.1 203.3 2.4 Aliphatic
    128 545 VVLVLAAPAAVP 12 57.3 195.0 2.3 Aliphatic
    129 561 AAVAIVLPAVVP 12 50.2 195.0 2.4 Aliphatic
    130 562 ALIAAIVPALVP 12 50.2 211.7 2.4 Aliphatic
    131 563 ALAVIVVPALAP 12 50.2 203.3 2.4 Aliphatic
    132 564 VAIALIVPALAP 12 50.2 211.7 2.4 Aliphatic
    133 565 VAIVLVAPAVAP 12 50.2 195.0 2.4 Aliphatic
    134 582 VAVALIVPALAP 12 50.2 203.3 2.4 Aliphatic
    135 583 AVILALAPIVAP 12 50.2 211.6 2.4 Aliphatic
    136 585 ALIVAIAPALVP 12 50.2 211.6 2.4 Aliphatic
    137 601 AAILIAVPIAAP 12 57.3 195.8 2.3 Aliphatic
    138 602 VIVALAAPVLAP 12 50.2 203.3 2.4 Aliphatic
    139 603 VLVALAAPVIAP 12 57.3 203.3 2.4 Aliphatic
    140 604 VALIAVAPAVVP 12 57.3 195.0 2.4 Aliphatic
    141 605 VIAAVLAPVAVP 12 57.3 195.0 2.4 Aliphatic
    142 622 ALIVLAAPVAVP 12 50.2 203.3 2.4 Aliphatic
    143 623 VAAAIALPAIVP 12 50.2 187.5 2.3 Aliphatic
    144 625 ILAAAAAPLIVP 12 50.2 195.8 2.2 Aliphatic
    145 643 LALVLAAPAIVP 12 50.2 211.6 2.4 Aliphatic
    146 645 ALAVVALPAIVP 12 50.2 203.3 2.4 Aliphatic
    147 661 AAILAPIVAALP 12 50.2 195.8 2.2 Aliphatic
    148 664 ILIAIAIPAAAP 12 54.9 204.1 2.3 Aliphatic
    149 665 LAIVLAAPVAVP 12 50.2 203.3 2.3 Aliphatic
    150 666 AAIAIIAPAIVP 12 50.2 195.8 2.3 Aliphatic
    151 667 LAVAIVAPALVP 12 50.2 203.3 2.3 Aliphatic
    152 683 LAIVLAAPAVLP 12 50.2 211.7 2.4 Aliphatic
    153 684 AAIVLALPAVLP 12 50.2 211.7 2.4 Aliphatic
    154 685 ALLVAVLPAALP 12 57.3 211.7 2.3 Aliphatic
    155 686 AALVAVLPVALP 12 57.3 203.3 2.3 Aliphatic
    156 687 AILAVALPLLAP 12 57.3 220.0 2.3 Aliphatic
    157 703 IVAVALVPALAP 12 50.2 203.3 2.4 Aliphatic
    158 705 IVAVALLPALAP 12 50.2 211.7 2.4 Aliphatic
    159 706 IVAVALLPAVAP 12 50.2 203.3 2.4 Aliphatic
    160 707 IVALAVLPAVAP 12 50.2 203.3 2.4 Aliphatic
    161 724 VAVLAVLPALAP 12 57.3 203.3 2.3 Aliphatic
    162 725 IAVLAVAPAVLP 12 57.3 203.3 2.3 Aliphatic
    163 726 LAVAIIAPAVAP 12 57.3 187.5 2.2 Aliphatic
    164 727 VALAIALPAVLP 12 57.3 211.6 2.3 Aliphatic
    165 743 AIAIALVPVALP 12 57.3 211.6 2.4 Aliphatic
    166 744 AAVVIVAPVALP 12 50.2 195.0 2.4 Aliphatic
    167 746 VAIIVVAPALAP 12 50.2 203.3 2.4 Aliphatic
    168 747 VALLAIAPALAP 12 57.3 195.8 2.2 Aliphatic
    169 763 VAVLIAVPALAP 12 57.3 203.3 2.3 Aliphatic
  • TABLE 14
    Rigidity/ Sturctural
    Sequence Flexibility Feature Hydropathy Residue
    ID Number aMTD Sequences Length (II) (AI) (GRAVY) Structure
    170 764 AVALAVLPAVVP 12 57.3 195.0 2.3 Aliphatic
    171 765 AVALAVVPAVLP 12 57.3 195.0 2.3 Aliphatic
    172 766 IVVIAVAPAVAP 12 50.2 195.0 2.4 Aliphatic
    173 767 IVVAAVVPALAP 12 50.2 195.0 2.4 Aliphatic
    174 783 IVALVPAVAIAP 12 50.2 203.3 2.5 Aliphatic
    175 784 VAALPAVALVVP 12 57.3 195.0 2.4 Aliphatic
    176 786 LVAIAPLAVLAP 12 41.3 211.7 2.4 Aliphatic
    177 787 AVALVPVIVAAP 12 50.2 195.0 2.4 Aliphatic
    178 788 AIAVAIAPVALP 12 57.3 187.5 2.3 Aliphatic
    179 803 AIALAVPVLALP 12 57.3 211.7 2.4 Aliphatic
    180 805 LVLIAAAPIALP 12 41.3 220.0 2.4 Aliphatic
    181 806 LVALAVPAAVLP 12 57.3 203.3 2.3 Aliphatic
    182 807 AVALAVPALVLP 12 57.3 203.3 2.3 Aliphatic
    183 808 LVVLAAAPLAVP 12 41.3 203.3 2.3 Aliphatic
    184 809 LIVLAAPALAAP 12 50.2 195.8 2.2 Aliphatic
    185 810 VIVLAAPALAAP 12 50.2 187.5 2.2 Aliphatic
    186 811 AVVLAVPALAVP 12 57.3 195.0 2.3 Aliphatic
    187 824 LIIVAAAPAVAP 12 50.2 187.5 2.3 Aliphatic
    188 825 IVAVIVAPAVAP 12 43.2 195.0 2.5 Aliphatic
    189 826 LVALAAPIIAVP 12 41.3 211.7 2.4 Aliphatic
    190 827 IAAVLAAPALVP 12 57.3 187.5 2.2 Aliphatic
    191 828 IALLAAPIIAVP 12 41.3 220.0 2.4 Aliphatic
    192 829 AALALVAPVIVP 12 50.2 203.3 2.4 Aliphatic
    193 830 IALVAAPVALVP 12 57.3 203.3 2.4 Aliphatic
    194 831 IIVAVAPAAIVP 12 43.2 203.3 2.5 Aliphatic
    195 832 AVAAIVPVIVAP 12 43.2 195.0 2.5 Aliphatic
    196 843 AVLVLVAPAAAP 12 41.3 219.2 2.5 Aliphatic
    197 844 VVALLAPLIAAP 12 41.3 211.8 2.4 Aliphatic
    198 845 AAVVIAPLLAVP 12 41.3 203.3 2.4 Aliphatic
    199 846 IAVAVAAPLLVP 12 41.3 203.3 2.4 Aliphatic
    200 847 LVAIVVLPAVAP 12 50.2 219.2 2.6 Aliphatic
    201 848 AVAIVVLPAVAP 12 50.2 195.0 2.4 Aliphatic
    202 849 AVILLAPLIAAP 12 57.3 220.0 2.4 Aliphatic
    203 850 LVIALAAPVALP 12 57.3 211.7 2.4 Aliphatic
    204 851 VLAVVLPAVALP 12 57.3 219.2 2.5 Aliphatic
    205 852 VLAVAAPAVLLP 12 57.3 203.3 2.3 Aliphatic
    206 863 AAVVLLPIIAAP 12 41.3 211.7 2.4 Aliphatic
    207 864 ALLVIAPAIAVP 12 57.3 211.7 2.4 Aliphatic
    208 865 AVLVIAVPAIAP 12 57.3 203.3 2.5 Aliphatic
    209 867 ALLVVIAPLAAP 12 41.3 211.7 2.4 Aliphatic
    210 868 VLVAAILPAAIP 12 54.9 211.7 2.4 Aliphatic
    211 870 VLVAAVLPIAAP 12 41.3 203.3 2.4 Aliphatic
    212 872 VLAAAVLPLVVP 12 41.3 219.2 2.5 Aliphatic
    213 875 AIAIVVPAVAVP 12 50.2 195.0 2.4 Aliphatic
    214 877 VAIIAVPAVVAP 12 57.3 195.0 2.4 Aliphatic
    215 878 IVALVAPAAVVP 12 50.2 195.0 2.4 Aliphatic
    216 879 AAIVLLPAVVVP 12 50.2 219.1 2.5 Aliphatic
    217 881 AALIVVPAVAVP 12 50.2 195.0 2.4 Aliphatic
    218 882 AIALVVPAVAVP 12 57.3 195.0 2.4 Aliphatic
    219 883 LAIVPAAIAALP 12 50.2 195.8 2.2 Aliphatic
  • TABLE 15
    Rigidity/ Sturctural
    Sequence Flexibility Feature Hydropathy Residue
    ID Number aMTD Sequences Length (II) (AI) (GRAVY) Structure
    220 885 LVAIAPAVAVLP 12 57.3 203.3 2.4 Aliphatic
    221 887 VLAVAPAVAVLP 12 57.3 195.0 2.4 Aliphatic
    222 888 ILAVVAIPAAAP 12 54.9 187.5 2.3 Aliphatic
    223 889 ILVAAAPIAALP 12 57.3 195.8 2.2 Aliphatic
    224 891 ILAVAAIPAALP 12 54.9 195.8 2.2 Aliphatic
    225 893 VIAIPAILAAAP 12 54.9 195.8 2.3 Aliphatic
    226 895 AIIIVVPAIAAP 12 50.2 211.7 2.5 Aliphatic
    227 896 AILIVVAPIAAP 12 50.2 211.7 2.5 Aliphatic
    228 897 AVIVPVAIIAAP 12 50.2 203.3 2.5 Aliphatic
    229 899 AVVIALPAVVAP 12 57.3 195.0 2.4 Aliphatic
    230 900 ALVAVIAPVVAP 12 57.3 195.0 2.4 Aliphatic
    231 901 ALVAVLPAVAVP 12 57.3 195.0 2.4 Aliphatic
    232 902 ALVAPLLAVAVP 12 41.3 203.3 2.3 Aliphatic
    233 904 AVLAVVAPVVAP 12 57.3 186.7 2.4 Aliphatic
    234 905 AVIAVAPLVVAP 12 41.3 195.0 2.4 Aliphatic
    235 906 AVIALAPVVVAP 12 57.3 195.0 2.4 Aliphatic
    236 907 VAIALAPVVVAP 12 57.3 195.0 2.4 Aliphatic
    237 908 VALALAPVVVAP 12 57.3 195.0 2.3 Aliphatic
    238 910 VAALLPAVVVAP 12 57.3 195.0 2.3 Aliphatic
    239 911 VALALPAVVVAP 12 57.3 195.0 2.3 Aliphatic
    240 912 VALLAPAVVVAP 12 57.3 195.0 2.3 Aliphatic
    52.6 ± 5.1 201.7 ± 7.8 2.3 ± 0.1
  • 3-4. Design of the peptides that did not satisfy at least one Critical Factor
  • To demonstrate that this invention of new hydrophobic CPPs—aMTDs, which satisfy all critical factors described above, are correct and rationally designed, the peptides which do not satisfy at least one critical factor have also been designed. Total of 31 rPeptides (rPs) are designed, developed and categorized as follows: no bending peptides, either no proline in the middle as well at the end and/or no central proline; rigid peptides (II<40); too much flexible peptides; aromatic peptides (aromatic ring presences); hydrophobic, with non-aromatic peptides but have amino acids other than A, V, L, I, P or additional proline residues; hydrophilic, but non-aliphatic peptides.
  • 3-4-1. Peptides that do not Satisfy the Bending Potential
  • Table 16 shows the peptides that do not have any proline in the middle (at 5′, 6′, 7′ or 8′) and at the end of the sequences. In addition, Table 16 describes the peptides that do not have proline in the middle of the sequences. All these peptides are supposed to have no-bending potential.
  • TABLE 16
    Proline Rigidity/ Sturctural
    rPeptide Position Flexibility Feature Hydropathy
    Group ID Sequences Length (PP) (II) (AI) (GRAVY)
    No-Bending Peptides 931 AVLIAPAILAAA 12 6 57.3 204.2 2.5
    (No Proline at 5, 6, 7 936 ALLILAAAVAAP 12  12 41.3 204.2 2.4
    or 8 and/or 12) 152 LAAAVAAVAALL 12 None 9.2 204.2 2.7
    27 LAIVAAAAALVA 12 None 2.1 204.2 2.8
    935 ALLILPAAAVAA 12 6 57.3 204.2 2.4
    670 ALLILAAAVAAL 12 None 25.2 236.6 2.8
    934 LILAPAAVVAAA 12 5 57.3 195.8 2.5
    37 TTCSQQQYCTNG 12 None 53.1 0.0 −1.1
    16 NNSCTTVTNGSQ 12 None 47.4 0.0 −1.4
    113 PVAVALLIAVPP 12 1, 11, 12 57.3 195.0 2.1
  • 3-4-2. Peptides that do not Satisfy the Rigidity/Flexibility
  • To prove that rigidity/flexibility of the sequence is a crucial critical factor, rigid (Avg. II: 21.8±6.6) and too high flexible sequences (Avg. II: 82.3±21.0) were also designed. Rigid peptides that instability index is much lower than that of new aMTDs (II: 41.3-57.3, Avg. II: 53.3±5.7) are shown in Table 17. Bending, but too high flexible peptides that II is much higher than that of new aMTDs are also provided in Table 18.
  • TABLE 17
    Proline Rigidity/ Sturctural
    Position Flexibility Feature Hydropathy
    Group rPeptide ID Sequences Length (PP) (II) (AI) (GRAVY)
    Rigid Peptides 226 ALVAAIPALAIP 12 6 20.4 195.8 2.2
    (II < 50) 6 VIAMIPAAFWVA 12 6 15.7 146.7 2.2
    750 LAIAAIAPLAIP 12 8, 12 22.8 204.2 2.2
    26 AAIALAAPLAIV 12 8 18.1 204.2 2.5
    527 LVLAAVAPIAIP 12 8, 12 22.8 211.7 2.4
    466 IIAAAAPLAIIP 12 7, 12 22.8 204.2 2.3
    167 VAIAIPAALAIP 12 6, 12 20.4 195.8 2.3
    246 VVAVPLLVAFAA 12 5 25.2 195.0 2.7
    426 AAALAIPLAIIP 12 7, 12 4.37 204.2 2.2
    606 AAAIAAIPIIIP 12 8, 12 4.4 204.2 2.4
    66 AGVLGGPIMGVP 12 7, 12 35.5 121.7 1.3
    248 VAAIVPIAALVP 12 6, 12 34.2 203.3 2.5
    227 LAAIVPIAAAVP 12 6, 12 34.2 187.5 2.2
    17 GGCSAPQTTCSN 12 6 51.6 8.3 −0.5
    67 LDAEVPLADDVP 12 6, 12 34.2 130.0 0.3
  • TABLE 18
    Proline Rigidity/ Sturctural
    rPeptide Position Flexibility Feature Hydropathy
    Group ID Sequences Length (PP) (II) (AI) (GARVY)
    Bending Peptides 692 PAPLPPVVILAV 12 1, 3, 5, 6 105.5 186.7 1.8
    but Too High 69 PVAVLPPAALVP 12 1, 6, 7, 12 89.4 162.5 1.6
    Flexibility 390 VPLLVPVVPVVP 12 2, 6, 9, 12 105.4 210.0 2.2
    350 VPILVPVVPVVP 12 2, 6, 9, 12 121.5 210.0 2.2
    331 VPVLVPLVPVVP 12 2, 6, 9, 12 105.4 210.0 2.2
    9 VALVPAALILPP 12 5, 11, 12 89.4 203.3 2.1
    68 VAPVLPAAPLVP 12 3, 6, 9, 12 105.5 162.5 1.6
    349 VPVLVPVVPVVP 12 2, 6, 9, 12 121.5 201.6 2.2
    937 VPVLVPLPVPVV 12 2, 6, 8, 10 121.5 210.0 2.2
    938 VPVLLPVVVPVP 12 2, 6, 10, 12 121.5 210.0 2.2
    329 LPVLVPVVPVVP 12 2, 6, 9, 12 121.5 210.0 2.2
    49 VVPAAPAVPVVP 12 3, 6, 9, 12 121.5 145.8 1.7
    772 LPVAPVIPIIVP 12 2, 5, 8, 12 79.9 210.8 2.1
    210 ALIALPALPALP 12 6, 9, 12 89.4 195.8 1.8
    28 AVPLLPLVPAVP 12 3, 6, 9, 12 89.4 186.8 1.8
    693 AAPVLPVAVPIV 12 3, 6, 10 82.3 186.7 2.1
    169 VALVAPALILAP 12 6, 12 73.4 211.7 2.4
    29 VLPPLPVLPVLP 12 3, 4, 6, 9, 12 121.5 202.5 1.7
    190 AAILAPAVIAPP 12 6, 11, 12 89.4 163.3 1.8
  • 3-4-3. Peptides that do not Satisfy the Structural Features
  • New hydrophobic CPPs—aMTDs are consisted with only hydrophobic and aliphatic amino acids (A, V, L, I and P) with average ranges of the indexes—AI: 180-220 and GRAVY: 2.1-2.6 (Table 9). Based on the structural indexes, the peptides which contain an aromatic residue (W, F or Y) are shown in Table 19 and the peptides which are hydrophobic with non-aromatic sequences but have amino acids residue other than A, V, L, I, P or additional proline residues are designed (Table 20). Finally, hydrophilic and/or bending peptides which are consisted with non-aliphatic amino acids are shown in Table 21.
  • TABLE 19
    Proline Rigidity/ Sturctural
    rPeptide Position Flexibility Feature Hydropathy
    Group ID Sequences Length (PP) (II) (AI) (GRAVY)
    Aromatic Peptides 30 WFFAGPIMLIWP 12 6, 12 9.2 105.8 1.4
    (Aromatic Ring 33 AAAILAPAFLAV 12 7 57.3 171.7 2.4
    Presences) 131 WIIAPVWLAWIA 12 5 51.6 179.2 1.9
    922 WYVIPVLPLVVP 12 8, 12 41.3 194.2 2.2
    71 FMWMWFPFMWYP 12 7, 12 71.3 0.0 0.6
    921 IWWFVVLPLVVP 12 8, 12 41.3 194.2 2.2
  • TABLE 20
    Proline Rigidity/ Sturctural
    rPeptide Position Flexibility Feature Hydropathy
    Group ID Sequences Length (PP) (II) (AI) (GARVY)
    Hydrophobic 436 VVMLVVPAVMLP 12 7.12 57.3 194.2 2.6
    but Non Aromatic 138 PPAALLAILAVA 12 1.2 57.3 195.8 2.2
    Peptides 77 PVALVLVALVAP 12 1.12 41.3 219.2 2.5
    577 MLMIALVPMIAV 12 8 18.9 195.0 2.7
    97 ALLAAPPALLAL 12 6.7 57.3 204.2 2.1
    214 ALIVAPALMALP 12 6.12 60.5 187.5 2.2
    59 AVLAAPVVAALA 12 6 41.3 187.5 2.5
    54 LAVAAPPVVALL 12 6.7 57.3 203.3 2.3
  • TABLE 21
    Proline Rigidity/ Sturctural
    Position Flexibility Feature Hydropathy
    Group rPeptide ID Sequences Length (PP) (II) (AI) (GRAVY)
    Hydrophilic Peptides 949 SGNSCQQCGNSS 12 None 41.7 0.0 −1.1
    but Non Aliphatic 39 CYNTSPCTGCCY 12 6 52.5 0.0 0.0
    19 YVSCCTYTNGSQ 12 None 47.7 0.0 −1.0
    947 CYYNQQSNNNNQ 12 None 59.6 0.0 −2.4
    139 TGSTNSPTCTST 12 7 53.4 0.0 −0.7
    18 NYCCTPTTNGQS 12 6 47.9 0.0 −0.9
    20 NYCNTCPTYGQS 12 7 47.4 0.0 −0.9
    635 GSTGGSQQNNQY 12 None 31.9 0.0 −1.9
    40 TYNTSCTPGTCY 12 8 49.4 0.0 −0.6
    57 QNNCNTSSQGGG 12 None 52.4 0.0 −1.6
    159 CYSGSTSQNQPP 12 11.12 51.0 0.0 −1.3
    700 GTSNTCQSNQNS 12 None 19.1 0.0 −1.6
    38 YYNQSTCGGQCY 12 None 53.8 0.0 −1.0
  • 3-5. Summary of Newly Designed Peptides
  • Total of 457 sequences have been designed based on the critical factors. Designed potentially best aMTDs (hydrophobic, flexible, bending, aliphatic and 12-A/a length peptides) that do satisfy all range/feature of critical factors are 316. Designed rPeptides that do not satisfy at least one of the critical factors are 141 that no bending peptide sequences are 26; rigid peptide (II<40) sequences are 23; too much flexible peptides are 24; aromatic peptides (aromatic ring presences) are 27; hydrophobic, but non-aromatic peptides are 23; and hydrophilic, but non-aliphatic peptides are 18.
  • 4. Preparation of Recombinant Report Proteins Fused to aMTDs and rPeptides
  • Recombinant proteins fused to aMTDs and others [rPeptides, reference hydrophobic CPP sequences (MTM and MTD)] were expressed in a bacterial system, purified with single-step affinity chromatography and prepared as soluble proteins in physiological condition. These recombinant proteins have been tested for the ability of their cell-permeability by utilizing flow cytometry and laser scanning confocal microscopy.
  • 4-1. Selection of Cargo Protein for Recombinant Proteins Fused to Peptide Sequences
  • For clinical/non-clinical application, aMTD-fused cargo materials would be biologically active molecules that could be one of the following: enzymes, transcription factors, toxic, antigenic peptides, antibodies and antibody fragments. Furthermore, biologically active molecules could be one of these following macromolecules: enzymes, hormones, carriers, immunoglobulin, membrane-bound proteins, transmembrane proteins, internal proteins, external proteins, secreted proteins, virus proteins, native proteins, glycoproteins, fragmented proteins, disulfide bonded proteins, recombinant proteins, chemically modified proteins and prions. In addition, these biologically active molecules could be one of the following: nucleic acid, coding nucleic acid sequence, mRNAs, antisense RNA molecule, carbohydrate, lipid and glycolipid.
  • According to these pre-required conditions, a non-functional cargo to evaluate aMTD-mediated protein uptake has been selected and called as Cargo A (CRA) that should be soluble and non-functional. The domain (A/a 289-840; 184 A/a length) is derived from protein S (Genbank ID: CP000113.1).
  • 4-2. Construction of Expression Vector and Preparation of Recombinant Proteins
  • Coding sequences for recombinant proteins fused to each aMTD are cloned Ndel (5′) and SalI (3′) in pET-28a(+) (Novagen, Darmstadt, Germany) from PCR-amplified DNA segments. PCR primers for the recombinant proteins fused to aMTD and rPeptides are SEQ ID NOs: 481˜797. Structure of the recombinant proteins is displayed in FIG. 1.
  • The recombinant proteins were forcedly expressed in E. coli BL21 (DE3) cells grown to an OD600 of 0.6 and induced for 2 hours with 0.7 mM isopropyl-β-D-thiogalactopyranoside (IPTG). The proteins were purified by Ni2+ affinity chromatography as directed by the supplier (Qiagen, Hilden, Germany) in natural condition. After the purification, purified proteins were dissolved in a physiological buffer such as DMEM medium.
  • TABLE 22
    Potentially Best aMTDs (Hydrophobic, 240
    Flexible, Bending, Aliphatic & Helical)
    Random Peptides 31
    No Bending Peptides (No Proline at 5 or 6 and/or 12) 02
    No Bending Peptides (No Central Proline) 01
    Rigid Peptides (II < 50) 09
    Too Much Flexible Peptides 09
    Aromatic Peptides (Aromatic Ring Presences) 01
    Hydrophobic, But Non-Aromatic Peptides 02
    Hydrophilic, But Non-Aliphatic Peptides 07
  • 4-3. Expression of aMTD- or Random Peptide (rP)-Fused Recombinant Proteins
  • Using the standardized six critical factors, 316 aMTD sequences have been designed. In addition, 141 rPeptides are also developed that lack one of these critical factors: no bending peptides: i) absence of proline both in the middle and at the end of sequence or ii) absence of proline either in the middle or at the end of sequence, rigid peptides, too much flexible peptides, aromatic peptides (aromatic ring presence), hydrophobic but non-aromatic peptides, and hydrophilic but non-aliphatic peptides (Table 22).
  • These rPeptides are devised to be compared and contrasted with aMTDs in order to analyze structure/sequence activity relationship (SAR) of each critical factor with regard to the peptides' intracellular delivery potential. All peptide (aMTD or rPeptide)-containing recombinant proteins have been fused to the CRA to enhance the solubility of the recombinant proteins to be expressed, purified, prepared and analyzed.
  • These designed 316 aMTDs and 141 rPeptides fused to CRA were all cloned (FIG. 2) and tested for inducible expression in E. coli (FIG. 3). Out of these peptides, 240 aMTDs were inducibly expressed, purified and prepared in soluble form (FIG. 4). In addition, 31 rPeptides were also prepared as soluble form (FIG. 4).
  • To prepare the proteins fused to rPeptides, 60 proteins were expressed that were 10 out of 26 rPeptides in the category of no bending peptides (Table 16); 15 out of 23 in the category of rigid peptides [instability index (II)<40] (Table 17); 19 out of 24 in the category of too much flexible peptides (Table 18); 6 out of 27 in the category of aromatic peptides (Table 19); 8 out of 23 in the category of hydrophobic but non-aromatic peptides (Table 20); and 12 out of 18 in the category of hydrophilic but non-aliphatic peptides (Table 21).
  • 4-4. Quantitative Cell-Permeability of aMTD-Fused Recombinant Proteins
  • The aMTDs and rPeptides were fluorescently labeled and compared based on the critical factors for cell-permeability by using flow cytometry and confocal laser scanning microscopy (FIGS. 5 to 8). The cellular uptake of the peptide-fused non-functional cargo recombinant proteins could quantitatively be evaluated in flow cytometry, while confocal laser scanning microscopy allows intracellular uptake to be assessed visually. The analysis included recombinant proteins fused to a negative control [rP38] that has opposite characteristics (hydrophilic and aromatic sequence: YYNQSTCGGQCY) to the aMTDs (hydrophobic and aliphatic sequences). Relative cell-permeability (relative fold) of aMTDs to the negative control was also analyzed (Table 23 and FIG. 9).
  • Table 23 shows Comparison Analysis of Cell-Permeability of aMTDs with a Negative Control (A: rP38).
  • TABLE 23
    Negative Control
    rP38
    aMTD 19.6 ± 1.6*
    The Average of 240 aMTDs (Best: 164.2)
    *Relative Fold (aMTD in Geo Mean in its comparison to rP38)
  • Relative cell-permeability (relative fold) of aMTDs to the reference CPPs [B: MTM12 (AAVLLPVLLAAP), C: MTD85 (AVALLILAV)] was also analyzed (Tables 40 and 41)
  • Table 24 shows Comparison Analysis of Cell-Permeability of aMTDs with a Reference CPP (B: MTM12).
  • TABLE 24
    MTM12
    aMTD 13.1 ± 1.1*
    The Average of 240 aMTDs (Best: 109.9)
    *Relative Fold (aMTD in Geo Mean in its comparison to MTM12)
  • Table 25 shows Comparison Analysis of Cell-Permeability of aMTDs with a Reference CPP (C: MTD85).
  • TABLE 25
    MTD85
    aMTD 6.6 ± 0.5*
    The Average of 240 aMTDs (Best: 55.5)
    *Relative Fold (aMTD in Geo Mean in its comparison to MTD85)
  • Geometric means of negative control (histidine-tagged rP38-fused CRA recombinant protein) subtracted by that of naked protein (histidine-tagged CRA protein) lacking any peptide (rP38 or aMTD) was standardized as relative fold of 1. Relative cell-permeability of 240 aMTDs to the negative control (A type) was significantly increased by up to 164 fold, with average increase of 19.6±1.6 (Table 26-31).
  • TABLE 26
    Proline Rigidity/ Sturctural Relative
    Sequence Position Flexibility Feature Hydropathy Ratio (Fold)
    ID Number aMTD Sequences Length (PP) (II) (AI) (GRAVY) A B C
    1 899 AVVIALPAVVAP 12 7 57.3 195.0 2.4 164.2 109.9 55.5
    2 908 VALALAPVVVAP 12 7 57.3 195.0 2.3 150.6 100.8 50.9
    3 910 VAALLPAVVVAP 12 6 57.3 195.0 2.3 148.5 99.4 50.2
    4 810 VIVLAAPALAAP 12 7 50.2 187.5 2.2 120.0 80.3 40.6
    5 904 AVLAVVAPVVAP 12 8 57.3 186.7 2.4 105.7 70.8 35.8
    6 321 IVAVALPALAVP 12 7 50.2 203.3 2.3 97.8 65.2 32.9
    7 851 VLAVVLPAVALP 12 7 57.3 219.2 2.5 96.6 64.7 32.7
    8 911 VALALPAVVVAP 12 6 57.3 195.0 2.3 84.8 56.8 28.7
    9 852 VLAVAAPAVLLP 12 7 57.3 203.3 2.3 84.6 56.6 28.6
    10 803 AIALAVPVLALP 12 7 57.3 211.7 2.4 74.7 50.0 25.3
    11 888 ILAVVAIPAAAP 12 8 54.9 187.5 2.3 71.0 47.5 24.0
    12 825 IVAVIVAPAVAP 12 8 43.2 195.0 2.5 69.7 46.6 23.6
    13 895 AIIIVVPAIAAP 12 7 50.2 211.7 2.5 60.8 40.7 20.6
    14 896 AILIVVAPIAAP 12 8 50.2 211.7 2.5 57.5 38.5 19.4
    15 727 VALAIALPAVLP 12 8 57.3 211.6 2.3 54.7 36.7 18.5
    16 603 VLVALAAPVIAP 12 8 57.3 203.3 2.4 54.1 36.1 18.2
    17 847 LVAIVVLPAVAP 12 8 50.2 219.2 2.6 50.2 33.4 16.9
    18 826 LVALAAPIIAVP 12 7 41.3 211.7 2.4 49.2 32.9 16.6
    19 724 VAVLAVLPALAP 12 8 57.3 203.3 2.3 47.5 31.8 16.1
    20 563 ALAVIVVPALAP 12 8 50.2 203.3 2.4 47.1 31.4 15.9
    21 811 AVVLAVPALAVP 12 7 57.3 195.0 2.3 46.5 31.1 15.7
    22 831 IIVAVAPAAIVP 12 7 43.2 203.3 2.5 46.3 31.0 15.7
    23 829 AALALVAPVIVP 12 8 50.2 203.3 2.4 44.8 30.0 15.2
    24 891 ILAVAAIPAALP 12 8 54.3 195.8 2.2 44.7 29.9 15.1
    25 905 AVIAVAPLVVAP 12 7 41.3 195.0 2.4 44.0 29.5 14.9
    26 564 VAIALIVPALAP 12 8 50.2 211.7 2.4 43.6 29.1 14.7
    27 124 IAVALPALIAAP 12 6 50.3 195.8 2.2 43.6 29.0 14.7
    28 827 IAAVLAAPALVP 12 8 57.3 187.5 2.2 43.0 28.8 14.6
    29 2 AAAVPLLAVVVP 12 5 41.3 195.0 2.4 40.9 27.2 13.8
    30 385 IVAIAVPALVAP 12 7 50.2 203.3 2.4 38.8 25.9 13.1
    31 828 IALLAAPIIAVP 12 7 41.3 220.0 2.4 36.8 24.6 12.4
    32 806 LVALAVPAAVLP 12 7 57.3 203.3 2.3 36.7 24.6 12.4
    33 845 AAVVIAPLLAVP 12 7 41.3 203.3 2.4 35.8 24.0 12.1
    34 882 AIALVVPAVAVP 12 7 57.3 195.0 2.4 35.0 23.4 11.8
    35 545 VVLVLAAPAAVP 12 8 57.3 195.0 2.3 34.6 23.1 11.7
    36 161 AVIALPALIAAP 12 6 57.3 195.8 2.2 34.5 23.0 11.6
    37 481 AIAIAIVPVALP 12 8 50.2 211.6 2.4 34.3 23.0 11.6
    38 900 ALVAVIAPVVAP 12 8 57.3 195.0 2.4 34.3 22.9 11.6
    39 223 AILAVPIAVVAP 12 6 57.3 203.3 2.4 33.0 22.1 11.2
    40 824 LIIVAAAPAVAP 12 8 50.2 187.5 2.3 32.8 21.9 11.1
    41 562 ALIAAIVPALVP 12 8 50.2 211.7 2.4 32.7 21.8 11.0
    42 222 ALLIAPAAVIAP 12 6 57.3 195.8 2.2 32.6 21.7 11.0
    43 61 VAALPVLLAALP 12 5 57.3 211.7 2.3 31.2 20.8 10.5
    44 582 VAVALIVPALAP 12 8 50.2 203.3 2.4 30.6 20.4 10.3
    45 889 ILVAAAPIAALP 12 7 57.3 195.8 2.2 30.3 20.3 10.3
    46 787 AVALVPVIVAAP 12 6 50.2 195.0 2.4 29.3 19.6 9.9
    47 703 IVAVALVPALAP 12 8 50.2 203.3 2.4 29.2 19.5 9.9
    48 705 IVAVALLPALAP 12 8 50.2 211.7 2.4 28.6 19.1 9.7
    49 885 LVAIAPAVAVLP 12 6 57.3 203.3 2.4 28.3 19.0 9.6
    50 3 AALLVPAAVLAP 12 6 57.3 187.5 2.1 27.0 18.0 9.1
    51 601 AAILIAVPIAAP 12 8 57.3 195.8 2.3 26.8 17.9 9.0
    52 843 AVLVLVAPAAAP 12 8 41.3 219.2 2.5 26.4 17.7 8.9
    53 403 AAALVIPAAILP 12 7 54.2 195.8 2.2 25.2 16.8 8.5
    54 544 IVALIVAPAAVP 12 8 43.1 203.3 2.4 23.4 15.6 7.9
    55 522 ALLVIAVPAVAP 12 8 57.3 203.3 2.4 22.7 15.2 7.7
  • TABLE 27
    Proline Rigidity/ Sturctural Relative
    Sequence Position Flexibility Feature Hydropathy Ratio (Fold)
    ID Number aMTD Sequences Length (PP) (II) (AI) (GRAVY) A B C
    56 805 LVLIAAAPIALP 12 8 41.3 220.0 2.4 22.3 14.9 7.6
    57 464 AVVILVPLAAAP 12 7 57.3 203.3 2.4 22.3 14.9 7.5
    58 405 LAAAVIPVAILP 12 7 54.9 211.7 2.4 22.2 14.8 7.5
    59 747 VALLAIAPALAP 12 8 57.3 195.8 2.2 22.0 14.8 7.5
    60 501 VIVALAVPALAP 12 8 50.2 203.3 2.4 21.5 14.4 7.3
    61 661 AAILAPIVAALP 12 6 50.2 195.8 2.2 21.4 14.3 7.2
    62 786 LVAIAPLAVLAP 12 6 41.3 211.7 2.4 21.2 14.2 7.2
    63 625 ILAAAAAPLIVP 12 8 50.2 195.8 2.2 20.9 13.9 7.0
    64 442 ALAALVPAVLVP 12 7 57.3 203.3 2.3 20.4 13.6 6.9
    65 912 VALLAPAVVVAP 12 6 57.3 195.0 2.3 19.9 13.3 6.7
    66 165 ALAVPVALAIVP 12 5 50.2 203.3 2.4 19.8 13.2 6.7
    67 422 VVAILAPLLAAP 12 7 57.3 211.7 2.4 19.6 13.1 6.6
    68 686 AALVAVLPVALP 12 8 57.3 203.3 2.3 19.5 13.1 6.6
    69 343 IVAVALPALVAP 12 7 50.2 203.3 2.3 19.4 12.9 6.5
    70 323 IVAVALPVALAP 12 7 50.2 203.3 2.3 19.1 12.8 6.4
    71 461 IAAVIVPAVALP 12 7 50.2 203.3 2.4 19.0 12.7 6.4
    72 21 AVALLPALLAVP 12 6 57.3 211.7 2.3 18.9 12.6 6.4
    73 404 LAAAVIPAAILP 12 7 54.9 195.8 2.2 18.9 12.6 6.4
    74 261 LVLVPLLAAAAP 12 5 41.3 211.6 2.3 18.5 12.3 6.2
    75 524 AVALIVVPALAP 12 8 50.2 203.3 2.4 18.3 12.2 6.2
    76 225 VAALLPAAAVLP 12 6 57.3 187.5 2.1 18.3 12.2 6.2
    77 264 LAAAPVVIVIAP 12 5 50.2 203.3 2.4 18.2 12.1 6.1
    78 1 AAALAPVVLALP 12 6 57.3 187.5 2.1 17.7 11.8 6.0
    79 382 AAALVIPAILAP 12 7 54.3 195.8 2.2 17.7 11.8 6.0
    80 463 AVAILVPLLAAP 12 7 57.3 211.7 2.4 17.6 11.7 5.9
    81 322 VVAIVLPALAAP 12 7 50.2 203.3 2.3 17.6 11.7 5.9
    82 503 AAIIIVLPAALP 12 8 50.2 223.0 2.4 17.6 11.8 5.9
    83 870 VLVAAVLPIAAP 12 8 41.3 203.3 2.4 16.6 11.1 5.6
    84 241 AAAVVPVLLVAP 12 6 57.3 195.0 2.4 16.6 11.0 5.6
    85 726 LAVAIIAPAVAP 12 8 57.3 187.5 2.2 16.5 11.0 5.6
    86 341 IVAVALPAVLAP 12 7 50.2 203.3 2.3 16.4 10.9 5.5
    87 542 ALALIIVPAVAP 12 8 50.2 211.6 2.4 16.2 10.8 5.5
    88 361 AVVIVAPAVIAP 12 7 50.2 195.0 2.4 16.0 10.7 5.4
    89 224 ILAAVPIALAAP 12 6 57.3 195.8 2.2 15.8 10.6 5.3
    90 482 ILAVAAIPVAVP 12 8 54.9 203.3 2.4 15.8 10.6 5.3
    91 64 AIVALPVAVLAP 12 6 50.2 203.3 2.4 15.8 10.6 5.3
    92 484 LAVVLAAPAIVP 12 8 50.2 203.3 2.4 15.6 10.4 5.3
    93 868 VLVAAILPAAIP 12 8 54.9 211.7 2.4 14.9 10.0 5.0
    94 541 LLALIIAPAAAP 12 8 57.3 204.1 2.1 14.8 9.9 5.0
    95 666 AAIAIIAPAIVP 12 8 50.2 195.8 2.3 14.7 9.9 5.0
    96 665 LAIVLAAPVAVP 12 8 50.2 203.3 2.3 14.7 9.9 5.0
    97 363 AVLAVAPALIVP 12 7 50.2 203.3 2.3 14.7 9.8 4.9
    98 242 AALLVPALVAAP 12 6 57.3 187.5 2.1 14.6 9.7 4.9
    99 384 VIVAIAPALLAP 12 7 50.2 211.6 2.4 14.0 9.4 4.7
    100 877 VAIIAVPAVVAP 12 7 57.3 195.0 2.4 14.0 9.4 4.7
    101 863 AAVVLLPIIAAP 12 7 41.3 211.7 2.4 13.8 9.3 4.7
    102 525 ALAIVVAPVAVP 12 8 50.2 195.0 2.4 13.8 9.2 4.7
    103 875 AIAIVVPAVAVP 12 7 50.2 195.0 2.4 13.8 9.2 4.7
    104 285 AIVLLPAAVVAP 12 6 50.2 203.3 2.4 13.3 8.9 4.5
    105 281 ALIVLPAAVAVP 12 6 50.2 203.3 2.4 13.3 8.9 4.5
    106 867 ALLVVIAPLAAP 12 8 41.3 211.7 2.4 13.2 8.8 4.4
    107 766 IVVIAVAPAVAP 12 8 50.2 195.0 2.4 12.9 8.6 4.4
    108 342 VIVALAPAVLAP 12 7 50.2 203.3 2.3 12.7 8.5 4.3
    109 881 AALIVVPAVAVP 12 7 50.2 195.0 2.4 12.7 8.5 4.3
    110 505 AIIIVIAPAAAP 12 8 50.2 195.8 2.3 12.4 8.3 4.2
  • TABLE 28
    Proline Rigidity/ Sturctural Relative
    Sequence Position Flexibility Feature Hydropathy Ratio (Fold)
    ID Number aMTD Sequences Length (PP) (II) (AI) (GRAVY) A B C
    111 763 VAVLIAVPALAP 12 8 57.3 203.3 2.3 12.3 7.2 4.2
    112 706 IVAVALLPAVAP 12 8 50.2 203.3 2.4 12.0 7.0 4.1
    113 687 AILAVALPLLAP 12 8 57.3 220.0 2.3 12.0 7.0 4.1
    114 643 LALVLAAPAIVP 12 8 50.2 211.6 2.4 11.8 7.9 4.0
    115 282 VLAVAPALIVAP 12 6 50.2 203.3 2.4 11.8 7.9 4.0
    116 543 LLAALIAPAALP 12 8 57.3 204.1 2.1 11.7 7.8 4.0
    117 325 IVAVALPAVALP 12 7 50.2 203.3 2.3 11.7 7.8 4.0
    118 846 IAVAVAAPLLVP 12 8 41.3 203.3 2.4 11.7 6.8 4.0
    119 383 VIVALAPALLAP 12 7 50.2 211.6 2.3 11.6 7.7 3.9
    120 381 VVAIVLPAVAAP 12 7 50.2 195.0 2.4 11.5 7.7 3.9
    121 808 LVVLAAAPLAVP 12 8 41.3 203.3 2.3 11.5 7.6 3.9
    122 865 AVLVIAVPAIAP 12 8 57.3 203.3 2.5 11.3 7.5 3.8
    123 725 IAVLAVAPAVLP 12 8 57.3 203.3 2.3 11.2 7.5 3.8
    124 844 VVALLAPLIAAP 12 7 41.3 211.8 2.4 11.2 7.5 3.8
    125 897 AVIVPVAIIAAP 12 5 50.2 203.3 2.5 11.2 7.5 3.8
    126 605 VIAAVLAPVAVP 12 8 57.3 195.0 2.4 11.0 7.4 3.7
    127 744 AAVVIVAPVALP 12 8 50.2 195.0 2.4 11.0 7.3 3.7
    128 221 AAILAPIVALAP 12 6 50.2 195.8 2.2 10.9 7.3 3.7
    129 622 ALIVLAAPVAVP 12 8 50.2 203.3 2.4 10.6 7.1 3.6
    130 401 AALAVIPAAILP 12 7 54.9 195.8 2.2 10.6 7.1 3.6
    131 324 IVAVALPAALVP 12 7 50.2 203.3 2.3 10.3 6.9 3.5
    132 878 IVALVAPAAVVP 12 7 50.2 195.0 2.4 10.3 6.9 3.5
    133 302 LALAPALALLAP 12 5 57.3 204.2 2.1 10.2 6.8 3.4
    134 635 ALLVAVLPAALP 12 8 57.3 211.7 2.3 10.2 5.9 3.4
    135 848 AVAIVVLPAVAP 12 8 50.2 195.0 2.4 10.0 6.7 3.4
    136 602 VIVALAAPVLAP 12 8 50.2 203.3 2.4 9.9 5.8 3.4
    137 788 AIAVAIAPVALP 12 8 57.3 187.5 2.3 9.8 6.6 3.3
    138 145 LLAVVPAVALAP 12 6 57.3 203.3 2.3 9.5 6.3 3.2
    139 11 VVALAPALAALP 12 6 57.3 187.5 2.1 9.5 6.3 3.2
    140 141 AVIVLPALAVAP 12 6 50.2 203.3 2.4 9.4 6.3 3.2
    141 521 LAALIVVPAVAP 12 8 50.2 203.3 2.4 9.4 6.3 3.2
    142 425 AVVAIAPVLALP 12 7 57.3 203.3 2.4 9.4 6.3 3.2
    143 365 AVIVVAPALLAP 12 7 50.2 203.3 2.3 9.3 6.2 3.1
    144 263 ALAVIPAAAILP 12 6 54.9 195.8 2.2 9.0 6.0 3.0
    145 345 ALLIVAPVAVAP 12 7 50.2 203.3 2.3 8.9 5.9 3.0
    146 850 LVIALAAPVALP 12 8 57.3 211.7 2.4 8.8 5.9 3.0
    147 144 VLAIVPAVALAP 12 6 50.2 203.3 2.4 8.8 5.9 3.0
    148 767 IVVAAVVPALAP 12 8 50.2 195.0 2.4 8.5 5.0 2.9
    149 185 AALVLPLIIAAP 12 6 41.3 220.0 2.4 8.5 5.7 2.9
    150 849 AVILLAPLIAAP 12 7 57.3 220.0 2.4 8.3 4.8 2.8
    151 864 ALLVIAPAIAVP 12 7 57.3 211.7 2.4 8.2 4.8 2.8
    152 162 AVVALPAALIVP 12 6 50.2 203.3 2.4 8.2 5.5 2.8
    153 164 LAAVLPALLAAP 12 6 57.3 195.8 2.1 8.2 5.5 2.8
    154 907 VAIALAPVVVAP 12 7 57.3 195.0 2.4 8.1 5.4 2.8
    155 444 LAAALVPVALVP 12 7 57.3 203.3 2.3 8.1 5.4 2.7
    156 443 ALAALVPVALVP 12 7 57.3 203.3 2.3 8.0 5.3 2.7
    157 901 ALVAVLPAVAVP 12 7 57.3 195.0 2.4 7.7 5.1 2.6
    158 887 VLAVAPAVAVLP 12 6 57.3 195.0 2.4 7.7 5.1 2.6
    159 746 VAIIVVAPALAP 12 8 50.2 203.3 2.4 7.6 4.4 2.6
    160 902 ALVAPLLAVAVP 12 5 41.3 203.3 2.3 7.6 5.1 2.6
    161 565 VAIVLVAPAVAP 12 8 50.2 195.0 2.4 7.5 5.0 2.5
    162 245 AAALAPVLALVP 12 6 57.3 187.5 2.1 7.5 5.0 2.5
    163 743 AIAIALVPVALP 12 8 57.3 211.6 2.4 7.4 4.9 2.5
    164 465 AVVILVPLAAAP 12 7 57.3 203.3 2.4 7.4 4.9 2.5
    165 104 AVVAAPLVLALP 12 6 41.3 203.3 2.3 7.3 4.9 2.5
  • TABLE 29
    Proline Rigidity/ Sturctural Relative
    Sequence Position Flexibility Feature Hydropathy Ratio (Fold)
    ID Number aMTD Sequences Length (PP) (II) (AI) (GRAVY) A B C
    166 707 IVALAVLPAVAP 12 8 50.2 203.3 2.4 7.3 4.9 2.5
    167 872 VLAAAVLPLVVP 12 8 41.3 219.2 2.5 7.3 4.9 2.5
    168 583 AVILALAPIVAP 12 8 50.2 211.6 2.4 7.3 4.8 2.4
    169 879 AAIVLLPAVVVP 12 7 50.2 219.1 2.5 7.2 4.8 2.4
    170 784 VAALPAVALVVP 12 5 57.3 195.0 2.4 7.1 4.7 2.4
    171 893 VIAIPAILAAAP 12 5 54.9 195.8 2.3 7.0 4.7 2.4
    172 13 AAALVPVVALLP 12 6 57.3 203.3 2.3 7.0 4.7 2.4
    173 809 LIVLAAPALAAP 12 7 50.2 195.8 2.2 7.0 4.7 2.4
    174 445 ALAALVPALVVP 12 7 57.3 203.3 2.3 6.9 4.6 2.3
    175 81 AALLPALAALLP 12 5 57.3 204.2 2.1 6.9 4.6 2.3
    176 667 LAVAIVAPALVP 12 8 50.2 203.3 2.3 6.9 4.5 2.3
    177 906 AVIALAPVVVAP 12 7 57.3 195.0 2.4 6.8 4.6 2.3
    178 483 ILAAAIIPAALP 12 8 54.9 204.1 2.2 6.8 4.5 2.3
    179 485 AILAAIVPLAVP 12 8 50.2 211.6 2.4 6.8 4.5 2.3
    180 421 AAILAAPLIAVP 12 7 57.3 195.8 2.2 6.7 4.5 2.3
    181 585 ALIVAIAPALVP 12 8 50.2 211.6 2.4 6.6 4.4 2.2
    182 424 AVVVAAPVLALP 12 7 57.3 195.0 2.4 6.6 4.4 2.2
    183 364 LVAAVAPALIVP 12 7 50.2 203.3 2.3 6.5 4.3 2.2
    184 402 ALAAVIPAAILP 12 7 54.9 195.8 2.2 6.4 4.3 2.2
    185 462 IAAVLVPAVALP 12 7 57.3 203.3 2.4 6.3 4.2 2.1
    186 265 VLAIAPLLAAVP 12 6 41.3 211.6 2.3 6.0 4.0 2.0
    187 301 VIAAPVLAVLAP 12 6 57.3 203.3 2.4 6.0 4.0 2.0
    188 183 LLAAPVVIALAP 12 6 57.3 211.6 2.4 6.0 4.0 2.0
    189 243 AAVLLPVALAAP 12 6 57.3 187.5 2.1 5.9 3.9 2.0
    190 664 ILIAIAIPAAAP 12 8 54.9 204.1 2.3 5.7 3.8 1.9
    191 783 IVALVPAVAIAP 12 6 50.2 203.3 2.5 5.7 3.8 1.9
    192 502 AIVALAVPVLAP 12 8 50.2 203.3 2.4 5.6 3.7 1.9
    193 262 ALIAVPAIIVAP 12 6 50.2 211.6 2.4 5.5 3.7 1.5
    194 683 LAIVLAAPAVLP 12 8 50.2 211.7 2.4 5.5 3.2 1.9
    195 830 IALVAAPVALVP 12 7 57.3 203.3 2.4 5.3 3.5 1.8
    196 764 AVALAVLPAVVP 12 8 57.3 195.0 2.3 5.0 3.4 1.7
    197 807 AVALAVPALVLP 12 7 57.3 203.3 2.3 5.0 3.3 1.7
    198 184 LAAIVPAIIAVP 12 6 50.2 211.6 2.4 4.8 3.2 1.6
    199 305 IALAAPILLAAP 12 6 57.3 204.2 2.2 4.8 3.2 1.6
    200 101 LVALAPVAAVLP 12 6 57.3 203.3 2.3 4.5 3.0 1.5
    201 304 AIILAPIAAIAP 12 6 57.3 204.2 2.3 4.4 3.0 1.5
    202 604 VALIAVAPAVVP 12 3 57.3 195.0 2.4 4.3 2.5 1.5
    203 645 ALAVVALPAIVP 12 8 50.2 203.3 2.4 4.3 2.9 1.5
    204 201 LALAVPALAALP 12 6 57.3 195.8 2.1 4.2 2.8 1.4
    205 163 LALVLPAALAAP 12 6 57.3 195.8 2.1 4.1 2.4 1.4
    206 832 AVAAIVPVIVAP 12 7 43.2 195.0 2.5 4.1 2.7 1.4
    207 182 ALIAPVVALVAP 12 6 57.3 203.3 2.4 4.0 2.7 1.4
    208 23 VVLVLPAAAAVP 12 6 57.3 195.0 2.4 4.0 2.6 1.3
    209 105 LLALAPAALLAP 12 6 57.3 204.1 2.1 4.0 2.6 1.3
    210 561 AAVAIVLPAVVP 12 8 50.2 195.0 2.4 3.9 2.6 1.3
    211 765 AVALAVVPAVLP 12 8 57.3 195.0 2.3 3.8 2.2 1.3
    212 684 AAIVLALPAVLP 12 8 50.2 211.7 2.4 3.5 2.1 1.2
    213 143 AVLAVPAVLVAP 12 6 57.3 195.0 2.4 3.3 2.2 1.1
    214 504 LIVALAVPALAP 12 8 50.2 211.7 2.4 3.3 2.2 1.1
    215 22 AVVLVPVLAAAP 12 6 57.3 195.0 2.4 3.1 2.1 1.1
    216 5 AAALLPVALVAP 12 6 57.3 187.5 21 3.1 2.1 1.0
    217 283 AALLAPALIVAP 12 6 50.2 195.8 2.2 3.1 2.0 1.0
    218 65 IAIVAPVVALAP 12 6 50.2 203.3 2.4 3.0 2.0 1.0
    219 883 LAIVPAAIAALP 12 6 50.2 195.8 2.2 3.0 2.0 1.0
    220 123 AAIIVPAALLAP 12 6 50.2 195.8 2.2 2.9 2.0 1.0
  • TABLE 30
    Proline Rigidity/ Sturctural Relative
    Sequence Position Flexibility Feature Hydropathy Ratio (Fold)
    ID Number aMTD Sequences Length (PP) (II) (AI) (GRAVY) A B C
    221 284 ALIAPAVALIVP 12 5 50.2 211.7 2.4 2.8 1.8 0.9
    222 205 ALALVPAIAALP 12 6 57.3 195.8 2.2 2.6 1.7 0.9
    223 42 VAALPVVAVVAP 12 5 57.3 186.7 2.4 2.5 1.7 0.8
    224 121 AIVALPALALAP 12 6 50.2 195.8 2.2 2.5 1.7 0.8
    225 25 IVAVAPALVALP 12 6 50.2 203.3 2.4 2.4 1.6 0.8
    226 24 IALAAPALIVAP 12 6 50.2 195.8 2.2 2.3 1.6 0.8
    227 204 LIAALPAVAALP 12 6 57.3 195.8 2.2 2.2 1.5 0.8
    228 12 LLAAVPAVLLAP 12 6 57.3 211.7 2.3 2.2 1.5 0.7
    229 43 LLAAPLVVAAVP 12 5 41.3 187.5 2.1 2.1 1.4 0.7
    230 103 ALIAAPILALAP 12 6 57.3 204.2 2.2 2.1 1.4 0.7
    231 82 AVVLAPVAAVLP 12 6 57.3 195.0 2.4 2.1 1.4 0.7
    232 4 ALALLPVAALAP 12 6 57.3 195.8 2.1 2.0 1.3 0.7
    233 85 LLVLPAAALAAP 12 5 57.3 195.8 2.1 1.9 1.3 0.7
    234 63 AALLVPALVAVP 12 6 57.3 203.3 2.3 1.9 1.3 0.7
    235 44 ALAVPVALLVAP 12 5 57.3 203.3 2.3 1.6 1.1 0.5
    236 84 AAVAAPLLLALP 12 6 41.3 195.8 2.1 1.5 1.0 0.5
    237 62 VALLAPVALAVP 12 6 57.3 203.3 2.3 1.4 0.9 0.5
    238 83 LAVAAPLALALP 12 6 41.3 195.8 2.1 1.4 0.9 0.5
    239 102 LALAPAALALLP 12 5 57.3 204.2 2.1 1.4 0.9 0.5
    240 623 VAAAIALPAIVP 12 8 50.2 187.5 2.3 0.8 0.6 0.3
    19.6 ± 13.1 ± 1.1 6.6 ±
    1.6 0.5
  • Moreover, compared to reference CPPs (B type: MTM12 and C type: MTD85), novel 240 aMTDs averaged of 13±1.1 (maximum 109.9) and 6.6±0.5 (maximum 55.5) fold higher cell-permeability, respectively (Tables 26-31).
  • TABLE 31
    Negative Control
    rP38 MTM12 MTD85
    aMTD 19.6 ± 1.6* 13.1 ± 1.1* 6.6 ± 0.5*
    The Average (Best: 164.2) (Best: 109.9) (Best: 55.5)
    of 240 aMTDs
    *Relative Fold (aMTD in Geo Mean in its comparison to rP38, MTM12 or MTD85)
  • In addition, cell-permeability of 31 rPeptides has been compared with that of 240 aMTDs (0.3±0.04; Tables 32 and 33).
  • TABLE 32
    Proline Rigidity/ Sturctural
    Position Flexibility Feature Hydropathy Relative Ratio
    Number ID Sequence Length (PP) (II) (AI) (GRAVY) to aMTD AVE
    1 692 PAPLPPVVILAV 12 1, 3, 5, 6 105.5 186.7 1.8 0.74
    2 26 AAIALAAPLAIV 12 8 18.1 204.2 2.5 0.65
    3 113 PVAVALLIAVPP 12 1, 11, 12 57.3 195.0 2.1 0.61
    4 466 IIAAAAPLAIIP 12 7, 12 22.8 204.2 2.3 0.52
    5 167 VAIAIPAALAIP 12 6, 12 20.4 195.8 2.3 0.50
    6 97 ALLAAPPALLAL 12 6, 7 57.3 204.2 2.1 0.41
    7 390 VPLLVPVVPVVP 12 2, 6, 9, 12 105.4 210.0 2.2 0.41
    8 426 AAALAIPLAIIP 12 7, 12 4.37 204.2 2.2 0.40
    9 214 ALIVAPALMALP 12 6, 12 60.5 187.5 2.2 0.33
    10 68 VAPVLPAAPLVP 12 3, 6, 9, 12 105.5 162.5 1.6 0.32
    11 39 CYNTSPCTGCCY 12 6 52.5 0.0 0.0 0.29
    12 934 LILAPAAVVAAA 12 5 57.3 195.8 2.5 0.28
    13 938 VPVLLPVVVPVP 12 2, 6, 10, 12 121.5 210.0 2.2 0.28
    14 329 LPVLVPVVPVVP 12 2, 6, 9, 12 121.5 210.0 2.2 0.23
    15 606 AAAIAAIPIIIP 12 8, 12 4.4 204.2 2.4 0.20
    16 49 VVPAAPAVPVVP 12 3, 6, 9, 12 121.5 145.8 1.7 0.18
    17 139 TGSTNSPTCTST 12 7 53.4 0.0 −0.7 0.17
    18 772 LPVAPVIPIIVP 12 2, 5, 8, 12 79.9 210.8 2 1 0.16
    19 921 IWWFVVLPLVVP 12 8, 12 41.3 194.2 2.2 0.14
    20 66 AGVLGGPIMGVP 12 7, 12 35.5 121.7 1.3 0.13
    21 693 AAPVLPVAVPIV 12 3, 6, 10 82.3 186.7 2.1 0.13
    22 18 NYCCTPTTNGQS 12 6 47.9 0.0 −0.9 0.10
    23 16 NNSCTTYTNGSQ 12 None 47.4 0.0 −1.4 0.08
    24 227 LAAIVPIAAAVP 12 6, 12 34.2 187.5 2.2 0.08
    25 17 GGCSAPQTTCSN 12 6 51.6 8.3 −0.5 0.08
    26 67 LDAEVPLADDVP 12 6, 12 34.2 130.0 0.3 0.08
    27 635 GSTGGSQQNNQY 12 None 31.9 0.0 −1.9 0.07
    28 29 VLPPLPVLPVLP 12 3, 4, 6, 9, 12 121.5 202.5 1.7 0.07
    29 57 QNNCNTSSQGGG 12 None 52.4 0.0 −1.6 0.06
    30 700 GTSNTCQSNQNS 12 None 19.1 0.0 −1.6 0.05
    31 38 YYNQSTCGGQCY 12 ND 53.8 0.0 −1.0 0.05
    AVE 0.3 ± 0.04
  • TABLE 33
    Relative Ratio to
    aMTD AVE*
    rPeptide 0.3 ± 0.04
    The Average of 31 aMTDs
    *Out of 240 aMTDs, average relative fold of aMTD had been 19.6 fold compared to type A (rP38).
  • In summary, relative cell-permeability of aMTDs has shown maximum of 164.0, 109.9 and 55.5 fold higher to rP38, MTM12 and MTD85, respectively. In average of total 240 aMTD sequences, 19.6±1.6, 13.1±1.1 and 6.6±0.5 fold higher cell-permeability are shown to the rP38, MTM12 and MTD85, respectively (Tables 26-31). Relative cell-permeability of negative control (rP38) to the 240 aMTDs is only 0.3±0.04 fold.
  • 4-5. Intracellular Delivery and Localization of aMTD-Fused Recombinant Proteins
  • Recombinant proteins fused to the aMTDs were tested to determine their intracellular delivery and localization by laser scanning confocal microscopy with a negative control (rP38) and previous published CPPs (MTM12 and MTD85) as the positive control references. NIH3T3 cells were exposed to 10 μM of FITC-labeled protein for 1 hour at 37, and nuclei were counterstained with DAPI. Then, cells were examined by confocal laser scanning microscopy (FIG. 7). Recombinant proteins fused to aMTDs clearly display intracellular delivery and cytoplasmic localization (FIG. 7) that are typically higher than the reference CPPs (MTM12 and MTD85). The rP38-fused recombinant protein did not show internalized fluorescence signal (FIG. 7a ). In addition, as seen in FIG. 8, rPeptides (his-tagged CRA recombinant proteins fused to each rPeptide) display lower- or non-cell-permeability.
  • 4-6. Summary of Quantitative and Visual Cell-Permeability of Newly Developed aMTDs
  • Histidine-tagged aMTD-fused cargo recombinant proteins have been greatly enhanced in their solubility and yield. Thus, FITC-conjugated recombinant proteins have also been tested to quantitate and visualize intracellular localization of the proteins and demonstrated higher cell-permeability compared to the reference CPPs.
  • In the previous studies using the hydrophobic signal-sequence-derived CPPs—MTS/MTM or MTDs, 17 published sequences have been identified and analyzed in various characteristics such as length, molecular weight, pI value, bending potential, rigidity, flexibility, structural feature, hydropathy, amino acid residue and composition, and secondary structure of the peptides. Based on these analytical data of the sequences, novel artificial and non-natural peptide sequences designated as advanced MTDs (aMTDs) have been invented and determined their functional activity in intracellular delivery potential with aMTD-fused recombinant proteins.
  • aMTD-fused recombinant proteins have promoted the ability of protein transduction into the cells compared to the recombinant proteins containing rPeptides and/or reference hydrophobic CPPs (MTM12 and MTD85). According to the results, it has been demonstrated that critical factors of cell-penetrating peptide sequences play a major role to determine peptide-mediated intracellular delivery by penetrating plasma membrane. In addition, cell-permeability can considerably be improved by following the rational that all satisfy the critical factors.
  • 5. Structure/Sequence Activity Relationship (SAR) of aMTDs on Delivery Potential
  • After determining the cell-permeability of novel aMTDs, structure/sequence activity relationship (SAR) has been analyzed for each critical factor in selected some of and all of novel aMTDs (FIGS. 13 to 16 and Table 34).
  • TABLE 34
    Rank of Rigidity/ Sturctural Relative Ratio Amino Acid
    Delivery Flexibility Feature Hydropathy (Fold) Composition
    Potential (II) (AI) (GRAVY) A B C A V I L
     1~10 55.9 199.2 2.3 112.7 75.5 38.1 4.0 3.5 0.4 2.1
    11~20 51.2 205.8 2.4 56.2 37.6 19.0 4.0 2.7 1.7 1.6
    21~30 49.1 199.2 2.3 43.6 28.9 14.6 4.3 2.7 1.4 1.6
    31~40 52.7 201.0 2.4 34.8 23.3 11.8 4.2 2.7 1.5 1.6
    41~50 53.8 201.9 2.3 30.0 20.0 10.1 4.3 2.3 1.1 2.3
    51~60 51.5 205.2 2.4 23.5 15.7 7.9 4.4 2.1 1.5 2.0
    222~231 52.2 197.2 2.3 2.2 1.5 0.8 4.5 2.1 1.0 2.4
    232~241 54.1 199.7 2.2 1.7 1.2 0.6 4.6 1.7 0.2 3.5
  • 5-1. Proline Position:
  • In regards to the bending potential (proline position: PP), aMTDs with its proline at 7′ or 8′ amino acid in their sequences have much higher cell-permeability compared to the sequences in which their proline position is at 5′ or 6′ (FIGS. 14a and 15a ).
  • 5-2. Hydropathy:
  • In addition, when the aMTDs have GRAVY (Grand Average of Hydropathy) ranging in 2.1-2.2, these sequences display relatively lower cell-permeability, while the aMTDs with 2.3-2.6 GRAVY are shown significantly higher one (FIGS. 14b and 15b ).
  • 5-3. rPeptide SAR:
  • To the SAR of aMTDs, rPeptides have shown similar SAR correlations in the cell-permeability, pertaining to their proline position (PP) and hydropathy (GRAVY). These results confirms that rPeptides with high GRAVY (2.4˜2.6) have better cell-permeability (FIG. 16).
  • 5-4. Analysis of Amino Acid Composition:
  • In addition to proline position and hydropathy, the difference of amino acid composition is also analyzed. Since aMTDs are designed based on critical factors, each aMTD-fused recombinant protein has equally two proline sequences in the composition. Other hydrophobic and aliphatic amino acids—alanine, isoleucine, leucine and valine —are combined to form the rest of aMTD peptide sequences.
  • Alanine: In the composition of amino acids, the result does not show a significant difference by the number of alanine in terms of the aMTD's delivery potential because all of the aMTDs have three to five alanines. In the sequences, however, four alanine compositions show the most effective delivery potential (geometric mean) (FIG. 13a ).
  • Leucine and Isoleucine: Also, the compositions of isoleucine and leucine in the aMTD sequences show inverse relationship between the number of amino acid (I and L) and delivery potential of aMTDs. Lower number of isoleucine and leucine in the sequences tends to have higher delivery potential (geometric mean) (FIGS. 13a and 13b ).
  • Valine: Conversely, the composition of valine of aMTD sequences shows positive correlation with their cell-permeability. When the number of valine in the sequence is low, the delivery potential of aMTD is also relatively low (FIG. 13b ).
  • Ten aMTDs having the highest cell-permeability are selected (average geometric mean: 2584±126). Their average number of valine in the sequences is 3.5; 10 aMTDs having relatively low cell-permeability (average geometric mean: 80±4) had average of 1.9 valine amino acids. The average number of valine in the sequences is lowered as their cell-permeability is also lowered as shown in FIG. 13b . Compared to higher cell-permeable aMTDs group, lower sequences had average of 1.9 in their valine composition. Therefore, to obtain high cell-permeable sequence, an average of 2-4 valines should be composed in the sequence.
  • 5-5. Conclusion of SAR Analysis:
  • As seen in FIG. 15, all 240 aMTDs have been examined for these association of the cell-permeability and the critical factors: bending potential (PP), rigidity/flexibility (II), structure feature (AI), and hydropathy (GRAVY), amino acid length and composition. Through this analysis, cell-permeability of aMTDs tends to be lower when their central proline position is at 5′ or 6′ and GRAVY is 2.1 or lower (FIG. 15). Moreover, after investigating 10 higher and 10 lower cell-permeable aMTDs, these trends are clearly shown to confirm the association of cell-permeability with the central proline position and hydropathy.
  • 6. Experimental Confirmation of Index Range/Feature of Critical Factors
  • The range and feature of five out of six critical factors have been empirically and experimentally determined that are also included in the index range and feature of the critical factors initially proposed before conducting the experiments and SAR analysis. In terms of index range and feature of critical factors of newly developed 240 aMTDs, the bending potential (proline position: PP), rigidity/flexibility (Instability Index: II), structural feature (Aliphatic Index: AI), hydropathy (GRAVY), amino acid length and composition are all within the characteristics of the critical factors derived from analysis of reference hydrophobic CPPs.
  • Therefore, our hypothesis to design and develop new hydrophobic CPP sequences as advanced MTDs is empirically and experimentally proved and demonstrated that critical factor-based new aMTD rational design is correct.
  • TABLE 35
    Summarized Critical Factors of aMTD
    Analysis of
    Newly Designed CPPs Experimental Results
    Critical Factor Range Range
    Bending Potential Proline presences in Proline presences in
    (Proline Position: PP) the middle (5′, 6′, 7′ the middle (5′, 6′, 7′
    or 8′) and at the or 8′) and at the
    end of peptides end of peptides
    Rigidity/Flexibility 40-60 41.3-57.3
    (Instability Index: II)
    Structural Feature 180-220 187.5-220.0
    (Aliphatic Index: AI)
    Hydropathy 2.1-2.6 2.2-2.6
    (Grand Average of
    Hydropathy GRAVY)
    Length  9-13 12
    (Number of Amino Acid)
    Amino acid Composition A, V, I, L, P A, V, I, L, P

    7. Discovery and Development of Protein-Based New Biotherapeutics with MITT Enabled by aMTDs for Protein Therapy
  • Total of 240 aMTD sequences have been designed and developed based on the critical factors. Quantitative and visual cell-permeability of 240 aMTDs (hydrophobic, flexible, bending, aliphatic and 12 a/a-length peptides) are all practically determined.
  • To measure the cell-permeability of aMTDs, rPeptides have also been designed and tested. As seen in FIGS. 13 to 15, there are vivid association of cell-permeability and the critical factors of the peptides. Out of these critical factors, we are able to configure that the most effective cell-permeable aMTDs have the amino acid length of 12; composition of A, V, L, I and P; multiple proline located at either 7′ or 8′ and at the end (12′); instability index ranged of 41.3-57.3; aliphatic index ranged of 187.5-220.0; and hydropathy (GRAVY) ranged of 2.2-2.6.
  • These examined critical factors are within the range that we have set for our critical factors; therefore, we are able to confirm that the aMTDs that satisfy these critical factors have relatively high cell-permeability and much higher intracellular delivery potential compared to reference hydrophobic CPPs reported during the past two decades.
  • It has been widely evident that many human diseases are caused by proteins with deficiency or over-expression that causes mutations such as gain-of-function or loss-of-function. If biologically active proteins could be delivered for replacing abnormal proteins within a short time frame, possibly within an hour or two, in a quantitative manner, the dosage may be regulated depending on when and how proteins may be needed. By significantly improving the solubility and yield of novel aMTD in this invention (Table 31), one could expect its practical potential as an agent to effectively deliver therapeutic macromolecules such as proteins, peptides, nucleic acids, and other chemical compounds into live cells as well as live mammals including human. Therefore, newly developed MITT utilizing the pool (240) of novel aMTDs can be used as a platform technology for discovery and development of protein-based biotherapeutics to apprehend intracellular protein therapy after determining the optimal cargo-aMTD relationship.
  • The following examples are presented to aid practitioners of the invention, to provide experimental support for the invention, and to provide model protocols. In no way are these examples to be understood to limit the invention.
  • Example 1. Development of Novel Advanced Macromolecule Transduction Domain (aMTD)
  • H-regions of signal sequences (HRSP)-derived CPPs (MTS/MTM and MTD) do not have a common sequence, a sequence motif, and/or a common structural homologous feature. In this invention, the aim is to develop improved hydrophobic CPPs formatted in the common sequence and structural motif that satisfy newly determined ‘critical factors’ to have a ‘common function,’ to facilitate protein translocation across the plasma membrane with similar mechanism to the analyzed CPPs.
  • The structural motif as follows:
  • In Table 9, universal common sequence/structural motif is provided as follows. The amino acid length of the peptides in this invention ranges from 9 to 13 amino acids, mostly 12 amino acids, and their bending potentials are dependent with the presence and location of proline in the middle of sequence (at 5′, 6′, 7′ or 8′ amino acid) and at the end of peptide (at 12′) for recombinant protein bending. Instability index (II) for rigidity/flexibility of aMTDs is II<40, grand average of hydropathy (GRAVY) for hydropathy is around 2.2, and aliphatic index (AI) for structural features is around 200 (Table 9). Based on these standardized critical factors, new hydrophobic peptide sequences, namely advanced macromolecule transduction domain peptides (aMTDs), in this invention have been developed and summarized in Tables 10 to 15.
  • Example 2. Construction of Expression Vectors for Recombinant Proteins Fused to aMTDs
  • Our newly developed technology has enabled us to expand the method for making cell-permeable recombinant proteins. The expression vectors were designed for histidine-tagged CRA proteins fused with aMTDs or rPeptides. To construct expression vectors for recombinant proteins, polymerase chain reaction (PCR) had been devised to amplify each designed aMTD or rPeptide fused to CRA.
  • The PCR reactions (100 ng genomic DNA, 10 pmol each primer, each 0.2 mM dNTP mixture, 1× reaction buffer and 2.5 U Pfu(+) DNA polymerase (Doctor protein, Korea) was digested on the restriction enzyme site between Nde I (5′) and Sal I (3′) involving 35 cycles of denaturation (95° C.), annealing (62° C.), and extension (72° C.) for 30 seconds each. For the last extension cycle, the PCR reactions remained for 5 minutes at 72° C. Then, they were cloned into the site of pET-28a(+) vectors (Novagen, Darmstadt, Germany). DNA ligation was performed using T4 DNA ligase at 4° C. overnight. These plasmids were mixed with competent cells of E. coli DH5-alpha strain on the ice for 10 minutes. This mixture was placed on the ice for 2 minutes after it was heat shocked in the water bath at 42° C. for 90 seconds. Then, the mixture added with LB broth media was recovered in 37° C. shaking incubator for 1 hour. Transformant was plated on LB broth agar plate with kanamycin (50 μg/mL) (Biopure, Johnson City, Tenn., USA) before incubating at 37° C. overnight. From a single colony, plasmid DNA was extracted, and after the digestion of Nde I and Sal I restriction enzymes, digested DNA was confirmed at 645 bp by using 1.2% agarose gels electrophoresis (FIG. 2). PCR primers for the CRA recombinant proteins fused to aMTD and random peptides (rPeptide) are summarized in Tables 23 to 30. Amino acid sequences of aMTD and rPeptide primers are shown in Tables 31 to 38.
  • Example 3. Inducible Expression, Purification and Preparation of Recombinant Proteins Fused to aMTDs and rPeptides
  • To express recombinant proteins, pET-28a(+) vectors for the expression of CRA proteins fused to a negative control [rPeptide 38 (rP38)], reference hydrophobic CPPs (MTM12 and MTD85) and aMTDs were transformed in E. coli BL21 (DE3) strains. Cells were grown at 37° C. in LB medium containing kanamycin (50 μg/ml) with a vigorous shaking and induced at OD600=0.6 by adding 0.7 mM IPTG (Biopure) for 2 hours at 37° C. Induced recombinant proteins were loaded on 15% SDS-PAGE gel and stained with Coomassie Brilliant Blue (InstantBlue, Expedeon, Novexin, UK) (FIG. 3).
  • The E. coli cultures were harvested by centrifugation at 5,000× rpm for 10 minutes, and the supernatant was discarded. The pellet was re-suspended in the lysis buffer (50 mM NaH2PO4, 10 mM Imidazol, 300 mM NaCl, pH 8.0). The cell lysates were sonicated on ice using a sonicator (Sonics and Materials, Inc., Newtown, Conn., USA) equipped with a probe. After centrifuging the cell lysates at 5,000× rpm for 10 minutes to pellet the cellular debris, the supernatant was incubated with lysis buffer-equilibrated Ni-NTA resin (Qiagen, Hilden, Germany) gently by open-column system (Bio-rad, Hercules, Calif., USA). After washing protein-bound resin with 200 ml wash buffer (50 mM NaH2PO4, 20 mM Imidazol, 300 mM NaCl, pH 8.0), the bounded proteins were eluted with elution buffer (50 mM NaH2PO4, 250 mM Imidazol, 300 mM NaCl, pH 8.0).
  • Recombinant proteins purified under natural condition were analyzed on 15% SDS-PAGE gel and stained with Coomassie Brilliant Blue (FIG. 4). All of the recombinant proteins were dialyzed for 8 hours and overnight against physiological buffer, a 1:1 mixture of cell culture medium (Dulbecco's Modified Eagle's Medium: DMEM, Hyclone, Logan, Utah, USA) and Dulbecco's phosphate buffered saline (DPBS, Gibco, Grand Island, N.Y., USA). From 316 aMTDs and 141 rPeptides cloned, 240 aMTD- and 31 rPeptide-fused recombinant proteins were induced, purified, prepared and analyzed for their cell-permeability.
  • Example 4. Determination of Quantitative Cell-Permeability of Recombinant Proteins
  • For quantitative cell-permeability, the aMTD- or rPeptide-fused recombinant proteins were conjugated to fluorescein isothiocyanate (FITC) according to the manufacturer's instructions (Sigma-Aldrich, St. Louis, Mo., USA). RAW 264.7 cells were treated with 10 μM FITC-labeled recombinant proteins for 1 hour at 37° C., washed three times with cold PBS, treated with 0.25% tripsin/EDTA (Sigma-Aldrich, St. Louis, Mo.) for 20 minutes at 37° C. to remove cell-surface bound proteins. Cell-permeability of these recombinant proteins were analyzed by flow cytometry (Guava, Millipore, Darmstadt, Germany) using the FlowJo cytometric analysis software (FIGS. 5 to 6). The relative cell-permeability of aMTDs were measured and compared with the negative control (rP38) and reference hydrophobic CPPs (MTM12 and MTD85) (Table 31).
  • Example 5. Determination of Cell-Permeability and Intracellular Localization of Recombinant Proteins
  • For a visual reference of cell-permeability, NIH3T3 cells were cultured for 24 hours on coverslip in 24-wells chamber slides, treated with 10 μM FITC-conjugated recombinant proteins for 1 hour at 37° C., and washed three times with cold PBS. Treated cells were fixed in 4% paraformaldehyde (PFA, Junsei, Tokyo, Japan) for 10 minutes at room temperature, washed three times with PBS, and mounted with VECTASHIELD Mounting Medium (Vector laboratories, Burlingame, Calif., USA), and counter stained with DAPI (4′,6-diamidino-2-phenylindole). The intracellular localization of the fluorescent signal was determined by confocal laser scanning microscopy (LSM700, Zeiss, Germany; FIGS. 7 and 8).
  • Example 6-1. Cloning of aMTD/SD-Fused SOCS3 Recombinant Protein
  • Full-length cDNA for human SOCS3 (SEQ ID NO: 815) was purchased from Origene (USA). Histidine-tagged human SOCS3 proteins were constructed by amplifying the SOCS3 cDNA (225 amino acids) using primers for aMTD fused to SOCS3 cargo. The PCR reactions (100 ng genomic DNA, 10 pmol each primer, each 0.2 mM dNTP mixture, 1× reaction buffer and 2.5 U Pfu(+) DNA polymerase (Doctor protein, Korea)) were digested on the restriction enzyme site between Nde I (5′) and Sal I (3′) involving 35 cycles of denaturing (95° C.), annealing (62° C.), and extending (72° C.) for 45 sec each. For the last extension cycle, the PCR reactions remained for 10 min at 72° C. The PCR products were subcloned into 6× His expression vector, pET-28a(+) (Novagen, Darmstadt, Germany). Coding sequence for SDA or SDB fused to C terminus of his-tagged aMTD-SOCS3 was cloned at BamHI (5′) and SalI (3′) in pET-28a(+) from PCR-amplified DNA segments and confirmed by DNA sequence analysis of the resulting plasmids.
  • TABLE 36
    Cargo SD Recombinant Protein 5′ Primers 3′ Primers
    SOCS3 HS3 5′- 5′-
    GGAATTCCATATGGTCACCCAC CCCGGATCCTAAA
    AGCAAGTTTCCCGCCGCC-3′ GCGGGGCATCGTAC
    TGGTCCAGGAA-3′
    HM165S3 5′- 5′-
    GGAATTCCATATGGCGCTGGCG CCCGGATCCTAAA
    GTGCCGGTGGCGCTGGCGATT GCGGGGCATCGTAC
    GTGCCGGTCACCCACAGCAAG TGGTCCAGGAA-3′
    TTTC-3′
    A HM165S3A 5′- 5′-
    GGAATTCCATATGGGGCTGGCG CGCGTCGACTTACC
    GTGCCGGTGGCGCTGGCGATT TCGGCTGCACCGG
    GTGCCGGTCACCCACAGCAAG ACGGCGATAC-3′
    TTTC-3′
    B HM165S3B 5′- 5′-
    GGAATTCCATATGGCGCTGGCG CGCGTCGACTTAAA
    GTGCCGGTGGCGCTGGCGATT GGGTTTCCGAAGGC
    GTGCCGGTCACCCACAGCAAG TTGGCTATCTT-3′
    TTTC-3′
    C HM165S3C 5′- 5′-
    GGAATTCCATATGGCGCTGGCG GCGTCGACTTAGGC
    GTGCCGGTGGCGCTGGCGATT CAGGTTAGCGTCGA
    GTGCCGGTCACCCACAGCAAG G-3′
    TTTC-3′
    D HM165S3D 5′- 5′-
    GGAATTCCATATGGCGCTGGCG GCGTCGACTTATTTT
    GTGCCGGTGGCGCTGGCGATT TTCTCGGACAGATA-
    GTGCCGGTCACCCACAGCAAG 3′
    TTTC-3′
    E HM165S3E 5′- 5′-
    GGAATTCCATATGGCGCTGGCG ACGCGTCGACTTAA
    GTGCCGGTGGCGCTGGCGATT CCTCCAATCTGTTC
    GTGCCGGTCACCCACAGCAAG GCGGTGAGCCTC-3′
    TTTC-3′
  • Example 6-2. Preparation of aMTD/SD-Fused SOCS3 Recombinant Protein
  • To determine a stable structure of the cell-permeable aMTD/SD-fused SOCS3 recombinant protein, a pET-28a(+) vector and an E. coli BL21-CodonPlus (DE3)-RIL were subjected to the following experiment.
  • Each of the recombinant expression vectors, HS3, HMS3, HMS3A, HMS3B, HMS3C, HMS3D, and HMS3E prepared in example 6-1 was transformed into E. coli BL21 CodonPlus(DE3)-RIL by a heat shock method, and then 600 ul of each was incubated in an LB medium (Biopure, Johnson City, Tenn., USA) containing 50 μg/ml of kanamycin at 37° C. for 1 hour. Thereafter, the recombinant protein gene-introduced E. coli was inoculated in 7 ml of LB medium, and then incubated at 37° C. overnight. The E. coli was inoculated in 700 ml of LB medium and incubated at 37° C. until OD600 reached 0.6. To this culture medium, 0.6 mM of isopropyl-β-D-thiogalactoside (IPTG, Gen Depot, USA) was added as a protein expression inducer, followed by further incubation at 37° C. for 3 hours. This culture medium was centrifuged at 4° C. and 8,000 rpm for 10 minutes and a supernatant was discarded to recover a cell pellet. The cell pellet thus recovered was suspended in a lysis buffer (100 mM NaH2PO4, 10 mM Tris-HCl, 8 M Urea, pH 8.0), and cells were disrupted by sonication (on/off time: 30 sec/30 sec, on time 2 hours, amplify 40%), and centrifuged at 15,000 rpm for 30 min to obtain a soluble fraction and an insoluble fraction.
  • This insoluble fraction was suspended in a denature lysis buffer (8 M Urea, 10 mM Tris, 100 mM Sodium phosphate) and purified by Ni2+ affinity chromatography as directed by the supplier(Qiagen, Hilden, Germany) and refolded by dialyzing with a refolding buffer (0.55 M guanidine HCl, 0.44 M L-arginine, 50 mM Tris-HCl, 150 mM NaCl, 1 mM EDTA, 100 mM NDSB, 2 mM reduced glutathione, and 0.2 mM oxidized glutathione). After purification, the proteins were put in a SnakeSkin Dialysis Tubing bag (pore size: 10000 mw, Thermo scientific, USA) and then they were dialyzed by physiological buffer (DMEM). The strain lysate where protein expression was not induced, the strain lysate where protein expression was induced by addition of IPTG, and purified proteins were loaded on SDS-PAGE to analyze protein expression characteristics and expression levels (FIGS. 19 and 20).
  • As shown in FIG. 19, it was confirmed that SOCS3 recombinant proteins showed high expression levels in the BL21CodonPlus(DE3)-RIL strain. SOCS3 recombinant proteins containing aMTD165 and solubilization domain (HM165S3A and HM165S3B) had little tendency to precipitate whereas recombinant SOCS3 proteins lacking a solubilization domain (HM165S3) or lacking an aMTD and a SD (HS3) were largely insoluble. Solubility of aMTD/SD-fused SOCS3 proteins was scored on a 5 point scale compared with that of SOCS3 proteins lacking the solubilization domain.
  • Example 6-3. Determination of Solubility/Yield of aMTD/SD-Fused SOCS3 Recombinant Proteins According to SD Type
  • To determine aMTD/SD-fused SOCS3 recombinant proteins having optimal cell-permeability, solubilization domains were replaced in the same manner as in Example 6-2 to prepare 5 kinds of aMTD/SD-fused SOCS3 recombinant proteins, and their solubility/yield were measured (FIG. 19).
  • As shown in FIG. 19, it was confirmed that the aMTD/SD-fused SOCS3 recombinant protein prepared by fusing with SDB among the different SDs showed the highest solubility/yield. Therefore, the SDB-fused iCP-SOCS3 recombinant protein was used in the subsequent experiment.
  • Example 6-4. Comparison Between aMTD/SD-Fused SOCS3 Recombinant Protein and Basic CPP/SD-Fused SOCS3 Recombinant Protein
  • To compare the solubility/yield, cell/tissue-permeability, mechanism of cytopermeability of aMTD/SD-fused SOCS3 recombinant proteins to those of conventional basic CPP/SD-fused SOCS3 recombinant proteins, cloning, preparation, and measurement of solubility/yield of the basic CPP/SD-fused SOCS3 recombinant proteins were performed in the same manner as in Examples 6-1 to 6-3 except for a known basic CPP (TAT or PolyR) being used instead of aMTD. Sequences of amino acids and nucleotides of basic CPP, and the primers used in this example are shown in FIG. 97.
  • The solubility/yield of aMTD165/SD-fused SOCS3 recombinant proteins was much higher than that of TAT/SD-fused SOCS3 or PolyR/SD-fused SOCS3 recombinant proteins (FIG. 98).
  • Example 7-1. Cell-Permeability Test
  • To examine cell-permeability of SOCS3 recombinant protein, SOCS3 recombinant proteins were conjugated to 5/6-fluorescein isothiocyanate (FITC). RAW 264.7 (KCLB, Seoul, South Korea) (FIG. 20) or NIH3T3 cells (KCLB, Seoul, South Korea) (FIG. 21) were treated with 10 μM FITC-labeled SOCS3 recombinant proteins and cultivated for 1 hr at 37° C.
  • In this regard, RAW 264.7 cells were cultured in a DMEM medium containing 10% fetal bovine serum (FBS, Hyclone, USA) and 500 mg/ml of 1% penicillin/streptomycin (Hyclone, USA).
  • After cultivation, the cells were washed three times with ice-cold PBS (Phosphate-buffered saline, Hyclone, USA) and treated with proteinase K (10 μg/mL, SIGMA, USA) to remove surface-bound proteins, and internalized proteins were measured by flow cytometry (FlowJo cytometric analysis software, Guava, Millipore, Darmstadt, Germany). Untreated cells (gray) and equimolar concentration of unconjugated FITC (FITC only, green)-treated cells were served as control (FIG. 20). Each of NIH3T3 cells was incubated for 1 hour at 37° C. with 10 μM FITC-labeled SOCS3 protein. For nuclear staining, a mixture of VECTASHIELD Mounting Medium (Vector Laboratories, Burlingame, Calif.) and DAPI (4′,6-diamidino-2-phenylindole) was added to NIH3T3 cells, and visualized using a confocal laser microscope (LSM700, Zeiss, Germany) (FIG. 21).
  • As shown in FIGS. 20 and 21, SOCS3 recombinant proteins containing aMTD165 (HM165S3, HM165S3A and HM165S3B) efficiently entered the cells (FIGS. 20 and 21) and were localized to various extents in cytoplasm (FIG. 21). In contrast, SOCS3 protein containing non-aMTD (HS3) did not appear to enter cells. While all SOCS3 proteins containing aMTD165 transduced into the cells, HM165S3B displayed more uniform cellular distribution, and protein uptake of HM165S3B was also very efficient.
  • Example 7-1-2. Comparison Between aMTD/SD-Fused SOCS3 Recombinant Protein and Basic CPP/SD-Fused SOCS3 Recombinant Protein
  • The cell-permeability of basic CPP/SD-fused SOCS3 recombinant proteins was assessed by the same method as used in Example 7-1 except for a known basic CPP (TAT or PolyR) being used instead of aMTD. The results of the assessment were shown in FIG. 99.
  • According to the results, all recombinant proteins exhibited cell-permeability. Among the proteins, aMTD/SD-fused SOCS3 recombinant protein (HM165S3B) showed the highest cell-permeability.
  • Example 7-2. Tissue-Permeability Test
  • To further investigate in vivo delivery of SOCS3 recombinant proteins, ICR mice (Doo-Yeol Biotech Co. Ltd., Seoul, Korea) were intraperitoneal (IP) injected with 600 μg/head of 10 μM FITC (Fluorescein isothiocyanate, SIGMA, USA)-labeled SOCS3 proteins and sacrificed after 2 hrs. From the mice, the liver, kidney, spleen, lung, heart, and brain were removed and washed with PBS, and then placed on a dry ice, and embedded with an O.C.T. compound (Sakura). After cryosectioning at 20 μm, tissue distributions of fluorescence-labeled-SOCS3 proteins in different organs was analyzed by fluorescence microscopy (Carl Zeiss, Gottingen, Germany)(FIG. 22).
  • As shown in FIG. 22, SOCS3 recombinant proteins fused to aMTD165 (HM165S3, HM165S3A and HM165S3B) were distributed to a variety of tissues (liver, kidney, spleen, lung, heart and, to a lesser extent, brain). Liver showed highest levels of fluorescent cell-permeable SOCS3 since intraperitoneal administration favors the delivery of proteins to this organ via the portal circulation. SOCS3 containing aMTD165 was detectable to a lesser degree in lung, spleen and heart. aMTD/SDB-fused SOCS3 recombinant protein (HM165S3B) showed the highest systemic delivery of SOCS3 protein to the tissues compared to the SOCS3 containing only aMTD (HM165S3) or aMTD/SDA (HM165S3A) proteins. These data suggest that SOCS3 protein containing both of aMTD165 and SDB leads to higher cell-/tissue-permeability due to the increase in solubility and stability of the protein, and it displayed a dramatic synergic effect on cell-/tissue-permeability.
  • Example 7-2-2. Comparison Between aMTD/SD-Fused SOCS3 Recombinant Protein and Basic CPP/SD-Fused SOCS3 Recombinant Protein
  • The tissue-permeability of basic CPP/SD-fused SOCS3 recombinant proteins was assessed by the same method as used in Example 7-2 except for a known basic CPP (TAT or PolyR) being used instead of aMTD. The results of the assessment were shown in FIG. 100.
  • According to the results, only aMTD/SD-fused SOCS3 recombinant protein (HM165S3B) exhibited superior cell-permeability.
  • Example 8-1 Biological Activity Test of iCP-SOCS3—Inhibition Activity of IFN-γ-Induced STAT Phosphorylation
  • It was examined whether the iCP-SOCS3 recombinant proteins prepared by fusion with combinations of aMTD and SD inhibits activation of the JAK/STAT-signaling pathway.
  • PANC-1 Cells (KCLB, Seoul, South Korea) were treated with 10 ng/ml IFN-γ (R&D systems, Abingdon, UK) for 15 min and treated with either DMEM (vehicle) or 10 μM aMTD/SD-fused SOCS3 recombinant proteins for 2 hrs. The cells were lysed in RIPA lysis buffer (Biosesang, Seongnam, Korea) containing proteinase inhibitor cocktail (Roche, Indianapolis, Ind., USA), incubated for 15 min at 4° C., and centrifuged at 13,000 rpm for 10 min at 4° C. Equal amounts of lysates were separated on 10% SDS-PAGE gels and transferred to a nitrocellulose membrane. The membranes were blocked using 5% skim milk in TBST and for western blot analysis incubated with the following antibodies: anti-phospho-STAT1 (Cell Signaling Technology, USA) and anti-phospho-STAT3 (Cell Signaling Technology, USA), then HRP conjugated anti-rabbit secondary antibody (Santacruz).
  • The well-known cytokine signaling inhibitory actions of SOCS3 are inflammation inhibition through i) inhibition of IFN-γ-mediated JAK/STAT and ii) inhibition of LPS-mediated cytokine secretion. The ultimate test of cell-penetrating efficiency is a determination of intracellular activity of SOCS3 proteins transported by aMTD. Since endogenous SOCS3 are known to block phosphorylation of STAT1 and STAT3 by IFN-γ-mediated Janus kinases (JAK) 1 and 2 activation, we demonstrated whether cell-permeable SOCS3 inhibits the phosphorylation of STATs. As shown in FIG. 23, All SOCS3 recombinant proteins containing aMTD (HM165S3, HM165S3A and HM165S3B), suppressed IFN-γ-induced phosphorylation of STAT1 and STAT3. In contrast, STAT phosphorylation was readily detected in cells exposed to HS3, which lacks the aMTD motif required for membrane penetration, indicating that HS3, which lacks an MTD sequence did not enter the cells, has no biological activity.
  • Example 8-2. Biological Activity Test of iCP-SOCS3
  • Peritoneal macrophages were obtained from C3H/HeJ mice (Doo-Yeol Biotech Co. Ltd. Korea) Peritoneal macrophages were incubated with 10 μM SOCS3 recombinant proteins (1:HS3, 2:HM165S3, 3:HM165S3A and 4:HM165S3B, respectively) for 1 hr at 37° C. and then stimulated them with LPS (Lipopolysaccharide)(500 ng/ml) and/or IFN-γ (100 U/ml) without removing iCP-SOCS3 proteins for 3, 6, or 9 hrs. The culture media were collected, and the extracellular levels of cytokine (TNF-α, IL-6) were measured by a cytometric bead array (BD Pharmingen, San Diego, Calif., USA) according to the manufacturer's instructions.
  • The effect of cell-permeable SOCS3 proteins on cytokines secretion was investigated. Treatment of C3H/HeJ primary peritoneal macrophages with SOCS3 proteins containing aMTD165 suppressed TNF-α and IL-6 secretion induced by the combination of IFN-γ and LPS by 50-90% during subsequent 9 hrs of incubation (FIG. 24). In particular, aMTD165/SDB-fused SOCS3 recombinant protein showed the greatest inhibitory effect on cytokine secretion. In contrast, cytokine secretion in macrophages treated with non-permeable SOCS3 protein (HS3) was unchanged, indicating that recombinant SOCS3 lacking the aMTD doesn't affect intracellular signaling. Therefore, we conclude that differences in the biological activities of HM165S3B as compared to HS3B are due to the differences in protein uptake mediated by the aMTD sequence. In light of solubility/yield, cell-/tissue-permeability, and biological effect, SOCS3 recombinant protein containing aMTD and SDB (HM165S3B) is a prototype of a new generation of improved cell-permeable SOCS3 (iCP-SOCS3), and will be selected for further evaluation as a potential anti-tumor agent.
  • Example 9. Preparation of Control Protein (Non-CP-SOCS3: HS3B Recombinant Protein)
  • As an experimental negative control, a SOCS3 recombinant protein having no cell permeability was prepared.
  • According to Example 6-2, SOCS3 recombinant proteins lacking SD (HM165S3) or both aMTD and SD (HS3) were found to be less soluble, produced lower yields, and showed tendency to precipitate when they were expressed and purified in E. coli. Therefore, we additionally designed and constructed SOCS3 recombinant protein containing only SDB (without aMTD165: HS3B) as a negative control (FIG. 25). Preparation, expression and purification, and measurement of solubility/yield of the recombinant proteins were performed in the same manner as in Examples 6-2 and 6-3.
  • TABLE 37
    Recombinant
    Cargo SD Protein 5′ Primers 3′ Primers
    SOCS3 B HS3B 5′- 5′-
    GGAATTCCATATGG CGCGTCGACTTAAA
    TCACCCAC GGGTTTCCGAAGGC
    AGCAAGTTTCCC TTGGCTATCTT-3′
    GCCGCC-3′
  • As expected, its solubility and yield increased compared to that of SOCS3 proteins lacking SDB (HS3; FIG. 26). Therefore, HS3B proteins were used as a control protein.
  • Example 10. Selection of aMTD for Cell-Permeability
  • After a basic structure of the stable recombinant proteins fused with combinations of aMTD and SD was determined, 22 aMTDs were selected for development of iCP-SOCS3 recombinant protein (Tables 38 and 39), based on the critical Factors, in order to examine which aMTD provides the highest cell-/tissue-permeability.
  • For comparison, 5 kinds of random peptides that do not satisfying one or more critical factors were selected (Table 40).
  • Solubility/yield and cell-permeability of 22 kinds of aMTD/SDB-fused SOCS3 recombinant proteins, prepared by using primers of Table 41 in the same manner as in Example 6-2, were analyzed according to Examples 6-3 and 7-1, respectively.
  • TABLE 41
    Amino Acid
    Cargo Sequence 5′ Primers 3′ Primers
    aMTD ID
    SOCS3 MTM AAVLLPVLLAAP GGAATTCCATATGGCGGCGGTGCTGC CGCGTCGACTTAAAGGGTTTCCGA
    TGCCGGTGCTGCTGGCGGCGCCGGT AGGCTTGGCTATCTT
    CACCCACAGCAAGTTTCCCGCCGCC
    44 ALAVPVALLVAP GGAATTCCATATGGCGCTGGCGGTGCC
    GGTGGCGCTGCTGGTGGCGCCGGTCA
    CCCACAGCAAGTTTCCCGCCGCC
    81 AALLPALAALLP GGAATTCCATATGGCGGCGCTGCTGCC
    GGCGCTGGCGGCGCTGCTGCCGGTCA
    CCCACAGCAAGTTTCCCGCCGCC
    123 AAIIVPAALLAP GGAATTCCATATGGCGGCGATTATTGTG
    CCGGCGGCGCTGCTGGCGCCGGTCAC
    CCACAGCAAGTTTCCCGCCGCC
    162 AVVALPAALIVP GGAATTCCATATGGCGGTGGTGGCGCT
    GCCGGCGGCGCTGATTGTGCCGGTCA
    CCCACAGCAAGTTTCCCGCCGCC
    281 ALIVLPAAVAVP GGAATTCCATATGGCGCTGATTGTGCT
    GCCGGCGGCGGTGGCGGTGCCGGTC
    ACCCACAGCAAGTTTCCCGCCGCC
    324 IVAVALPAALVP GGAATTCCATATGATTGTGGCGGTGGC
    GCTGCCGGCGGCGCTGGTGCCGGTCA
    CCCACAGCAAGTTTCCCGCCGCC
    364 LVAAVAPALIVP GGAATTCCATATGCTGGTGGCGGCGGT
    GGCGCCGGCGCTGATTGTGCCGGTCA
    CCCACAGCAAGTTTCCCGCCGCC
    365 AVIVVAPALLAP GGAATTCCATATGGCGGTGATTGTGGT
    GGCGCCGGCGCTGCTGGCGCCGGTCA
    CCCACAGCAAGTTTCCCGCCGCC
    622 ALIVLAAPVAVP GGAATTCCATATGGCGCTGATTGTGCT
    GGCGGCGCCGGTGGCGGTGCCGGTC
    ACCCACAGCAAGTTTCCCGCCGCC
    662 ALAVILAPVAVP GGAATTCCATATGGCGCTGGCGGTGAT
    TCTGGCGCCGGTGGCGGTGCCGGTCA
    CCCACAGCAAGTTTCCCGCCGCC
    563 ALAVIVVPALAP GGAATTCCATATGGCGCTGGCGGTGAT
    TGTGGTGCCGGCGCTGGCGCCGGTCA
    CCCACAGCAAGTTTCCCGCCGCC
    899 AVVIALPAVVAP GGAATTCCATATGGCGGTGGTGATTGC
    GCTGCCGGCGGTGGTGGCGCCGGTCA
    CCCACAGCAAGTTTCCCGCCGCC
    897 AVIVPVAIIAAP GGAATTCCATATGGCGGTGATTGTGCC
    GGTGGCGATTATTGCGGCGCCGGTCA
    CCCACAGCAAGTTTCCCGCCGCC
    623 VAAAIALPAIVP GGAATTCCATATGGTGGCGGCGGCGAT
    TGCGCTGCCGGCGATTGTGCCGGTCA
    CCCACAGCAAGTTTCCCGCCGCC
    908 VALALAPVVVAP GGAATTCCATATGGTGGCGCTGGCGCT
    GGCGCCGGTGGTGGTGGCGCCGGTCA
    CCCACAGCAAGTTTCCCGCCGCC
    911 VALALPAVVVAP GGAATTGCATATGGTGGCGCTGGCGCT
    GCCGGCGGTGGTGGTGGCGCCGGTCA
    CCCACAGCAAGTTTCCCGCCGCC
    2 AAAVPLLAVVVP GGAATTCCATATGGCGGCGGCGGTGC
    CGCTGCTGGCGGTGGTGGTGCCGGTC
    ACCCACAGCAAGTTTCCCGCCGCC
    904 AVLAVVAPVVAP GGAATTCCATATGGCGGTGCTGGCGGT
    GGTGGCGCCGGTGGTGGCGCCGGTCA
    CCCACAGCAAGTTTCCCGCCGCC
    481 AIAIAIVPVALP GGAATTCCATATGGCGATTGCGATTGC
    GATTGTGCCGGTGGCGCTGCCGGTCA
    CCCACAGCAAGTTTCCCGCCGCC
    787 AVALVPVIVAAP GGAATTGCATATGGCGGTGGCGCTGGT
    GCCGGTGATTGTGGCGGCGCCGGTCA
    CCCACAGCAAGTTTCCCGCCGCC
    264 LAAAPVVIVIAP GGAATTCCATATGCTGGCGGCGGCGC
    CGGTGGTGATTGTGATTGCGCCGGTCA
    CCCACAGCAAGTTTCCCGCCGCC
    363 AVLAVAPALIVP GGAATTCCATATGGCGGTGCTGGCGGT
    GGCGCCGGCGCTGATTGTGCCGGTCA
    CCCACAGCAAGTTTCCCGCCGCC
    121 AIVALPALALAP GGAATTGGATATGGGGATTGTGGCGCT
    GCCGGCGCTGGCGCTGGCGCCGGTCA
    CCCACAGCAAGTTTCCCGCCGCC
    rPeptide
    ID
    SOCS3 921 IWWFVVLPLVVP GGAATTCCATATGATTTGGTCGTTTGTG CGCGTCGACTTAAAGGGTTTCCGAA
    GTGCTGCCGCTGGTGGTGCCGGTCAC GGCTTGGCTATCTT
    CCACAGCAAGTTTCCCGCCGCC
    16 NNSCTTYTNGSQ GGAATTCCATATGAACAACAGCTGCAC
    CACCTATACCAACGGCAGCCAGGTCA
    CCCACAGCAAGTTTCCCGCCGCC
    67 LDAEVPLADDVP GGAATTCCATATGCTGGATGCGGAAGT
    GCCGCTGGCGGATGATGTGCCGGTCA
    CCCACAGCAAGTTTCCCGCCGCC
    29 VLPPLPVLPVLP GGAATTCCATATGGTGCTGCCGCCGCT
    GCCGGTGCTGCCGGTGCTGCCGGTCA
    CCCACAGCAAGTTTCCCGCCGCC
    700 GTSNTCQSNQNS GGAATTCCATATGGGCACCAGCAACAC
    CTGCCAGAGCAACCAGAACAGCGTCA
    CCCACAGCAAGTTTCCCGCCGCC
  • As shown in FIGS. 27 to 34, it was confirmed that most of the aMTD/SDB-fused SOCS3 recombinant proteins showed high solubility and yield and high cell permeability by aMTD. However, Random peptide-SOCS3-SDB recombinant protein showed remarkably low cell permeability.
  • Example 11-1. Investigation of Biological Activity for Determination of Optimal aMTD-Fused SOCS3 Recombinant Protein—1
  • Four kinds of aMTD/SD-fused SOCS3 recombinant proteins having high cell permeability and one kind of aMTD/SD-fused SOCS3 recombinant protein having the lowest cell permeability were selected, and their biological activity was analyzed.
  • PANC-1 cells (pancreatic carcinoma cell line) were seeded in a 16-well chamber slide at a density of 5×103 cells/well, and then treated with 10 uM of aMTD/SD-fused SOCS3 for 24 hours. Apoptotic cells were analyzed using terminal dUTP nick-end labeling (TUNEL) assay with In Situ Cell Death Detection kit TMR red (Roche, 4056 Basel, Switzerland). Cells were treated with either 10 μM SOCS3 recombinant protein or buffer alone for 16 hrs with 2% fetal bovine serum (Hyclone, Logan, Utah, USA). Treated cells were washed with cold PBS two times, fixed in 4% paraformaldehyde (PFA, Junsei, Tokyo, Japan) for 1 hr at room temperature, and incubated in 0.1% Triton X-100 for 2 min on the ice. Cells were washed with cold PBS twice, and treated TUNEL reaction mixture for 1 hr at 37° C. in dark, washed cold PBS three times and observed by fluorescence microscopy (Nikon, Tokyo, Japan).
  • As shown in FIG. 35, most of the aMTD/SDB-fused SOCS3 recombinant proteins induced cell death of pancreatic carcinoma cells, and of them, aMTD165 or aMTD324-fused SOCS3 recombinant protein induced death of the largest number of cancer cells.
  • Example 11-2. Investigation of Biological Activity for Determination of Optimal aMTD-Fused SOCS3 Recombinant Protein—2
  • AGS cells (gastric carcinoma cell line) (American Type Culture Collection; ATCC) were seeded in a 12-well plate at a density of 1×105 cells/well, and then treated with 10 uM of aMTD/SD-fused SOCS3 for 14 hours. Cancer cell death was analyzed by Annexin V analysis. Annexin V/7-Aminoactinomycin D (7-AAD) staining was performed using flow cytometry according to the manufacturer's guidelines (BD Pharmingen, San Diego, Calif., USA). Briefly, cells were washed three times with ice-cold PBS. The cells were then resuspended in 100 μl of binding buffer and incubated with 1 μl of 7-AAD and 1 μl of annexin V-PE for 30 min in the dark at 37° C. Flow cytometric analysis was immediately performed using a guava easyCyte™ 8 Instrument (Merck Millipore, Darmstadt, Germany).
  • As shown in FIG. 36, most of the aMTD/SDB-fused SOCS3 recombinant proteins induced cell death of gastric carcinoma cells, and of them, aMTD165 or aMTD281-fused SOCS3 recombinant protein induced death of the largest number of cancer cells.
  • Example 11-3. Investigation of Biological Activity for Determination of Optimal aMTD-Fused SOCS3 Recombinant Protein—3
  • AGS cells (gastric cancer cell line) were seeded in a 12 well plate at a density of 2.5×105 per well, grown to 90% confluence. Confluent AGS cells were incubated with 10 μM HM#S3B in serum-free medium for 2 hrs prior to changing the growth medium (DMEM/F12, Hyclone, Logan, Utah, USA) and washed twice with PBS, and the monolayer at the center of the well was “wounded” by scraping with a sterilized white pipette tip. Cells were cultured for an additional 24 hrs and cell migration was observed by phase contrast microscopy (Nikon, ECLIPSE Ts2). The migration was quantified by counting the number of cells that migrated from the wound edge into the clear area.
  • As shown in FIG. 37, most of the aMTD/SDB-fused SOCS3 recombinant proteins inhibited cell migration of gastric carcinoma cells, and of them, aMTD165 or aMTD904-fused SOCS3 recombinant protein showed the most effective inhibition of cancer cell migration.
  • Solubility/yield, permeability, and biological activity of 22 kinds of the aMTD-fused recombinant proteins were examined in Examples 10 to 11-3, and as a result, the aMTD165/SDB-fused SOCS3 recombinant protein was found to show the most excellent effect (FIG. 38). Therefore, the aMTD165-fused recombinant protein was used in the subsequent experiment.
  • Example 12-1. Preparation of iCP-SOCS3 Recombinant Protein and Investigation of Equivalence Thereof-1
  • To develop a new drug as an anticancer agent, His-tag-removed iCP-SOCS3 recombinant protein was prepared and equivalence of His-Tag+, −iCP-SOCS3 was investigated.
  • Histidine-tag free human SOCS3 proteins were constructed by amplifying the SOCS3 cDNA (225 amino acids) for aMTD fused to SOCS3 cargo. The PCR reactions (100 ng genomic DNA, 10 pmol each primer, each 0.2 mM dNTP mixture, 1× reaction buffer and 2.5 U Pfu(+) DNA polymerase (Doctor protein, Korea)) were digested on the restriction enzyme site between Nde I (5′) and Sal I (3′) involving 35 cycles of denaturing (95° C.), annealing (62° C.), and extending (72° C.) for 45 sec each. For the last extension cycle, the PCR reactions remained for 10 min at 72° C. The PCR products were subcloned into pET-26b(+) (Novagen, Darmstadt, Germany). Coding sequence for SDB fused to C terminus of aMTD-SOCS3 was cloned at BamHI (5′) and SalI (3′) in pET-26b(+) from PCR-amplified DNA segments and confirmed by DNA sequence analysis of the resulting plasmids.
  • TABLE 42
    Recombinant
    Cargo SD Protein 5′ Primers 3′ Primers
    SOCS3 HS3 5′- 5′-
    GGAATTCCATATGGTCACCCAC CCCGGATCCTTAAA
    AGCAAGTTTCCCGCCGCC-3′ GCGGGGCATCGTAC
    TGGTCCAGGAA-3′
    HM165S3 5′- 5′-
    GGAATTCCATATGGCGCTGGCG CCCGGATCCTTAAA
    GTGCCGGTGGCGCTGGCGATT GCGGGGCATCGTAC
    GTGCCGGTCACCCACAGCAAG TGGTCCAGGAA-3′
    TTTC-3′
    A HM165S3A 5′- 5′-
    GGAATTCCATATGGCGCTGGCG CGCGTCGACTTACC
    GTGCCGGTGGCGCTGGCGATT TCGGCTGCACCGGC
    GTGCCGGTCACCCACAGCAAG ACGGCGATAC-3′
    TTTC-3′
    B HM165S3B 5′- 5′-
    GGAATTCCATATGGCGCTGGCG CGCGTCGACTTAAA
    GTGCCGGTGGCGCTGGCGATT GGGTTTCCGAAGGC
    GTGCCGGTCACCCACAGCAAG TTGGCTATCTT-3′
    TTTC-3′
    C HM165S3C 5′- 5′-
    GGAATTCCATATGGCGCTGGCG GCGTCGACTTAGGC
    GTGCCGGTGGCGCTGGCGATT CAGGTTAGCGTCGA
    GTGCCGGTCACCCACAGCAAG G-3′
    TTTC-3′
    D HM165S3D 5′- 5′-
    GGAATTCCATATGGCGCTGGCG GCGTCGACTTATTTT
    GTGCCGGTGGCGCTGGCGATT TTCTCGGACAGATA-
    GTGCCGGTCACCCACAGCAAG 3′
    TTTC-3′
    E HM165S3E 5′- 5′-
    GGAATTCCATATGGCGCTGGCG ACGCGTCGACTTAA
    GTGCCGGTGGCGCTGGCGATT CCTCCAATCTGTTC
    GTGCCGGTCACCCACAGCAAG GCGGTGAGCCTC-3′
    TTTC-3′
    B M165S3B 5′- 5′-
    GGAATTCCATATGGCGCTGGCG CGCGTCGACTTAAA
    GTGCCGGTGGCGCTGGCGATT GGGTTTCCGAAGGC
    GTGCCGGTCACCCACAGCAAG TTGGCTATCTT-3′
    TTTC-3′
  • Expression, purification and solubility/yield were measured in the same manner as in Example 6-2 to 6-3, and as a result, his-tag-removed M165S3B was found to have high solubility/yield (FIG. 39).
  • Example 12-2. Preparation of iCP-SOCS3 Recombinant Protein and Investigation of Equivalence Thereof-2
  • In the same manner as in Example 7-1, RAW264.7 cells were treated with FITC-labeled HS3B, HM165S3B, and M165S3B proteins, and cell permeability was evaluated.
  • As shown in FIG. 40, both HM165S3B and M165S3B were found to have high cell permeability.
  • Example 12-3. Preparation of iCP-SOCS3 Recombinant Protein and Investigation of Equivalence Thereof-3
  • To investigate biological activity equivalence of the HM165S3B and M165S3B recombinant proteins, induction of apoptosis of gastric carcinoma cell line (AGS) was analyzed by Annexin V staining in the same manner as in Example 11-2, and inhibition of migration was analyzed in the same manner as in Example 11-3.
  • As shown in FIG. 41, it was confirmed that both HM165S3B and M165S3B showed high anticancer efficacy and M165S3B exhibited efficacy equivalent to or higher than HM165S3B.
  • Example 12-4. Preparation of iCP-SOCS3 Recombinant Protein and Investigation of Equivalence Thereof-4
  • In silico MHC class II binding analysis using iTope™ (ANTITOPE.LTD) revealed changing the V28 pl anchor residue in SDB sequence to L makes this region human germline and as such both MHC class II binding peptides within this region would be expected to be low risk due to T cell tolerance.
  • To prepare humanized SDB domain, iCP-SOCS3 was prepared in the same manner as in Example 6-1, except that SDB′ having a substitution of valine with leucine at an amino acid position 28 was used (FIG. 94). Further, protein purification was performed in the same manner as in Example 6-2 using the primer as below (Table 43).
  • TABLE 43
    Recombinant
    Cargo SD Protein 5′ Primers 3′ Primers
    SOCS3 HS3 5′- 5′-
    GGAATTCCATATGGTCACCCAC CCCGGATCCTTAAA
    AGCAAGTTTCCCGCCGCC-3′ GCGGGGCATCGTAC
    TGGTCCAGGAA-3′
    HM165S3 5′- 5′-
    GGAATTCCATATGGCGCTGGCG CCCGGATCCTTAAA
    GTGCCGGTGGCGCTGGCGATT GCGGGGCATCGTAC
    GTGCCGGTCACCCACAGCAAG TGGTCCAGGAA-3′
    TTTC-3′
    A HM165S3A 5′- 5′-
    GGAATTCCATATGGCGCTGGCG CGCGTCGACTTACC
    GTGCCGGTGGCGCTGGCGATT TCGGCTGCACCGGC
    GTGCCGGTCACCCACAGCAAG ACGGCGATAC-3′
    TTTC-3′
    B HM165S3B 5′- 5′-
    GGAATTCCATATGGCGCTGGCG CGCGTCGACTTAAA
    GTGCCGGTGGCGCTGGCGATT GGGTTTCCGAAGGC
    GTGCCGGTCACCCACAGCAAG TTGGCTATCTT-3′
    TTTC-3′
    C HM165S3C 5′- 5′-
    GGAATTCCATATGGCGCTGGCG GCGTCGACTTAGGC
    GTGCCGGTGGCGCTGGCGATT CAGGTTAGCGTCGA
    GTGCCGGTCACCCACAGCAAG G-3′
    TTTC-3′
    D HM165S3D 5′- 5′-
    GGAATTCCATATGGCGCTGGCG GCGTCGACTTATTTT
    GTGCCGGTGGCGCTGGCGATT TTCTCGGACAGATA-
    GTGCCGGTCACCCACAGCAAG 3′
    TTTC-3′
    E HM165S3E 5′- 5′-
    GGAATTCCATATGGCGCTGGCG ACGCGTCGACTTAA
    GTGCCGGTGGCGCTGGCGATT CCTCCAATCTGTTC
    GTGCCGGTCACCCACAGCAAG GCGGTGAGCCTC-3′
    TTTC-3′
    B* HM165S3B* 5′- 5′-
    GGAATTCCATATGGCGCTGGCG CGCGTCGACTTAAA
    GTGCCGGTGGCGCTGGCGATT GGGTTTCCGAAGGC
    GTGCCGGTCACCCACAGCAAG TTGGCTATCTT-3′
    TTTC-3′
  • As shown in FIG. 44, both HM165S3B and HM165S3B′(V28L) were found to have high solubility/yield.
  • Example 12-5. Preparation of iCP-SOCS3 Recombinant Protein and Investigation of Equivalence Thereof-5
  • In the same manner as in Example 7-1, RAW264.7 cells were treated with FITC-labeled HM165S3B and HM165S3B′(V28L) proteins, and cell permeability was evaluated.
  • As shown in FIG. 45, both HM165S3B and HM165S3B′(V28L) were found to have high cell permeability.
  • Example 12-6. Preparation of iCP-SOCS3 Recombinant Protein and Investigation of Equivalence Thereof-6
  • To investigate biological activity equivalence of the HM165S3B and HM165S3B′(V28L) recombinant proteins, anti-proliferative activity was examined and induction of apoptosis of gastric carcinoma cell line (AGS) was analyzed by Annexin V staining in the same manner as in Example 11-2, and inhibition of migration was analyzed in the same manner as in Example 11-3.
  • Antiproliferative activity were evaluated with the CellTiter-Glo Cell Viability Assay. AGS cells (3×103/well) were seeded in 96 well plates. The next day, cells were treated with DMEM (vehicle) or 10 μM HM165S3B, HM165S3B′(V28L) for 96 hrs in the presence of serum (2%). Proteins were replaced daily. Cell growth and survival were evaluated with the CellTiter-Glo Cell Viability Assay (Promega, Madison, Wis.). Measurements using a Luminometer (Turner Designs, Sunnyvale, Calif.) were conducted following the manufacturer's protocol.
  • It was confirmed that both HM165S3B and HM165S3B′(V28L) showed high anti-proliferative effects on gastric carcinoma cells (FIG. 46), and also effects of inducing apoptosis (FIG. 47) and of inhibiting migration of gastric carcinoma cells (FIG. 48), and in particular, HM165S3B′(V28L) exhibited efficacy equivalent to or higher than HM165S3B.
  • Example 12-7. Preparation of iCP-SOCS3 Recombinant Protein and Investigation of Equivalence Thereof-7
  • iCP-SOCS3 of BS3M165 structure was prepared in the same manner as in Example 6-1, and B′S3M165 iCP-SOCS3 was also prepared by humanized SDB domain (FIG. 49a ).
  • TABLE 44
    Recombinant
    Cargo SD Protein 5′ Primers 3′ Primers
    SOCS3 B BS3M165 5′-GGAATTCCATA 5′-
    TGATGGCAGAACAA ACGCGTCGACTTAC
    AGCGAC-3′ GCCAGCGCCACCG
    GCACCGCCAGCGC
    AATCACCGGAAGCG
    GGGCATCGTACTGG
    TCCAG-3′
    B* B*S3M165 5′-GGAATTCCATA 5′-
    TGATGGCAGAACAA ACGCGTCGACTTAC
    AGCGAC-3′ GCCAGCGCCACCG
    GCACCGCCAGCGC
    AATCACCGGAAGCG
    GGGCATCGTACTGG
    TCCAG-3′
  • Expressions and purifications of iCP-SOCS3 recombinant protein (BS3M165, B′S3M165) in E. coli (bottom) were analyzed in the same manner as in Examples 6-2 and 6-3, respectively, and shown in FIG. 49b . Further, E. coli codon-optimized iCP-SOCS3 was prepared.
  • Example 13. Test of Biological Activity of iCP-SOCS3—Inhibition Activity of IFN-γ-Induced STAT Phosphorylation
  • Whether iCP-SOCS3 (HM165S3B) recombinant protein inhibits activation of the JAK/STAT-signaling pathway was examined by the method of Example 8-1.
  • PANC-1 Cells (KCLB, Seoul, South Korea) were treated with 10 ng/ml IFN-γ (R&D systems, Abingdon, UK) for 15 min and treated with either DMEM (vehicle) or 1, 5, 10 μM aMTD/SD-fused SOCS3 recombinant proteins for 2 hrs. The cells were lysed in RIPA lysis buffer (Biosesang, Seongnam, Korea) containing proteinase inhibitor cocktail (Roche, Indianapolis, Ind., USA), incubated for 15 min at 4° C., and centrifuged at 13,000 rpm for 10 min at 4° C. Equal amounts of lysates were separated on 10% SDS-PAGE gels and transferred to a nitrocellulose membrane. The membranes were blocked using 5% skim milk in TBST and for western blot analysis incubated with the following antibodies: anti-phospho-STAT3 (Cell Signaling Technology, USA), then HRP conjugated anti-rabbit secondary antibody (Santacruz).
  • The well-known cytokine signaling inhibitory actions of SOCS3 are inflammation inhibition through i) inhibition of IFN-γ-mediated JAK/STAT and ii) inhibition of LPS-mediated cytokine secretion. The ultimate test of cell-penetrating efficiency is a determination of intracellular activity of SOCS3 proteins transported by aMTD. Since endogenous SOCS3 are known to block phosphorylation of STAT3 by IFN-γ-mediated Janus kinases (JAK) 1 and 2 activation, we demonstrated whether cell-permeable SOCS3 inhibits the phosphorylation of STATs. As shown in FIG. 50, iCP-SOCS3 (HM165S3B) suppressed IFN-γ-induced phosphorylation of STAT3 in dose dependent manner. In contrast, STAT phosphorylation was readily detected in cells exposed to HS3B, which lacks the aMTD motif required for membrane penetration, indicating that HS3B, which lacks an MTD sequence did not enter the cells, has no biological activity.
  • Example 14. Investigation of aMTD-Mediated Intracellular Delivery Mechanism
  • The mechanism of aMTD165-mediated intracellular delivery was investigated.
  • (1) RAW 264.7 cells were pretreated with 100 mM EDTA for 3 hours, and then treated with 10 μM of iCP-SOCS3 (HM165S3B) recombinant protein for 1 hour, followed by flow cytometry in the same manner as in Example 7-1 (FIG. 51A).
  • (2) RAW 264.7 cells were pretreated with 5 μg/ml of proteinase K for 10 minutes, and then treated with 10 μM of iCP-SOCS3 (HM165S3B) recombinant protein for 1 hour, followed by flow cytometry (FIG. 51B).
  • (3) RAW 264.7 cells were pretreated with 20 μM taxol for 30 minutes, and then treated with 10 μM of iCP-SOCS3 (HM165S3B) recombinant protein for 1 hour, followed by flow cytometry (FIG. 52A).
  • (4) RAW 264.7 cells were pretreated with 1 mM ATP and 10 μM antimycin singly or in combination for 2 hours, and then treated with 10 μM of iCP-SOCS3 (HM165S3B) recombinant protein for 1 hour, followed by flow cytometry (FIG. 52B).
  • (5) RAW 264.7 cells were left at 4° C. and 37° C. for 1 hour, respectively, and then treated with 10 μM of iCP-SOCS3 (HM165S3B) recombinant protein for 1 hour, followed by flow cytometry (FIG. 53).
  • The aMTD-mediated intracellular delivery of SOCS3 protein did not require protease-sensitive protein domains displayed on the cell surface (FIG. 51B), microtubule function (FIG. 52A), or ATP utilization (FIG. 52B), since aMTD165-dependent uptake, compare to HS3 and HS3B, was essentially unaffected by treating cells with proteinase K, taxol, or the ATP depleting agent, antimycin. Conversely, iCP-SOCS3 (HM165S3B) proteins uptake was blocked by treatment with EDTA and low temperature (FIGS. 51A and 53), indicating the importance of membrane integrity and fluidity for aMTD-mediated protein transduction.
  • Moreover, whether cells treated with iCP-SOCS3 (HM165S3B) protein could transfer the protein to neighboring cells were also tested.
  • For this, RAW 264.7 cells were treated with 10 μM of FITC-labeled iCP-SOCS3 (HM165S3B) recombinant protein for 1 hour. Thereafter, these cells were co-cultured with PerCP-Cy5.5-CD14-stained RAW 264.7 cells for 2 hours. Cell-to-cell protein transfer was assessed by flow cytometry, scoring for CD14/FITC double-positive cells. Efficient cell-to-cell transfer of HM165S3B, but not HS3 or HS3B (FIG. 54), suggests that SOCS3 recombinant proteins containing aMTD165 are capable of bidirectional passage across the plasma membrane.
  • Example 14-2. Investigation of Basic CPP-Mediated Intracellular Delivery Mechanism
  • The mechanism of basic CPP (TAT and PolyR)-mediated intracellular delivery was also investigated in the same manner as in Example 7-1 and Example 14.
  • As shown in FIG. 101, it was confirmed that aMTD165/SD-fused SOCS3 recombinant proteins are independent to cell surface receptor (FIG. 101A) and the cell-permeability of aMTD165/SD-fused SOCS3 recombinant proteins is not due to endocytosis (FIG. 101B).
  • Whether cells treated with aMTD165/SD-fused SOCS3, TAT/SD-fused SOCS3, and PolyR/SD-fused SOCS3 could transfer the protein to neighboring cells were also tested on a molecular level in the same manner as in Example 13.
  • For this, RAW 264.7 cells were treated with 5 μM of FITC-labeled HM165S3B, HTS3B for 2 hour and washed with PBS two times. Thereafter, they were seeded on PANC-1 cell, incubated for 2 hours and treated with 20 ng/ml of IFN-γ for 15 minutes, followed by Western blotting in the same manner as in Example 8-1. And Cell-to-cell protein transfer was assessed by flow cytometry.
  • As shown in FIG. 102, efficient cell-to-cell transfer of HM165S3B, but not HTS3B or HRS3B, suggests that only SOCS3 recombinant proteins containing aMTD165 are capable of bidirectional passage across the plasma membrane.
  • Moreover, as shown in FIG. 103, phospho-STAT3 was only reduced in cells treated with HM165S3B.
  • Example 15. Investigation of Bioavailability of iCP-SOCS3
  • To investigate BA of the iCP-SOCS3 (HM165S3B) recombinant proteins, ICR mice (Doo-Yeol Biotech Co. Ltd., Seoul, Korea) were intravenous (IV) injected with 600 μg/head of 10 μM FITC (Fluorescein isothiocyanate, SIGMA, USA)-labeled SOCS3 recombinant proteins (HS3B, HM165S3B) and after 15 min, 30 min, 1H, 2H, 4H, 8H, 12H, 16H, 24H, 36H, 48H, mice of each group were sacrificed. From the mice, peripheral blood mononuclear cells (PBMCs), splenocytes, and hepatocytes were separated.
  • Further, the spleen was removed and washed with PBS, and then placed on a dry ice and embedded in an O.C.T. compound (Sakura). After cryosectioning at 20 μm, tissue distributions of fluorescence-labeled-SOCS3 proteins in different organs was analyzed by fluorescence microscopy (Carl Zeiss, Gottingen, Germany).
  • Isolation of PBMC
  • After anesthesia with ether, ophthalmectomy was performed and the blood was collected therefrom using a 1 ml syringe. The collected blood was immediately put in an EDTA tube and mixed well. The blood was centrifuged at 4,000 rpm and 4° C. for 5 minutes, and plasma was discarded and only buffy coat was collected in a new microtube. 0.5 ml of RBC lysis buffer (Sigma) was added thereto, followed by vortexing. The microtube was left at room temperature for 5 minutes, and then centrifuged at 4,000 rpm and 4° C. for 5 minutes. 0.3 ml of PBS was added to a pellet, followed by pipetting and flow cytometry (FlowJo cytometric analysis software, Guava, Millipore, Darmstadt, Germany).
  • Isolation of Splenocytes and Hepatocytes
  • Mice were laparotomized and the spleen or liver were removed. The spleen or liver thus removed was separated into single cells using a Cell Strainer (SPL, Korea). These cells were collected in a microtube, followed by centrifugation at 4,000 rpm and 4° C. for 5 minutes. 0.5 ml of RBC lysis buffer was added thereto, followed by vortexing. The microtube was left at room temperature for 5 minutes, and then centrifuged at 4,000 rpm and 4° C. for 5 minutes. 0.5 ml of PBS was added to a pellet, followed by pipetting and flow cytometry (FlowJo cytometric analysis software, Guava, Millipore, Darmstadt, Germany).
  • As shown in FIG. 55, in PBMCs, the maximum permeability of iCP-SOCS3 was observed at 30 minutes, and in splenocytes, the maximum permeability of iCP-SOCS3 was observed at 2 hours and maintained up to 16 hours. In hepatocytes, the maximum permeability of iCP-SOCS3 was observed at 15 minutes and maintained up to 16 hours. As shown in FIG. 56, in the pancreas tissue, very high distribution of iCP-SOCS3 was observed at 2 hours, and maintained up to 8 hours. Therefore, it can be seen that iCP-SOCS3 is rapidly delivered from blood to various tissues within 2 hours, and maintained up to 8-16 hours depending on the tissues.
  • Example 16. Investigation of Anticancer Efficacy of iCP-SOCS3 Recombinant Protein
  • To develop the iCP-SOCS3 recombinant protein as a therapeutic agent for solid tumors, its permeability to solid tumors (gastric cancer, colorectal cancer, breast cancer, glioblastoma cell line) and various tissues was investigated.
  • AGS (gastric cancer cell line), HCT116 (colorectal cancer cell line), MDA-MB-231 (breast cancer cell line), and U-87 MG (glioblastoma cell line) cells (Korean cell line bank, Korea) were seeded in a 12 well plate, grown to 90% confluence. Cells were treated with 10 μM FITC-labeled iCP-SOCS3 recombinant proteins and cultivated for 1 hr at 37° C.
  • After cultivation, the cells were treated with proteinase K (10 μg/mL, SIGMA, USA) and washed three times with ice-cold PBS (Phosphate-buffered saline, Hyclone, USA) to remove surface-bound proteins, and internalized proteins were measured by flow cytometry (FlowJo cytometric analysis software, Guava, Millipore, Darmstadt, Germany). Untreated cells (gray) and equimolar concentration of unconjugated FITC (FITC only, green)-treated cells were served as control (FIG. 57).
  • The permeability to various tissues was analyzed at 2 hours after protein injection by the method described in Example 7-3.
  • As shown in FIG. 57, iCP-SOCS3 recombinant proteins (FITC-HM165S3SB) promoted the transduction into cultured various cancer cells. In contrast, SOCS3 proteins containing non-aMTD (FITC-HS3 and FITC-HS3B) did not appear to enter cells.
  • In addition, iCP-SOCS3 recombinant proteins (FITC-HM165S3SB) enhanced the systemic delivery to various tissues after intraperitoneal injection (FIG. 58). Therefore, these data indicate that iCP-SOCS3 protein could be intracellularly delivered and distributed to the various cancer cells and various tissues, contributing for beneficial biotherapeutic effects.
  • Example 17. Investigation of Anticancer Efficacy of iCP-SOCS3 Recombinant Protein Example 17-1. Investigation of Anti-Cancer Efficacy of iCP-SOCS3 Recombinant Protein in Gastric Cancer Cells
  • To develop the iCP-SOCS3 recombinant protein as a mechanism-specific therapeutic agent for solid tumors, SOCS3 levels endogenously expressed and activation of JAK/STAT-signaling pathway were investigated in different gastric cancer cell lines and normal cells, and normal hepatic cells.
  • Example 17-1-1. Analysis of Hypermethylation Level in Gastric Cancer Cells
  • SOCS3 expression is suppressed due to methylation in cancer cells, and therefore, inflammation or cancer development is increased. Further, to investigate the effect of cell permeable SOCS3, a cancer cell line where endogenous SOCS3 expression is suppressed should be selected or applied to a model, and therefore, the present experiments were performed.
  • Genomic DNA was extracted from cancer cell line using an Exgene™ Tissue SV mini kit (Geneall®, Korea). DNA was quantified, and experiments were performed using 500 ng of gDNA and an EZ DNA Methylation-Gold™ kit (ZYMO Research, Orange, Calif., USA) according to the manufacturer's instructions. DNA was used to perform PCR, and methylation and unmethylation of endogenous SOCS3 were qualitatively analyzed by electrophoresis. In this regard, the primers used are as follows.
  • Unmethyl-F was 5′-tag tgt gta agt tgt agg aga gtg g-3′(SEQ ID NO: 816), Unmethyl-R was 5′-cta aac ata aaa aaa taa cac taa tcc aaa-3′ (SEQ ID NO: 817), Methyl-F was 5′-gta gtg cgt aag ttg tag gag agc-3′ (SEQ ID NO: 818), Methyl-R was 5′-gta aaa aaa taa cgc taa tcc gaa-3′ (SEQ ID NO: 819), PCR was performed for 30 cycles consisting of pre-denaturation at 95° C. for 5 minutes, denaturation at 95° C. for 30 seconds, annealing at 60° C. for 45 seconds, extension at 72° C. for 1 minute, and final extension at 72° C. for 8 minutes.
  • As shown in FIG. 95, unmethylation of SOCS3 was observed in HEK293 and HaCaT cells which are normal cells, whereas hypermethylation of the promoter region of SOCS3 gene was observed in MNK75, MKN74, MKN45, MKN28, AGS, STKM2, NCI-N87 which are gastric cancer cell lines (U: unmethylated SOCS3, M: methylated SOCS3). These results indicate that SOCS3 is silenced by hypermethylation in gastric cancer cell lines.
  • Example 17-1-2. Analysis of Expression Level of Endogenous SOCS3 mRNA in Gastric Cell Line
  • SOCS3 mRNA expression levels in cancer cells were analyzed by RT-PCR. mRNAs were isolated from normal cell lines and cancer cell lines according to a method provided in a manufacturer's sheet of Hybrid-R (Geneall, Korea), and PCR was performed using SOCS3 primer F 5′-cct act gaa ccc tcc tcc ga-3′ (SEQ ID NO: 820) and SOCS3 primer R 5′-gca get ggg tga ctt tct ca-3′ (SEQ ID NO: 821) for 30 cycles consisting of denaturing (95° C.), annealing (60° C.), and extending (72° C.) for 45 seconds each.
  • It was confirmed that high expression levels of SOCS3 were observed in the normal HEK293 whereas low expression levels of SOCS3 were observed in the gastric cancer cell lines, except MKN74 (FIG. 59).
  • Example 17-1-3. Analysis of Endogenous SOCS3 and JAK/STAT Signaling Activation Status in Gastric Cell Line
  • JAK/STAT3 activation in cancer cells was analyzed by Western blot analysis. The normal HEK293 cells, and gastric cancer cell lines, MKN75, MKN74, MKN45, MKN28, AGS, STKM2, and NCI-N87 were washed with PBS, and then the cells were lysed in RIPA lysis buffer (Biosesang, Seongnam, Korea) containing protease inhibitor cocktail (Roche, Indianapolis, Ind., USA), incubated for 15 min at 4° C., and centrifuged at 13,000 rpm for 10 min at 4° C. to isolate proteins, followed by Western blotting in the same manner as in Example 8-1.
  • As shown in FIG. 60, high expression levels of SOCS3 and low levels of phosphorylations of p-JAK1, p-JAK2, p-STAT1, and p-STAT3 were observed in normal HEK293 cells. Low expression levels of SOCS3 gene were observed in the gastric cancer cell lines, compared to the normal cell line, and high levels of p-JAK1 and p-JAK2 in MKN75, high levels of p-STAT1 in MKN75, MKN74, MKN28, and NCI-N87, and high levels of p-STAT3 in MKN75, MKN45, AGS, STKN2, and NCI-N87. These results suggest a possibility of developing a mechanism-specific anticancer agent, because SOCS3-deficient cancer cells can be replenished with cell permeable proteins, and activated JAK/STAT-signaling can be negatively regulated.
  • Example 17-2. Investigation of Anti-Cancer Efficacy of iCP-SOCS3 Recombinant Protein
  • To develop the iCP-SOCS3 recombinant protein as a mechanism-specific therapeutic agent for solid tumors, SOCS3 levels endogenously expressed and activation of JAK/STAT-signaling pathway were investigated in different colorectal cell lines, normal cells, and normal hepatic cells.
  • Example 17-2-1. Analysis of Hypermethylation Level in Colorectal Cancer Cells
  • SOCS3 expression is suppressed due to methylation in cancer cells, and therefore, inflammation or cancer development is increased. Further, to investigate the effect of cell permeable SOCS3, a cancer cell line where endogenous SOCS3 expression is suppressed should be selected or applied to a model, and therefore, the present experiments were performed.
  • Genomic DNA was extracted from cancer cell line using an Exgene™ Tissue SV mini kit (Geneall®, Korea). DNA was quantified, and experiments were performed using 500 ng of gDNA and an EZ DNA Methylation-Gold™ kit (ZYMO Research, Orange, Calif., USA) according to the manufacturer's instructions. DNA was used to perform PCR, and methylation and unmethylation of endogenous SOCS3 were qualitatively analyzed by electrophoresis. In this regard, the primers used are as follows.
  • Unmethyl-F was 5′-tag tgt gta agt tgt agg aga gtg g-3′(SEQ ID NO: 816), Unmethyl-R was 5′-cta aac ata aaa aaa taa cac taa tcc aaa-3′ (SEQ ID NO: 817), Methyl-F was 5′-gta gtg cgt aag ttg tag gag agc-3′ (SEQ ID NO: 818), Methyl-R was 5′-gta aaa aaa taa cgc taa tcc gaa-3′ (SEQ ID NO: 819), PCR was performed for 30 cycles consisting of pre-denaturation at 95° C. for 5 minutes, denaturation at 95° C. for 30 seconds, annealing at 60° C. for 45 seconds, extension at 72° C. for 1 minute, and final extension at 72° C. for 8 minutes.
  • As shown in FIG. 96, unmethylation of SOCS3 was observed in HEK293 and HaCaT cells which are normal cells, whereas hypermethylation of the promoter region of SOCS3 gene was observed in HCT116, SW480, RKO, HT29 which are colorectal cell lines (U: unmethylated SOCS3, M: methylated SOCS3). These results indicate that SOCS3 is silenced by hypermethylation in colorectal cancer cell lines.
  • Example 17-2-2. Analysis of Expression Level of Endogenous SOCS3 mRNA in Colorectal Cancer Cell Line
  • SOCS3 mRNA expression levels in cancer cells were analyzed by RT-PCR. mRNAs were isolated from normal cell lines and cancer cell lines according to a method provided in a manufacturer's sheet of Hybrid-R (Geneall, Korea), and PCR was performed using SOCS3 primer F 5′-cct act gaa ccc tcc tcc ga-3′ (SEQ ID NO: 820) and SOCS3 primer R 5′-gca get ggg tga ctt tct ca-3′ (SEQ ID NO: 821) for 30 cycles consisting of denaturing (95° C.), annealing (60° C.), and extending (72° C.) for 45 seconds each.
  • The results of electrophoresis showed that high expression levels of SOCS3 were observed in the normal HEK293 cells whereas low expression levels of SOCS3 gene were observed in the colorectal cancer cell lines, SW480 and HT29 (FIG. 61).
  • Example 17-2-3. Analysis of Expression of Endogenous SOCS3 and JAK/STAT Signaling Activation Status in Colorectal Cancer Cell Line
  • JAK/STAT3 activation in cancer cells was analyzed by Western blot analysis. The normal HEK293 cells, and colorectal cancer cell lines, HCT116, SW480, RKO, and HT29 were washed with PBS, and then the cells were lysed in RIPA lysis buffer (Biosesang, Seongnam, Korea) containing proteinase inhibitor cocktail (Roche, Indianapolis, Ind., USA), incubated for 15 min at 4° C., and centrifuged at 13,000 rpm for 10 min at 4° C. to isolate proteins, followed by western blotting in the same manner as in Example 8-1.
  • As shown in FIG. 62, high expression level of SOCS3 and low phosphorylation levels of p-JAK1, p-JAK2, p-STAT1, and p-STAT3 were observed in normal HEK293 cells. Low expression level of SOCS3 gene was observed in the colorectal cancer cell lines, compared to the normal cell line, and similar levels of p-JAK1, p-JAK2, and p-STAT3 were observed in HCT116 and HT29. These results suggest a possibility of developing a mechanism-specific anticancer agent, because SOCS3-deficient cancer cells can be replenished with cell permeable proteins, and activated JAK/STAT-signaling can be negatively regulated.
  • Example 17-3. Investigation of Anticancer Efficacy of iCP-SOCS3 Recombinant Protein (Glioblastoma Cells)
  • To develop the iCP-SOCS3 recombinant protein as a mechanism-specific therapeutic agent for solid tumors, SOCS3 levels endogenously expressed and activation of JAK/STAT-signaling pathway were investigated in different glioblastoma cells, normal cells, and normal hepatic cells.
  • Example 17-3-1. Analysis of Hypermethylation Level in Glioblastoma Cell Line
  • SOCS3 expression is suppressed due to methylation in cancer cells, and therefore, inflammation or cancer development is increased. Further, to investigate the effect of cell permeable SOCS3, a cancer cell line where endogenous SOCS3 expression is suppressed should be selected or applied to a model, and therefore, the present experiments were performed.
  • Genomic DNA was extracted from cancer cell line using an Exgene™ Tissue SV mini kit (Geneall®, Korea). DNA was quantified, and experiments were performed using 500 ng of gDNA and an EZ DNA Methylation-Gold™ kit (ZYMO Research, Orange, Calif., USA) according to the manufacturer's instructions. DNA was used to perform PCR, and methylation and unmethylation of endogenous SOCS3 were qualitatively analyzed by electrophoresis. In this regard, the primers used are as follows.
  • Unmethyl-F was 5′-tag tgt gta agt tgt agg aga gtg g-3′(SEQ ID NO: 816), Unmethyl-R was 5′-cta aac ata aaa aaa taa cac taa tcc aaa-3′ (SEQ ID NO: 817), Methyl-F was 5′-gta gtg cgt aag ttg tag gag agc-3′ (SEQ ID NO: 818), and Methyl-R was 5′-gta aaa aaa taa cgc taa tcc gaa-3′ (SEQ ID NO: 819). PCR was performed for 30 cycles consisting of pre-denaturation at 95° C. for 5 minutes, denaturation at 95° C. for 30 seconds, annealing at 60° C. for 45 seconds, and extension at 72° C. for 1 minute, and then final extension at 72° C. for 8 minutes.
  • As shown in FIG. 63, unmethylation of SOCS3 was observed in HEK293 and HaCaT cells which are normal cells, whereas hypermethylation of the promoter region of SOCS3 gene was observed in U-87 MG, U-118 MG, T98G, LN229 which are glioblastoma cell lines (U: unmethylated SOCS3, M: methylated SOCS3). These results indicate that SOCS3 is silenced by hypermethylation in glioblastoma cell lines.
  • Example 17-3-2. Analysis of Expression of Endogenous SOCS3 and JAK/STAT Signaling Activation Status in Glioblastoma Cells
  • JAK/STAT3 activation in cancer cells was analyzed by Western blot analysis. The normal HaCaT and HEK293 cells, and glioblastoma cells, U-87 MG, U-118 MG, T98G, and LN229 were washed with PBS, and then the cells were lysed in RIM lysis buffer (Biosesang, Seongnam, Korea) containing protease inhibitor cocktail (Roche, Indianapolis, Ind., USA), incubated for 15 min at 4° C., and centrifuged at 13,000 rpm for 10 min at 4° C. to isolate proteins, followed by western blotting in the same manner as in Example 8-1.
  • As shown in FIG. 64, high expression levels of SOCS3 and low phosphorylation levels of p-STAT1, p-STAT3, and p-p65 were observed in normal HaCaT and HEK293 cells. Low expression level of SOCS3 gene was observed in the glioblastoma cell lines, compared to the normal cell line, and high levels of p-STAT1 and p-STAT3 were observed in LN229 and T98G. High levels of P-p65 were observed in all 4 types of glioblastoma cell lines. These results suggest a possibility of developing a mechanism-specific anticancer agent, because SOCS3-deficient cancer cells can be replenished with cell permeable proteins, and activated JAK/STAT-signaling can be negatively regulated.
  • Example 17-4. Investigation of Anticancer Efficacy of iCP-SOCS3 Recombinant Protein
  • To develop the iCP-SOCS3 recombinant protein as a mechanism-specific therapeutic agent for solid tumors, SOCS3 levels endogenously expressed and activation of JAK/STAT-signaling pathway were investigated in different breast cancer cell lines, normal cells, and normal hepatic cells.
  • Example 17-4-1. Analysis of Expression Level of Endogenous SOCS3 mRNA in Breast Cancer Cell Line
  • SOCS3 mRNA expression levels in cancer cells were analyzed by RT-PCR. mRNAs were isolated from normal cell lines and cancer cell lines according to a method provided in a manufacturer's sheet of Hybrid-R (Geneall, Korea), and PCR was performed using SOCS3 primer F 5′-cct act gaa ccc tcc tcc ga-3′ (SEQ ID NO: 820) and SOCS3 primer R 5′-gca get ggg tga ctt tct ca-3′ (SEQ ID NO: 821) for 30 cycles consisting of denaturing (95° C.), annealing (60° C.), and extending (72° C.) for 45 seconds each.
  • The results of electrophoresis showed that high expression levels of SOCS3 were observed in the normal HEK293 cells whereas low expression levels of SOCS3 were observed in the breast cancer cell lines, except MDA-MB-231 (FIG. 65).
  • Example 17-4-2. Analysis of Hypermethylation Level in Breast Cancer Cell Line
  • SOCS3 expression is suppressed due to methylation in cancer cells, and therefore, inflammation or cancer development is increased. Further, to investigate the effect of cell permeable SOCS3, a cancer cell line where endogenous SOCS3 expression is suppressed should be selected or applied to a model, and therefore, the present experiments were performed.
  • Genomic DNA was extracted from cancer cell line using an Exgene™ Tissue SV mini kit (Geneall®, Korea). DNA was quantified, and experiments were performed using 500 ng of gDNA and an EZ DNA Methylation-Gold™ kit (ZYMO Research, Orange, Calif., USA) according to the manufacturer's instructions. DNA was used to perform PCR, and methylation and unmethylation of endogenous SOCS3 were qualitatively analyzed by electrophoresis. In this regard, the primers used are as follows.
  • Unmethyl-F was 5′-tag tgt gta agt tgt agg aga gtg g-3′(SEQ ID NO: 816), Unmethyl-R was 5′-cta aac ata aaa aaa taa cac taa tcc aaa-3′ (SEQ ID NO: 817), Methyl-F was 5′-gta gtg cgt aag ttg tag gag agc-3′ (SEQ ID NO: 818), and Methyl-R was 5′-gta aaa aaa taa cgc taa tcc gaa-3′ (SEQ ID NO: 819). PCR was performed for 30 cycles consisting of pre-denaturation at 95° C. for 5 minutes, denaturation at 95° C. for 30 seconds, annealing at 60° C. for 45 seconds, and extension at 72° C. for 1 minute, and then final extension at 72° C. for 8 minutes.
  • As shown in FIG. 66, unmethylation of SOCS3 was observed in HEK293 cells which are normal cells, whereas hypermethylation of the promoter region of SOCS3 gene was observed in breast cancer cell lines, MDA-MB-231, MCF7, SK-BR3, and T47D (U: unmethylated SOCS3, M: methylated SOCS3). These results indicate that SOCS3 is silenced by hypermethylation in breast cancer cell lines.
  • Example 17-4-3. Analysis of Expression of Endogenous SOCS3 and JAK/STAT Signaling Activation Status in Breast Cancer Cell Line
  • JAK/STAT3 activation in cancer cells was analyzed by Western blot analysis. The normal HEK293 cells, and breast cancer cell lines, MDA-MB-231, MCF7, SK-BR3, and T47D were washed with PBS, and then the cells were lysed in RIPA lysis buffer (Biosesang, Seongnam, Korea) containing protease inhibitor cocktail (Roche, Indianapolis, Ind., USA), incubated for 15 min at 4° C., and centrifuged at 13,000 rpm for 10 min at 4° C. to isolate proteins, followed by western blotting in the same manner as in Example 8-1.
  • As shown in FIG. 67, high expression levels of SOCS3 and low phosphorylation levels of p-STAT1 and p-STAT3 were observed in normal HEK293 cells. Low expression level of SOCS3 gene was observed in the breast cancer cell lines, compared to the normal cell line. Further, high levels of p-STAT1 were observed in SK-BR3 and T47D, and high levels of p-STAT3 were observed in MDA-MB-231 and T47D. These results suggest a possibility of developing a mechanism-specific anticancer agent, because SOCS3-deficient cancer cells can be replenished with cell permeable proteins, and activated JAK/STAT-signaling can be negatively regulated.
  • Example 18. Investigation of Anti-Cancer Efficacy of iCP-SOCS3 Example 18-1. Investigation of Anti-Cancer Efficacy (Anti-Proliferative Activity) of iCP-SOCS3
  • In order to develop the iCP-SOCS3 recombinant protein as a therapeutic agent for solid tumors, efficacy of iCP-SOCS3 on proliferation of solid tumors (gastric cancer, colorectal cancer, breast cancer, and glioblastoma cell lines) was investigated.
  • Cells originated from human gastric cancer cells (AGS, MKN45, MKN75, NCI-N87), colorectal cancer cells (HCT116), glioblastoma (U-87 MG), breast cancer cells (MDA-MB-231), and mouse fibroblast (NIH3T3) were purchased from ATCC (Manassas, Va., USA) and KCLB (Seoul, Korea). All the cells were maintained as recommended by the supplier. These cells (3×103/well) were seeded in 96 well plates. The next day, cells were treated with DMEM (vehicle) or 10 μM recombinant SOCS3 proteins for 96 hrs in the presence of serum (2%). Proteins were replaced daily. Cell growth and survival were evaluated with the CellTiter-Glo Cell Viability Assay (Promega, Madison, Wis.). Measurements using a Luminometer (Turner Designs, Sunnyvale, Calif.) were conducted following the manufacturer's protocol.
  • Since the endogenous level of SOCS3 protein is reduced in solid tumor patient, and SOCS3 negatively regulates cell growth and motility in cultured solid tumor cells, we investigated whether iCP-SOCS3 inhibits cell viability through SOCS3 intracellular replacement in solid tumor cells. As shown in FIG. 68, SOCS3 recombinant proteins containing aMTD165 significantly suppressed cancer cell proliferation. HM165S3B (iCP-SOCS3) protein was the most cytotoxic to gastric cancer cells—over 40% in 10 μM treatment (p<0.01)—especially compared to vehicle alone (i.e. exposure of cells to culture media without recombinant proteins; left).
  • As shown in FIG. 69, SOCS3 recombinant proteins containing aMTD165 significantly suppressed cancer cell proliferation. HM165S3B (iCP-SOCS3) protein was the most cytotoxic to colorectal cancer cells—over 60% in 10 μM treatment (p<0.01)—especially compared to vehicle alone (i.e. exposure of cells to culture media without recombinant proteins; left).
  • As shown in FIG. 70, SOCS3 recombinant proteins containing aMTD165 significantly suppressed cancer cell proliferation. HM165S3B (iCP-SOCS3) protein was the most cytotoxic to glioblastoma cells—over 80% in 10 μM treatment (p<0.01)—especially compared to vehicle alone (i.e. exposure of cells to culture media without recombinant proteins; left).
  • As shown in FIG. 71, SOCS3 recombinant proteins containing aMTD165 significantly suppressed cancer cell proliferation. HM165S3B (iCP-SOCS3) protein was the most cytotoxic to breast cancer cells—over 40% in 10 μM treatment (p<0.01)—especially compared to vehicle alone (i.e. exposure of cells to culture media without recombinant proteins; left).
  • However, neither cell-permeable SOCS3 protein adversely affected the cell viability of non-cancer cells (NIH3T3) even after exposing these cells to equal concentrations (10 μM) of protein over 4 days. These results suggest that the iCP-SOCS3 protein is not overly toxic to normal cells and selectively kills tumor cells, and would have a great ability to inhibit cell survival-associated phenotypes in various cancer without any severe aberrant effects as a protein-based biotherapeutics.
  • Example 18-2. Investigation of Anti-Cancer Efficacy (Cell Migration Inhibition) of iCP-SOCS3
  • In order to develop the iCP-SOCS3 recombinant protein as a therapeutic agent for solid tumors, efficacy of iCP-SOCS3 on migration and metastasis of solid tumors (gastric cancer, colorectal cancer, breast cancer, and glioblastoma cell lines) was investigated.
  • Cells were seeded into 12-well plates, grown to 90% confluence, and incubated with 10 μM Non-CP-SOCS3 (HS3B), iCP-SOCS3 (HM165S3B) in serum-free medium for 2 hrs prior to changing the growth medium (recommended medium by the supplier). The cells were washed twice with PBS, and the monolayer at the center of the well was “wounded” by scraping with a pipette tip. Cells were cultured for an additional 24-48 hrs and cell migration was observed by phase contrast microscopy (Nikon, ECLIPSE Ts2). The migration was quantified by counting the number of cells that migrated from the wound edge into the clear area.
  • As shown in FIGS. 72-75, iCP-SOCS3 (HM165S3B) suppressed the repopulation of wounded monolayer although SOCS3 protein lacking aMTD165 (HS3B) had no effect on the cell migration, in AGS (FIG. 72), HCT116 (FIG. 73), U-87 MG (FIG. 74), and MDA-MB-231 (FIG. 75), respectly. In normal cells (NIH3T3), iCP-SOCS3 (HM165S3B) had no effect on the cell migration.
  • Transwell Migration Assay
  • The lower surface of Transwell inserts (Costar) was coated with 0.1% gelatin, and the membranes were allowed to dry for 1 hr at room temperature. The Transwell inserts were assembled into a 24-well plate, and the lower chamber was filled with growth media containing 10% FBS and FGF2 (40 ng/ml). Cells were added to each upper chamber at a density of 5×105, and the plate was incubated at 37° C. in a 5% CO2 incubator for 24 hrs. Migrated cells were stained with 0.6% hematoxylin and 0.5% eosin and counted.
  • As shown in FIGS. 76 (left) and 77, AGS (FIG. 76) and U-87 MG (FIG. 77) cells treated with iCP-SOCS3 (HM165S3B) protein also showed significant inhibitory effect on their Transwell migration compared with untreated cells (Vehicle) and non-permeable SOCS3 protein-treated cells.
  • Invasion Assay
  • The lower surface of Transwell inserts (Costar) was coated with 0.1% gelatin, the upper surface of Transwell inserts was coated with matrigel (40 μg per well; BD Pharmingen, San Diego, Calif., USA), and the membranes were allowed to dry for 1 hr at room temperature. The Transwell inserts were assembled into a 24-well plate, and the lower chamber was filled with growth media containing 10% FBS and FGF2 (40 ng/ml). Cells (5×105) were added to each upper chamber, and the plate was incubated at 37° C. in a 5% CO2 incubator for 24 hrs. Migrated cells were stained with 0.6% hematoxylin and 0.5% eosin and counted.
  • As shown in FIG. 76 (right), AGS treated with iCP-SOCS3 (HM165S3B) protein caused remarkable decrease in invasion compared with the control proteins. Taken together, these data indicate that iCP-SOCS3 contributes to inhibit tumorigenic activities of solid tumors.
  • Example 18-3. Investigation of Anti-Cancer Efficacy (Induction of Apoptosis) of iCP-SOCS3
  • To further determine the effect of iCP-SOCS3 on the tumorigenicity of solid tumors, we subsequently investigated whether iCP-SOCS3 regulates apoptosis in various cancer cells, such as gastric cancer cells (AGS and MKN45), colorectal cancer cells (HCT116), glioblastoma cells (U-87 MG), and breast cancer cells (MDA-MB-231).
  • Annexin V/7-Aminoactinomycin D (7-AAD) staining was performed using flow cytometry according to the manufacturer's guidelines (BD Pharmingen, San Diego, Calif., USA). Briefly, 1×106 cells were washed three times with ice-cold PBS. The cells were then resuspended in 100 μl of binding buffer and incubated with 1 μl of 7-AAD and 1 μl of Annexin V-7-AAD for 30 min in the dark at 4° C. Flow cytometric analysis was immediately performed using a guava easyCyte™ 8 Instrument (Merck Millipore, Darmstadt, Germany). In this regard, gemcitabine (0.1˜10 μM) was used as a reference drug.
  • As shown in FIG. 78, 10 uM of iCP-SOCS3 (HM165S3B) protein did not induce apoptosis in normal cell lines (NIH3T3 and HEK293). Whereas, as shown in FIGS. 79-82, 10 uM iCP-SOCS3 (HM165S3B) was efficient inducer of apoptosis in cancer cells, as assessed by Annexin V staining. Consistently, no changes in Annexin V staining were observed in cancer cells treated with HS3B compared to untreated cell (Vehicle).
  • Example 18-4. Investigation of Anti-Cancer Efficacy (Induction of Apoptosis) of iCP-SOCS3
  • To further determine the effect of iCP-SOCS3 on the tumorigenicity of solid tumors, we subsequently investigated whether iCP-SOCS3 regulates apoptosis in colorectal cancer cells.
  • Apoptotic cells were analyzed using terminal dUTP nick-end labeling (TUNEL) assay with In Situ Cell Death Detection kit TMR red (Roche, 4056 Basel, Switzerland). Cells were treated for 24 hr with 10 μM HS3B or HM165S3B proteins with 2% fetal bovine serum and apoptotic cells were visualized by TUNEL staining. Treated cells were washed with cold PBS two times, fixed in 4% paraformaldehyde (PFA, Junsei, Tokyo, Japan) for 1 hr at room temperature, and incubated in 0.1% Triton X-100 for 2 min on the ice. Cells were washed with cold PBS twice, and treated TUNEL reaction mixture for 1 hr at 37° C. in dark, washed cold PBS three times and observed by fluorescence microscopy (Nikon, Tokyo, Japan).
  • iCP-SOCS3 (HM165S3B) protein was considerably efficient inducer of apoptosis in HCT116 cells (FIG. 83), as assessed by a fluorescent terminal dUTP nick-end labeling (TUNEL) assay. Consistently, no changes in TUNEL was observed in HCT116 cells treated with HS3B compared to untreated cell (Vehicle).
  • Example 18-5. Investigation of Anti-Cancer Efficacy (Inhibition of Cell Cycle Progression) of iCP-SOCS3
  • To further determine the effect of iCP-SOCS3 on the tumorigenicity of solid tumors, we subsequently investigated whether iCP-SOCS3 regulates cell cycle progression in various cancer cells, such as gastric cancer cells (AGS), colorectal cancer cells (HCT116), glioblastoma cells (U-87 MG, and breast cancer cells (MDA-MB-231).
  • Cancer cells and normal cells (NIH3T3, HaCaT, and HEK293) were treated with 10 uM protein (Non-CP-SOCS3 (HS3B) and iCP-SOSC3 (HM165S3B)) for 8 hrs. After the treatment, cells were washed twice with cold PBS and re-suspended in 1 ml cold PBS, fixed in cold 70% ethanol, washed with cold PBS twice and re-suspended in PI master mix (PI 10 ug/ml (sigma), 50 ug/ml RNase A (invitrogen) in staining buffer) at a final cell density of 2×105 cell/ml. The cell mixtures were incubated 30 min in the dark at 4° C. Flow cytometric analysis was immediately performed using a guava easyCyte™ 8 Instrument (Merck Millipore, Darmstadt, Germany).
  • iCP-SOCS3 (HM165S3B) protein efficiently inhibited cell cycle progression in various cancer cells, such as AGS, HCT116, U-87 MG, and MDA-MB-231 (FIGS. 85 to 88), while iCP-SOCS3 (HM165S3B) protein did not inhibit cell cycle progression in normal cells (FIG. 84).
  • Example 18-6. Investigation of Anti-Cancer Efficacy (Effect on Molecular Mechanism-Mode of Action) of iCP-SOCS3
  • To further determine the effect of iCP-SOCS3 on the tumorigenicity of solid tumors, we subsequently investigated whether iCP-SOCS3 regulates apoptosis in breast cancer cells (MDA-MB-231).
  • Cells were treated for 24 hr with 10 μM HS3B or HM165S3B proteins, lysed in RIPA lysis buffer containing proteinase inhibitor cocktail, incubated for 15 min at 4° C., and centrifuged at 13,000 rpm for 10 min at 4° C. Equal amounts of lysates were separated on 15% SDS-PAGE gels and transferred to a nitrocellulose membrane. The membranes were blocked using 5% skim milk or 5% Albumin in TBST and incubated with the following antibodies: anti-Bcl-2 (Santa Cruz biotechnology), anti-Cleaved Caspase 3 (Cell Signaling Technology), then HRP conjugated anti-mouse or anti-rabbit secondary antibody. The expression of each protein was determined by immunoblotting with indicated antibodies. An antibody against β-actin was used as a loading control.
  • MDA-MB-231 cells treated with iCP-SOCS3 (HM165S3B) protein dramatically reduced the expression of anti-apoptotic protein such as B-cell lymphoma 2 (Bcl-2) and increased the level of cleaved cysteine-aspartic acid protease (caspase-3) at 24 hr (FIG. 89). These results indicate that iCP-SOCS3 induces apoptosis of breast cancer cells and may suppress the cancer progression by this pathway.
  • Example 19. Investigation of Anti-Cancer Efficacy of iCP-SOCS3 in Xenograft Model (Cell-Derived Xenograft (CDX)-Model) Example 19-1. Investigation of Anti-Cancer Efficacy of iCP-SOCS3 in Gastric Cancer Cell (NCI-N87)-Derived Xenograft (CDX) Model
  • In the following experiments, Western blotting was performed in the same manner as in Example 8-1, and RT-PCR and IHC were performed by the following method.
  • RT-PCR
  • Tumor tissues were finely minced using a homogenizer according to the manufacturer's protocol of Hybrid-R (geneall, Korea), and then mRNA was isolated therefrom. 1 ug of mRNA thus separated was used to synthesize cDNA using an Accupower RT Premix (Bioneer, Korea). PCR was performed using 2 ul of cDNA and the primers of Table 45. PCR was performed using an Accupower PCR Premix (Bioneer, Korea) for 30 cycles consisting of denaturing (95° C.), annealing (60° C.), and extending (72° C.) for 45 seconds each.
  • TABLE 45
    Genes Forward Sequence Reverse Sequence
    Cyclin E CCGTTTACAAGCTAAGCAGC GTGGTTCCAAGTCAGAATGC
    Cyclin TCAGTACTTGAGGCGACAAGG CTCCCTAATTGCTTGCTGAGG
    A1
    Bax CCCTTTTGCTTCAGGGTTTC GCCACTCGGAAAAAGACCTC
    FAK TGGTGAAAGCTGTCATCGAG CTGGGCCAGTTTCATCTTGT
    p21 CAGCGGAACAAGGAGTCAGA AGAAACGGGAACCAGGACAC
    p27 GATAATCCCGCTCTGAATGC GCTTCTCTTAGTGCTGTAGC
    VEGF CTTCAAGCCATCCTGTGTGC ACGCGAGTCTGTGTTTTTGC
    HIF-1α ATCAGACACCTAGTCCTTCCG TTGAGGACTTGCGCTTTCAGG
    GAPDH AAGGGTCATCATCTCTGCCC GTGATGGCATGGACTGTGGT
  • Immunohistochemistry (IHC)
  • Tissue samples were fixed in 4% Paraformaldehyde (Duksan, South Korea) for 3 days, dehydrated, cleared with xylene and embedded in Paraplast. Sections (6 μm thick) of tumor were placed onto poly-L-lysine coated slides. To block endogenous peroxidase activity, sections were incubated for 15 min with 3% H2O2 in methanol. After washing three times with PBS, slides were incubated for 30 min with blocking solution (5% fetal bovine serum in PBS). Mouse anti-Bax antibody (sc-7480, Santa Cruz Biotechnology, SantaCruz, Calif., USA) and rabbit anti-VEGF (ab46154, Abcam, Cambridge, UK) were diluted 1:1000 (to protein concentration 0.1 μg/ml) in blocking solution, applied to sections, and incubated at 4° C. for 24 hrs. After washing three times with PBS, sections were incubated with biotinylated mouse and rabbit IgG (Vector Laboratories, Burlingame, Calif., USA) at a 1:1000 dilution for 1 hr at room temperature, then incubated with avidin-biotinylated peroxidase complex using a Vectorstain ABC Kit (Vector Laboratories, Burlingame, Calif., USA) for 30 min at room temperature. After the slides are reacted with oxidized 3, 3-diaminobenzidine as a chromogen, they were counterstained with Harris hematoxylin (Sigma-Aldrich, USA). Permanently mounted slides were observed and photographed using a microscope equipped with a digital imaging system (ECLIPSE Ti, Nikon, Japan).
  • Anti-tumor activity of iCP-SOCS3 against human cancer xenografts was assessed. Female Balb/cnu/nu mice (5-week-old, Doo-Yeol Biotech Co. Ltd. Korea) were subcutaneously implanted with Hep3B2.1-7 tumor block (1 mm3) into the left back side of the mouse. Tumor-bearing mice were intravenously administered with 600 μg/head of iCP-SOCS3 (HM165S3B) or the control proteins (HS3B) for 21 days and observed for 2 weeks following the termination of the treatment. After protein treatment, mice were killed, and six organs (brain, heart, lung, liver, kidney, and spleen) from each were collected and kept in a suitable fixation solution until the next step. Tumor size was monitored by measuring the longest (length) and shortest dimensions (width) once a day with a dial caliper, and tumor volume was calculated as width2×length×0.5.
  • As shown in FIG. 90, HM165S3B protein significantly suppressed the tumor growth (p<0.05) during the treatment and the effect persisted for at least 2 weeks after the treatment was terminated (65% inhibition at day 35, respectively). Whereas, the growth of HS3B-treated tumors increased, matching the rates observed in control mice (Vehicle). These results suggest that iCP-SOCS3 inhibits the growth of established tumors as well as the tumor growth of gastric cancer cells.
  • mRNA levels were analyzed using the tumor tissues removed at 2 weeks (Day 35) after termination of drug administration to analyze changes in expressions of major biomarker genes by iCP-SOCS3.
  • Expressions of cell cycle (cyclinA1) and angiogenesis (VEGF)-related genes were significantly regulated by treatment of iCP-SOCS3.
  • Accordingly, it was confirmed that iCP-SOCS3 is able to inhibit development of gastric cancer through expressions of various factors regulating cancer activity such as cell cycle, angiogenesis, etc.
  • Further, tumor tissues removed at 2 weeks (Day 35) after termination of drug administration were used to perform Western blotting and IHC, and increase or decrease in the expressions of cancer activity regulating factors by iCP-SOCS3 was observed.
  • In iCP-SOCS3-treated tumor tissues, expressions of a cell cycle regulator, p21 and apoptosis inducers, Bax and cleaved caspase-3 were increased. Further, expression of an angiogenesis inducer VEGF was also remarkably decreased (FIG. 90). Accordingly, it was demonstrated that cell cycle, apoptosis, and angiogenesis are also regulated by iCP-SOCS3 in-vivo.
  • Example 19-2. Investigation of Anti-Cancer Efficacy of iCP-SOCS3 in Colorectal Cancer Cell (HCT116)-Derived Xenograft (CDX) Model
  • The following experiments were performed in the same manner as in Example 19-1.
  • As shown in FIG. 91, HM165S3B protein significantly suppressed the tumor growth (p<0.05) during the treatment and the effect persisted for at least 2 weeks after the treatment was terminated (79% at day 35, respectively). Whereas, the growth of HS3B-treated tumors increased, matching the rates observed in control mice (Vehicle). These results suggest that iCP-SOCS3 inhibits the growth of established tumors as well as the tumor growth of colorectal cancer cells.
  • mRNA levels were analyzed using the tumor tissues removed at 2 weeks (Day 35) after termination of drug administration to analyze changes in expressions of major biomarker genes by iCP-SOCS3.
  • Expressions of apoptosis (Bax) and angiogenesis (VEGF)-related genes were significantly regulated by treatment of iCP-SOCS3.
  • Accordingly, it was confirmed that iCP-SOCS3 is able to inhibit development of colorectal cancer through expressions of various factors regulating cancer activity such as apoptosis, angiogenesis, etc.
  • Further, tumor tissues removed at 2 weeks (Day 35) after termination of drug administration were used to perform Western blotting and IHC, and increase or decrease in the expressions of cancer activity regulating factors by iCP-SOCS3 was observed.
  • In iCP-SOCS3-treated tumor tissues, expressions of a cell cycle regulator, p21 and an apoptosis inducer, Bax were increased. Further, expression of an angiogenesis inducer VEGF was also remarkably decreased (FIG. 91). Accordingly, it was demonstrated that cell cycle, apoptosis, and angiogenesis are also regulated by iCP-SOCS3 in-vivo.
  • Example 19-3. Investigation of Anti-Cancer Efficacy of iCP-SOCS3 in Glioblastoma Cell (U-87 MG)-Derived Xenograft (CDX) Model
  • The following experiments were performed in the same manner as in Example 19-1.
  • As shown in FIG. 92, HM165S3B protein significantly suppressed the tumor growth (p<0.05) during the treatment and the effect persisted for at least 2 weeks after the treatment was terminated (76% at day 42, respectively). Whereas, the growth of HS3B-treated tumors increased, matching the rates observed in control mice (Vehicle). These results suggest that iCP-SOCS3 inhibits the growth of established tumors as well as the tumor growth of glioblastoma cells.
  • mRNA levels were analyzed using the tumor tissues removed at 3 weeks (Day 42) after termination of drug administration to analyze changes in expressions of major biomarker genes by iCP-SOCS3.
  • Expressions of apoptosis (Bax), cell cycle (CDK4, CDK2, cyclinE, cyclinAl), angiogenesis (HIF1a, VEGF), and migration (FAK)-related genes were significantly regulated by treatment of iCP-SOCS3.
  • Expressions of NSE, FSTL-1, and GFAT which are biomarker genes of glioblastoma were remarkably decreased by iCP-SOCS3.
  • Expressions of VEGF and angiopoietin I which are biomarker genes of angiogenesis were significantly decreased by treatment of iCP-SOCS3.
  • Accordingly, it was confirmed that iCP-SOCS3 is able to inhibit development of glioblastoma through expressions of various factors regulating cancer activity such as apoptosis, cell cycle, migration, angiogenesis, etc.
  • Example 19-4. Investigation of Anti-Cancer Efficacy of iCP-SOCS3 in Breast Cancer Cell (MDA-MB-231)-Derived Xenograft (CDX) Model
  • The following experiments were performed in the same manner as in Example 19-1.
  • As shown in FIG. 93, HM165S3B protein significantly suppressed the tumor growth (p<0.05) during the treatment and the effect persisted for at least 2 weeks after the treatment was terminated (63% at day 35, respectively). Whereas, the growth of HS3B-treated tumors increased, matching the rates observed in control mice (Vehicle). These results suggest that iCP-SOCS3 inhibits the growth of established tumors as well as the tumor growth of breast cancer cells. mRNA levels were analyzed using the tumor tissues removed at 2 weeks (Day 35) after termination of drug administration to analyze changes in expressions of major biomarker genes by iCP-SOCS3.
  • Expressions of cell cycle (p21, p2′7, cyclinE), proliferation (PCNA), and angiogenesis (HIF1a, VEGF)-related genes were significantly regulated by treatment of iCP-SOCS3.
  • Accordingly, it was confirmed that iCP-SOCS3 is able to inhibit development of breast cancer through expressions of various factors regulating cancer activity such as cell cycle, angiogenesis, etc., indicating characteristics of iCP-SOCS3 as a mechanism-specific therapeutic agent targeting solid tumors.
  • Further, tumor tissues removed at 2 weeks (Day 35) after termination of drug administration were used to perform Western blotting and IHC, and increase or decrease in the expressions of cancer activity regulating factors by iCP-SOCS3 was observed.
  • In iCP-SOCS3-treated tumor tissues, expression of a cell cycle regulator, p21 was increased and expressions of angiogenesis inducers, VEGF and CD31 were remarkably decreased (FIG. 93). Accordingly, it was demonstrated that cell cycle and angiogenesis are also regulated by iCP-SOCS3 in-vivo.
  • Statistical Analysis
  • Statistical analysis and graphic presentation have been performed using GraphPad Prism 5.01 software (GraphPad, La Jolla, Calif., USA). All experimental data are presented as means±SEM. Statistical significance was analyzed by the Student's t-test or ANOVA method. Experimental differences between groups were assessed using paired Student's t-tests. For animal experiments, ANOVA was used for comparing between and within groups to determine the significance. Differences with p<0.05 are considered to be statistically significant.
  • Those skilled in the art to which the present invention pertains will appreciate that the present invention may be implemented in different forms without departing from the essential characteristics thereof. Therefore, it should be understood that the disclosed embodiments are not limitative, but illustrative in all aspects. The scope of the present invention is made to the appended claims rather than to the foregoing description, and all variations which come within the range of equivalency of the claims are therefore intended to be embraced therein.

Claims (20)

1. An improved Cell-Permeable (iCP)-SOCS3 recombinant protein, which comprises a SOCS3 protein and an advanced macromolecule transduction domain (aMTD) being composed of 9-13 amino acid sequences and having improved cell and/or tissue permeability,
wherein the aMTD is fused to one end or both ends of the SOCS3 protein and has the following features of:
(a) being composed of 3 or more amino acids sequences selected from the group consisting of Ala, Val, Ile, Leu, and Pro;
(b) having proline as amino acid sequences corresponding to any one or more of positions 5 to 8, and 12 of its amino acid sequence; and
(c) having an instability index of 40-60; an aliphatic index of 180-220; and a grand average of hydropathy (GRAVY) of 2.1-2.6, as measured by Protparam.
2. The iCP-SOCS3 recombinant protein according to claim 1, wherein one or more solubilization domain (SD)(s) are further fused to the end(s) of one or more of the SOCS3 protein and the aMTD.
3. The iCP-SOCS3 recombinant protein according to claim 1, wherein the aMTD is composed of 12 amino acid sequences and represented by the following general formula:
Figure US20170137482A1-20170518-P00999
Here, X(s) independently refer to Alanine (A), Valine (V), Leucine (L) or Isoleucine (I); and Proline (P) can be positioned in one of U(s) (either 5′, 6′, 7′ or 8′); and the remaining U(s) are independently composed of A, V, L or I, P at the 12′ is Proline.
4. The iCP-SOCS3 recombinant protein according to claim 1, wherein the SOCS3 protein has an amino acid sequence of SEQ ID NO: 814.
5. The iCP-SOCS3 recombinant protein according to claim 4, wherein the SOCS3 protein is encoded by a polynucleotide sequence of SEQ ID NO: 815.
6. The iCP-SOCS3 recombinant protein according to claim 1, wherein the SOCS3 protein further comprises a ligand selectively binding to a receptor of a cell, a tissue, or an organ.
7. The iCP-SOCS3 recombinant protein according to claim 1, wherein the aMTD has an amino acid sequence selected from the group consisting of SEQ ID NOs: 1-240 and 822.
8. The iCP-SOCS3 recombinant protein according to claim 7, wherein the aMTD is encoded by a polynucleotide sequence selected from the group consisting of SEQ ID NOs: 241-480 and 823.
9. The iCP-SOCS3 recombinant protein according to claim 2, wherein the SD(s) have an amino acid sequence independently selected from the group consisting of SEQ ID NOs: 798, 799, 800, 801, 802, 803, and 804.
10. The iCP-SOCS3 recombinant protein of claim 9, wherein the SD(s) are encoded by a polynucleotide sequence independently selected from the group consisting of SEQ ID NOs: 805, 806, 807, 808, 809, 810, and 811.
11. The iCP-SOCS3 recombinant protein according to claim 1, wherein the iCP-SOCS3 recombinant protein has a histidine-tag affinity domain additionally fused to one end thereof.
12. The iCP-SOCS3 recombinant protein according to claim 11, wherein the histidine-tag affinity domain has an amino acid sequence of SEQ ID NO: 812.
13. The iCP-SOCS3 recombinant protein of claim 12, wherein the histidine-tag affinity domain is encoded by a polynucleotide sequence of SEQ ID NO: 813.
14. The iCP-SOCS3 recombinant protein according to claim 1, wherein the fusion is formed via a peptide bond or a chemical bond.
15. The iCP-SOCS3 recombinant protein according to claim 1, wherein the iCP-SOCS3 recombinant protein is used for the treating, preventing, or delaying the onset of, solid tumor.
16. A polynucleotide sequence encoding the iCP-SOCS3 recombinant protein of claim 1.
17. A recombinant expression vector comprising the polynucleotide sequence of claim 16.
18. A transformant transformed with the recombinant expression vector of claim 17.
19. A preparing method of the iCP-SOCS3 recombinant protein of claim 1 comprising:
preparing the recombinant expression vector of claim 17;
preparing a transformant using the recombinant expression vector;
culturing the transformant; and
recovering the recombinant protein expressed by the culturing.
20. A method of treating, preventing, or delaying the onset of, solid tumor in a subject comprising:
identifying a subject in need of treating, preventing, or delaying the onset of, solid tumor; and
administering to the subject a therapeutically effective amount of the iCP-SOCS3 recombinant protein of claim 1.
US15/361,701 2014-08-27 2016-11-28 Cell-permeable (icp)-socs3 recombinant protein and uses thereof Abandoned US20170137482A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
US15/361,701 US20170137482A1 (en) 2014-08-27 2016-11-28 Cell-permeable (icp)-socs3 recombinant protein and uses thereof
US16/426,751 US10961292B2 (en) 2014-08-27 2019-05-30 Cell-permeable (ICP)-SOCS3 recombinant protein and uses thereof

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201462042493P 2014-08-27 2014-08-27
US14/838,304 US20160060314A1 (en) 2014-08-27 2015-08-27 Development of a Protein-Based Biotherapeutic Agent That Penetrates Cell-Membrane and Induces Anti-Tumor Effect in Solid Tumors - Improved Cell-Permeable Suppressor of Cytokine Signaling (iCP-SOCS3) Proteins, Polynucleotides Encoding the Same, and Anti-Tumor Compositions Comprising the Same
US15/361,701 US20170137482A1 (en) 2014-08-27 2016-11-28 Cell-permeable (icp)-socs3 recombinant protein and uses thereof

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US14/838,304 Continuation-In-Part US20160060314A1 (en) 2014-08-27 2015-08-27 Development of a Protein-Based Biotherapeutic Agent That Penetrates Cell-Membrane and Induces Anti-Tumor Effect in Solid Tumors - Improved Cell-Permeable Suppressor of Cytokine Signaling (iCP-SOCS3) Proteins, Polynucleotides Encoding the Same, and Anti-Tumor Compositions Comprising the Same
US14/838,304 Continuation US20160060314A1 (en) 2014-08-27 2015-08-27 Development of a Protein-Based Biotherapeutic Agent That Penetrates Cell-Membrane and Induces Anti-Tumor Effect in Solid Tumors - Improved Cell-Permeable Suppressor of Cytokine Signaling (iCP-SOCS3) Proteins, Polynucleotides Encoding the Same, and Anti-Tumor Compositions Comprising the Same

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US16/426,751 Division US10961292B2 (en) 2014-08-27 2019-05-30 Cell-permeable (ICP)-SOCS3 recombinant protein and uses thereof

Publications (1)

Publication Number Publication Date
US20170137482A1 true US20170137482A1 (en) 2017-05-18

Family

ID=55401724

Family Applications (15)

Application Number Title Priority Date Filing Date
US14/838,280 Abandoned US20160060311A1 (en) 2014-08-27 2015-08-27 Development of Protein-Based Biotherapeutics That Penetrates Cell-Membrane and Induces Anti-Lung Cancer Effect - Improved Cell-Permeable Suppressor of Cytokine Signaling (iCP-SOCS3) Proteins, Polynucleotides Encoding the Same, and Anti-Lung Cancer Compositions Comprising the Same
US14/838,295 Abandoned US20160060313A1 (en) 2014-08-27 2015-08-27 Development of Protein-Based Biotherapeutics That Penetrates Cell-Membrane and Induces Anti-Angiogenic Effect - Improved Cell-Permeable Suppressor of Cytokine Signaling (iCP-SOCS3) Proteins, Polynucleotides Encoding the Same, and Anti-Angiogenic Compositions Comprising the Same
US14/838,318 Abandoned US20160060319A1 (en) 2014-08-27 2015-08-27 Development of Protein-Based Biotherapeutics That Induced Osteogenesis for Bone Healing Therapy: Cell-Permeable BMP2 and BMP7 Recombinant Proteins (CP-BMP2 & CP-BMP7), Polynucleotides Encoding the Same and Pro-osteogenic Compositions Comprising the Same
US14/838,260 Abandoned US20160060310A1 (en) 2014-08-27 2015-08-27 Development of Protein-Based Biotherapeutics That Penetrates Cell-Membrane and Induces Anti-Hepatocellular Carcinoma Effect - Improved Cell-Permeable Suppressor of Cytokine Signaling (iCP-SOCS3) Proteins, Polynucleotides Encoding the Same, and Anti-Hepatocellular Carcinoma Compositions Comprising the Same
US14/838,304 Abandoned US20160060314A1 (en) 2014-08-27 2015-08-27 Development of a Protein-Based Biotherapeutic Agent That Penetrates Cell-Membrane and Induces Anti-Tumor Effect in Solid Tumors - Improved Cell-Permeable Suppressor of Cytokine Signaling (iCP-SOCS3) Proteins, Polynucleotides Encoding the Same, and Anti-Tumor Compositions Comprising the Same
US14/838,288 Abandoned US20160060312A1 (en) 2014-08-27 2015-08-27 Development of Protein-Based Biotherapeutics That Penetrates Cell-Membrane and Induces Anti-Pancreatic Cancer Effect - Improved Cell-Permeable Suppressor of Cytokine Signaling (iCP-SOCS3) Proteins, Polynucleotides Encoding the Same, and Anti-Pancreatic Cancer Compositions Comprising the Same
US15/361,701 Abandoned US20170137482A1 (en) 2014-08-27 2016-11-28 Cell-permeable (icp)-socs3 recombinant protein and uses thereof
US15/408,230 Abandoned US20170198019A1 (en) 2014-08-27 2017-01-17 Cell-permeable (icp)-socs3 recombinant protein and uses thereof
US15/408,123 Active US10781241B2 (en) 2014-08-27 2017-01-17 Cell-permeable (iCP)-SOCS3 recombinant protein and uses thereof
US15/432,662 Active US10385103B2 (en) 2014-08-27 2017-02-14 Cell-permeable (ICP)-SOCS3 recombinant protein and uses thereof
US15/631,982 Active US10787492B2 (en) 2014-08-27 2017-06-23 Cell-permeable (iCP)-SOCS3 recombinant protein and uses thereof
US15/884,884 Active US10774123B2 (en) 2014-08-27 2018-01-31 Cell-permeable bone morphogenetic protein (CP-BMP) recombinant protein and use thereof
US16/426,864 Active US10975132B2 (en) 2014-08-27 2019-05-30 Cell-permeable (ICP)-SOCS3 recombinant protein and uses thereof
US16/426,751 Active US10961292B2 (en) 2014-08-27 2019-05-30 Cell-permeable (ICP)-SOCS3 recombinant protein and uses thereof
US16/831,520 Active 2036-01-18 US11279743B2 (en) 2014-08-27 2020-03-26 Cell-permeable bone morphogenetic protein (CPBMP) recombinant protein and use thereof

Family Applications Before (6)

Application Number Title Priority Date Filing Date
US14/838,280 Abandoned US20160060311A1 (en) 2014-08-27 2015-08-27 Development of Protein-Based Biotherapeutics That Penetrates Cell-Membrane and Induces Anti-Lung Cancer Effect - Improved Cell-Permeable Suppressor of Cytokine Signaling (iCP-SOCS3) Proteins, Polynucleotides Encoding the Same, and Anti-Lung Cancer Compositions Comprising the Same
US14/838,295 Abandoned US20160060313A1 (en) 2014-08-27 2015-08-27 Development of Protein-Based Biotherapeutics That Penetrates Cell-Membrane and Induces Anti-Angiogenic Effect - Improved Cell-Permeable Suppressor of Cytokine Signaling (iCP-SOCS3) Proteins, Polynucleotides Encoding the Same, and Anti-Angiogenic Compositions Comprising the Same
US14/838,318 Abandoned US20160060319A1 (en) 2014-08-27 2015-08-27 Development of Protein-Based Biotherapeutics That Induced Osteogenesis for Bone Healing Therapy: Cell-Permeable BMP2 and BMP7 Recombinant Proteins (CP-BMP2 & CP-BMP7), Polynucleotides Encoding the Same and Pro-osteogenic Compositions Comprising the Same
US14/838,260 Abandoned US20160060310A1 (en) 2014-08-27 2015-08-27 Development of Protein-Based Biotherapeutics That Penetrates Cell-Membrane and Induces Anti-Hepatocellular Carcinoma Effect - Improved Cell-Permeable Suppressor of Cytokine Signaling (iCP-SOCS3) Proteins, Polynucleotides Encoding the Same, and Anti-Hepatocellular Carcinoma Compositions Comprising the Same
US14/838,304 Abandoned US20160060314A1 (en) 2014-08-27 2015-08-27 Development of a Protein-Based Biotherapeutic Agent That Penetrates Cell-Membrane and Induces Anti-Tumor Effect in Solid Tumors - Improved Cell-Permeable Suppressor of Cytokine Signaling (iCP-SOCS3) Proteins, Polynucleotides Encoding the Same, and Anti-Tumor Compositions Comprising the Same
US14/838,288 Abandoned US20160060312A1 (en) 2014-08-27 2015-08-27 Development of Protein-Based Biotherapeutics That Penetrates Cell-Membrane and Induces Anti-Pancreatic Cancer Effect - Improved Cell-Permeable Suppressor of Cytokine Signaling (iCP-SOCS3) Proteins, Polynucleotides Encoding the Same, and Anti-Pancreatic Cancer Compositions Comprising the Same

Family Applications After (8)

Application Number Title Priority Date Filing Date
US15/408,230 Abandoned US20170198019A1 (en) 2014-08-27 2017-01-17 Cell-permeable (icp)-socs3 recombinant protein and uses thereof
US15/408,123 Active US10781241B2 (en) 2014-08-27 2017-01-17 Cell-permeable (iCP)-SOCS3 recombinant protein and uses thereof
US15/432,662 Active US10385103B2 (en) 2014-08-27 2017-02-14 Cell-permeable (ICP)-SOCS3 recombinant protein and uses thereof
US15/631,982 Active US10787492B2 (en) 2014-08-27 2017-06-23 Cell-permeable (iCP)-SOCS3 recombinant protein and uses thereof
US15/884,884 Active US10774123B2 (en) 2014-08-27 2018-01-31 Cell-permeable bone morphogenetic protein (CP-BMP) recombinant protein and use thereof
US16/426,864 Active US10975132B2 (en) 2014-08-27 2019-05-30 Cell-permeable (ICP)-SOCS3 recombinant protein and uses thereof
US16/426,751 Active US10961292B2 (en) 2014-08-27 2019-05-30 Cell-permeable (ICP)-SOCS3 recombinant protein and uses thereof
US16/831,520 Active 2036-01-18 US11279743B2 (en) 2014-08-27 2020-03-26 Cell-permeable bone morphogenetic protein (CPBMP) recombinant protein and use thereof

Country Status (3)

Country Link
US (15) US20160060311A1 (en)
EP (4) EP3341400B1 (en)
WO (6) WO2017034349A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20180195047A1 (en) * 2015-08-10 2018-07-12 Cellivery Therapeutics, Inc. Cell-permeable reprogramming factor (icp-rf) recombinant protein and use thereof
US20180237485A1 (en) * 2014-08-27 2018-08-23 Cellivery Therapeutics, Inc. Cell-permeable bone morphogenetic protein (cp-bmp) recombinant protein and use thereof
JP7450983B2 (en) 2020-02-18 2024-03-18 セリベリー セラピューティクス,インコーポレーテッド Improved cell-permeable nuclear transport inhibitor synthetic peptides and their uses for suppressing cytokine storm or inflammatory diseases

Families Citing this family (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20170029798A1 (en) 2015-07-27 2017-02-02 Cellivery Therapeutics, Inc. Development of Improved Cell-Permeable (iCP) Parkin Recombinant Protein as a Protein-Based Anti-Neurodegenerative Agent for the Treatment of Parkinson's Disease-Associated Phenotypes by Utilizing BBB-Penetrating Protein Delivery System MITT, Enabled by Advanced Macromolecule Transduction Domain (aMTD)
EP3337815B1 (en) * 2015-08-18 2020-12-16 Cellivery Therapeutics, Inc. Cell-permeable (cp)- socs3 recombinant protein and uses thereof
EP3443001A4 (en) 2016-04-11 2020-04-29 Obsidian Therapeutics, Inc. Regulated biocircuit systems
AU2017298385A1 (en) * 2016-07-19 2019-03-07 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Oncolytic viruses targeting STAT3
WO2018062866A2 (en) * 2016-09-28 2018-04-05 Cellivery Therapeutics, Inc. CELL-PERMEABLE (CP)-Cas9 RECOMBINANT PROTEIN AND USES THEREOF
KR101944517B1 (en) 2016-12-27 2019-02-01 서울대학교산학협력단 Dual functional peptide for cell penetration and bone tissue regeneration and use of the same
CN108727469B (en) * 2017-04-17 2021-10-29 北京翼方生物科技有限责任公司 Novel cell-penetrating peptide for mediating drug delivery and application thereof
CN109411465B (en) * 2017-08-17 2022-04-15 联华电子股份有限公司 Semiconductor structure and design method of virtual pattern layout
EP3806888B1 (en) 2018-06-12 2024-01-31 Obsidian Therapeutics, Inc. Pde5 derived regulatory constructs and methods of use in immunotherapy
EP3870600A1 (en) 2018-10-24 2021-09-01 Obsidian Therapeutics, Inc. Er tunable protein regulation
US20220117989A1 (en) * 2019-01-18 2022-04-21 Children's Medical Center Corporation Compositions and methods for inducing or supplementing socs3 to abrogate tumor growth and proliferative retinopathy
WO2021046451A1 (en) 2019-09-06 2021-03-11 Obsidian Therapeutics, Inc. Compositions and methods for dhfr tunable protein regulation
IL308018A (en) 2021-04-30 2023-12-01 Kalivir Immunotherapeutics Inc Oncolytic viruses for modified mhc expression

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090209458A1 (en) * 2004-03-04 2009-08-20 Vanderbilt University Cell-penetrating socs polypeptides that inhibit cytokine-induced signaling

Family Cites Families (33)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU1205599A (en) 1997-11-03 1999-05-24 Incyte Pharmaceuticals, Inc. Suppressor of cytokine signaling
US7892532B2 (en) * 1999-04-30 2011-02-22 Warsaw Orthopedic, In Emory University Intracellular delivery of osteoinductive proteins and peptides
US20030104622A1 (en) 1999-09-01 2003-06-05 Robbins Paul D. Identification of peptides that facilitate uptake and cytoplasmic and/or nuclear transport of proteins, DNA and viruses
ATE321782T1 (en) 1999-09-27 2006-04-15 Elan Corp MEMBRANE-TRANSLOCING PEPTIDE AS A DRUG TRANSPORT SYSTEM
AU2003244340A1 (en) 2002-02-05 2003-09-02 Indian Institute Of Technology Method for specific integration of t7 rna polymerase gene in the chromosome of corynebacterial and the resultant corynebacteria-t7 promoter based shuttle vector system.
JP4188909B2 (en) 2002-03-29 2008-12-03 クレアゼン インコーポレーテッド Cytoplasmic residual cell membrane permeation peptide and uses thereof {CytoplasmicTransductionPeptides and Usetherof}
US6835810B2 (en) 2002-05-13 2004-12-28 Geneshuttle Biopharma, Inc. Fusion protein for use as vector
US7897394B2 (en) * 2006-09-21 2011-03-01 Intrexon Corporation Endoplasmic reticulum localization signals
KR20080044710A (en) * 2006-11-17 2008-05-21 김정문 Non-activated polypeptides having a function of tissue regeneration and improved cell permeability, and use thereof
KR20090103957A (en) * 2007-01-29 2009-10-01 주식회사 프로셀제약 Novel macromolecule transduction domains and methods for identification and uses thereof
WO2009032148A1 (en) 2007-08-29 2009-03-12 Tufts University Methods of making and using a cell penetrating peptide for enhanced delivery of nucleic acids, proteins, drugs, and adenovirus to tissues and cells, and compositions and kits
WO2009139599A2 (en) 2008-05-16 2009-11-19 주식회사 프로셀제약 Cell-permeable p27 recombinant protein, a polynucleotide that codes the same and an anti-cancer composition containing the same as an active ingredient
US20110229525A1 (en) * 2010-03-12 2011-09-22 Vanderbilt University Modulation of cytokine signaling
WO2012050402A2 (en) * 2010-10-14 2012-04-19 주식회사 프로셀제약 Cell-permeable recombinant parkin protein and a pharmaceutical composition for treating degenerative brain diseases containing the same
CA2819416C (en) 2010-12-02 2019-04-30 Bionor Immuno As Peptide scaffold design
CA2834577A1 (en) 2011-05-23 2012-11-29 Phylogica Limited Method of determining, identifying or isolating cell-penetrating peptides
KR101258279B1 (en) 2011-11-23 2013-04-25 주식회사 프로셀제약 Development of the macromolecule transduction domain with improved cell permeability and its applications
US9259481B2 (en) * 2011-11-23 2016-02-16 Procell Therapeutics Inc. Development of novel macromolecule transduction domain with improved cell permeability and method for using same
US20150315643A1 (en) * 2012-12-13 2015-11-05 Baylor Research Institute Blood transcriptional signatures of active pulmonary tuberculosis and sarcoidosis
US20160122793A1 (en) 2013-05-24 2016-05-05 Novo Nordisk A/S Fusion Protease
JP6621409B2 (en) * 2013-11-22 2019-12-18 ミナ セラピューティクス リミテッド C / EBPα small molecule activated RNA composition
WO2015112904A1 (en) * 2014-01-24 2015-07-30 University Of Florida Research Foundation, Inc. Socs mimetics for the treatment of diseases
KR101643718B1 (en) * 2014-07-16 2016-07-28 한국항공우주연구원 Containing and charging apparatus of pole type for unmanned vtol aircraft and method for containing and charging unmanned vtol aircraft using the same
KR101694161B1 (en) * 2014-07-16 2017-01-09 엘지전자 주식회사 Lighting device and lighting system includes the lighting device
KR20160009456A (en) * 2014-07-16 2016-01-26 계명대학교 산학협력단 Baby protection apparatus of vehicle seat
WO2016028036A1 (en) * 2014-08-17 2016-02-25 Cellivery Therapeutics, Inc. Advanced macromolecule transduction domain (amtd) sequences for improvement of cell-permeability, polynucleotides encoding the same, method to identify the unique features of amtds comprising the same, method to develop the amtd sequences comprising the same
US20160060311A1 (en) 2014-08-27 2016-03-03 Daewoong Jo Development of Protein-Based Biotherapeutics That Penetrates Cell-Membrane and Induces Anti-Lung Cancer Effect - Improved Cell-Permeable Suppressor of Cytokine Signaling (iCP-SOCS3) Proteins, Polynucleotides Encoding the Same, and Anti-Lung Cancer Compositions Comprising the Same
US20160068825A1 (en) * 2014-09-04 2016-03-10 Daewoong Jo Development of Protein-Based Biotherapeutics That Penetrate Cell-Membrane and Induce Anti-Cancer Effect- Cell-Permeable Glutathione Peroxidase7 (CP-GPX7) in Gastrointestinal Track (GIT), Polynucleotides Encoding the Same, and Anti-Cancer Compositions Comprising the Same
US20160083441A1 (en) * 2014-09-24 2016-03-24 Daewoong Jo Development of Protein-Based Biotherapeutics That Penetrate Cell-Membrane and Induce Anti-Cancer Effect - Cell-Permeable Trefoil Factor 1 (CP-TFF1) in Gastrointestinal Track (GIT) Cancer, Polynucleotides Encoding The Same, and Anti-Cancer Compositions Comprising The Same
US20170029798A1 (en) * 2015-07-27 2017-02-02 Cellivery Therapeutics, Inc. Development of Improved Cell-Permeable (iCP) Parkin Recombinant Protein as a Protein-Based Anti-Neurodegenerative Agent for the Treatment of Parkinson's Disease-Associated Phenotypes by Utilizing BBB-Penetrating Protein Delivery System MITT, Enabled by Advanced Macromolecule Transduction Domain (aMTD)
EP3334755B1 (en) * 2015-08-10 2020-04-08 Cellivery Therapeutics, Inc. Improved cell-permeable reprogramming factor (icp-rf) recombinant protein and use thereof
EP3334756B1 (en) * 2015-08-10 2020-06-17 Cellivery Therapeutics, Inc. Improved cell-permeable cre (icp-cre) recombinant protein and use thereof
EP3337815B1 (en) * 2015-08-18 2020-12-16 Cellivery Therapeutics, Inc. Cell-permeable (cp)- socs3 recombinant protein and uses thereof

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090209458A1 (en) * 2004-03-04 2009-08-20 Vanderbilt University Cell-penetrating socs polypeptides that inhibit cytokine-induced signaling

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20180237485A1 (en) * 2014-08-27 2018-08-23 Cellivery Therapeutics, Inc. Cell-permeable bone morphogenetic protein (cp-bmp) recombinant protein and use thereof
US10385103B2 (en) 2014-08-27 2019-08-20 Cellivery Therapeutics, Inc. Cell-permeable (ICP)-SOCS3 recombinant protein and uses thereof
US10774123B2 (en) * 2014-08-27 2020-09-15 Cellivery Therapeutics, Inc. Cell-permeable bone morphogenetic protein (CP-BMP) recombinant protein and use thereof
US10781241B2 (en) * 2014-08-27 2020-09-22 Cellivery Therapeutics, Inc. Cell-permeable (iCP)-SOCS3 recombinant protein and uses thereof
US10787492B2 (en) 2014-08-27 2020-09-29 Cellivery Therapeutics, Inc. Cell-permeable (iCP)-SOCS3 recombinant protein and uses thereof
US10961292B2 (en) 2014-08-27 2021-03-30 Cellivery Therapeutics, Inc. Cell-permeable (ICP)-SOCS3 recombinant protein and uses thereof
US10975132B2 (en) 2014-08-27 2021-04-13 Cellivery Therapeutics, Inc. Cell-permeable (ICP)-SOCS3 recombinant protein and uses thereof
US11279743B2 (en) 2014-08-27 2022-03-22 Cellivery Therapeutics, Inc. Cell-permeable bone morphogenetic protein (CPBMP) recombinant protein and use thereof
US20180195047A1 (en) * 2015-08-10 2018-07-12 Cellivery Therapeutics, Inc. Cell-permeable reprogramming factor (icp-rf) recombinant protein and use thereof
US10508265B2 (en) * 2015-08-10 2019-12-17 Cellivery Therapeutics, Inc. Cell-permeable reprogramming factor (iCP-RF) recombinant protein and use thereof
JP7450983B2 (en) 2020-02-18 2024-03-18 セリベリー セラピューティクス,インコーポレーテッド Improved cell-permeable nuclear transport inhibitor synthetic peptides and their uses for suppressing cytokine storm or inflammatory diseases

Also Published As

Publication number Publication date
EP3341396A4 (en) 2019-03-06
WO2017034333A1 (en) 2017-03-02
EP3341400A4 (en) 2018-09-12
WO2017034349A1 (en) 2017-03-02
EP3341394B1 (en) 2021-07-28
EP3341395A4 (en) 2018-08-08
US20170190754A1 (en) 2017-07-06
EP3341394A4 (en) 2019-01-09
EP3341394A1 (en) 2018-07-04
EP3341400A1 (en) 2018-07-04
US11279743B2 (en) 2022-03-22
US20200299348A1 (en) 2020-09-24
US10385103B2 (en) 2019-08-20
US10961292B2 (en) 2021-03-30
US20160060310A1 (en) 2016-03-03
US20190359669A1 (en) 2019-11-28
WO2017034347A1 (en) 2017-03-02
US20180237485A1 (en) 2018-08-23
US10787492B2 (en) 2020-09-29
US20160060311A1 (en) 2016-03-03
US20170198019A1 (en) 2017-07-13
EP3341396A1 (en) 2018-07-04
EP3341400B1 (en) 2020-10-21
US20160060319A1 (en) 2016-03-03
US10975132B2 (en) 2021-04-13
EP3341396B1 (en) 2021-04-07
US20160060312A1 (en) 2016-03-03
US20170226168A1 (en) 2017-08-10
US10774123B2 (en) 2020-09-15
WO2017034344A1 (en) 2017-03-02
EP3341395A1 (en) 2018-07-04
US20180051060A1 (en) 2018-02-22
EP3341395B1 (en) 2023-11-29
US20190338000A1 (en) 2019-11-07
US20160060313A1 (en) 2016-03-03
US20160060314A1 (en) 2016-03-03
WO2017034330A1 (en) 2017-03-02
WO2017034335A1 (en) 2017-03-02
US10781241B2 (en) 2020-09-22

Similar Documents

Publication Publication Date Title
US10961292B2 (en) Cell-permeable (ICP)-SOCS3 recombinant protein and uses thereof
JP2022078262A (en) Cell penetrating peptide, conjugate comprising the same, and composition comprising the same
JP2020178689A (en) Cell penetrating peptides, conjugates and compositions comprising same
KR102201430B1 (en) Cell penetrating peptide, conjugate comprising same, and composition comprising conjugate
US10662419B2 (en) Cell-permeable (ICP) parkin recombinant protein and use thereof
US10689424B2 (en) Cell-permeable (CP)-Δ SOCS3 recombinant protein and uses thereof
US10669531B2 (en) Cell-permeable Cre (iCP-Cre) recombinant protein and use thereof
EP2868326A1 (en) Peptide inhibitors of TEAD/YAP-TAZ interaction
JP7312948B2 (en) ATF5 peptide variants and their uses
JP5700409B2 (en) Novel anticancer chimeric peptide targeting Hsp90
US9683025B2 (en) Methods and compositions for treating cancer and inflammatory diseases
US20160068825A1 (en) Development of Protein-Based Biotherapeutics That Penetrate Cell-Membrane and Induce Anti-Cancer Effect- Cell-Permeable Glutathione Peroxidase7 (CP-GPX7) in Gastrointestinal Track (GIT), Polynucleotides Encoding the Same, and Anti-Cancer Compositions Comprising the Same
US20220354957A1 (en) Peptides targeting macrophages, and conjugates, compositions, and uses thereof
CA3153060A1 (en) Peptides targeting macrophages, and conjugates, compositions, and uses thereof

Legal Events

Date Code Title Description
AS Assignment

Owner name: JO, DAEWOONG, TENNESSEE

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:JO, DAEWOONG;CHOI, YOUNGSIL;LEE, KUYSOOK;AND OTHERS;REEL/FRAME:040690/0568

Effective date: 20161125

Owner name: CELLIVERY THERAPEUTICS, INC., KOREA, REPUBLIC OF

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:JO, DAEWOONG;CHOI, YOUNGSIL;LEE, KUYSOOK;AND OTHERS;REEL/FRAME:040690/0568

Effective date: 20161125

AS Assignment

Owner name: CELLIVERY THERAPEUTICS, INC., KOREA, REPUBLIC OF

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:JO, DAEWOONG;REEL/FRAME:041061/0842

Effective date: 20170123

AS Assignment

Owner name: CELLIVERY THERAPEUTICS, INC., KOREA, REPUBLIC OF

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:JO, DAEWOONG;REEL/FRAME:041685/0876

Effective date: 20170308

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION