NZ627445B2 - Anticancer fusion protein - Google Patents

Anticancer fusion protein Download PDF

Info

Publication number
NZ627445B2
NZ627445B2 NZ627445A NZ62744512A NZ627445B2 NZ 627445 B2 NZ627445 B2 NZ 627445B2 NZ 627445 A NZ627445 A NZ 627445A NZ 62744512 A NZ62744512 A NZ 62744512A NZ 627445 B2 NZ627445 B2 NZ 627445B2
Authority
NZ
New Zealand
Prior art keywords
seq
domain
sequence
fusion protein
protein
Prior art date
Application number
NZ627445A
Other versions
NZ627445A (en
Inventor
Sebastian Dominik Pawlak
Jerzy Szczepan Pieczykolan
Piotr Kamil Rozga
Bart?Omiej Maciej ?Erek
Original Assignee
Adamed Pharma Sa
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from PL397595A external-priority patent/PL223487B1/en
Application filed by Adamed Pharma Sa filed Critical Adamed Pharma Sa
Publication of NZ627445A publication Critical patent/NZ627445A/en
Publication of NZ627445B2 publication Critical patent/NZ627445B2/en

Links

Classifications

    • 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/005Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from viruses
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/32Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Bacillus (G)
    • 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
    • C07K14/43504Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates
    • C07K14/43563Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates from insects
    • 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
    • C07K14/43504Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates
    • C07K14/43563Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates from insects
    • C07K14/43572Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates from insects from bees
    • 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
    • C07K14/461Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from fish
    • 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
    • C07K14/463Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from amphibians
    • 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
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/525Tumour necrosis factor [TNF]
    • 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
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/70575NGF/TNF-superfamily, e.g. CD70, CD95L, CD153, CD154
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • 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
    • 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
    • C07K7/086Bombesin; Related peptides
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2760/00MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA ssRNA viruses negative-sense
    • C12N2760/00011Details
    • C12N2760/16011Orthomyxoviridae
    • C12N2760/16033Use of viral protein as therapeutic agent other than vaccine, e.g. apoptosis inducing or anti-inflammatory
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N7/00Viruses; Bacteriophages; Compositions thereof; Preparation or purification thereof
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/14Hydrolases (3)
    • C12N9/16Hydrolases (3) acting on ester bonds (3.1)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y301/00Hydrolases acting on ester bonds (3.1)
    • C12Y301/04Phosphoric diester hydrolases (3.1.4)
    • C12Y301/04003Phospholipase C (3.1.4.3)

Abstract

Disclosed is a fusion protein comprising: domain (a) which is a functional fragment of the sequence of soluble hTRAIL protein, which fragment begins with an amino acid in the range from hTRAIL95 to hTRAIL121, inclusive, and ends with the amino acid at the position hTRAIL281, or a homolog of said functional fragment having at least 70% sequence identity, preferably 85% identity, and at least one domain (b) which is the sequence of a cytolytic effector peptide with an amphipathic alpha-helix conformation forming pores in the cell membrane, wherein the sequence of the domain (b) is attached at the C-terminus and/or N-terminus of domain (a). nctional fragment having at least 70% sequence identity, preferably 85% identity, and at least one domain (b) which is the sequence of a cytolytic effector peptide with an amphipathic alpha-helix conformation forming pores in the cell membrane, wherein the sequence of the domain (b) is attached at the C-terminus and/or N-terminus of domain (a).

Description

Anticancer fusion protein The invention relates to the field of therapeutic fusion proteins, especially recombinant fusion proteins. More ularly, the invention relates to fusion proteins comprising the fragment of a sequence of the soluble human TRAIL protein and a sequence of a peptide forming pores in the cell or mitochondrial membrane, pharmaceutical compositions containing them, their use in therapy, especially as anticancer agents, and to polynucleotide ces encoding the fusion proteins, expression vectors containing the cleotide sequences, and host cells containing these expression s.
TRAIL (Tumor Necrosis Factor- Related Apoptosis Inducing Ligand) protein, a member of the cytokines family, also known as ApoZL (ApoZ-ligand), is a potent activator of apoptosis in tumor cells and in cells infected by viruses. TRAIL is a ligand naturally occurring in the body. TRAIL protein, its amino acid sequence, coding DNA sequence SEQ. No.s and protein expression systems were disclosed for the first time in EP0835305A1.
TRAIL protein exerts its anticancer activity by binding to pro-apoptotic surface TRAIL receptors 1 and 2 (TRAIL-R1/R2) and subsequent activation of these receptors. These receptors, also known as DR4 and DR5 (death receptor 4 and death receptor 5), are members of the TNF receptor family and are overexpressed by different types of cancer cells. Activation of these ors can induce external signaling y of suppressor gene p53-independent apoptosis, which by activated e-8 leads to the activation of executive caspases and thereby degradation of c acids. Caspase-8 ed upon TRAIL activation may also cause the release of truncated Bid protein, which is translocated to mitochondria, where it stimulates the release of cytochrome c, thus ctly amplifying the apoptotic signal from death receptors.
TRAIL acts ively on tumor cells, essentially without inducing apoptosis in healthy cells which show resistance to this protein. Therefore, the enormous potential of TRAIL was recognized as an anticancer agent which acts on a wide range of different types of s, including hematologic malignancies and solid tumors, while sparing normal cells and exerting potentially relatively little side effects.
TRAIL protein is a type II membrane n having the length of 281 amino acids, and its extracellular region sing amino acid residues 114-281 upon cleavage by proteases forms soluble sTRAIL molecule of 20 kDa size, which is also biologically active. Both forms, TRAIL and sTRAIL, are capable of ring apoptosis via interaction with TRAIL receptors present on target cells. Strong antitumor activity and very low systemic ty of soluble part of TRAIL molecule was demonstrated using cell lines tests. Also, preliminary human clinical studies with recombinant human soluble TRAIL (rhTRAIL) having amino acid sequence corresponding to amino acids 114-281 of hTRAIL, known under the INN dulanermin, showed its good tolerance and absence of dose limiting toxicity.
Toxic effects of recombinant TRAIL protein on liver cells reported up to now appear to be associated with the ce of modification, i.e. polyhistidine tags, while untagged TRAIL showed no systemic toxicity.
Fragments of TRAIL r than 114-281 are also able to bind with membrane death receptors and induce apoptosis via these receptors, for example, as recently reported in EP 1 688 498 for recombinant circularly permuted mutant of 122-281hTRAIL.
However, in further clinical trials on patients the actual effectiveness of TRAIL as a monotherapy proved to be low. Also problematic was primary or acquired resistance to TRAIL shown by many cancer cells (see for example W02007/022214). ance may be due to various isms and may be specific for a cancer type and/or patient-dependent (Thorburn A, Behbakht K, Ford H. TRAIL receptor-targeted therapeutics: resistance mechanisms and strategies to avoid them. Drug Resist Updat 2008; 11: 17-24). This resistance limits the usefulness of TRAIL as an anticancer agent. Although the ism of resistance to TRAIL has not been fully tood, it is believed that it may st itself at different levels of TRAIL-induced apoptosis pathway, ranging from the level of cell e ors to the executive caspases within the signaling pathway.
To overcome this low efficiency and the resistance of tumors to TRAIL, various combination therapies with radio- and chemotherapeutic agents were designed, which resulted in synergistic apoptotic effect (W02009/ 002947; A. Almasan and 3O A. Ashkenazi, Cytokine Growth Factor Reviews 14 (2003) 337-348; RK Srivastava, Neoplasis, Vol 3, No. 6, 2001, 535-546, Soria JC et al., J. Clin. Oncology, Vol 28, No. 9 (2010), p. 1527-1533). The use of rhTRAIL for cancer treatment in combination with selected conventional chemotherapeutic agents (paclitaxel, latin) and monoclonal anti-VEGF dies are described in W02009/ 140469. However, such a combination necessarily implies well-known deficiencies of conventional herapy or radiotherapy. Prior art is silent, however, about any data suggesting abolishing of cell resistance to TRAIL obtained by fusing TRAIL protein with other proteins or fragments thereof.
Moreover, the problem connected with TRAIL therapy appeared to be its low stability and rapid elimination from the body after administration.
The effect of destruction of cancer cells and tion of tumor proliferation as a result of disintegration ntinuity) of the cell ne or mitochondrial membrane is known. There are also attempts to use substances with cytolytic effect capable of ne disintegration both as an anti-cancer therapy and adjunct anti-cancer therapy.
Many natural and synthetic peptides and proteins having cytolytic activity are known. Cytolytic peptides are also bed as pore-forming peptides or cytolysins. Interactions of pore forming peptides with the surface of the membrane may be based on nonspecific electrostatic ctions of the positively charged peptide with negatively charged e of cell membrane.
These peptides are generally of cationic character, so that they are capable of electrostatic interactions with surfaces with predominantly vely charged particles. Upon contact and interaction of a cytolytic peptide with lipids on the cell surface, and after penetration inside the cell with the lipids on the surface of the ondrial membrane, interruption of the uity of the cell ne occurs, followed by formation of small size transmembrane pores, by which leakage of the ts of the cytoplasm, including ions, outside the cell occurs, resulting in rapid and irreversible electrolyte imbalance in the cell, cell lysis and death.
The interactions of pore-forming peptides with the surface of the membrane may also include interactions with specific receptors present on the surface.
Known naturally occurring cytolytic peptides of bacterial, plant or mammalian origin capable of forming pores in the cell membrane are often called hemolysines, because they cause lysis of red blood cells and other otic cells. These toxins include cecropin A and B, aurein 1.2, citropin 1.1, defensin 3O (HNP-Z), lactoferricin B, lesin, PR-39, cytolysins of Enterococcus faecalis, delta hemolysin, diphtheria toxin, cytolysin of Vibrio cholerae, toxin from Actinia equina, granulysin, lytic peptides from Streptococcus intermedius, lentiviral lytic peptides, leukotoxin of Actinobacillus actinomycetemcomitans, magainin, melittin, lymphotoxin, enkephalin, paradaxin, perforin (in particular the N-terminal fragment thereof), ngolysin O (PFO/theta toxin) from Clostridium perfringens, and streptolysins. Their usefulness as medicaments is limited by their ability to cause hemolysis.
Natural cytolytic peptides are described, for example, in R. czyk et al., Postepy Hig. Med. Dosw., 2009; 63: 360-368 There are also known tic cytolytic pore-forming peptides. They are designed to be devoid of hemolytic properties, to be less immunogenic, or to have surfaces enabling high binding specificity to ar targets such as for example VEGFR (vascular endothelial growth factor receptor) family receptors and the receptors of the EGFR (epidermal growth factor receptor) family. They are often hybrids of natural cytolytic peptides fragments, such as a hybrid of cecropin A fragment and magainin 2 CA (1-8) MA (1-12) fragment or a hybrid of cecropin A fragment and melittin CAMEL (CA (1-7) MEL (2-9)) fragment. There are also known synthetic cytolytic es D-K4-L2-R9 and D-K6-L9, consisting of amino acids lysine, arginine and leucine, part of which is in the form of D-amino acids. There are also known synthetic chimeric peptides RGD-4CD(KLAKLAK)2, which contains the RGD motif binding with integrin (1,433 and an effector domain composed of D-amino acids KLAKLAKKLAKLAK, and PTD-5D(KLAKLAK)2 containing PTD-5 motif which allows penetration into the cells and an effector domain composed of D-amino acids KLAKLAKKLAKLAK (see, for e.g., R. Smolarczyk et al., Postepy Hig. Med. Dosw., 2009, 63: 360-368). Other well-known cytolytic synthetic peptides are described, for example, in Regen et al., m.
Biophys. Res. Commun. 159: 566-571, 1989.
The destruction of the ne occurring after adhering of the peptide to the membrane may occur by the mechanism of "barrel staves" (barrel-stave model), the ism of a nut-like shape" (toroidal-pore model) or a “carpet” mechanism (see, for e.g., R. Smolarczyk et al., Postepy Hig. Med. Dosw., 2009; 63: 360-368).
The mechanism of "barrel staves" is observed for amphipathic peptides with helical mation having a length of at least 23 amino acids. For 3O example, peptides which cause the destruction of the membrane by the mechanism of "barrel staves" are idin A, alameticin, perforin, pilosulin, synthetic peptides with repeated KLAK motifs, cathelicidin, peptides isolated from Entamoeba histolytica, orins and ins. Peptides, which cause the destruction of the membrane by the dal pore model” include, for example, melittin and magainin. For example, peptides which cause the destruction of the membrane by the “carpet” model are cecropins A and B.
Disintegration of cell membrane with formation of pores may be also caused by interaction of peptides of a high positive charge with negatively charged membrane components. Such properties show, among others, granulysins, analogs and derivatives of melittin, peptides comprising K(L)xR motif, tachyplesin, bombesin, magainin and viscotoxin.
The formation of pores in the membrane of the target cell may also be asso- ciated with enzymatic activity of peptides. The enzymatic activity of phospho- lipase A is shown, for example, by phospholipases with specific phospho- diesterase activity against phosphatidylcholine and omyelin, hemolysins and cytolysins having nonspecific cytolytic activity, or hemolysins and cytolysins having cytolytic activity against biological nes containing, for example, cholesterol. This type of enzymatic activity ing in the formation of pores in the cell or mitochondrial membrane is exhibited by listeriolysin, equinatoxin, phospholipase PC-PLC and alpha-toxin from Clostridium ngens.
There are also known conjugates and chimeras of pore-forming es with domains capable to target to tumor cells. For targeting, there are used antigens, carbohydrate moieties or growth factor receptors, overexpressed on the surface of tumor cells. Targeted delivery provides high levels of orming peptide on the cell surface which is necessary for cytolytic activity.
The use of targeted pore forming actinoporins is described in Panchal RG. et al., Poreforming proteins and their application in biotechnology. Curr Pharm Biotechnol 2002, 3299-115; Panchal RG: Novel therapeutic strategies to selectively kill cancer cells. Biochem Pharmacol 1998, 55:247-252 and in Hoskin DW, Ramamoorthy A: Studies on ncer activities of antimicrobial peptides.
Biochim Biophys Acta-Biomembr 2008, 1778:357-87.
It is also known that pore-forming peptides and ns may be endowed with the ability to direct to the tumor associated antigens and receptors by means of appropriate genetic cation as well as by chemical joining to the suitable ligands or antibodies. Such modifications are described for toxin of 3O Bacillus thuringiensis, equinatoxin II from Actinia , sticholysin | of dactyla helianthus and diphtheria toxin of Corynebacterium diphtheriae (Soletti RC., iation of anticancer-drug cytotoxicity by sea anemone pore- forming ns in human glioblastoma cells. Anti-Cancer Drugs 2008, 19:517- 525; Pederzolli C,: Biochemical and cytotoxic properties of ates of transferrin with toxin-ll, a cytolysin from a sea anemone. Bioconjugate Chem 1995, 62166-173, van der Spek JC.: Fusion protein toxins based on diphtheria toxin: Selective targeting of growth factor ors of eukaryotic cells. Appl Chimeric Genes Hybrid Proteins Pt B 2000, 327:239-249).
Also bed is a fusion protein consisting of pore forming sticholysin toxin I and a monoclonal antibody directed against a tumor specific antigen C2, and its usefulness in the treatment in a colon cancer cell line model (Tejuca M. et al., Construction of an immunotoxin with the pore forming protein St1 and/or C5, a monoclonal antibody against a colon cancer cell line, Int. Immunopharmacol. 2004, 4:731-744). A number of fusion proteins sing diphtheria toxin and interleukin-2 or EGF, and their potential to destroy the cell overexpressing the target ors is also described (Murphy JR, van der Spek JC, Targeting diph- theria-toxin to growth-factor receptors, Semin Cancer Biol 1995, 267).
There is also known the use of cleavage sites recognized by ic proteases in fusion proteins molecules comprising cytolytic peptides in order to enable the release of effector proteins in the tumor environment and, consequently, their alization into tumor cells. For example, Panchal R. et al. (Nat Biotechnol 1996, 14:852-856) disclosed alpha-hemolysins sing in their sequence a cleavage site recognized by sin B, which is activated by a protease present in the tumor environment.
There are also known modified proaerolysins (PA), inactive precursors of bacterial cytolytic pore-forming proteins, activated when cleaved by protease of prostate cancer cells (PSA) (Williams S.A. et al., JNCI J. Natl. Cancer Inst. (2007) 99 (5): 376-385). 771B1 discloses conjugates, including fusion proteins, of pore-forming cytolytic peptides with an dy or antigen as an t selectively binding on a tumor cell, and linkers enabling the selective activation of the cytolytic peptide in the tumor environment, such as, for example cleavage site ized by enzymes such as proteases, in particular proteases overexpressed specifically in the tumor environment.
Barua et al. (Cancer Letters 293 (2010) 240—253) reported that prostate cancer 3O cell lines resistant to TRAIL and insensitive to treatment with death receptor agonist antibodies DR4 and DR5 become sensitive to these antibodies after pre- treatment of these cells with synthetic ic amphipathic lytic e KLA containing KLAK sequences.
The present invention provides fusion proteins with anti-cancer properties, which contain a domain derived from TRAIL and a domain of a cytolytic effector peptide with pore-forming properties against cell and/or mitochondrial membranes of mammalian cells.
Each of the two domains of the protein of the invention has different functions.
Due to the presence of a domain derived from hTRAIL, proteins according to the invention are directed ively to cancer cells, wherein the elements of the protein exert their effects. In ular, TRAIL domain after binding with a cell may exert its activity of triggering apoptosis, and the or e the ty of forming pores in cell and/or mitochondrial membrane and causing lysis of the cancer cell.
Delivery of the protein of the invention into the tumor environment allows to minimize toxicity and side effects against healthy cells in the body, as well as reduction of the ncy of administration of a ment. In addition, targeted therapy with the use of proteins according to the invention allows to avoid the problem of low efficiency of previously known nonspecific therapies based on the pores formation in the cell or mitochondrial membrane with the use of plant or bacterial toxins, caused by high toxicity and by the necessity of administering high doses.
It turned out that in many cases fusion proteins of the invention are more potent than soluble hTRAIL and its variants including the fragment of a sequence.
Until now, effector peptides used in the fusion n of the invention have not been used in medicine as such because of unfavorable kinetics, rapid degradation by nonspecific proteases and accumulation in the body caused by lack of proper sequence of activation of pathways, which is necessary to enable the proper action of the effector peptide at target site. Incorporation of the effector es into the fusion protein allows their selective delivery to the site where their action is desirable. Furthermore, the attachment of the or peptide ses the mass of n, which results in prolonged half-life and increased retention of protein in the tumor and its enhanced efficiency.
Novel fusion proteins have also at least reduced or limited, or even substantially 3O eliminated ytic activity compared to their individual natural tic peptides.
Additionally, in many cases, novel fusion proteins also overcome natural or induced resistance to TRAIL. Most likely, overcoming the resistance is due to destabilization of the cell membrane potential as a result of the fusion proteins binding to lipids of cell or mitochondrial membrane and formation of pores, which causes leakage of divalent ions outside the cell. As a consequence of binding to the lipids of mitochondrial membrane, the release of cytochrome C, SMAC/Diablo protein and AIF factor into the cytoplasm occurs, which causes proapoptotic e activation in the affected cell. Degradation of the mitochondrial nes leads also to the activation of caspase-9, resulting in the induction of apoptosis.
Description of Figures.
Fig. 1 presents tumor volume changes (% of initial stage) in be.Cg-foxn1(nu)/J mice burdened with lung cancer A549 treated with fusion protein of the invention of Ex. 11a compared to rhTRAIL114-281; Fig. 2 presents tumor growth inhibition values (%TG|) in be.Cg-foxn1(nu)/J mice burdened with lung cancer A549 treated with fusion protein of the invention of Ex. 11a compared to rhTRAIL114-281; Fig. 3 presents tumor volume changes (% of initial stage) in Crl:SHO- PrkdcscidHrhr mice burdened with lung cancer A549 treated with fusion protein of the invention of Ex. 11a compared to L114-281; Fig. 4 presents tumor growth inhibition values (%TG|) in Crl:SHO-PrkdcscidHrhr mice burdened with lung cancer A549 treated with fusion protein of the invention of Ex. 11a ed to rhTRAIL114-281; Fig. 5 ts tumor volume changes (% of initial stage) in Crl:SHO- PrkdcscidHrhr mice nu ed with lung cancer NCI-H460-Luc2 treated with fusion n of the invention of Ex. 11a compared to rhTRAIL114-281; Fig. 6 presents tumor growth inhibition values (%TG|) in Crl:SHO-PrkdcscidHrhr mice burdened with lung cancer NCl-H460-Luc2 treated with fusion protein of the invention of Ex. 11a ed to rhTRAIL114-281; Fig. 7 ts tumor volume changes (% of initial stage) in foxn1(nu)/J mice burdened with prostate cancer PC3 treated with fusion protein of the invention of Ex. 11a compared to rhTRAIL114-281; Fig. 8 presents tumor growth inhibition values (%TG|) in be.Cg-foxn1(nu)/J mice burdened with te cancer PC3 treated with fusion protein of the invention of Ex. 11a compared to rhTRAIL114-281; Fig. 9 presents tumor volume changes (% of initial stage) in Crl:SHO- PrkdcscidHrhr mice burdened with pancreas cancer PANC-1 treated with fusion protein of the invention of Ex. 11a compared to rhTRAIL114-281; Fig. 10 presents tumor growth inhibition values (%TG|) in Crl:SHO-PrkdcscidHrhr mice ed with pancreas cancer PANC-1 treated with fusion protein of the invention of Ex. 11a compared to rhTRAIL114-281; Fig. 11 presents tumor volume changes (% of initial stage) in Crl:SHO- PrkdcscidHrhr mice burdened with colon cancer HCT116 treated with fusion protein of the invention of Ex. 11a compared to rhTRAIL114-281; Fig. 12 ts tumor growth inhibition values (%TG|) in Crl:SHO-PrkdcscidHrhr mice burdened with colon cancer HCT116 treated with fusion n of the invention of Ex. 11a compared to rhTRAIL114-281; Fig. 13 presents tumor volume changes (% of initial stage) in Crl:SHO- PrkdcscidHrhr mice burdened with colon cancer SW620 treated with fusion protein of the ion of Ex. 16a compared to rhTRAIL114-281; Fig. 14 presents tumor growth inhibition values (%TG|) in Crl:SHO-PrkdcscidHrhr mice burdened with colon cancer SW620 treated with fusion protein of the invention of Ex. 16a compared to rhTRAIL114-281; Fig. 15 presents tumor volume changes (% of initial stage) in O- PrkdcscidHrhr mice burdened with colon cancer Col0205 treated with fusion protein of the invention of Ex. 16a compared to rhTRAIL114-281; Fig. 16 presents tumor growth inhibition values (%TG|) in Crl:SHO-PrkdcscidHrhr mice burdened with colon cancer 5 treated with fusion protein of the invention of Ex. 16a compared to rhTRAIL114-281; Fig. 17 presents tumor volume s (% of initial stage) in Crl:SHO- PrkdcscidHrhr mice burdened with uterine a MES-SA/Dx5 treated with fusion protein of the invention of Ex. 11a compared to rhTRAIL114-281; Fig. 18 presents tumor growth inhibition values (%TG|) in Crl:SHO-PrkdcscidHrhr mice burdened with uterine a MES-SA/Dx5 treated with fusion protein of the ion of Ex. 11a compared to rhTRAIL114-281; Fig. 19 presents tumor volume changes (% of initial stage) in Crl:SHO- PrkdcscidHrhr mice burdened with pancreatic carcinoma MIA Paca-Z treated with fusion protein of the invention of EX. 16b compared to rhTRAIL114-281; Fig. 20 ts tumor growth inhibition values (%TG|) in Crl:SHO-PrkdcscidHrhr mice burdened with pancreatic carcinoma MIA Paca-Z treated with fusion protein of the invention of Ex. 16b compared to rhTRAIL114-281; Fig. 21 presents tumor volume changes (% of l stage) in Crl:SHO-PrkdcscidHrhr mice burdened with colon cancer HCT1 16 treated with fusion protein of the invention of Ex. 16b compared to rhTRAIL114-281; Fig. 22 presents tumor growth inhibition values (%TGI) in Crl:SHO-PrkdcscidHrhr mice ed with colon cancer HCT1 16 treated with fusion n of the invention of Ex. 16 b compared to rhTRAILII4-281; Fig. 23 presents tumor volume changes (% of initial stage) in Crl:SHO-PrkdcscidHrhr mice burdened with hepatocellular carcinoma HepG2 d with fusion protein of the invention of Ex. 16b compared to rhTRAILI 14-281; Fig. 24 presents tumor growth inhibition values (%TGI) in Crl:SHO-PrkdcscidHrhr mice burdened with hepatocellular carcinoma HepG2 d with fusion protein of the invention of Ex. 16b compared to LII4-281; Fig. 25 presents tumor volume changes (% of initial stage) in Crl:SHO-PrkdcscidHrhr mice burdened with hepatoma PLC/PRF/5 treated with fusion protein of the invention of Ex. 16 b compared to rhTRAIL1 14-281; and Fig. 26 presents tumor growth inhibition values (%TGI) in Crl:SHO-PrkdcscidHrhr mice ed with hepatoma PLC/PRF/5 treated with fusion protein of the invention of Ex. 16 b compared to rhTRAILI 14-281.
Detailed Description of the Invention The invention relates to a fusion protein comprising: - domain (a) which is a functional fragment of the sequence of e hTRAIL protein, which fragment begins with an amino acid at a position not lower than hTRAIL95 and ends with the amino acid hTRAIL281 or a homolog of said functional nt having at least 70% sequence identity, preferably 85% identity, and - at least one domain (b) which is the sequence of a cytolytic or peptide forming pores in the cell membrane, wherein the sequence of the domain (b) is attached at the C-terminus and/or N-terminus of domain (a).
In an embodiment the invention relates to a fusion protein comprising: domain (a) which is a functional nt of the sequence of soluble hTRAIL protein, which fragment begins with an amino acid in the range from hTRAIL95 to hTRAIL121, ive, and ends with the amino acid at the position hTRAIL281, or a homolog of said (10567700_1):GGG functional nt having at least 70% sequence identity, preferably 85% identity, and at least one domain (b) which is the sequence of a cytolytic effector peptide with an amphipathic alpha-helix conformation forming pores in the cell membrane, wherein the sequence of the domain (b) is ed at the C-terminus and/or N-terminus of domain (a).
The term "peptide" in accordance with the invention should be understood as a molecule built from plurality of amino acids linked together by means of a peptide bond. Thus, the term "peptide" according to the invention es oligopeptides, polypeptides and proteins. (10567700_1):GGG 1111 In the t invention, the amino acid sequences of peptides will be presented in a conventional manner adopted in the art in the direction from inus (N- end) of the peptide towards its C-terminus (C-end). Any sequence will thus have its N-terminus on the left side and inus on the right side of its linear tation.
The term “a functional soluble fragment of the sequence of soluble hTRAIL n” should be understood as denoting any such fragment of soluble hTRAIL protein that is capable of inducing apoptotic signal in ian cells upon binding to its receptors on the surface of the cells.
It will be also appreciated by a skilled person that the existence of at least 70% or 85% homology of the TRAIL sequence is known in the art.
It should be understood that domain (b) of the or peptide in the fusion protein of the invention is neither hTRAIL protein nor a part or fragment of hTRAIL protein.
The fusion protein of the invention incorporates at least one domain (b) of the effector peptide, attached at the C-terminus and/or or at the N-terminus of domain (a).
By sequence hTRAIL it is understood the known sequence of hTRAIL published in the GenBank database under accession number P505591 as well as in EP0835305A1 and presented in the Sequence Listing of the present ion as SEQ. No. 90.
In a particular embodiment, domain (a) is the fragment of TRAIL sequence, beginning with an amino acid from the range of hTRAIL95 to hTRAIL121, inclusive, and ending with the amino acid TRAIL 281.
In particular, domain (a) may be selected from the group consisting of ces corresponding to hTRAIL95-281, hTRAIL114-281, 115-281, hTRAIL119-281 and hTRAIL121-281. It will be evident to those d in the art that hTRAIL95- 281, hTRAIL114-281, hTRAIL115-281, hTRAIL116-281, hTRAIL119-281 and hTRAIL121-281 represent a fragment of human TRAIL protein starting with amino acid denoted with the number 95, 114, 115, 116, 119 and 121, respectively, and ending with the last amino acid 281, in the known sequence of TRAIL.
In another particular embodiment, domain (a) is the homolog of a functional fragment of soluble TRAIL protein sequence beginning at amino acid position not lower than hTRAIL95 and ending at amino acid hTRAIL281, the sequence of which is at least in 70%, preferably in 85%, identical to original sequence. 1212 In specific variants of this embodiment domain (a) is the homolog of a fragment ed from the group consisting of sequences corresponding to hTRAIL95-281, hTRAIL114-281, hTRAIL115-281, hTRAIL116-281, hTRAIL119-281 and hTRAIL121- 281.
It should be understood that the homolog of a TRAIL fragment is a variation/modification of the amino acid sequence of this nt, wherein at least one amino acid is changed, including 1 amino acid, 2 amino acids, 3 amino acids, 4 amino acids, 5 amino acids, 6 amino acids, and not more than 15% of amino acids, and wherein the fragment of a modified sequence has preserved functionality of the TRAIL sequence, i.e. the ability to bind to cell surface death ors and induce apoptosis in mammalian cells. Modification of the amino acid sequence may include, for example, tution, deletion and/or addition of amino acids.
Preferably, the homolog of TRAIL fragment having modified sequence shows modified affinity to the death receptors DR4 (TRAIL-R1) or DR5 (TRAIL-R2) in comparison with the native fragment of TRAIL.
The term "modified ty" refers to increased affinity and/or affinity with altered receptor selectivity.
Preferably, the homolog of the fragment of TRAIL having modified sequence shows increased affinity to the death receptors DR4 and DR5 compared to native nt of TRAIL.
Particularly preferably, the homolog of a nt of TRAIL having modified ce shows increased affinity to the death receptor DR5 in ison with the death receptor DR4, i .e. increased selectivity DR5/DR4.
Also ably, the homolog of a fragment of TRAIL having modified sequence shows an increased selectivity towards the death receptors DR4 and/or DR5 in relation to the affinity towards the receptors DR1 (TRAIL-R3) and/or DR2 (TRAIL- R4).
Modifications of TRAIL resulting in increased affinity and/or selectivity towards the death receptors DR4 and DR5 are known to those skilled in the art. For e, Tur V, van der Sloot AM, Reis CR, Szegezdi E, Cool RH, Samali A, Serrano L, Quax WJ. DR4-selective tumor necrosis factor-related apoptosis- ng ligand (TRAIL) variants obtained by structure-based design. J. Biol.
Chem. 2008 Jul 18;283(29):20560-8, describe D218H mutation having increased selectivity towards DR4, and Gasparian ME, Chernyak BV, Dolgikh DA, Yagolovich AV, Popova EN, a AM, Moshkovskii SA, Kirpichnikov MP. Generation of new 1313 TRAIL mutants DR5-A and DR5-B with improved selectivity to death or 5, Apoptosis. 2009 Jun;14(6):778-87, be D269H mutation having reduced affinity towards DR4. hTRAIL mutants resulting in increased affinity towards one receptor selected from DR4 and DR5 compared with DR1 and DR2 receptors and increased affinity towards or DR5 compared with DR4 are also described in W02009077857 and W02009066174.
Suitable mutations are one or more mutations in the positions of native hTRAL selected from the group consisting of amino acids 131, 149, 159, 193, 199, 201, 204, 204, 212, 215, 218 and 251, in particular ons involving substitution of an amino acid with a basic amino acid such as lysine, histidine or arginine, or an acidic amino acid such as glutamic acid or aspargic acid. In particular, one or more mutations selected from the group consisting of G131R, G131K, R149I, R149M, R149N, R149K, S159R, Q193H, Q193K, N199H, N199R, K201H, K201R, K204E, K204D, K204L, K204Y, K212R, $215E, S215H, $215K, $215D, D218Y, D218H, K251D, K251E and K251Q, as described in W02009066174, may be mentioned.
Suitable mutations are also one or more mutations in the positions of native hTRAIL selected from the group consisting of amino acids 195, 269 and 214, particularly mutations involving substitution of an amino acid with a basic amino acid such as lysine, histidine or arginine. In particular, one or more mutations selected from the group consisting of D269H, E195R, and T214R, as bed in W02009077857, may be mentioned.
In a ular ment, the domain (a) which is the homolog of a fragment of hTRAIL, is selected from D218H mutant of the native TRAIL sequence, as bed in W02009066174, or the Y189N-R191K-Q193R-H264R-I266R-D269H mutant of the native TRAIL sequence, as described in Gasparian ME et al. tion of new TRAIL mutants DR5-A and DR5-B with improved selectivity to death receptor 5, sis. 2009 Jun; 14(6): 778-87.
Domain (a), i.e. the fragment of TRAIL, is a domain responsible for binding of 3O the construct of the fusion protein to death receptors on the surface of a cell.
Furthermore, domain (a) upon binding will exert its known agonistic activity, i.e. activation of sic pathway of apoptosis.
Domain (b) of the fusion protein of the invention is the domain of an effector peptide with cytolytic activity against eukaryotic cell.
In particular embodiments of the fusion protein of the invention, the effector peptide of domain (b) of the fusion protein is a peptide having pore-forming 1414 ty against cancer cells, selected from the group consisting of SEQ. No. 34 to SEQ. No. 56, and SEQ. No. 125 to SEQ. No. 132.
For the peptide with cytolytic activity it is meant a peptide having the ability of forming pores in the cell membrane, and after penetration into the cell, also in the mitochondrial membrane, thereby disrupting the uity of the membrane. As a result of the disruption of membrane, a leakage of the ts of the cytoplasm, including ions, outside the cell occurs, which causes rapid and irreversible electrolyte imbalance in the cell, and its destruction (cell lysis).
The ability of a peptide to form pores in the cell or mitochondrial membrane and thus g cell lysis a can be ined by a method of testing permeabilization of cell nes known to those skilled in the art, for example by measuring the release from the cell of intracellular substances which previously had been d to the cell, e.g. of ATP or radiolabelled , or by measuring the uptake of a dye, such as trypan blue, which does not occur when the cells are intact.
The cytolytic effector peptide of the invention may be either a natural peptide or a synthetic peptide.
Natural cytolytic pore-forming peptide may be bacterial exotoxin such as alpha— HL, aerolysin, perfringolysin, lysin, streptolysin O, listeriolysin, Bacillus thuringensis toxin, orin of Bacillus thuringensis, lytic molecules from E.coli such as hemolysin or colicin.
Natural cytolytic pore-forming peptide may be also an otic e such as human granulysin, pilosulins family, including pilosulin 1 and pilosulin-5 from the venom of the Australian ants Myrmecia Pilosula, magainin such as magainin- 2 from the skin of African frog Xenopus laevis, aurein 1.2 from the skin of African frog Litoria raniformis, citropin 1.1 from the skin of the tree frog Litoria a, melittin from the venom of the honey bee Apis mellifera, defensins, such as alpha-defensin and beta-defensin isolated from human cells, lactoferricins, such as lactoferricin B from cow's milk, tachyplesin from 3O leukocytes of the crab Tachypleus tridentatus, cecropins A and B, or pleurocidins isolated from the Pleuronectes americanus.
Synthetic cytolytic pore-forming peptide may be known cytolytic peptide such as the hybryd of cecropin A fragment and magainin 2 fragment CA(1-8)MA(1-12), the hybryd of cecropin A fragment and fragment of melittin CAMEL (CA(1- 7)MEL(2-9)), synthetic cytolytic peptides consisting of positively charge amino acids lysine, ne and leucine, the part of which are in the form of D-amino 1515 acids such as D-K4-L2-R9 and D-K6-L9, and peptides containing domain composed of D-amino acid motif KLAKLAK or repetitions thereof, for example (KLAKLAK)2, synthetic hybrid peptides of two lytic peptides such as hybrids in- bombesin and cecropin-melittin, synthetic fusion peptides containing the synthetic cytolytic peptide and domain binding to the receptor present on the cell surface other than TRAIL or or domain that allows penetration into the cell, or lytic peptides based on amphipathic helix model consisting of KLLLK and KLLK series or modified and/or truncated peptides (preferably in the form of fusions with transduction or ing domains) of ian origin.
The effector peptide of domain (b) of the fusion protein of the invention may be a peptide forming pores in the cell or ondrial membrane by direct interactions of the peptides having high positive charge with the negatively charged membrane.
Exemplary sequences of the effector e in this embodiment are designated as SEQ. No. 34 (an active form of human granulysin), SEQ. No. 35 (15-amino acids synthetic lytic peptide), SEQ. No. 38 (peptide from tachyplesin), SEQ. No. 39 (fusion peptide bombesin-magainin 2), SEQ. No. 40 (magainin-2), SEQ. No. 42 ino acids hybrid e in-melittin), SEQ. No. 53 toxin A3 (VtA3)), and SEQ. No. 56 (fusion peptide comprising an EGF inhibitor and synthetic lytic peptide), SEQ. No. 132 (melittin), SEQ. No. 129 and SEQ. No. 131 (fusion peptide comprising bombesin and truncated ns of BMAP27 (B27) or BMAP28 (B28), SEQ. No. 130 (17-amino acids synthetic peptide).
The effector peptide of domain (b) of the fusion protein of the invention may be a pore-forming peptide possessing amphipathic alpha-helixes conformation enabling interactions with biological membranes.
Exemplary sequences of the effector peptide in this embodiment are designated as SEQ. No. 36 (pilosulin-1), SEQ. No. 37 (pilosulin-5), SEQ. No. 41 (14-amino acids synthetic lytic peptide), SEQ. No. 43 (27-amino acids peptide FF/CAP-18), SEQ. No. 44 (BAMP-28 peptide), SEQ. No. 45 (the ue of isoform C of lytic 3O e from Entamoeba histolytica), SEQ. No. 46 (the ue of isoform A of lytic e from Entamoeba histolytica), SEQ. No. 47 (the analogue of isoform B of lytic peptide from Entamoeba histolytica), SEQ. No. 48 (the fragment of HA2 domain of influenza virus hemagglutinin), SEQ. No. 54 (the active fragment of human perforin), SEQ. No. 55 (parasporin-2 from Bacillus thuringensis), SEQ.
No. 125 synthetic fusion peptide with KLLK motif, SEQ. No. 126, and SEQ. No. 127 (pleurocidin analogues), SEQ. No. 128 synthetic peptide with KLLK motif. 1616 The effector peptide of domain (b) of the fusion protein of the invention may be a pore-forming peptide with enzymatic activity selected from the group of phospholipases, hemolysins or cytolysins. Exemplary sequences of the effector peptide in this embodiment are designated as SEQ. No. 49 (N-terminal domain of alpha-toxin from Clostridium perfringens with phospholipase C activity), SEQ.
No. 50 (Listeriolysin O), SEQ. No. 51 (phospholipase PC-PLC), and SEQ. No. 52 (equinatoxin EqTx-II).
The effector peptide of domain (b) with pore-forming activity against eukaryotic cell may be a peptide which is an active form of human granulysin, belonging to the saponin-like family, which exhibit strong g ability to ne lipids (P. M. Lydyard, A. Whelan, M. W. , ie wyklady: Immunologia”, Wydawnictwo Naukowe PWN 2001). Due to ability to bind to membranes ysin is able to degrade mitochondrial membranes. This process leads to the e into the cytoplasm of cytochrome C, n SMAC/Diablo and the AIF factor, which causes activation of apoptotic cascade and activation of caspase-9, also resulting in the induction of apoptosis. Granulysin also activates Bid proteins to the form tBid, which is directly involved in the formation of pores in the membranes of mitochondria (Zhang et al, The Journal of Immunology, 182: 6993-7000, 2009).
In particular, such an effector peptide is no acids peptide presented in the attached sequence listing as SEQ. No. 34.
Another effector peptide of domain (b) with pore-forming activity t eukaryotic cell may be a synthetic cytolytic peptide composed of leucine (L) and lysine (K) and structurally resembling natural lytic peptides from bee venom or a peptide from Ameba histolitica (Makovitzki, A., Suppression of Human Solid Tumor Growth in Mice by umor and Systemic Inoculation of Histidine-Rich and endent Host Defense—like Lytic Peptides, cancer Research, 2009).
Due to the high content of said amino acids the peptide has strong positive charge allowing its selective interaction with membranes of tumor transformed 3O cells and penetration into their structure with formation of pores according to the "barrel staves" mechanism.
In particular, such an effector peptide is 15-amino acids peptide presented in the attached sequence listing as SEQ. No. 35. r effector peptide of domain (b) with pore-forming ty against eukaryotic cell may be a peptide pilosulin-1, which is a cationic molecule derived from venom of the lian ant Myrmecia Pilosula. Pilosulin 1 is a 1717 peptide with high content of lysine and ne regularly repeated in a sequence. Due to the high content of these amino acids, the peptide has a strong positive charge allowing its selective interaction with membranes of cancer cells and formation of pores through the "barrel staves" mechanism (Kourie et al., Am J Physiol Cell Physiol, 278: 1063 — 1087, 2000).
In particular, such an effector peptide is 56-amino acids peptide ted in the attached sequence g as SEQ. No. 36.
Another effector peptide of domain (b) with pore-forming activity t eukaryotic cell may be peptide pilosulin-5, responsible for ionic interactions with the cell membrane resulting in the formation of pores, and as a consequence inhibition of tumor growth. Pilosulin 5 is the peptide belonging to the pilosulins family derived from the venom of Australian ant Myrmecia Pilosula. This peptide has in its structure cyclically repeated pattern of amino acids lysine, alanine, and aspartic acid, imparting a positive charge, which can potentiate interactions with tumor cells e.
In particular, such an or e is 100-amino acids e presented in the attached sequence listing as SEQ. No. 37.
Another effector peptide of domain (b) with pore-forming activity against otic cell may be tachyplesin, cationic peptide isolated from leukocytes of the crab Tridentatus Tachypleus. After ation inside eukaryotic cells tachyplesin exhibits high affinity to ondrial nes and causes their destabilization through the "barrel staves" mechanism and leakage from the mitochondria into the cytoplasm of factors such as cytochrome C, protein SMAC/DIABLO and AIF factor, which leads to cell death (Chen, Y., et al., RGD- Tachyplezin inhibits tumor growth. Cancer Res, 2001. 61(6): p. 2434-8; Ouyang, G.L., et al., Effects of tachyplesin on proliferation and differentiation of human hepatocellular carcinoma SMMC-7721 cells. World J Gastroenterol, 2002. 8(6): p. 1053-8.).
In particular, such an effector peptide is 17-amino acids peptide presented in 3O the attached sequence listing as SEQ. No. 38.
Another effector peptide of domain (b) may be a fusion e bombesin- magainin with cytolytic activity against eukaryotic cell.
Magainins family consists of 21 - 27 amino acid polypeptides isolated from the skin of the African frog Xenopus . The amphipathic alpha-helical structure of the peptides allows them to form pores in the cell membrane through the "barrel staves" mechanism zaki et al, Biochim Biophys Acta, 1327:119-130, 1818 1997). Bombesin, a 14-amino acids peptide with high positive charge isolated from the skin of frogs, belongs to the group of tumor-homing peptides, and as such exhibits high affinity to the surface of some types of solid tumors and blood cancers, which are characterized by a strong negative charge of the cell membrane (Moody et al, Peptides, 1983; volume 4: 683-686). Bombesin is able to bind to cellular receptors for neuromedin B, a close homologue of bombesin existing in the human body, which are highly expressed on the surface of tumor cells. This significantly increases the level of specificity of bombesin and its accumulation in the tissues occupied by the tumor, while minimizing systemic ty. Conjugates of toxic peptides with bombesin exhibit enhanced antitumor activity compared to the individual proteins (Huawei et al, Mol. Pharmaceutics, 2010; 22586-596). Magainin is characterized by limited binding properties to tumor cells receptors and consequently its cytotoxicity is manifested only at high concentrations. It was shown that fusion peptide comprising magainin and bombesin allows to increase specificity and xicity against tumor cells (Liu S. et al., Enhancement of cytotoxicity of antimicrobial peptide magainin II in tumor cells by in-targeted delivery. Acta Pharmacol Sin. 2011 Jan;32(1):79-88).
In particular, such an effector peptide is 40-amino acids e presented in the attached sequence listing as SEQ. No. 39. r effector peptide of domain (b) may be also a fusion peptide comprising in and truncated versions of BMAP27 (BZ7) or BMAP28 (328) with cytolytic activity against otic cell. Such chimeric peptide reveals xic activity and decreased systemic toxicity (Cai H. et al., ive apoptotic killing of solid and logic tumor cells by bombesin-targeted delivery of mitochondria- disrupting peptides, Mol. Pharmaceutics, 2010, 7(2), pp. 586—596).
In ular, such effector peptides are 31-amino acids peptide presented in the attached sequence g as SEQ. No. 129 and 29 amino acids peptide presented in the ed sequence listing as SEQ. No.131. 3O Another effector peptide of domain (b) may be magainin-2 peptide with cytolytic activity against eukaryotic cell, forming pores in cell and mitochondrial membrane. In ular, such an effector peptide is 23-amino acids peptide presented in the attached sequence listing as SEQ. No. 40.
Another tic effector peptide of domain (b) with activity against eukaryotic cell may be a synthetic lytic peptide. Synthetic peptides of formula (KLAKKLA)n (where n is a number of repetitions of the motif) as amphipathic and alpha- 1919 helical proteins after penetration into the cell are selectively accumulated in the negatively charged mitochondrial membrane, causing formation of pores and destabilization of electrostatic ial of mitochondria, thereby selectively eliminating cells of selected cancer cell lines (Javadpour et al, J Med Chem, 39:3107-13, 1996).
In particular, such an effector peptide is 14-amino acids peptide presented in the attached ce listing as SEQ. No. 41.
Another effector peptide of domain (b) with cytolytic activity against eukaryotic cell may be an another synthetic lytic peptide which disintegrates the cell membrane in a detergent-like manner. (Papo N, Shai Y. New lytic peptides based on the D,L-amphipathic helix motif preferentially kill tumor cells compared to normal cells. Biochemistry. 2003 Aug 12;42(31):9346-54).
In particular, such an effector peptide is 17-amino acids e presented in the attached sequence listing as SEQ. No. 128.
Another effector e of domain (b) with cytolytic activity against eukaryotic cell may be cecropin-melittin hybrid e, which causes formation of pores in cell membrane and consequently leads to inhibition of tumor growth. Cecropin A contains in its structure a large number of positively charged amino acids such as lysine, leucine, and alanine (Quellette, A., J., and Selsted, M., E., 1996, FASEB.
J., 10(11), 1280-9), due to which forms pores in the membrane of eukaryotic cells. In addition, cecropin A has toxic activity, destroying the ure of cell cytoskeleton structure by destabilizing the structure of microtubules (Jaynes, J. et al., 1989, Peptide Res., 2 (2), 157-60).
Melittin is a peptide constructed of 25 amino acids, having high positive charge and alpha helical structure, and therefore ly interacts with the membranes of tumor cells and forming pores by the “barrel stave” ism (Smolarczyk, R. et al Peptydy — nowa klasa lekow przeciwnowotworowych, Postepy Hig and Med. Doswiadczalnej, 2009, 63: 360-368). Additionally, melittin stimulates membrane enzyme phospholipase A2, responsible for decomposition 3O of ne phospholipids, which results in the release of fatty acids which are components of the lipid bilayer of the cell membrane. Synthetic ic peptide cecropin A\melittin ed from genetic fusion of positively charged N — terminus of cecropin peptide with hydrophobic N-terminus of melittin peptide exhibits greater cytotoxic activity against target cells and is devoid of haemolytic activity characteristic for in and melittin , H., G. et al (1989) FEBS Lett 259, 6; Andreu, D. et al (1992) FEBS Lett. 296, 190-194). 2020 In particular, such an effector peptide is 26-amino acids peptide presented in the attached sequence listing as SEQ. No. 42. r effector peptide of domain (b) with pore forming activity is the peptide FF/CAP18 bed by Isogai E. in “Antimicrobial and Lipopolysaccharide- Binding Activities of C-Terminal Domain of Human CAP18 Peptides to Genus Leptospira”, The Journal of Applied Research, Vol. 4, No. 1, 2004, 180-185).
FF/CAP18 is the ue of 27-amino acids inal sequence of human cathelicidin hCAP18109.135, which was ed by replacement of 2 amino acid residues with phenylalanines. FF/CAP18 has strongly cationic character, increased in relation to the native sequence due to incorporated modification, and strongly binds to otic cell membranes. Once bound to the surface of the membrane, FF/CAP18 forms channels and ionic pores, leading to destabilization of electrostatic balance of cells. In addition, after penetration inside the cell, the analog builds into the mitochondrial membrane to form ion channels, thus destabilizing the electrostatic potential of mitochondria and leading to the release from mitochondria to cytosol factors such as cytochrome C, iablo or AIF factor, which initiates the process of apoptosis.
In particular, such an effector peptide is no acids peptide presented in the attached sequence listing as SEQ. No. 43.
Another effector peptide of domain (b) with pore forming activity is a peptide 8 with strong positive charge belonging to the cathelicidins family. This peptide is also the structural analogue of human histatins, a group of 12 peptides with a mass below 4 kDa produced by salivary glands cells and exhibiting antibacterial and antifungal properties (W. Kamysz et al., yny - bialka liniowe bogate w histydyne, Nowa Stomatologia 2004). The N-terminal domain of BAMP-28 e is strongly positively charged and is responsible for docking to the cell membrane, whereas the C-terminal part is responsible for the xic activity. (Hugosson, M., D. et al., 1994). Antibacterial peptides and ondrial presequences affect ondrial coupling, ation and 3O protein import. Eur. J. Biochem. 223:1027—1033.). The mechanism of BMAP-28 peptide activity is primarily based on the formation of pores in cell and mitochondrial membranes (A. Risso et al., BMAP-28, an Antibiotic Peptide of Innate Immunity, Induces Cell Death h Opening of the Mitochondrial Permeability tion Pore, MOLECULAR AND CELLULAR BIOLOGY, Mar. 2002, p. 1926—1935).
In particular, such an effector peptide is 27-amino acids peptide presented in the attached sequence listing as SEQ. No. 44. 2121 Another effector peptide of domain (b) with pore forming activity is the analogue of isoform A of lytic e from Entamoeba histolytica responsible for accumulation on the cell surface resulting in pores formation, which in consequence leads to inhibition of tumor growth. Three isoforms A, B, and C of the peptides of Entamoeba histolytica have been identified, located in the granular cytoplasm of the parasite. These are 77-amino acids polypeptides stabilized by three sulfide s, containing in the secondary structure four amphipathic helices e, M et al EMBO J. 11, 506, 1992). These peptides have lytic properties against eukaryotic cells (Leippe, M. and Miller- Eberhard, H.J. logy 87, 5-18, 1994). The third helix in the structure of these peptides has a length suitable for penetration of the cell membrane and formation of pores. Based on the amino acid sequences comprising only the third helix domain of all three isoforms, a series of analog synthetic peptides (A3, B3 and C3) has been constructed having the pore-forming and cytotoxic activity t the human cancer cell lines and characterized with low hemolytic activity (Andra et al., FEBS Letters 385: 96-100,1996).
In ular, such an effector peptide is 24-amino acids peptide presented in the attached sequence g as SEQ. No. 45.
Another effector peptide of domain (b) with pore forming ty against eukaryotic cell is an analogue of isoform B of lytic peptide from Entamoeba histolytica.
In particular, such an effector peptide is 24-amino acids peptide presented in the ed sequence listing as SEQ. No. 46.
Another effector peptide of domain (b) with pore g activity against eukaryotic cell is an analogue of isoform C of lytic peptide from Entamoeba histolytica.
In particular, such an effector peptide is 24-amino acids peptide presented in the attached sequence listing as SEQ. No. 47.
Another effector peptide of domain (b) with pore forming activity against 3O eukaryotic cell is a homologue of ZO-amino acids N-terminal nt, so called on peptide”, of HA2 domain of influenza virus haemagglutinin, responsible for ction of viral capsid with host cell membrane (intercalation) resulting in formation of pores in the cell membrane of the host. Haemagglutinin (HA) of the influenza virus is a homotrimeric glycoprotein responsible for fusion of viral capsid with host cell membrane. Two domains can be distinguished in the structure of the protein, HA1 responsible for or binding and H2 responsible 2222 for interactions with cell membrane. In the structure of HA2 domain only N- terminal part (20 amino acids), so-called "fusion peptide", directly intercalates into the structure of the cell ne (DUrrer P et al., J Biol Chem 271213417— 13421, 1996). Structural analysis of fusion peptide homologues showed that its activity is associated with conformational change leading to the formation of amphipathic alpha helices, which are capable of endosome membrane perforation (Takahashi 5., Biochemistry 29: 6257-6264, 1990). Therefore, derivatives of "fusion peptide" can be used as effective carriers of biologically active substance providing an efficient and quick "escape" from endosomes.
In ular, such an effector peptide is 12-amino acids peptide presented in the attached sequence listing as SEQ. No. 48. r effector peptide of domain (b) with pore g activity against eukaryotic cell is N-terminal domain of alpha toxin from idium perfringens with phospholipase C activity t atidylcholine and sphingomyelin from cell nes, allowing formation of pores. N-terminal domain of the alpha-toxin of Clostridium perfringens includes the active center of phospho- lipase C.
In particular, such an effector peptide is 247-amino acids peptide ted in the attached sequence listing as SEQ. No. 49.
Another effector peptide of domain (b) with pore forming activity against otic cell is a fragment of listeriolysin O, a cholesterol-dependent pore- forming peptide belonging to the group of sins secreted by pathogen and activated after endosome environment acidification pf P, Portnoy DA. iolysin O: a phagosome-specific lysin. Microbes Infect. 2007 Aug; 9( 10): 7. Epub 2007 May 7). It has been shown that listeriolysin O in the form of a fusion protein with targeting protein can specifically eliminate tumor cells (Bergelt S, Frost S, Lilie H. Listeriolysin O as cytotoxic component of an immunotoxin. Protein Sci. 2009 Jun;18(6):1210-20).
In particular, such an effector e is 468-amino acids peptide presented in 3O the attached sequence listing as SEQ. No. 50.
Another effector peptide of domain (b) with pore forming activity against eukaryotic cell is an active fragment of phospholipase PC-PLC. olipase PC- PLC is responsible for efficient lysis of vacuoles in primary endothelial cells and acts synergistically with listeriolysin in the lysis of primary and secondary vacuole. A substrate for PC-PLC is phosphatidylcholine (Smith GA, Marquis H, Jones S, Johnston NC, Portnoy DA, Goldfine H. The two distinct phospholipases C 2323 of Listeria monocytogenes have overlapping roles in escape from a vacuole and cell-to-cell spread. Infect Immun. 1995 Nov;63(11):4231-7).
In particular, such an effector peptide is 288-amino acids peptide presented in the attached sequence listing as SEQ. No. 51.
Another effector peptide of domain (b) with pore forming activity against eukaryotic cell is equinatoxin protein. Equinatoxin EqTx-II is a pore-forming cytolysin isolated from a equina anemone, characterized by a high, non- ic toxicity with respect to mammalian cells.
In particular, such an effector peptide is 179-amino acids peptide presented in the attached sequence listing as SEQ. No. 52.
Another effector peptide of domain (b) with pore forming activity against eukaryotic cell is viscotoxin A3 (VtA3). Viscotoxin A3 is one of thionins from mistletoe (Viscum album). Structurally it consists of a 46-amino acids chain with three disulfide bridges typical for the family (Coulon A. et al., Comparative membrane interaction study of viscotoxins A3, A2 and B from toe (Viscum album) and connections with their structures. m J. 2003 Aug 15;374(Pt 1):71-8). Toxic properties of oxin on cancer cell lines are known (Tabiasco J. et al Mistletoe viscotoxins increase natural killer cell-mediated cytotoxicity.
Eur J Biochem. 2002 May;269(10):2591-600). The exact molecular mechanism of action has not been described for VtA3. It is known, however, that it involves ion of ion ls and damage of membrane structures by permeabilization. A strong positive charge of the molecule favors binding to both the nucleic acids and phospholipids (Giudici M, Pascual R, de la Canal L, PfL'iller K, ler U, Villalain J. ction of viscotoxins A3 and B with membrane model systems: implications to their mechanism of action. Biophys J. 2003 Aug;85 (2):971-81).
In particular, such an effector peptide is 46-amino acids peptide presented in the attached sequence listing as SEQ. No. 53. r or peptide of domain (b) with pore forming activity against 3O eukaryotic cell is a peptide which is an active fragment of human in. The use of proteins of human origin which are "invisible" to the immune system, including human perforin, can solve the problem of limited clinical ness of protein as containing toxins of bacterial, animal or plant origin because of generated strong immunogenicity (Frankel AE. Reducing the immune response to immunotoxin. Clin Cancer Res. 2004 Jan 1;10(1 Pt 1):13-5). N-terminal 34-amino acids fragment of human perforin forming nonspecifically pores in the cell 2424 membrane retains selective cytotoxic activity of the whole protein (Liu CC, Walsh CM, Young JD. Perforin: ure and function. Immunol Today.1995 Apr;16(4):194-201). A perforin fragment fused to an antibody targeting to cancer cells retains selective cytotoxic activity of the whole protein (Wan L. Expression, purification, and ing of a novel immunotoxin containing humanized single- chain fragment variable antibody against CTLA4 and the N-terminal fragment of human perforin. Protein Expr. Purif. 2006 Aug;48(2):307-13. Epub 2006 Mar 9).
In particular, such an effector e is 33-amino acids peptide presented in the attached sequence listing as SEQ. No. 54. r effector peptide of domain (b) with pore-forming activity against eukaryotic cell is parasporin-Z from Bacillus gensis. Parasporins family comprises 13 different toxins belonging to subgroups PS1, PSZ- PS3, PS4 (Ohba M.
Parasporin, a new anticancer protein group from Bacillus thuringiensis.
Anticancer Res. 2009 Jan;29(1):427—33). Parasporin-Z exerts specificity against cancer cells 4, Jurkat, HL60, HepGZ, ) and exists in a form of 37- kDa protoxin activated by cutting off by proteinase K a portion of N and C- terminal nts of respectively 51 and 36 amino acids. Key action of parasporin-2 consist of oligomerization within the cell membrane to form pores having a diameter of about 3 nm, ing in increasing its permeability. Effects of parasporin-2 activity depend on the type of cell lines tested and include ion of so-called "blebbs" or bulges caused by the outflow of the cytoplasm from the cells and their lysis (HepGZ and 3 cells) or formation of vacuole- like structures resulting in the burst of cells (MOLT-4) (Kitada S. Cytocidal actions of parasporin-2,an anti-tumor crystal toxin from Bacillus thuringiensis. J.
Biol. Chem. 2006 Sep 8;281(36):26350-60). In addition, activity of parasporin-2 leads to the destruction of the structure of microtubules, actin filaments lement, ntation of mitochondria and endoplasmic reticulum (Akiba T. Crystal structure of the parasporin-Z Bacillus thuringiensis toxin that recognizes cancer cells. J. Mol. Biol. 2009 Feb 13;386(1):121-33). 3O In particular, such an effector peptide is 251-amino acids peptide presented in the attached sequence g as SEQ. No. 55.
Another effector peptide of domain (b) with pore-forming activity against euka- ryotic cell is a fusion protein comprising synthetic lytic peptide and peptide inhi- bitor of the EGF receptor on the cell surface. Binding of an inhibitor of EGFR on the cell surface allows location of the lytic peptide to the cell surface, and addi- tionally ts localized intracellularly receptor tyrosine . Inhibition of kinase activity results in the lack of cascade of biochemical signals leading to 2525 the release of the cytoplasmic Ca2+ ions and to activation of RAS signaling path- way g to increased activity of glycolyse pathways, protein synthesis and thus causes decreased cell proliferation and limited tumor progression (Carpenter G, Cohen S., (May 1990). rmal growth factor". The l of Biological Chemistry 265 (14): 7709—12). Synthetic peptides comprising repeated leucine and lysine residues as amphipathic and helical proteins selectively elimi- nate ed cancer cell lines (Javadpour et al., J. Med. Chem., 39:3107-13, 1996) In particular, such an effector peptide is 32-amino acids peptide presented in the attached sequence listing as SEQ. No. 56.
Fusion peptide presented in the attached sequence listing as SEQ. No. 56, comprising an EGF inhibitor and synthetic lytic peptide, is novel and has not been described before.
Another effector peptide of domain (b) with pore-forming activity against eukaryotic cell is a fusion protein sing synthetic lytic peptide with KLLK motif and a peptide being antagonist of PDGF or on the cell surface. g of an inhibitor of PDGF on the cell surface allows location of the lytic peptide to the cell surface, and additionally binding affects cell proliferation and angiogenesis (Ostman A. et al., PDGF Receptors as Targets in Tumor Treatment, Adv. Cancer Res., 2007;97:247-74.) In particular, such an effector peptide is 39-amino acids peptide presented in the ed sequence listing as SEQ. No. 125.
Fusion peptide presented in the attached sequence listing as SEQ. No. 125 and a fusion variant of PDGF nist and tic lytic peptide of SEQ. No. 125 is novel and has not been described before.
Another effector peptide of domain (b) with orming activity against eukaryotic cell is a n being analogue of pleurocidin. Pleurocidins are cationic a-helical proteins interacting with cell ne (Cole AM et al., Isolation and characterization of pleurocidin, an antimicrobial peptide in the 3O skin secretions of winter flounder. J Biol Chem.1997 May 2;272(18):12008-13).
Pleurocidin-like peptides are active against breast carcinoma cells, including drug-resistant and slow-growing breast cancer cells. ie AL et al., Pleurocidin-family cationic antimicrobial peptides are cytolytic for breast carcinoma cells and prevent growth of tumor xenografts. Breast Cancer Res. 2011 Oct 24;13(5):R102). 2626 In particular, such effector peptides are 25-amino acids peptide presented in the attached sequence listing as SEQ. No. 126 and 26 amino acids peptide presented in the attached sequence listing as SEQ. No. 127.
Another effector peptide of domain (b) with pore-forming ty against eukaryotic cell is a 327 peptide — a truncated version of BMAP27 protein isolated form bovine myeloid cells belonging to cathelicidin family. (Donati M. et al., Activity of Cathelicidin Peptides t Chlamydia spp., Antimicrob Agents Chemother. 2005 March; 49(3): 1201—1202).
In particular, such or peptides are no acids peptide presented in the attached sequence listing as SEQ. No. 130.
Upon binding to TRAIL receptors present on the surface of cancer cells, the fusion n will exert a double effect. Domain (a), that is a functional fragment of TRAIL or its homolog with preserved functionality, will exert its known agonistic activity, i.e. g to death receptors on the cell surface and activation of sic pathway of apoptosis. The effector e of the domain (b) of the fusion protein will be able to potentially exert its action extra- cellularly or intracellularly in parallel to the activity of TRAIL domain.
In the fusion protein according to the ion, the antitumor TRAIL activity is potentiated by formation of pores in the cell or mitochondrial membrane ing in disturbance of electrostatic charge of the cell, leakage of ions from the cytoplasm or destabilization of electrostatic potential mitochondria and release into the cytoplasm of factors such as cytochrome C, SMAC/DIABLO or factor AIF, which in turn activates internally induced apoptosis synergistic with the signal from the attachment of TRAIL to functional receptors of DR series.
The new fusion proteins also exhibit at least a reduced or limited, or even substantially ated haemolytic activity characteristic for the individual natural cytolytic peptides.
In one of the ments of the invention, domain (a) and domain (b) are linked by at least one domain (c) comprising the sequence of a cleavage site 3O recognized by proteases present in the cell nment, especially in the tumor cell environment, e.g. such as metalloprotease, urokinase or furin.
Sequences recognized by protease may be selected from: a sequence recognized by metalloprotease MMP Pro Leu Gly Leu Ala Gly Glu Pro/ PLGLAGEP, or fragment thereof which with the 2727 last amino acid of the sequence to which is attached forms a sequence recognized by metalloprotease MMP, - a sequence recognized by urokinase uPA Arg Val Val Arg/ RWR, or fragment thereof, which with the last amino acid of the sequence to which is attached forms a sequence recognized by urokinase, and combinations thereof, or a sequence recognized by furin Arg Gln Pro Arg/ RQPR, Arg Gln Pro Arg Gly/RQPRG, Arg Lys Lys Arg/RKKR) or others atypical sequences recognized by furin disclosed by M. Gordon et al., in Inf. and Immun, 1995, 63, No. 1, p. 82-87, or native sequences recognized by furin Arg His Arg Gln Pro Arg Gly Trp Glu Gln Leu/RHRQPRGWEQL or GlnProArgGlyTrpGluGln / HRQPRGWEQ) or fragment thereof, which with the last amino acid of the sequence to which is attached forms a sequence recognized by furin.
In one of the embodiments of the invention, the protease cleavage site is a combination of the sequence ized by metalloprotease MMP and/or a sequence recognized by urokinase uPA and/or a sequence recognized by furin located next to each other in any order.
Preferably, in one of the embodiments domain (c) is a sequence recognized by furin ed from Arg Val Val Arg Pro Leu Gly Leu Ala Gly/ RWRPLGLAG and Pro Leu Gly Leu Ala Gly Arg Val Val Arg/PLGLAGRWR. ses metalloprotease MMP, urokinase uPA and furin are overexpressed in the tumor environment. The presence of the sequence recognized by the protease enables the cleavage of domain (a) from domain (b), i.e. the release of the functional domain (b) and thus its rated activation.
Activation of the effector peptide — onal domain (b) after internalization of the fusion protein into the cell may occur cifically by a ge of domain (a) from domain (b) of the fusion protein of the invention by lisosomal enzymes (non-specific proteases).
The presence of the protease cleavage site, by allowing quick release of the effector peptide, increases the s of transporting the peptide to the place of its action as a result of cutting off from the hTRAIL nt by means of protease overexpressed in the tumor environment before random degradation of the fusion protein by non-specific proteases occurs.
Additionally, a transporting domain (d) may be attached to domain (b) of the effector peptide of the fusion n of the invention. 2828 Domain (d) may be selected from the group consisting of: (d1) polyhistidine sequence transporting through the cell ne, consisting of 6, 7 , 8, 9, 10 or 11 ine residues (His/H); (d2) polyarginine sequence transporting through the cell membrane, consisting of 6, 7 , 8, 9, 10 or 11 arginine residues (Arg/R), (d3) PD4 transporting sequence (protein uction domain 4) Tyr Ala Arg Ala Ala Ala Arg Gln Ala Arg Ala /YARAAARQARA, (d4) a transporting ce consisting of transferrin receptor binding sequence Thr His Arg Pro Pro Met Trp Ser Pro Val Trp Pro /THRPPMWSPVWP, (d5) PD5 transporting sequence (protein transduction domain 5, TAT protein) Tyr Gly Arg Lys Lys Arg Arg Gln Arg Arg Arg /YGRKKRRQRRR, or fragments thereof, which with the last amino acid of the sequence to which they are attached form sequences of transporting s (d1) or (d2); and - combinations thereof.
The combination of domains, e.g. (d1) and (d2), may comprise in particular the combination (d1)/(d2) and (d2)/(d1).
Furthermore, the ation of domains, e.g. (d1) and (d2), may e domains located next to each other and connected to one end of domain (b) and/or domains linked to different ends of domain (b).
It should be understood that in the case when the fusion protein has both the transporting domain (d) attached to domain (b) and domain (c) of the cleavage site n domains (a) and (b), then domain (c) is located in such a manner that after cleavage of the construct transporting domain (d) remains attached to domain (b). In other words, if the fusion protein contains both the transporting domain (d) and the cleavage site domain (c), then domain (d) is located between domain (b) and domain (c), or is located at the end of domain (b) opposite to the place of attachment of domain (d).
The invention comprises also a variant, wherein domain (d) is located between two (c) domains, that is the variant wherein after cleavage of the construct transporting domain, preferably the ocation domain, is not attached neither to the TRAIL domain nor to the effector peptide domain. 2929 The invention does not comprise such a variant in which domain (d) is located between domain (c) and domain (a), that is the variant wherein after cleavage of the uct transporting domain remains attached to the TRAIL domain.
In another embodiment, between domain (a) and domain (b) there is additionally located domain (e) comprising a sequence suitable for ment of a PEG molecule to the fusion protein (pegylation linker). Such a linker may be known sequence Ala Ser Gly Cys Gly Pro Glu Gly/ASGCGPEG or fragments thereof, which with the last amino acid of the sequence to which it is attached forms a sequence suitable for attachment of a PEG molecule. The pegylation linker may be also selected from the group of the following: Ala Ala Cys Ala Ala/AACAA, Ser Gly Gly Cys Gly Gly Ser/SGGCGGS, and Ser Gly Cys Gly Ser/SGCGS, or fragment thereof, which with the last amino acid of the sequence to which it is attached forms a sequence suitable for attachment of a PEG molecule Preferably, the sequence of pegylation linker is Ala Ser Gly Cys Gly Pro Glu Gly/ ASGCGPEG.
Apart from main onal elements of the fusion protein and the cleavage site domain(s), the fusion proteins of the invention may contain a neutral sequence/sequences of a flexible steric linker. Such steric linkers are well known and described in the literature. Their incorporation into the ce of the fusion protein is intended to provide the correct folding of proteins produced by the s of its overexpression in the host cells. In particular, steric linker may be a glycine, glycine-serine or glycine-cysteine-alanine linker.
In particular, steric linker may be a combination of glycine and serine residues such as Gly Gly Gly Gly Ser/GGGGS or any fragment thereof acting as steric , for example a fragment Gly Gly Gly GS, Gly Gly Gly/GGG or Gly Gly Gly Gly/GGGG, Gly Gly Ser Gly Gly, Gly Gly Ser Gly/GGSG, Gly Ser Gly/GSG or Ser Gly Gly/SGG, or combinations thereof.
In other embodiment, the steric linker may be any combination of glycine, serine and alanine residues such as Ala Ser Gly Gly/ASGG or any nt f acting as steric linker, for example Ala Ser Gly/ASG. It is also possible to use the combination of steric linkers, for e the sequence Gly Gly Gly Ser Gly / GGGGS or any nt thereof acting as steric linker, for example the fragment Gly Gly Gly/GGG, with another fragment acting as steric linker. In such 3030 a case the steric linker may be a combination of glycine, serine and alanine residues such as Gly Gly Gly Ser Ala Ser Gly Gly/GGGSASGG, Gly Gly Ser Gly Gly Gly Ser Gly Gly Gly GSGGG, Gly Gly Ser Gly Gly Gly Gly Gly Ser/GGSGGGGGS or Gly Gly Gly Gly Gly Gly Ser/ GGGGGGS. In still another embodiment, steric linker may be a combination of serine and histidine residues Ser His His Ser/SHHS or Ser His His Ala Ser/SHHAS.
In still another embodiment, the steric linker may be also ed from single amino acid residues such as single glycine or cysteine residue, in ular one or two up to four glycine or cysteine residues.
In another embodiment, the linker may also be formed by a fragment of steric linkers described above, which with the terminal amino acid of the sequence to which it is attached forms a steric linker sequence.
In another embodiment, the steric linker may promote the formation and stabilization of the structure of the trimer of the fusion protein of the invention, thus increasing its half-life in the blood circulation system and preventing from deasociation which may affect activity of the protein after administration into the blood circulation . In this case the linker is a combination of cysteine and alanine, for example, a fragment Cys Cys Ala Ala Ala Ala Ala Cys/ AC or Cys Cys Ala Ala Ala Ala Ala Cys / CAACAAAC or fragments thereof, which is the terminal amino acid sequence to which it is attached and forms a steric linker sequence stabilising the trimer structure.
In addition, the steric linker may also be useful for activation of functional domain (b), occurring in a non-specific manner. Activation of domain (b) in a non-specific manner may be performed by cutting off the domain (a) from the domain (b) of the fusion protein according to the invention due to pH-dependent hydrolysis of the steric linker.
Particular embodiments of the fusion protein of the invention are fusion proteins sing a pore-forming peptide ed from the group of es represented by: 3O SEQ. No. 34, SEQ. No. 35; SEQ. N0. 36, SEQ. No. 37, SEQ. No. 38, SEQ. No. 39, SEQ. No. 40, SEQ. No. 41, SEQ. No. 42, SEQ. No. 43, SEQ. No. 44, SEQ. No. 45, SEQ. No. 46, SEQ. No. 47, SEQ. No. 48, SEQ. No. 49, SEQ. No. 50, SEQ. No. 51, SEQ. No. 52, SEQ. No. 53, SEQ. N0. 54, SEQ. No. 55m SEQ. No. 56, SEQ. No. 125, SEQ. No. 126, SEQ. No. 127, SEQ. No. 128, SEQ. No. 129, SEQ. No. 130, SEQ. No. 131, and SEQ. No. 132. 3131 A detailed description of the ure of representative fusion proteins mentioned above are shown in the Examples presented below.
In accordance with the present invention, by the fusion protein it is meant a single protein molecule containing two or more proteins or fragments thereof, covalently linked via peptide bond within their respective peptide chains, without additional chemical linkers.
The fusion protein can also be alternatively described as a protein construct or a chimeric protein. According to the present invention, the terms “construct” or “chimeric protein”, if used, should be understood as referring to the fusion protein as d above.
For a person skilled in the art it will be apparent that the fusion n thus d can be synthesized by known methods of chemical synthesis of es and ns.
The fusion protein can be sized by methods of chemical peptide synthesis, especially using the techniques of peptide synthesis in solid phase using suitable resins as carriers. Such techniques are conventional and known in the art, and described inter alia in the monographs, such as for example Bodanszky and zky, The Practice of Peptide Synthesis, 1984, Springer- Verlag, New York, Stewart et al., Solid Phase Peptide Synthesis, 2nd Edition, 1984, Pierce Chemical Company.
The fusion protein can be synthesized by the methods of chemical synthesis of peptides as a continuous protein. Alternatively, the individual fragments (domains) of protein may be synthesized separately and then combined together in one continuous peptide via a peptide bond, by condensation of the amino terminus of one peptide nt from the carboxyl terminus of the second peptide. Such techniques are conventional and well known.
Preferably, however, the fusion protein of the invention is a inant protein, generated by methods of gene expression of a polynucleotide sequence ng the fusion protein in host cells.
For cation of the structure of the resulting peptide known methods of the analysis of amino acid composition of peptides may be used, such as high resolution mass spectrometry technique to determine the molecular weight of the peptide. To confirm the peptide sequence, protein sequencers can also be used, which tially e the peptide and identify the sequence of amino acids. 3232 A further aspect of the invention is a polynucleotide sequence, particularly DNA sequence, encoding the fusion protein as d above. ably, the polynucleotide sequence, particularly DNA, according to the ion, encoding the fusion n as defined above, is a sequence optimized for expression in E. coli.
Another aspect of the invention is also an expression vector containing the polynucleotide sequence, ularly DNA sequence of the invention as defined above.
Another aspect of the invention is also a host cell comprising an expression vector as defined above.
A preferred host cell for expression of fusion proteins of the invention is an E. coli cell.
Methods for generation of recombinant proteins, including fusion proteins, are well known. In brief, this que consists in generation of polynucleotide molecule, for example DNA molecule encoding the amino acid sequence of the target protein and directing the expression of the target protein in the host.
Then, the target protein encoding polynucleotide molecule is incorporated into an appropriate expression vector, which ensures an efficient expression of the polypeptide. Recombinant expression vector is then introduced into host cells for transfection/transformation, and as a result a transformed host cell is produced. This is followed by a culture of transformed cells to overexpress the target protein, purification of obtained proteins, and optionally g off by ge the tag sequences used for expression or purification of the protein.
Suitable ques of sion and purification are described, for example in the monograph Goeddel, Gene Expression Technology, Methods in Enzymology 185, Academic Press, San Diego, CA (1990), and A. Staron et al., Advances iol., 2008, 47, 2, 1983-1995.
Cosmids, plasmids or modified viruses can be used as expression vectors for the introduction and replication of DNA sequences in host cells. Typically plasmids are used as expression vectors. Suitable plasmids are well known and commercially ble.
Expression vector of the invention comprises a cleotide molecule encoding the fusion protein of the invention and the necessary regulatory sequences for transcription and translation of the coding sequence incorporated into a suitable host cell. Selection of regulatory sequences is dependent on the type of host 3333 cells and can be easily carried out by a person skilled in the art. Examples of such regulatory sequences are transcriptional er and enhancer or RNA polymerase binding sequence, ribosome binding sequence, containing the transcription initiation signal, inserted before the coding sequence, and transcription terminator sequence, inserted after the coding sequence. er, depending on the host cell and the vector used, other sequences may be introduced into the expression vector, such as the origin of replication, additional DNA restriction sites, enhancers, and sequences allowing ion of transcription.
The expression vector will also comprise a marker gene sequence, which confers d phenotype to the transformed cell and enables specific selection of transformed cells. Furthermore, the vector may also contain a second marker sequence which allows to distinguish cells transformed with recombinant plasmid containing inserted coding sequence of the target protein from those which have taken up the plasmid without insert. Most often, typical otic ance s are used, however, any other reporter genes known in the field may be used, whose presence in a cell (in vivo) can be easily ined using autoradiography ques, spectrophotometry or bio- and chemiluminescence.
For example, depending on the host cell, reporter genes such as B-galactosidase, B-glucuronidase, rase, chloramphenicol acetyltransferase or green scent protein may be used.
Furthermore, the sion vector may contain signal sequence, transporting ns to the appropriate cell compartment, e.g. periplasma, where folding is facilitated. Additionally a sequence encoding a label/tag, such as HisTag attached to the N-terminus or GST attached to the C-terminus, may be present, which facilitates subsequent purification of the protein produced using the ple of affinity, via affinity chromatography on a nickel column. Additional sequences that protect the protein against proteolytic degradation in the host cells, as well as sequences that increase its solubility may also be present. 3O Auxiliary element attached to the sequence of the target protein may block its activity, or be detrimental for another reason, such as for example due to toxicity. Such element must be removed, which may be accomplished by enzymatic or chemical cleavage. In particular, a six-histidine tag HisTag or other markers of this type attached to allow protein purification by affinity tography should be removed, because of its described effect on the liver toxicity of soluble TRAIL protein. Heterologous expression systems based on various nown host cells may be used, including prokaryotic cells: bacterial, 3434 such as Escherichia coli or Bacillus subtilis, yeasts such as Saccharomyces cervisiae or Pichia pastoris, and eukaryotic cell lines (insect, mammalian, plant).
Preferably, due to the ease of culturing and genetic manipulation, and a large amount of obtained product, the E. coli expression system is used. Accordingly, the polynucleotide sequence containing the target sequence encoding the fusion protein of the invention will be optimized for expression in E. coli, i.e. it will n in the coding sequence codons optimal for expression in E. coli, selected from the possible ce variants known in the state of art. Furthermore, the expression vector will contain the above described elements suitable for E. coli attached to the coding sequence.
Accordingly, in a preferred embodiment of the invention a polynucleotide sequence sing a sequence ng a fusion n of the invention, optimized for expression in E. coli is selected from the group of polynucleotide ces consisting of: SEQ. No. 57; SEQ. No. 58; SEQ. No. 59; SEQ. No. 60; SEQ. No. 61; SEQ. No. 62; SEQ. No. 63; SEQ. No. 64; SEQ. No. 65; SEQ. No. 66; SEQ. No. 67; SEQ. No. 68; SEQ. No. 69; SEQ. No. 70; SEQ. No. 71; SEQ. No. 72; SEQ. No. 73; SEQ. No. 74; SEQ. No. 75; SEQ. No. 76; SEQ. No. 77; SEQ. No. 78 ; SEQ. No. 79; SEQ. No. 80; SEQ. No. 81; SEQ. No. 82, SEQ. No. 83; SEQ. No. 84; SEQ. No. 85; SEQ. No. 86; SEQ. No. 87; SEQ. No. 88 SEQ. No. 89; SEQ. No. 108; SEQ. No. 109; SEQ. No. 110 ; SEQ. No. 111; SEQ. No. 112; SEQ. No. 113; SEQ. No. 114, SEQ. No. 115; SEQ.
No. 116; SEQ. No. 117; SEQ. No. 118; SEQ. No. 119;SEQ. No. 120 SEQ. No. 121; SEQ. No. 122; SEQ. No. 123 and SEQ. No. 124, which encode fusion proteins having amino acid sequences corresponding to amino acid sequences selected from the group consisting of amino acid sequences, respectively: SEQ. No. 1; SEQ. No. 2; SEQ. No. 3; SEQ. No. 4; SEQ. No. 5; SEQ. No. 6; SEQ. No. 7; SEQ. No. 8; SEQ. No. 9; SEQ. No. 10; SEQ. No. 11; SEQ. No. 12; SEQ. No. 13; SEQ. No. 14; SEQ. No. 15; SEQ. No. 16; SEQ. No. 17; SEQ. No. 18; SEQ. No. 19; 3O SEQ. No. 20; SEQ. No. 21; SEQ. No. 22 ; SEQ. No. 23; SEQ. No. 24; SEQ. No. 25; SEQ. No. 26, SEQ No. 27; SEQ No. 28; SEQ No 29; SEQ No 30; SEQ No.31; SEQ. No. 32 SEQ No. 33; SEQ No. 91; SEQ No 92; SEQ No 93; SEQ. No. 94; SEQ. No. 95; SEQ No. 96; SEQ No. 97, SEQ No 98; SEQ No 99; SEQ. No. 100; SEQ. No. 101; SEQ. No. 102; SEQ. No. 103 SEQ. No. 104; SEQ. No. 105; SEQ. No. 106, and SEQ. No. 107 3535 In a preferred embodiment, the invention provides also an expression vector suitable for transformation of E. coli, comprising the polynucleotide sequence selected from the group of polynucleotide sequences SEQ. No. 57 to SEQ. No. 87 and SEQ. No. 108 to SEQ. No. 124 indicated above, as well as E. coli cell transformed with such an expression . ormation, i.e. uction of a DNA sequence into bacterial host cells, particularly E. coli, is usually performed on the competent cells, prepared to take up the DNA for example by treatment with calcium ions at low temperature (4°C), and then subjecting to the heat-shock (at 37-42°C) or by electroporation.
Such techniques are well known and are usually determined by the manufacturer of the expression system or are described in the literature and manuals for laboratory work, such as is et al., Molecular Cloning. Cold Spring Harbor, N.Y., 1982).
The procedure of overexpression of fusion proteins of the invention in E. coli expression system will be further described below.
The invention also provides a pharmaceutical composition ning the fusion protein of the invention as defined above as an active ingredient and a le pharmaceutically acceptable carrier, diluent and conventional ary components. The pharmaceutical composition will contain an ive amount of the fusion protein of the invention and pharmaceutically acceptable auxiliary components ved or dispersed in a carrier or diluent, and preferably will be in the form of a pharmaceutical composition formulated in a unit dosage form or formulation containing a plurality of doses. Pharmaceutical forms and methods of their formulation as well as other components, carriers and diluents are known to the skilled person and described in the literature. For example, they are described in the monograph Remington's ceutical Sciences, ed. 20, 2000, Mack Publishing y, Easton, USA.
The terms "pharmaceutically acceptable carrier, diluent, and auxiliary ingredient" comprise any solvents, dispersion media, surfactants, idants, 3O stabilizers, preservatives (e.g. antibacterial agents, antifungal ), isotonizing agents, known in the art. The pharmaceutical composition of the invention may contain various types of carriers, diluents and excipients, ing on the chosen route of administration and desired dosage form, such as liquid, solid and l forms for oral, parenteral, inhaled, topical, and whether that selected form must be sterile for administration route such as by injection. The preferred route of administration of the pharmaceutical composition according to the invention is parenteral, including injection routes 3636 such as intravenous, intramuscular, subcutaneous, intraperitoneal, umoral, or by single or continuous enous infusions.
In one embodiment, the pharmaceutical composition of the invention may be administered by injection directly to the tumor. In another embodiment, the ceutical composition of the invention may be administered intravenously.
In yet another embodiment, the pharmaceutical composition of the invention can be administered subcutaneously or intraperitoneally. A pharmaceutical composition for parenteral administration may be a solution or dispersion in a pharmaceutically acceptable aqueous or non-aqueous medium, ed to an appropriate pH and isoosmotic with body fluids, if necessary, and may also contain antioxidants, buffers, bacteriostatic agents and soluble substances, which make the composition compatible with the tissues or blood of recipient.
Other components, which may included in the composition, are for example water, alcohols such as ethanol, polyols such as ol, propylene glycol, liquid polyethylene , lipids such as triglycerides, vegetable oils, liposomes.
Proper fluidity and the particles size of the substance may be provided by coating substances, such as lecithin, and surfactants, such as hydroxypropyl- celulose, rbates, and the like. le isotonizing agents for liquid parenteral compositions are, for example, sugars such as glucose, and sodium chloride, and combinations thereof.
Alternatively, the pharmaceutical composition for administration by injection or infusion may be in a powder form, such as a lyophilized powder for reconstitution immediately prior to use in a suitable carrier such as, for example, sterile pyrogen-free water.
The pharmaceutical composition of the invention for eral administration may also have the form of nasal administration, including solutions, sprays or aerosols. Preferably, the form for intranasal administration will be an aqueous on and will be isotonic or buffered o maintain the pH from about 5.5 to about 6.5, so as to maintain a character similar to nasal secretions. er, it 3O will n preservatives or stabilizers, such as in the well-known intranasal preparations.
The composition may contain various idants which delay oxidation of one or more components. Furthermore, in order to prevent the action of microorganisms, the composition may contain various antibacterial and anti- fungal , including, for example, and not limited to, parabens, chloro- butanol, thimerosal, sorbic acid, and similar known substances of this type. In 3737 general, the pharmaceutical composition of the invention can include, for example at least about 0.01 wt% of active ingredient. More particularly, the composition may n the active ingredient in the amount from 1% to 75% by weight of the composition unit, or for example from 25% to 60% by weight, but not d to the indicated values. The actual amount of the dose of the composition according to the present invention administered to patients, including man, will be determined by physical and physiological s, such as body weight, severity of the condition, type of disease being treated, previous or concomitant therapeutic interventions, the patient and the route of administration. A suitable unit dose, the total dose and the concentration of active ingredient in the composition is to be determined by the treating physician.
The composition may for example be administered at a dose of about 1 microgram/kg of body weight to about 1000 mg/kg of body weight of the t, for example in the range of 5 mg/kg of body weight to 100 mg/kg of body weight or in the range of 5 mg/kg of body weight to 500 mg/kg of body weight. The fusion protein and the compositions containing it exhibit anticancer or antitumor and can be used for the treatment of cancer diseases. The invention also provides the use of the fusion protein of the invention as defined above for treating cancer diseases in mammals, including humans. The invention also provides a method of treating neoplastic/cancer diseases in mammals, including humans, comprising administering to a subject in need of such treatment an anit-neoplastic/anticancer effective amount of the fusion protein of the invention as defined above, optionally in the form of appropriate pharmaceutical ition.
The fusion protein of the ion can be used for the treatment of hematologic malignancies such as leukaemia, granulomatosis, myeloma and other logic malignancies. The fusion protein can also be used for the treatment of solid tumors such as breast cancer, lung cancer, including non-small cell lung cancer, colon cancer, atic cancer, ovarian cancer, r , prostate cancer, kidney cancer, brain cancer, and the like. Appropriate route of administration of the fusion protein in the treatment of cancer will be in particular parenteral route, which consists in administering the fusion protein of the invention in the form of injections or ons, in the composition and form appropriate for this stration route. The invention will be bed in more detail in the following general procedures and examples of specific fusion proteins. 3838 l procedure for overexpression of the fusion protein Preparation of a plasmid Amino acid sequence of a target fusion protein was used as a template to generate a DNA sequence encoding it, comprising codons optimized for expression in Escherichia coli. Such a procedure allows to increase the efficiency of further step of target protein synthesis in Escherichia coli. Resulting nucleotide sequence was then tically synthesized. Additionally, the cleavage sites of restriction enzymes Ndel (at the 5'-end of leading strand) and Xhol (at the 3'-end of leading strand) were added to the resulting gene encoding the target protein. These were used to clone the gene into the vector pET28a (Novagen). They may be also be used for cloning the gene encoding the protein to other vectors. Target protein expressed from this construct can be optionally equipped at the N-terminus with a polyhistidine tag (six histidines), preceded by a site recognized by thrombin, which subsequently serves to its purification via affinity chromatography. Some targets were expressed t any tag, in particular without ine tag, and those were subsequently ed on SP Sepharose. The tness of the resulting construct was confirmed firstly by restriction analysis of isolated plasmids using the enzymes Ndel and Xhol, followed by automatic sequencing of the entire reading frame of the target n. The primers used for sequencing were complementary to the sequences of T7 promoter (5'-TAATACGACTCACTATAGG-3') and T7 ator (5'- GCTAG'I'I'A'I'I'GCTCAGCGG-3') present in the vector. Resulting plasmid was used for overexpression of the target fusion protein in a commercial E. coli strain, which was transformed according to the manufacturer's recommendations. es obtained on the selection medium (LB agar, kanamycin 50 ug/ml, 1% glucose) were used for ing an overnight culture in LB liquid medium supplemented with kanamycin (50 ug/ml) and 1% glucose. After about 15h of growth in shaking incubator, the cultures were used to ate the appropriate culture. 3O Overexpression and purification of fusion proteins - general procedure A LB medium with kanamycin (30 ug/ml) and 100 uM zinc sulfate was ated with ght culture. The culture was incubated at 37°C until the optical density (OD) at 600 nm reached 0.60-0.80. Then IPTG was added to the final concentration in the range of 0.25 -1mM. After incubation (3.5 - 20h) with shaking at 25°C the e was centrifuged for 25 min at 6,000 g. Bacterial pellets were resuspended in a buffer containing 50 mM KH2P04, 0.5 M NaCl, 10 mM imidazole, pH 7.4. The suspension was sonicated on ice for 8 minutes (40% 3939 amplitude, 15-second pulse, 10 s interval). The resulting extract was clarified by fugation for 40 s at 20000 g, 4°C. Ni-Sepharose (GE Healthcare) resin was pre-treated by equilibration with buffer, which was used for preparation of the bacterial cells extract. The resin was then incubated overnight at 4°C with the supernatant obtained after centrifugation of the extract. Then it was loaded into chromatography column and washed with 15 to 50 volumes of buffer 50 mM KHZPO4, 0.5 M NaCl, 20 mM imidazole, pH 7.4. The obtained protein was eluted from the column using imidazole gradient in 50 mM KHZPO4 buffer with 0.5 M NaCl, pH 7.4. Obtained fractions were analyzed by SDS- PAGE. Appropriate fractions were combined and dialyzed overnight at 4°C against 50 mM Tris , pH 7.2, 150 mM NaCl, 500 mM L-arginine, 0.1 mM ZnSO4, 0.01% Tween 20, and at the same time Histag, if present, was cleaved with thrombin (1250). After the cleavage, thrombin was separated from the target fusion protein expressed with His tag by purification using Benzamidine SepharoseTM resin. Purification of target fusion proteins expressed t Histag was performed on SP Sepharose. The purity of the t was analyzed by SDS-PAGE electrophoresis (Maniatis et al, Molecular Cloning. Cold Spring Harbor, NY, 1982). pression and purification of fusion proteins - general procedure B LB medium with kanamycin (30 ug/ml) and 100 uM zinc sulfate was inoculated with overnight culture. Cultures were incubated at 37°C until optical density (OD) at 600 nm reached 0.60-0.80. Then IPTG was added to the final concentration in the range 0.5 -1mM. After 20h tion with shaking at 25°C the e was centrifuged for 25 min at 6000 g. Bacterial cells after overexpression were disrupted in a French Press in a buffer containing 50 mM KHZPO4, 0.5 M NaCl, 10 mM imidazole, 5mM beta-mercaptoethanol, 0.5mM PMSF (phenylmethylsulphonyl fluoride), pH 7.8. Resulting extract was clarified by centrifugation for 50 minutes at 8000 g. The Ni-Sepharose resin was incubated overnight with the obtained supernatant. Then the resin with bound protein was packed into the chromatography column. To wash-out the fractions containing non-binding proteins, the column was washed with 15 to 50 volumes of buffer 50 mM KHZPO4, 0.5 M NaCl, 10 mM imidazole, 5mM beta-mercaptoethanol, 0.5mM PMSF (phenylmethylsulphonyl fluoride), pH 7.8. Then, to ut the majority of proteins binding specifically with the bed, the column was washed with a buffer ning 50 mM KHZPO4, 0.5 M NaCl, 500 mM imidazole, 10% glycerol, 0.5 mM PMSF, pH 7.5. Obtained ons were analyzed by SDS-PAGE (Maniatis et al, Molecular Cloning. Cold Spring , NY, 1982). The ons containing 4040 the target protein were combined and, if the protein was expressed with histidine tag, cleaved with thrombin (1U per 4 mg of protein, 8h at 16°C) to remove polyhistidine tag. Then the fractions were ed against formulation buffer (500 mM L-arginine, 50 mM Tris, 2.5 mM ZnSO4, pH 7.4).
In this description Examples of proteins ally expressed with histidine tag that was uently removed are designated with superscript a) next to the Example number. Proteins that were originally expressed without histidine tag are designated with superscript b) next to the Example number.
Characterization of fusion proteins by 2-D electrophoresis In order to r characterize obtained proteins and to select precisely chro- matographic conditions, isoelectric points of the proteins were determined. For this purpose, two-dimensional electrophoresis (Z-D) method was used, in two stages according to the ing schedule.
Step 1. |soelectrofocusing of proteins in a pH gradient and denaturing conditions.
Protein preparations at concentrations of 1 - 2 mg/ ml were precipitated by mixing in a 1:1 ratio with a precipitation solution containing 10% trichloroacetic acid and 0.07% beta-mercaptoethanol in acetone. The mixture was incubated for min at -20°C and then fuged for 25 min at 15,000 g and 4°C. The supernatant was removed and the pellet was washed twice with cold acetone with 0.07% beta-mercaptoethanol. Then the residues of acetone were evaporated until no detectable odour. The n pellet was suspended in 250 ml of rehydration buffer 8M urea, 1% CHAPS, 15 mM DTI', 0.5% yte (GE Healthcare) with a e of pH 3-11 or 6-11, depending on the strip subsequently used. The protein on was placed in a ceramic chamber for isoelectrofocusing, followed by 13 cm DryStrip (GE Healthcare) with appropriate pH profile (3-11 or 6-11). The whole was d with a layer of mineral oil. The chambers were placed in the Ettan IPGphor ||| apparatus, where isoelectrofocusing was conducted according to the following program assigned to the dimensions of the strip and the pH profile: 3O 16h dehydration at 20° C.
Focusing in the electric field at a fixed pH gradient 4141 2h 30min gradient 1000 — 8000 V Then, the strip ning the focused proteins was washed for 1 min in deionised water, stained with sie Brilliant and then decolorized and archived as an image to mark the location of proteins. Discoloured strip was equilibrated 2 x 15 min with a buffer of the following composition: 50mM Tris- HCl pH 8.8, 6M urea, 1% D'I'I', 2% SDS, 30% glycerol.
Step 2. Separation in a second direction by SDS-PAGE.
The strip was placed over the 12.5% polyacrylamide gel containing a single well per standard size and then separation was performed in an apparatus for SDS- PAGE, at a voltage of 200V for 3 hours. The gel was stained with sie ant then archived with the applied scale. Proteins were identified by determining its weight on the basis of the standard of size, and its IPI was read for the scale of 6-11 on the basis of the curves provided by the manufacturer (GE Healthcare) (ratio of pH to % of length of the strip from the end marked as anode) or a scale of 3-11 on the basis of the curve determined experimentally by means of isoelectrofocusing calibration kit (GE Healthcare).
Examples The representative examples of the fusion proteins of the ion are shown in the following es.
In the examples the amino acids sequences of fusion proteins are written from N-terminus to C-terminus of the protein. In the Examples, by TRAIL is always meant hTRAIL.
The following ations of amino acids sequences components are used, wherein next to the three-letter designation, the equivalent single-letter designation is given.
LINKER1: steric linker Gly Gly /GG LINKERZ: steric linker Gly Gly Gly / GGG 4242 L|NKER3: steric linker Gly Ser Gly /GSG L|NKER4: steric linker Gly Gly Gly Gly Ser/GGGGS L|NKER5: steric linker Gly Gly Gly Gly Gly Ser/GGGGGS L|NKER6: steric linker Gly Gly Ser Gly Gly/GGSGG L|NKER7: steric linker Gly Gly Gly Ser Gly Gly Gly/ GGGSGGG L|NKER8: steric linker Gly Gly Gly Gly Ser Gly /GGGGSG L|NKER9: steric linker Gly Gly Gly Ser Gly Gly Gly Gly Gly Ser/ GGGSGGGGGS : steric linker Gly Gly Gly Gly Ser Gly Gly Gly Gly / GGGGSGGGG L|NKER11: steric linker Gly Ser Gly Gly Gly Ser Gly Gly Gly/ GSGGGSGGG 12: steric linker Cys Ala Ala Cys Ala Ala Ala Cys/ CAACAAAC L|NKER13: steric linker Cys Ala Ala Ala Cys Ala Ala Cys/ AC L|NKER 14: steric linker Cys/C L|NKER 15: steric linker Gly/G L|NKER16: steric linker Ser Gly Gly/SGG FURIN: sequence cleaved by furin Arg Lys Lys Arg / RKKR FURIN.NAT: native sequence cleaved by furin His Arg Gln Pro Arg Gly Trp Glu Gln / HRQPRGWEQ UROKIN: sequence cleaved by urokinase Arg Val Val Arg / RWR MMP: sequence cleaved by oprotease Pro Leu Gly Leu Ala Gly/ PLGLAG PEG1: pegylation linker Ala Ser Gly Cys Gly Pro Glu/ASGCGPE PEGZ: pegylation linker Ala Ser Gly Cys Gly Pro Glu Gly/ ASGCGPEG TRANS1: transporting sequence His His His His His His H TRANSZ: transporting sequence Arg Arg Arg Arg Arg Arg Arg/ RRRRRRR TRANS3: transporting sequence Arg Arg Arg Arg Arg Arg Arg Arg/ RRRRRRRR TRANS4: transporting sequence Tyr Ala Arg Ala Ala Ala Arg Gln Ala Arg Ala /YARAAARQARA TRANSS: transporting sequence Thr His Arg Pro Pro Met Trp Ser Pro Val Trp Pro MWSPVWP TRANS6: transporting sequence Tyr Gly Arg Lys Lys Arg Arg Gln Arg Arg Arg /YGRKKRRQRRR 4343 Example 1. Fusion protein of SEQ. No. 1 The protein of SEQ. No. 1 is a fusion protein having the length of 258 amino acids and the mass of 29.5 kDa, n domain (a) is the sequence of TRAIL121- 281, and domain (b) of the effector peptide is the 83-amino acids active form of human granulysin (SEQ. No. 34) attached at the C-terminus of domain (a).
Additionally, between domain (a) and domain (b) there are sequentially incorporated steric linker G, steric linker (GSG), metalloprotease MMP cleavage site (PLGLAG) and urokinase cleavage site (RWR). Thus, the structure of the fusion protein of the invention is as follows: (TRAIL121-281)-L|NKER15-LlNKER3-MMP-UROKIN-(SEQ. No. 34) The amino acid sequence and the DNA encoding sequence sing codons optimized for expression in E. coli are, respectively SEQ. No. 1 and SEQ. No. 57, as shown in the ed Sequence Listing.
The amino acid sequence SEQ. No. 1 of the structure described above was used as a template to generate its coding DNA ce SEQ. No. 57. A plasmid ning the coding sequence of DNA was generated and overexpression of the fusion protein was carried out in accordance with the general procedures described above. Overexpression was performed according to the general procedure A, using E. coli Tuner (DE3) strain from Novagen. The protein was separated by electrophoresis in ance with the general procedure described above. The protein was expressed with histidine tag.
Example 2. Fusion protein of SEQ. No. 2 The protein of SEQ. No. 2 is a fusion protein having the length of 261 amino acids and the mass of 30.09 kDa, wherein domain (a) is the sequence of TRAIL119-281, and domain (b) of the effector peptide is the no acids active form of human granulysin (SEQ. No. 34) attached at the N-terminus of domain (a).
Additionally, between domain (b) and domain (a) there are sequentially incorporated urokinase cleavage site (RWR), metalloprotease cleavage site (PLGLAG) and steric linker (GGGGS). Thus, the ure of the fusion protein of the invention is as follows: (SEQ. N0. 34)-UROKlN-MMP-LINKER4-(TRAIL119-281) The amino acid sequence and the DNA encoding ce comprising codons optimized for expression in E. coli are, respectively SEQ. No. 2 and SEQ. No. 58, as shown in the attached Sequence Listing. 4444 The amino acid sequence SEQ. No. 2 of the structure described above was used as a template to generate its coding DNA sequence SEQ. No. 58. A plasmid containing the coding ce of DNA was generated and pression of the fusion protein was carried out in accordance with the general procedures described above. Overexpression was performed according to the general procedure A, using E. coli Tuner (0E3) strain from Novagen. The protein was separated by electrophoresis in ance with the general procedure described above. The protein was expressed both with histidine tag (Ex. 2a) and without histidine tag (Ex. 2").
Example 3. Fusion protein of SEQ. No. 3 The protein of SEQ. No. 3 is a fusion n having the length of 186 amino acids and the mass of 21.5 kDa, n domain (a) is the sequence of TRAIL121- 281, and domain (b) of the effector peptide is the synthetic 15-amino acids lytic peptide (SEQ. No. 35) ed at the C-terminus of domain (a). onally, n domain (a) and domain (b) there are sequentially incorporated metalloprotease cleavage site (PLGLAG) and urokinase ge site (RWR). Thus, the structure of the fusion protein of the invention is as follows: (TRAIL121-281)-MMP-UROK|N-(SEQ. No. 35) The amino acid sequence and the DNA encoding sequence comprising codons optimized for expression in E. coli are, respectively SEQ. No. 3 and SEQ. No. 59, as shown in the attached Sequence Listing.
The amino acid sequence SEQ. No. 3 of the structure described above was used as a template to generate its coding DNA sequence SEQ. No. 59. A plasmid containing the coding sequence of DNA was generated and overexpression of the fusion protein was carried out in accordance with the general procedures described above. Overexpression was performed according to the general procedure A, using E. coli Tuner (0E3) strain from Novagen. The protein was separated by electrophoresis in accordance with the general procedure 3O described above. The protein was sed with histidine tag.
Example 4. Fusion protein of SEQ. No. 4 The protein of SEQ. No. 4 is a fusion protein having the length of 227 amino acids and the mass of 25.7 kDa, wherein domain (a) is the sequence of TRAIL121- 281, and domain (b) of the or peptide is the 56-amino acids pilosulin-1 (SEQ. No. 36) attached at the N-terminus of domain (a). 4545 Additionally, between domain (b) and domain (a) there are sequentially incorpo- rated urokinase cleavage site (RWR) and metalloprotease cleavage site (PLGLAG). Thus, the structure of the fusion protein of the ion is as follows: (SEQ. No. 36)-UROKIN-MMP-(TRAIL 121-281) The amino acid sequence and the DNA encoding sequence comprising codons optimized for expression in E. coli are, tively SEQ. No. 4 and SEQ. No. 60, as shown in the attached ce Listing.
The amino acid sequence SEQ. No. 4 of the structure described above was used as a template to te its coding DNA ce SEQ. No. 60. A plasmid containing the coding sequence of DNA was generated and overexpression of the fusion protein was carried out in accordance with the general procedures described above. Overexpression was performed according to the general procedure A, using E. coli Tuner (DE3) strain from n. The protein was separated by electrophoresis in accordance with the general procedure described above. The protein was expressed with histidine tag.
Example 5. Fusion protein of SEQ. No. 5 The protein of SEQ. No. 5 is a fusion protein having the length of 264 amino acids and the mass of 29.5kDa, wherein domain (a) is the ce of TRAIL95- 281, and domain (b) of the effector peptide is the 56-amino acids pilosulin-1 (SEQ. No. 36), and is attached at the C-terminus of domain (a).
Additionally, between domain (a) and domain (b) there are sequentially incorpo- rated steric linker (CAACAAC), steric linker (GGG), metalloprotease cleavage site (PLGLAG) and ase cleavage site (RWR). Thus, the structure of the fusion protein of the invention is as follows: (TRAIL95-281)-L|NKER12-LlNKER2-MMP-UROKIN-(SEQ. No. 36) The amino acid sequence and the DNA ng sequence comprising codons optimized for expression in E. coli are, respectively SEQ. No. 5 and SEQ.
No. 61 as shown in the attached Sequence Listing.
The amino acid sequence SEQ. No. 5 of the structure described above was used as a template to generate its coding DNA ce SEQ. No. 61. A plasmid containing the coding ce of DNA was generated and overexpression of the fusion protein was carried out in accordance with the general ures described above. Overexpression was performed according to the general procedure A, using E. coli Tuner (DE3) strain from Novagen. The protein was 4646 separated by electrophoresis in accordance with the general procedure described above. The protein was expressed with histidine tag.
Example 6. Fusion protein of SEQ. No. 6 The protein of SEQ. No. 6 is a fusion protein having the length of 299 amino acids and the mass of 33.2 kDa, wherein domain (a) is the sequence of TRAIL95- 281, and domain (b) of the effector e is the 90-amino acids peptide pilosulin 5 (SEQ. No. 37) attached at the inus of domain (a).
Additionally, between domain (a) and domain (b) there are sequentially incorpo- rated steric linker (GSG), steric linker (CAACAAAC), metalloprotease cleavage site (PLGLAG), urokinase cleavage site (RWR) and steric linker (G). Thus, the ure of the fusion protein of the invention is as follows: (TRAIL95-281)-L|NKER3-LINKER12-MMP-UROKIN-LINKER15-(SEQ. No. 37) The amino acid ce and the DNA encoding sequence comprising codons optimized for expression in E. coli are, respectively SEQ. No. 6 and SEQ. No. 62 as shown in the attached Sequence Listing.
The amino acid sequence SEQ. No. 6 of the structure described above was used as a template to generate its coding DNA sequence SEQ. No. 62. A plasmid containing the coding sequence of DNA was generated and pression of the fusion protein was carried out in accordance with the general procedures described above. Overexpression was performed according to the general ure A, using E. coli Tuner (DE3) strain from Novagen. The protein was separated by electrophoresis in accordance with the general ure described above. The protein was expressed both with histidine tag (Ex. 6a) and without ine tag (Ex. 6").
Example 7. Fusion protein of SEQ. No. 7 The protein of SEQ. No. 7 is a fusion protein having the length of 224 amino acids and the mass of 25.6 kDa, wherein domain (a) is the sequence of TRAIL95- 281, and domain (b) of the effector e is the 17-amino acids active peptide from lesin (SEQ. No. 38) attached at the C-terminus of domain (a).
Additionally, between domain (a) and domain (b) there are sequentially incorpo- rated steric linker (CAACAAAC), steric linker (GG), metalloprotease cleavage site (PLGLAG) and urokinase cleavage site (RWR). Thus, the structure of the fusion protein of the invention is as follows (TRAIL95-281)-L|NKER12-LINKER1-MMP-UROK|N-(SEQ. N0. 38) 4747 The amino acid sequence and the DNA encoding sequence comprising codons optimized for expression in E. coli are, respectively SEQ. No. 7 and SEQ. No. 63 as shown in the attached Sequence Listing.
The amino acid sequence SEQ. No. 7 of the structure described above was used as a template to generate its coding DNA ce SEQ. No. 63. A plasmid containing the coding sequence of DNA was ted and overexpression of the fusion protein was carried out in accordance with the general procedures bed above. Overexpression was performed according to the general procedure A, using E. coli Tuner (0E3) strain from Novagen. The protein was separated by electrophoresis in accordance with the l procedure described above. The protein was expressed both with histidine tag (Ex. 7a) and without histidine tag (Ex. 7b).
Example 8. Fusion protein of SEQ. No. 8 The n of SEQ. No. 8 is a fusion protein having the length of 202 amino acids and the mass of 23.8 kDa, wherein domain (a) is the sequence of TRAIL 114-281, and domain (b) of the or peptide is the 17-amino acids active peptide from tachyplesin (SEQ. No. 38) attached at the inus of domain (a).
Additionally, between domain (a) and domain (b) there are sequentially incorpo- rated metalloprotease cleavage site G), urokinase cleavage site (RWR) and 7-arginine transporting sequence (RRRRRRR). Thus, the structure of the fusion protein of the invention is as follows: (TRAIL114-281)-MMP-UROKlN-TRANSZ-(SEQ. No. 38) The amino acid sequence and the DNA encoding sequence comprising codons optimized for expression in E. coli are, respectively SEQ. No. 8 and SEQ. No. 64 as shown in the attached Sequence Listing.
The amino acid sequence SEQ. No. 8 of the structure described above was used as a template to generate its coding DNA sequence SEQ. No. 64. A plasmid containing the coding sequence of DNA was generated and overexpression of the fusion protein was carried out in accordance with the general procedures described above. Overexpression was performed ing to the l procedure A, using E. coli Tuner (0E3) strain from Novagen. The protein was separated by electrophoresis in accordance with the general procedure described above. The protein was sed both with histidine tag (Ex. 83) and t histidine tag (Ex. 8b). 4848 Example 9. Fusion protein of SEQ. No. 9 The protein of SEQ. No. 9 is a fusion protein having the length of 243 amino acids and the mass of 27.6 kDa, wherein domain (a) is the sequence of TRAIL 95- 281, and domain (b) of the effector e is a 38-amino acids fusion peptide bombesin-magainin (SEQ. No. 39) attached at the N-terminus of domain (a).
Additionally, between domain (b) and domain (a) there are sequentially incorporated urokinase cleavage site (RWR), metalloprotease cleavage site G) and steric linker (CAAACAAC). Thus, the structure of the fusion protein of the invention is as follows: (SEQ. No. 39)-UROKlN-MMP-LINKER13-(TRAIL95-281) The amino acid sequence and the DNA encoding ce comprising codons optimized for expression in E. coli are, respectively SEQ. No. 9 and SEQ. No. 65 as shown in the ed Sequence Listing.
The amino acid sequence SEQ. No. 9 of the structure described above was used as a template to generate its coding DNA sequence SEQ. No. 65. A plasmid containing the coding sequence of DNA was generated and overexpression of the fusion protein was carried out in accordance with the general procedures described above. Overexpression was performed ing to the general procedure A, using E. coli Tuner (DE3) strain from Novagen. The protein was separated by electrophoresis in accordance with the general procedure described above. The protein was expressed with histidine tag.
Example 10. Fusion protein of SEQ. No. 10 The protein of SEQ. No. 10 is a fusion protein having the length of 196 amino acids and the mass of 22.4 kDa, wherein domain (a) is the sequence of TRAIL 119-281, and domain (b) of the or peptide is the 23-amino acids magainin- 2 (SEQ. No. 40) attached at the N-terminus of domain (a).
Additionally, between domain (b) and domain (a) there are tially incorporated urokinase cleavage site (RWR) and metalloprotease cleavage site (PLGLAG). Thus, the ure of the fusion protein of the invention is as follows: (SEQ. N0. 40)-UROK|N-MMP-(TRAIL119-281) The amino acid ce and the DNA encoding sequence sing codons optimized for expression in E. coli are, respectively SEQ. No. 10 and SEQ. No. 66 as shown in the attached Sequence Listing. 4949 The amino acid sequence SEQ. No. 10 of the structure described above was used as a template to generate its coding DNA sequence SEQ. No. 66. A plasmid containing the coding ce of DNA was generated and overexpression of the fusion protein was carried out in accordance with the l procedures described above. Overexpression was performed according to the general procedure A, using E. coli Tuner (0E3) strain from Novagen. The protein was separated by electrophoresis in ance with the general ure described above. The protein was expressed with histidine tag.
Example 11. Fusion protein of SEQ. No. 11 The protein of SEQ. No. 11 is a fusion protein having the length of 202 amino acids and the mass of 23 kDa, wherein domain (a) is the sequence of TRAIL 121- 281, and domain (b) of the effector e is the synthetic 14-amino acids lytic peptide (SEQ. No. 41) attached at the C-terminus of domain (a).
Additionally, between domain (a) and domain (b) there are sequentially incorpo- rated steric linker (GGGGSGGGG), urokinase cleavage site (RWR), metalloprote- ase cleavage site (PLGLAG) and 8-arginine transporting sequence (RRRRRRRR).
Thus, the structure of the fusion protein of the invention is as s: (TRAIL121-281)-L|NKER10-UROKIN-MMP-TRANS3-(SEQ. No. 41) The amino acid sequence and the DNA encoding sequence comprising codons optimized for expression in E. coli are, respectively SEQ. No. 11 and SEQ. No. 67 as shown in the attached Sequence Listing.
The amino acid sequence SEQ. No. 11 of the structure described above was used as a template to generate its coding DNA sequence SEQ. No. 67. A plasmid containing the coding sequence of DNA was ted and overexpression of the fusion protein was carried out in accordance with the general procedures described above. Overexpression was performed according to the general ure A, using E. coli Tuner (0E3) strain from Novagen. The protein was separated by electrophoresis in accordance with the general procedure described above. The n was expressed both with histidine tag (Ex. 11a) and 3O without histidine tag (Ex. 11b).
Example 12. Fusion protein of SEQ. No. 12 The protein of SEQ. No. 12 is a fusion protein having the length of 205 amino acids and the mass of 23,2 kDa, n domain (a) is the sequence of TRAIL 121-281, and domain (b) of the effector peptide is the synthetic 14-amino acids lytic peptide (SEQ. No. 41) attached at the C-terminus of domain (a). 5050 Additionally, between domain (a) and domain (b) there are sequentially o- rated steric linker (66), sequence of pegylation linker (ASGCGPEG), steric linker ce (GGG), metalloprotease cleavage site (PLGLAG), urokinase cleavage site (RWR) and polyarginine transporting sequence (RRRRRRR). Thus, the structure of the fusion protein of the invention is as follows: (TRAIL 121-281)-L|NKER1-PEGZ-LlNKER2-MMP-UROK|N-TRANSZ-(SEQ. No. 41) The amino acid sequence and the DNA encoding sequence comprising codons optimized for expression in E. coli are, respectively SEQ. No. 12 and SEQ. No. 68 as shown in the attached Sequence Listing.
The amino acid sequence SEQ. No. 12 of the ure described above was used as a template to generate its coding DNA sequence SEQ. No. 68. A plasmid ning the coding sequence of DNA was generated and overexpression of the fusion protein was carried out in accordance with the general procedures described above. Overexpression was performed according to the l procedure A, using E. coli Tuner (DE3) strain from Novagen. The protein was separated by electrophoresis in ance with the l procedure described above. The protein was expressed with histidine tag.
Example 13. Fusion protein of SEQ. No. 13 The protein of SEQ. No. 13 is a fusion protein having the length of 228 amino acids and the mass of 25.9 kDa, wherein domain (a) is the sequence of TRAIL 95- 281, and domain (b) of the effector peptide is the 14-amino acids lytic peptide (SEQ. No. 41) attached at the C-terminus of domain (a).
Additionally, between domain (a) and domain (b) there are sequentially incorpo- rated steric linker (GGGGSGGGG), urokinase cleavage site (RWR), metallopro- tease cleavage site (PLGLAG) and 8-arginine transporting sequence (RRRRRRRR).
Thus, the structure of the fusion protein of the invention is as follows: (TRAIL 95-281)-L|NKER10-UROKIN-MMP-TRANS3-(SEQ. No. 41) The amino acid sequence and the DNA encoding sequence comprising codons optimized for expression in E. coli are, respectively SEQ. No. 13 and SEQ. No. 69 as shown in the ed Sequence Listing.
The amino acid sequence SEQ. No. 13 of the structure described above was used as a template to generate its coding DNA sequence SEQ. No. 69. A plasmid containing the coding sequence of DNA was generated and overexpression of the fusion n was d out in accordance with the general procedures described above. Overexpression was performed according to the general 5151 procedure A, using E. coli Tuner (DE3) strain from Novagen. The protein was separated by electrophoresis in accordance with the general procedure described above. The protein was expressed both with histidine tag (Ex. 13a) and t histidine tag (Ex. 13b).
Example 14. Fusion protein of SEQ. No. 14 The protein of SEQ. No. 14 is a fusion protein having the length of 192 amino acids and the mass of 22.1 kDa, wherein domain (a) is the sequence of TRAIL 121-281, and domain (b) of the effector peptide is the synthetic 14-amino acids lytic peptide (SEQ. No. 41) attached at the N-terminus of domain (a).
Additionally, n domain (b) and domain (a) there are sequentially incorpo- rated steric linker (C), urokinase cleavage site (RVVR) and metalloprotease cleavage site (PLGLAG). Additionally at the N-terminus of effector peptide is attached polyhistidine transporting domain (HHHHHH). Thus, the structure of the fusion protein of the invention is as s: TRANS1-(SEQ. N0. 41)-L|NKER14-UROK|N-MMP-(TRA|L121-281) The amino acid sequence and the DNA encoding sequence sing codons optimized for expression in E. coli are, tively SEQ. No. 14 and SEQ. No. 70 as shown in the attached Sequence Listing.
The amino acid ce SEQ. No. 14 of the structure described above was used as a template to generate its coding DNA sequence SEQ. No. 70. A d containing the coding sequence of DNA was generated and overexpression of the fusion protein was carried out in accordance with the general ures described above. Overexpression was performed according to the general procedure A, using E. coli Tuner (DE3) strain from Novagen. The protein was separated by electrophoresis in accordance with the general procedure bed above. The protein was sed with histidine tag.
Example 15. Fusion protein of SEQ. No. 15 The protein of SEQ. No. 15 is a fusion protein having the length of 200 amino acids and the mass of 23.3 kDa, wherein domain (a) is the sequence of TRAIL 121-281, and domain (b) of the effector peptide is the 14-amino acids lytic peptide (SEQ. No. 41) is attached at the N-terminus of domain (a).
Additionally, between domain (b) and domain (a) there are sequentially incorpo- rated steric linker (C), 8-arginine transporting sequence (RRRRRRRR), urokinase cleavage site (RVVR) and oprotease cleavage site (PLGLAG). Additionally, ine transporting sequence (HHHHHH) is attached at the N-terminus of the 5252 effector peptide. Thus, the structure of the fusion protein of the invention is as follows: TRANS1-(SEQ. No. 41)-L|NKER14-TRANS3-UROKIN-MMP-(TRAIL 121-281) The amino acid sequence and the DNA encoding sequence comprising codons optimized for expression in E. coli are, respectively SEQ. No. 15 and SEQ. No. 71 as shown in the attached Sequence Listing.
The amino acid sequence SEQ. No. 15 of the structure described above was used as a template to generate its coding DNA sequence SEQ. No. 71. A d ning the coding sequence of DNA was generated and overexpression of the fusion protein was carried out in accordance with the general procedures described above. Overexpression was med according to the general procedure A, using E. coli Tuner (DE3) strain from n. The n was separated by electrophoresis in accordance with the general procedure described above. The n was expressed with histidine tag.
Example 16. Fusion protein of SEQ. No. 16 The protein of SEQ. No. 16 is a fusion protein having the length of 202 amino acids and the mass of 23,1 kDa, wherein domain (a) is the sequence of TRAIL 121-281, and domain (b) of the effector peptide is the synthetic 14-amino acids lytic peptide (SEQ. No. 41) attached at the C-terminus of domain (a).
Additionally, between domain (a) and domain (b) there are sequentially incorpo- rated steric linker (GGGGSGGGG), metalloprotease cleavage site (PLGLAG), uro- kinase cleavage site (RVVR) and 8-arginine transporting sequence (RRRRRRRR).
Thus, the structure of the fusion protein of the invention is as follows: (TRAIL 1)-L|NKER10-MMP-UROKIN TRANS3-(SEQ. No. 41) The amino acid sequence and the DNA encoding sequence sing codons optimized for expression in E. coli are, respectively SEQ. No. 16 and SEQ. No. 72 as shown in the attached Sequence Listing.
The amino acid sequence SEQ. No. 16 of the structure bed above was used as a template to generate its coding DNA sequence SEQ. No. 72. A d containing the coding sequence of DNA was ted and overexpression of the fusion protein was carried out in accordance with the general procedures described above. Overexpression was performed according to the l procedure A, using E. coli Tuner (DE3) strain from Novagen. The protein was separated by electrophoresis in accordance with the general procedure 5353 bed above. The protein was expressed both with histidine tag (Ex. 16a) and without histidine tag (Ex. 16").
Example 17. Fusion protein of SEQ. No. 17 The protein of SEQ. No. 17 is a fusion protein having the length of 208 amino acids and the mass of 23.5kDa, wherein domain (a) is the sequence of TRAIL 116- 281, and domain (b) of the effector e is the 26-amino acids hybride peptide cecropin A-melittin (SEQ. No. 42) attached at the N-terminus of domain (a).
Additionally, between domain (b) and domain (a) there are sequentially incorpo- rated urokinase cleavage site (RWR) and metalloprotease cleavage site (PLGLAG). Thus, the structure of the fusion protein of the invention is as follows: (SEQ. N0. 42)-UROKlN-MMP-(TRAIL116-281) The amino acid sequence and the DNA encoding sequence comprising codons optimized for sion in E. coli are, respectively SEQ. No. 17 and SEQ. No. 73 as shown in the attached Sequence Listing.
The amino acid sequence SEQ. No. 17 of the structure described above was used as a template to generate its coding DNA sequence SEQ. No. 73. A plasmid ning the coding sequence of DNA was generated and overexpression of the fusion protein was carried out in accordance with the general procedures described above. pression was performed according to the general procedure A, using E. coli Tuner (DE3) strain from n. The protein was separated by electrophoresis in accordance with the l procedure described above. The protein was expressed with ine tag.
Example 18. Fusion protein of SEQ. No. 18 The protein of SEQ. No. 18 is a fusion protein having the length of 203 amino acids and the mass of 23.6 kDa, wherein domain (a) is the sequence of TRAIL 116-281, and domain (b) of the effector peptide is the 27-amino acids e hCAP-18/LL-37 (SEQ. No. 43) attached at the N-terminus of domain (a).
Additionally between domain (b) and domain (a) there are sequentially incorpo- rated urokinase cleavage site (RWR) and oprotease cleavage site (PLGLAG). Thus, the structure of the fusion protein of the invention is as follows: (SEQ. N0. 43)-UROKlN-MMP-(TRAIL116-281) 5454 The amino acid sequence and the DNA encoding sequence comprising codons optimized for expression in E. coli are, respectively SEQ. No. 18 and SEQ. No. 74 as shown in the attached ce Listing.
The amino acid sequence SEQ. No. 18 of the structure described above was used as a te to te its coding DNA ce SEQ. No. 74. A plasmid containing the coding sequence of DNA was generated and overexpression of the fusion protein was d out in accordance with the general procedures described above. pression was performed according to the general procedure A, using E. coli Tuner (0E3) strain from Novagen. The protein was separated by electrophoresis in accordance with the general procedure described above. The protein was expressed with histidine tag.
Example 19. Fusion protein of SEQ. No. 19 The n of SEQ. No. 19 is a fusion protein having the length of 203 amino acids and the mass of 23,3 kDa, wherein domain (a) is the sequence of TRAIL 1, and domain (b) of the effector peptide is the 27-amino acids peptide BAMP-28 (SEQ. No. 44) attached at the N-terminus of domain (a).
Additionally, between domain (b) and domain (a) there are sequentially incorpo- rated urokinase cleavage site (RWR) and metalloprotease cleavage site (PLGLAG). Thus, the structure of the fusion protein of the invention is as follows: (SEQ. No. 44)-UROKIN-MMP-(TRAIL116-281) The amino acid ce and the DNA encoding sequence comprising codons optimized for expression in E. coli are, respectively SEQ. No. 19 and SEQ. No. 75 as shown in the attached Sequence Listing.
The amino acid sequence SEQ. No. 19 of the structure described above was used as a template to generate its coding DNA sequence SEQ. No. 75. A d containing the coding sequence of DNA was generated and overexpression of the fusion protein was carried out in accordance with the general procedures described above. Overexpression was performed according to the general procedure A, using E. coli Tuner (0E3) strain from Novagen. The protein was separated by electrophoresis in accordance with the general procedure described above. The protein was sed with histidine tag.
Example 20. Fusion protein of SEQ. No. 20 The n of SEQ. No. 20 is a fusion protein having the length of 200 amino acids and the mass of 22.8 kDa, wherein domain (a) is the sequence of TRAIL 5555 121-281, and domain (b) of the effector peptide is the 24-amino acids analogue of isoform C of the lytic peptide from Entamoeba histolytica (SEQ. No. 45) attached at the C-terminus of domain (a).
Additionally, between domain (a) and domain (b) there are sequentially incorpo- rated steric linker (GGSGG), metalloprotease ge site (PLGLAG) and uroki- nase cleavage site (RVVR). Thus, the structure of the fusion protein of the invention is as follows: (TRAIL121-281)-L|NKER6-MMP-UROKlN-(SEQ. No. 45) The amino acid sequence and the DNA encoding sequence comprising codons optimized for expression in E. coli are, respectively SEQ. No. 20 and SEQ. No. 76 as shown in the attached Sequence Listing.
The amino acid sequence SEQ. No. 20 of the structure described above was used as a template to te its coding DNA sequence SEQ. No. 76. A plasmid containing the coding sequence of DNA was generated and overexpression of the fusion protein was carried out in accordance with the general procedures described above. Overexpression was performed according to the general procedure A, using E. coli Tuner (DE3) strain from Novagen. The protein was separated by electrophoresis in accordance with the general procedure described above. The protein was sed with ine tag.
Example 21. Fusion protein of SEQ. No. 20 The n of SEQ. No. 20 is a fusion protein having the length of 200 amino acids and the mass of 22.8 kDa, wherein domain (a) is the sequence of TRAIL 121-281, and domain (b) of the effector peptide is the 24-amino acids analogue of isoform A of the lytic peptide from eba ytica (SEQ. No. 46) attached at the inus of domain (a).
Additionally, between domain (a) and domain (b) there are sequentially incorpo- rated steric linker (GGSGG), metalloprotease cleavage site G) and uroki- nase cleavage site (RVVR). Thus, the structure of the fusion protein of the invention is as follows: 121-281)-L|NKER6-MMP-UROKIN-(SEQ. No. 46) The amino acid sequence and the DNA encoding sequence comprising codons optimized for expression in E. coli are, respectively SEQ. No. 21 and SEQ. No. 77 as shown in the ed Sequence Listing.
The amino acid sequence SEQ. No. 21 of the structure described above was used as a template to generate its coding DNA sequence SEQ. No. 77. A plasmid 5656 containing the coding sequence of DNA was ted and overexpression of the fusion protein was carried out in accordance with the general procedures described above. Overexpression was performed ing to the general ure A, using E. coli Tuner (DE3) strain from Novagen. The protein was separated by electrophoresis in ance with the general procedure described above. The protein was sed with histidine tag.
Example 22. Fusion protein of SEQ. No. 22 The protein of SEQ. No. 22 is a fusion protein having the length of 200 amino acids and the mass of 22.8 kDa, wherein domain (a) is the sequence of TRAIL 121-281, and domain (b) of the or peptide is the 24-amino acids analogue of isoform B of a lytic peptide from Entamoeba histolytica (SEQ. No. 47) attached at the inus of domain (a).
Additionally, between domain (a) and domain (b) there are sequentially o- rated steric linker (GGSGG), metalloprotease cleavage site G) and urokinase cleavage site (RWR). Thus, the structure of the fusion protein of the invention is as follows: (TRAIL121-281)-L|NKER6-MMP-UROKlN-(SEQ. N0. 47) The amino acid sequence and the DNA encoding sequence comprising codons optimized for expression in E. coli are, respectively SEQ. No. 22 and SEQ. No. 78 as shown in the attached ce Listing.
The amino acid sequence SEQ. No. 22 of the structure described above was used as a template to generate its coding DNA sequence SEQ. No. 78. A plasmid containing the coding sequence of DNA was generated and overexpression of the fusion protein was carried out in accordance with the general procedures described above. Overexpression was performed according to the general procedure A, using E. coli Tuner (0E3) strain from Novagen. The protein was separated by electrophoresis in accordance with the general procedure described above. The protein was expressed with histidine tag.
Example 23. Fusion protein of SEQ. No. 23 The protein of SEQ. No. 23 is a fusion protein having the length of 190 amino acids and the mass of 22.1 kDa, wherein domain (a) is the sequence of TRAIL 121-281, and domain (b) of the effector peptide is the 12-amino acids fragment of HA2 domain of influenza virus haemagglutinin (SEQ. No. 48) attached at the N-terminus of domain (a). 5757 Additionally, between domain (b) and domain (a) there are sequentially incorpo- rated 7-arginine transporting sequence (RRRRRRR), urokinase cleavage site (RWR) and metalloprotease cleavage site (PLGLAG). Thus, the ure of the fusion protein of the invention is as follows: (SEQ. No. 48)-TRANSZ-UROKlN-MMP-(TRA|L121-281) The amino acid sequence and the DNA encoding ce comprising codons optimized for expression in E. coli are, respectively SEQ. No. 23 and SEQ. No. 79 as shown in the attached Sequence g.
The amino acid sequence SEQ. No. 23 of the structure bed above was used as a template to generate its coding DNA sequence SEQ. No.79. A plasmid ning the coding sequence of DNA was generated and overexpression of the fusion protein was carried out in accordance with the general procedures described above. Overexpression was performed according to the general procedure B, using E. coli BL21 (DE3) or Tuner (DE3) strain from Novagen. The protein was ted by electrophoresis in accordance with the general procedure bed above. The protein was expressed both with histidine tag (Ex. 23a) and without histidine tag (Ex. 23").
Example 24. Fusion protein of SEQ. No. 24 The protein of SEQ. No. 24 is a fusion protein having the length of 429 amino acids and the mass of 48.6 kDa, wherein domain (a) is the sequence of TRAIL 121 -281, and domain (b) of the effector peptide is the 247-amino acids nt of N-terminal domain of alpha-toxin from Clostridium perfringens with phospholipase C activity (SEQ. No. 49) attached at the N-terminus of domain (a).
Additionally, between domain (b) and domain (a) there are sequentially incorpo- rated steric linker (G), steric linker (GGGGGS), pegylation linker (ASGCGPE) and steric linker (GGGGGS). Thus, the structure of the fusion protein of the invention is as follows: (SEQ. No. 49)-LINKER15-LlNKER5-PEGZ-LINKER5-(TRAIL121-281) The amino acid sequence and the DNA encoding sequence comprising codons optimized for expression in E. coli are, tively SEQ. No. 24 and SEQ. No. 80 as shown in the attached Sequence Listing.
The amino acid sequence SEQ. No. 24 of the structure described above was used as a template to te its coding DNA sequence SEQ. No. 80. A plasmid containing the coding sequence of DNA was generated and overexpression of the fusion protein was carried out in ance with the general procedures 5858 described above. Overexpression was performed according to the general procedure A, using E. coli Tuner (DE3) strain from Novagen. The protein was separated by electrophoresis in accordance with the general procedure bed above. The protein was expressed with histidine tag.
Example 25. Fusion protein of SEQ. No. 25 The protein of SEQ. No. 25 is a fusion protein having the length of 658 amino acids and the mass of 73 kDa, wherein domain (a) is the sequence of TRAIL 121- 281, and domain (b) of the effector peptide is the 468-amino acids peptide listeriolysin O (SEQ. No. 50) attached at the N-terminus of domain (a).
Additionally, between domain (b) and domain (a) there are tially incorpo- rated steric linker (G), steric linker (GGGGSGGGGGS), furin cleavage site (RKKR), pegylation linker PEG) and steric linker sequence (GGGGGS).
Thus, the structure of the fusion protein of the invention is as follows: (SEQ. N0. 50)-LINKER15-LlNKER9-FURIN-PEGZ-LINKER5-(TRAIL121-281) The amino acid sequence and the DNA encoding sequence comprising codons zed for expression in E. coli are, respectively SEQ. No. 25 and SEQ. No. 81 as shown in the attached Sequence Listing.
The amino acid sequence SEQ. No. 25 of the structure described above was used as a template to generate its coding DNA sequence SEQ. No. 81. A d containing the coding sequence of DNA was generated and pression of the fusion protein was carried out in accordance with the general procedures described above. Overexpression was performed according to the general ure A, using E. coli Tuner (DE3) strain from Novagen. The protein was separated by ophoresis in accordance with the general procedure described above. The protein was expressed with histidine tag.
Example 26. Fusion protein of SEQ. No. 26 The protein of SEQ. No. 26 is a fusion protein having the length of 478 amino acids and the mass of 54 kDa, wherein domain (a) is the sequence of TRAIL 121- 281, and domain (b) of the effector peptide is the 289-amino acids phospholipase PC-PLC (SEQ. No. 51) attached at the N-terminus of domain (a).
Additionally, n domain (b) and domain (a) there are tially incorpo- rated steric linker GGGGS), furin cleavage site (RKKR), pegylation linker (ASGCGPEG) and steric linker (GGGGGS). Thus, the structure of the fusion protein of the invention is as follows: (SEQ. No. 51)-L|NKER9-FURlN-PEGZ-LINKER5-(TRAIL121-281) 5959 The amino acid sequence and the DNA encoding sequence comprising codons optimized for expression in E. coli are, respectively SEQ. No. 26 and SEQ. No. 82 as shown in the attached Sequence Listing.
The amino acid sequence SEQ. No. 26 of the structure described above was used as a template to generate its coding DNA ce SEQ. No. 82. A plasmid containing the coding sequence of DNA was generated and overexpression of the fusion protein was carried out in ance with the general procedures described above. Overexpression was performed according to the general procedure A, using E. coli Tuner (DE3) strain from Novagen. The protein was separated by electrophoresis in accordance with the general ure described above. The protein was expressed with histidine tag.
Example 27. Fusion protein of SEQ. No. 27 The protein of SEQ. No. 27 is a fusion protein having the length of 361 amino acids and the mass of 40.2 kDa, wherein domain (a) is the sequence of TRAIL 121-281, and domain (b) of the effector peptide is the 179-amino acids equinatoxin EqTx-II (SEQ. No. 52) ed at the N-terminus of domain (a).
Additionally, n domain (b) and domain (a) there are sequentially incorpo- rated steric linker (GGGGS), furin cleavage site (RKKR), pegylation linker PE) and steric linker (GGGGS). Thus, the structure of the fusion protein of the invention is as follows: (SEQ. No. NKER4-FURIN-PEG1-L|NKER4-(TRAIL121-281) The amino acid sequence and the DNA encoding sequence sing codons optimized for expression in E. coli are, tively SEQ. No. 27 and SEQ. No. 83 as shown in the attached Sequence Listing.
The amino acid sequence SEQ. No. 27 of the structure described above was used as a template to generate its coding DNA sequence SEQ. No. 83. A plasmid containing the coding sequence of DNA was generated and overexpression of the fusion protein was carried out in accordance with the general procedures described above. Overexpression was performed according to the general procedure B, using E. coli BL21 (DE3) or Tuner (DE3) strain from Novagen. The protein was separated by electrophoresis in accordance with the l procedure described above. The protein was expressed with histidine tag.
Example 28. Fusion n of SEQ. No. 28 The protein of SEQ. No. 28 is a fusion n having the length of 227 amino acids and the mass of 25.5 kDa, wherein domain (a) is the sequence of TRAIL 6060 116-281, and domain (b) of the effector peptide is the 46-amino acids peptide viscotoxin A3 (VtA3) (SEQ. No. 53) attached at the N-terminus of domain (a).
Additionally, between domain (b) and domain (a) there are sequentially incorpo- rated oprotease cleavage site (PLGLAG), urokinase cleavage site (RWR) and steric linker (GGSGG). Thus, the structure of the fusion protein of the invention is as follows: (SEQ. No. 53)-MMP-UROKlN-LINKER6-(TRAIL116-281) The amino acid sequence and the DNA encoding sequence sing codons optimized for expression in E. coli are, respectively SEQ. No. 28 and SEQ. No. 84 as shown in the attached Sequence Listing.
The amino acid sequence SEQ. No. 28 of the structure described above was used as a template to generate its coding DNA sequence SEQ. No. 84. A plasmid containing the coding sequence of DNA was generated and overexpression of the fusion protein was carried out in accordance with the general procedures described above. Overexpression was performed according to the general procedure A, using E. coli Tuner (DE3) strain from n. The protein was separated by electrophoresis in accordance with the l procedure described above. The protein was expressed with ine tag.
Example 29. Fusion n of SEQ. No. 29 The protein of SEQ. No. 29 is a fusion n having the length of 224 amino acids and the mass of 24.9 kDa, wherein domain (a) is the sequence of TRAIL 121-281, and domain (b) of the or peptide is the 46-amino acids peptide viscotoxin A3 (VtA3) (SEQ. No. 53) attached at the C-terminus of domain (a).
Additionally, between domain (a) and domain (b) there are sequentially incorpo- rated steric linker (GGGSGGG), metalloprotease cleavage site (PLGLAG), uroki- nase cleavage site (RVVR). Thus, the ure of the fusion protein of the invention is as follows: 121-281)-L|NKER7-MMP-UROKIN-(SEQ. No. 53) The amino acid sequence and the DNA encoding sequence comprising codons optimized for expression in E. coli are, respectively SEQ. No. 29 and SEQ. No. 85 as shown in the attached Sequence Listing.
The amino acid sequence SEQ. No. 29 of the structure described above was used as a template to generate its coding DNA sequence SEQ. No. 85. A d containing the coding sequence of DNA was ted and overexpression of the fusion protein was carried out in accordance with the general procedures 6161 described above. Overexpression was performed according to the general procedure A, using E. coli Tuner (DE3) strain from Novagen. The n was ted by electrophoresis in accordance with the general procedure described above. The n was expressed with ine tag.
Example 30. Fusion protein of SEQ. No. 30 The protein of SEQ. No. 30 is a fusion protein having the length of 200 amino acids and the mass of 22.5 kDa, wherein domain (a) is the sequence of TRAIL 1, and domain (b) of the effector peptide is the 33-amino acids active fragment of human perforin (SEQ. No. 54) attached at the C-terminus of domain (a).
Additionally, between domain (a) and domain (b) is incorporated steric linker sequence G). Thus, the structure of the fusion protein of the invention is as follows: (TRAIL121-281)-L|NKER8—(SEQ. N0. 54) The amino acid sequence and the DNA encoding sequence comprising codons optimized for expression in E. coli are, tively SEQ. No. 30 and SEQ. No. 86 as shown in the attached Sequence Listing.
The amino acid sequence SEQ. No. 30 of the structure described above was used as a template to generate its coding DNA sequence SEQ. No. 86. A plasmid containing the coding sequence of DNA was generated and overexpression of the fusion protein was carried out in accordance with the l procedures described above. Overexpression was performed according to the general procedure A, using E. coli Tuner (DE3) strain from Novagen. The protein was separated by electrophoresis in accordance with the general procedure described above. The protein was expressed with histidine tag.
Example 31. Fusion protein of SEQ. No. 31 The protein of SEQ. No. 31 is a fusion n having the length of 210 amino acids and the mass of 23.5 kDa, wherein domain (a) is the sequence of TRAIL 121-281, and domain (b) of the or peptide is the 33-amino acids active fragment of human perforin (SEQ. No. 54) attached at the C-terminus of domain (3)- Additionally, between domain (a) and domain (b) there are sequentially incorpo- rated steric linker (GGGGSG), urokinase cleavage site (RWR), AND metallo- protease cleavage site (PLGLAG). Thus, the structure of the fusion n of the invention is as follows: 6262 (TRAIL121-281)-L|NKER8-UROK|N-MMP-(SEQ. No.54) The amino acid sequence and the DNA encoding ce comprising codons optimized for expression in E. coli are, respectively SEQ. No. 31 and SEQ. No. 87 as shown in the attached Sequence Listing.
The amino acid sequence SEQ. No. 31 of the ure described above was used as a template to generate its coding DNA sequence SEQ. No.87. A plasmid containing the coding sequence of DNA was generated and overexpression of the fusion protein was d out in accordance with the general procedures described above. Overexpression was performed according to the general procedure B, using E. coli BL21 (DE3) or Tuner (DE3) strain from Novagen. The protein was separated by electrophoresis in accordance with the general procedure described above. The protein was expressed with histidine tag. e 32. Fusion protein of SEQ. No. 32 The protein of SEQ. No. 32 is a fusion protein having the length of 436 amino acids and the mass of 48 kDa, n domain (a) is the sequence of TRAIL 116- 281, and domain (b) of the effector peptide is the 251-amino acids parasporin-2 from Bacillus thuringensis (SEQ. No. 55) attached at the N-terminus of domain (3)- Additionally, between domain (b) and domain (a) there are sequentially incorpo- rated urokinase cleavage site (RWR), metalloprotease cleavage site (PLGLAG) and steric linker (GSGGGSGGG). Thus, the structure of the fusion protein of the invention is as follows: (SEQ. No. OKIN-MMP-LINKER11-(TRAIL116-281) The amino acid sequence and the DNA encoding sequence comprising codons optimized for expression in E. coli are, tively SEQ. No. 32 and SEQ. No. 88 as shown in the attached Sequence Listing.
The amino acid ce SEQ. No. 32 of the structure described above was used as a template to generate its coding DNA sequence SEQ. No.88. A plasmid containing the coding sequence of DNA was generated and overexpression of the fusion protein was carried out in accordance with the general procedures described above. Overexpression was performed according to the general procedure B, using E. coli BL21 (DE3) or Tuner (DE3) strain from Novagen. The n was separated by ophoresis in accordance with the general procedure described above. The protein was sed with histidine tag.
Example 33. Fusion protein of SEQ. No. 33 6363 The protein of SEQ. No. 33 is a fusion protein having the length of 215 amino acids and the mass of 24,3 kDa, wherein domain (a) is the sequence of TRAIL 121-281, and domain (b) of the effector peptide is the 32-amino acids fusion peptide comprising EGF inhibitor and synthetic lytic e (SEQ. No. 56), ed at the C-terminus of domain (a).
Additionally, between domain (a) and domain (b) there are sequentially incorpo- rated steric linker (G), steric linker (CAACAAAC), steric linker (GGG), o- protease cleavage site G), and urokinase cleavage site (RWR). Thus, the structure of the fusion protein of the invention is as follows: (TRAIL121-281)-L|NKER15-LINKER12-LINKER2-MMP-UROKIN-(SEQ. No. 56) The amino acid sequence and the DNA encoding ce comprising codons optimized for expression in E. coli are, respectively SEQ. No. 33 and SEQ. No. 89 as shown in the attached Sequence g.
The amino acid sequence SEQ. No. 33 of the structure bed above was used as a template to generate its coding DNA ce SEQ. No. 89. A d containing the coding sequence of DNA was generated and overexpression of the fusion protein was carried out in accordance with the general procedures described above. Overexpression was performed according to the general procedure A, using E. coli Tuner (DE3) strain from Novagen. The protein was separated by electrophoresis in ance with the general procedure described above. The protein was expressed with histidine tag.
Example 34 Fusion protein of SEQ. No. 91 The protein of SEQ. No. 91 is a fusion protein having the length of 223 amino acids and the mass of 25.2 kDa, wherein domain (a) is the sequence of TRAIL 121-281, and domain (b) of the effector peptide is the 39-amino acids fusion peptide comprising PDGFR inhibitor and synthetic lytic peptide (SEQ. No. 125), attached at the N-terminus of domain (a).
Additionally, between domain (b) and domain (a) there are sequentially incorpo- rated urokinase cleavage site (RVVR) and metalloprotease cleavage site (PLGLAG), steric linker (GG), steric linker (CAAACAAC) and steric linker (566).
Thus, the structure of the fusion protein of the invention is as follows: (SEQ. No. 125)-UROK|N-MMP- LINKER1-L|NKER13-LINKER16-(TRAIL121-281) The amino acid sequence and the DNA encoding sequence comprising codons optimized for expression in E. coli are, respectively SEQ. No. 91 and SEQ. No. 108 as shown in the attached Sequence Listing. 6464 The amino acid ce SEQ. No. 91 of the structure described above was used as a template to generate its coding DNA sequence SEQ. No. 108. A plasmid containing the coding sequence of DNA was generated and overexpression of the fusion protein was carried out in accordance with the general procedures described above. Overexpression was performed according to the l procedure A, using E. coli Tuner (0E3) strain from Novagen. The protein was separated by ophoresis in ance with the general procedure described above. The protein was expressed without histidine tag.
Example 35 Fusion n of SEQ. No. 92 The protein of SEQ. No. 92 is a fusion protein having the length of 223 amino acids and the mass of 25.6 kDa, wherein domain (a) is the sequence of TRAIL 95- 281, and domain (b) of the effector peptide is the 14-amino acids fusion synthetic lytic peptide (SEQ. No. 41), attached at the inus of domain (a).
Additionally, between domain (b) and domain (a) there are sequentially incorpo- rated polyarginine transporting domain (RRRRRRR), furin cleavage site (RKKR), steric linker (GGG),and steric linker AAC). Thus, the structure of the fusion protein of the ion is as follows: (SEQ. No. 41)-TRANSZ-FURIN-LlNKERZ-LINKER13-(TRAIL95-281) The amino acid sequence and the DNA encoding sequence comprising codons optimized for expression in E. coli are, respectively SEQ. No. 92 and SEQ. No. 109 as shown in the attached Sequence Listing.
The amino acid sequence SEQ. No. 92 of the structure described above was used as a template to te its coding DNA sequence SEQ. No. 109. A plasmid containing the coding sequence of DNA was generated and overexpression of the fusion protein was carried out in accordance with the general procedures described above. pression was performed according to the general procedure A, using E. coli Tuner (0E3) strain from Novagen. The protein was ted by electrophoresis in accordance with the l procedure described above.
The protein was expressed without histidine tag.
Example 36 Fusion protein of SEQ. No. 93 The protein of SEQ. No. 93 is a fusion protein having the length of 232 amino acids and the mass of 26.7 kDa, wherein domain (a) is the sequence of TRAIL 95- 281, and domain (b) of the effector peptide is the 14-amino acids fusion synthetic lytic peptide (SEQ. No. 41), attached at the N-terminus of domain (a). 6565 Additionally, between domain (b) and domain (a) there are sequentially incorpo- rated polyarginine transporting domain (RRRRRRR), furin cleavage site (RKKR), native furin cleavage site (HRQPRGWEQ) steric linker (GGG),and steric linker (CAAACAAC). Thus, the ure of the fusion protein of the invention is as follows: (SEQ. No. 41)-TRANSZ-FURlN-FURN.NAT-LINKER2-LINKER13- (TRAIL95-281) The amino acid sequence and the DNA encoding sequence comprising codons optimized for expression in E. coli are, respectively SEQ. No. 93 and SEQ. No. 110 as shown in the attached Sequence Listing.
The amino acid sequence SEQ. No. 93 of the structure described above was used as a template to generate its coding DNA sequence SEQ. No. 110. A plasmid containing the coding sequence of DNA was generated and overexpression of the fusion protein was carried out in accordance with the general procedures described above. pression was performed according to the general procedure A, using E. coli Tuner (DE3) strain from Novagen. The protein was separated by ophoresis in accordance with the l procedure described above. The protein was expressed without histidine tag. e 37 Fusion protein of SEQ. No. 94 The protein of SEQ. No. 94 is a fusion protein having the length of 207 amino acids and the mass of 23 kDa, n domain (a) is the sequence of TRAIL 121- 281, and domain (b) of the effector peptide is the 14-amino acids fusion synthetic lytic peptide (SEQ. No. 41), attached at the inus of domain (a).
Additionally, between domain (a) and domain (b) there are tially incorpo- rated steric linker (GGGGSGGGG), metalloprotease cleavage site (PLGLAG), urokinase cleavage site (RWR), transporting domain (YARAAARQARA) and steric linker (66). Thus, the ure of the fusion protein of the invention is as follows: (TRAIL121-281)-L|NKER10-MMP-UROKlN-TRANS4-LINKER1-(SEQ. No. 41) The amino acid sequence and the DNA encoding sequence comprising codons optimized for expression in E. coli are, respectively SEQ. No. 94 and SEQ. No. 111 as shown in the attached Sequence Listing.
The amino acid sequence SEQ. No. 94 of the structure described above was used as a template to generate its coding DNA sequence SEQ. No. 111. A plasmid containing the coding sequence of DNA was generated and pression of the fusion protein was d out in accordance with the general procedures 6666 described above. Overexpression was performed according to the general procedure A, using E. coli Tuner (DE3) strain from Novagen. The protein was separated by electrophoresis in accordance with the general procedure described above. The protein was expressed without histidine tag.
Example 38 Fusion protein of SEQ. No. 95 The n of SEQ. No. 95 is a fusion protein having the length of 218 amino acids and the mass of 24.4 kDa, wherein domain (a) is the sequence of TRAIL 1, and domain (b) of the effector e is the 25-amino acids cidine analogue (SEQ. No. 126), attached at the inus of domain (a).
Additionally, between domain (a) and domain (b) there are sequentially incorpo- rated steric linker (GGGGSGGGG), metalloprotease cleavage site G), urokinase cleavage site (RWR), transporting domain (YARAAARQARA) and steric linker (GG). Thus, the structure of the fusion n of the invention is as follows: (TRAIL121-281)-L|NKER10-MMP-UROKlN-TRANS4-LINKER1-(SEQ. No. 126) The amino acid sequence and the DNA encoding sequence comprising codons zed for expression in E. coli are, respectively SEQ. No. 95 and SEQ. No. 112 as shown in the attached Sequence Listing.
The amino acid sequence SEQ. No. 95 of the structure bed above was used as a template to generate its coding DNA sequence SEQ. No. 112. A plasmid containing the coding sequence of DNA was generated and overexpression of the fusion protein was carried out in accordance with the general procedures described above. Overexpression was performed according to the general procedure A, using E. coli Tuner (DE3) strain from Novagen. The n was separated by electrophoresis in accordance with the general procedure described above. The protein was expressed without histidine tag.
Example 39 Fusion protein of SEQ. No. 96 The protein of SEQ. No. 96 is a fusion protein having the length of 219 amino acids and the mass of 24.5 kDa, wherein domain (a) is the sequence of TRAIL 121-281, and domain (b) of the or peptide is the 26-amino acids pleurocidine analogue (SEQ. No. 127), attached at the C-terminus of domain (a).
Additionally, between domain (a) and domain (b) there are sequentially incorpo- rated steric linker (GGGGSGGGG), metalloprotease cleavage site (PLGLAG), urokinase cleavage site (RWR), transporting domain (YARAAARQARA) and steric 6767 linker (66). Thus, the structure of the fusion protein of the invention is as follows: (TRAIL121-281)-L|NKER10-MMP-UROKlN-TRANS4-LINKER1-(SEQ. No. 127) The amino acid sequence and the DNA encoding sequence comprising codons optimized for sion in E. coli are, respectively SEQ. No. 96 and SEQ. No. 113 as shown in the attached Sequence Listing.
The amino acid sequence SEQ. No. 96 of the ure described above was used as a template to generate its coding DNA sequence SEQ. No. 113. A plasmid containing the coding sequence of DNA was ted and overexpression of the fusion protein was carried out in accordance with the general procedures described above. Overexpression was performed according to the general procedure A, using E. coli Tuner (DE3) strain from Novagen. The protein was separated by electrophoresis in accordance with the general procedure described above. The protein was expressed without histidine tag.
Example 40 Fusion protein of SEQ. No. 97 The protein of SEQ. No. 97 is a fusion protein having the length of 212 amino acids and the mass of 23.9 kDa, wherein domain (a) is the ce of TRAIL 121-281, and domain (b) of the effector peptide is the 17-amino acids synthetic lytic peptide (SEQ. No. 128), attached at the C-terminus of domain (a). onally, between domain (a) and domain (b) there are sequentially o- rated steric linker GGGG), metalloprotease cleavage site (PLGLAG), urokinase ge site (RWR), transporting domain (THRPPMWSPVWP) and steric linker (666). Thus, the structure of the fusion protein of the invention is as s: (TRAIL121-281)-L|NKER10-MMP-UROKlN-TRANSS-LINKERZ-(SEQ. No. 127) The amino acid sequence and the DNA encoding sequence sing codons optimized for expression in E. coli are, respectively SEQ. No. 97 and SEQ. No. 114 as shown in the attached Sequence Listing.
The amino acid sequence SEQ. No. 97 of the structure described above was used as a template to generate its coding DNA sequence SEQ. No. 114. A plasmid containing the coding sequence of DNA was ted and overexpression of the fusion protein was carried out in accordance with the general procedures des- cribed above. Overexpression was performed according to the general procedure A, using E. coli Tuner (DE3) strain from Novagen. The protein was separated by 6868 electrophoresis in ance with the general procedure described above. The protein was expressed without ine tag.
Example 41 Fusion protein of SEQ. No. 98 The protein of SEQ. No. 98 is a fusion protein having the length of 207 amino acids and the mass of 23.3 kDa, wherein domain (a) is the sequence of TRAIL 121-281, and domain (b) of the effector peptide is the 14-amino acids synthetic lytic peptide (SEQ. No. 41 ), attached at the C-terminus of domain (a).
Additionally, between domain (a) and domain (b) there are sequentially incorpo- rated steric linker (GGGGSGGGG), metalloprotease cleavage site (PLGLAG), urokinase cleavage site (RWR), transporting domain (YGRKKRRQRRR) and steric linker (66). Thus, the structure of the fusion protein of the invention is as follows: (TRAIL121-281)-L|NKER10-MMP-UROKlN-TRANS6-LINKER1-(SEQ. No. 41) The amino acid sequence and the DNA encoding sequence comprising codons zed for expression in E. coli are, respectively SEQ. No. 98 and SEQ. No. 115 as shown in the attached Sequence Listing.
The amino acid sequence SEQ. No. 98 of the ure described above was used as a template to generate its coding DNA sequence SEQ. No. 115. A plasmid containing the coding sequence of DNA was generated and overexpression of the fusion protein was carried out in accordance with the general procedures described above. Overexpression was performed according to the l procedure A, using E. coli Tuner (DE3) strain from Novagen. The protein was separated by electrophoresis in accordance with the l procedure described above. The protein was expressed t histidine tag.
Example 42 Fusion protein of SEQ. No. 99 The protein of SEQ. No. 99 is a fusion protein having the length of 207 amino acids and the mass of 24 kDa, wherein domain (a) is the sequence of TRAIL 121- 281, and domain (b) of the effector peptide is the no acids synthetic peptide (SEQ. No. 129), attached at the C-terminus of domain (a). 3O Additionally, between domain (a) and domain (b) there are tially incorpo- rated steric linker (GGGGSGGGG), metalloprotease cleavage site (PLGLAG), and ase cleavage site (RWR). Thus, the structure of the fusion protein of the invention is as follows: (TRAIL121-281)-L|NKER10-MMP-UROKlN-(SEQ. N0. 129) 6969 The amino acid sequence and the DNA encoding sequence comprising codons optimized for expression in E. coli are, respectively SEQ. No. 99 and SEQ. No. 116 as shown in the attached Sequence Listing.
The amino acid sequence SEQ. No. 99 of the structure described above was used as a template to generate its coding DNA ce SEQ. No. 116. A plasmid containing the coding ce of DNA was generated and overexpression of the fusion protein was carried out in accordance with the general procedures bed above. Overexpression was performed according to the general procedure A, using E. coli Tuner (0E3) strain from Novagen. The n was separated by electrophoresis in accordance with the general procedure described above. The protein was expressed without histidine tag.
Example 43 Fusion protein of SEQ. No. 100 The protein of SEQ. No. 100 is a fusion protein having the length of 210 amino acids and the mass of 24.1 kDa, wherein domain (a) is the sequence of TRAIL 121-281, and domain (b) of the effector peptide is the no acids synthetic peptide (SEQ. No. 130), attached at the C-terminus of domain (a).
Additionally, between domain (a) and domain (b) there are sequentially incorpo- rated steric linker (GGGGSGGGG), metalloprotease cleavage site (PLGLAG), and urokinase cleavage site (RWR), orting domain (YGRKKRRQRRR) and steric linker (66). Thus, the ure of the fusion protein of the invention is as follows: (TRAIL121-281)-L|NKER10-MMP-UROKIN-TRANSé-LINKER1-(SEQ. No. 130) The amino acid sequence and the DNA encoding sequence comprising codons optimized for expression in E. coli are, respectively SEQ. No. 100 and SEQ. No. 117 as shown in the attached Sequence Listing.
The amino acid sequence SEQ. No. 100 of the structure described above was used as a template to generate its coding DNA sequence SEQ. No. 117. A plasmid containing the coding sequence of DNA was generated and overexpression of the fusion protein was carried out in ance with the general procedures described above. Overexpression was performed according to the l procedure A, using E. coli Tuner (0E3) strain from n. The protein was separated by electrophoresis in ance with the general procedure described above. The protein was expressed without histidine tag.
Example 44 Fusion protein of SEQ. No. 101 7070 The protein of SEQ. No. 101 is a fusion protein having the length of 211 amino acids and the mass of 23.7 kDa, n domain (a) is the sequence of TRAIL 121-281, and domain (b) of the effector peptide is the 29-amino acids synthetic lytic e (SEQ. No. 131 ), attached at the C-terminus of domain (a).
Additionally, between domain (a) and domain (b) there are tially incorpo- rated steric linker (GGGGSGGGG), metalloprotease cleavage site (PLGLAG), and urokinase cleavage site (RVVR) and steric linker (66). Thus, the structure of the fusion protein of the invention is as follows: (TRAIL121-281)-L|NKER10-MMP-UROKIN-LINKER1-(SEQ. No. 131) The amino acid sequence and the DNA encoding ce comprising codons optimized for expression in E. coli are, tively SEQ. No. 101 and SEQ. No. 118 as shown in the attached Sequence g.
The amino acid sequence SEQ. No. 101 of the structure described above was used as a template to generate its coding DNA sequence SEQ. No. 118. A plasmid containing the coding sequence of DNA was generated and overexpression of the fusion protein was carried out in accordance with the general procedures described above. Overexpression was performed ing to the general procedure A, using E. coli Tuner (DE3) strain from Novagen. The protein was separated by electrophoresis in accordance with the general procedure described above. The protein was expressed without ine tag.
Example 45 Fusion protein of SEQ. No. 102 The protein of SEQ. No. 102 is a fusion protein having the length of 234 amino acids and the mass of 26.2 kDa, wherein domain (a) is the sequence of TRAIL 121-281, and two domains (b) of the effector peptide are the 25-amino acids melittin peptide (SEQ. No. 132) and 14-amino acids synthetic lytic peptide (SEQ.
No. 41 ), attached tially at the C-terminus of domain (a).
Additionally, between domain (a) and domain (b) there are sequentially incorpo- rated steric linker (GGGGSGGGG), metalloprotease cleavage site (PLGLAG), and urokinase cleavage site (RWR) and steric linker (66). Additionally a steric linker (GGGGS) is orated between two effector domains.
Thus, the structure of the fusion protein of the invention is as follows: (TRAIL121-281)-L|NKER10-MMP-UROKlN-LINKER1-(SEQ.No.132)-LINKER4-(SEQ.No. 41) The amino acid sequence and the DNA ng sequence comprising codons optimized for expression in E. coli are, respectively SEQ. No. 102 and SEQ. No. 119 as shown in the ed Sequence Listing. 7171 The amino acid sequence SEQ. No. 102 of the structure described above was used as a template to te its coding DNA ce SEQ. No. 119. A plasmid containing the coding sequence of DNA was generated and overexpression of the fusion protein was carried out in ance with the general procedures described above. Overexpression was performed according to the general procedure A, using E. coli Tuner (DE3) strain from Novagen. The protein was ted by electrophoresis in accordance with the general procedure described above. The protein was expressed t histidine tag.
Example 46 Fusion protein of SEQ. No. 103 The protein of SEQ. No. 103 is a fusion protein having the length of 205 amino acids and the mass of 23 kDa, wherein domain (a) is the sequence of TRAIL 121- 281, and domain (b) of the effector peptide is the 25-amino acids melittin peptide (SEQ. No. 132), attached at the inus of domain (a).
Additionally, between domain (a) and domain (b) there are tially incorpo- rated steric linker GGGG), metalloprotease cleavage site (PLGLAG) and urokinase cleavage site (RWR). Thus, the structure of the fusion protein of the invention is as follows: (TRAIL121-281)-L|NKER10-MMP-UROKIN-(SEQ.N0.132) The amino acid sequence and the DNA encoding sequence comprising codons optimized for expression in E. coli are, respectively SEQ. No. 103 and SEQ. No. 120 as shown in the attached Sequence Listing.
The amino acid sequence SEQ. No. 103 of the ure described above was used as a template to generate its coding DNA sequence SEQ. No. 120. A plasmid containing the coding sequence of DNA was generated and overexpression of the fusion protein was carried out in accordance with the general procedures described above. Overexpression was performed according to the general procedure A, using E. coli Tuner (0E3) strain from Novagen. The protein was ted by electrophoresis in accordance with the general procedure described above. The protein was expressed without histidine tag.
Example 47 Fusion protein of SEQ. No. 104 The protein of SEQ. No. 104 is a fusion protein having the length of 215 amino acids and the mass of 24.5 kDa, wherein domain (a) is the sequence of TRAIL 121-281, and domain (b) of the effector peptide is the no acids melittin e (SEQ. No. 132), attached at the C-terminus of domain (a). 7272 Additionally, between domain (a) and domain (b) there are sequentially incorpo- rated steric linker (GGGGSGGGG), metalloprotease cleavage site (PLGLAG), urokinase cleavage site (RWR), polyarginine transporting domain (RRRRRRRR) and steric linker (66). Thus, the structure of the fusion protein of the invention is as follows: (TRAIL121-281)-L|NKER10-MMP-UROKlN-TRANS3-LINKER1-(SEQ.N0.132) The amino acid ce and the DNA encoding sequence comprising codons optimized for expression in E. coli are, respectively SEQ. No. 104 and SEQ. No. 121 as shown in the attached ce g.
The amino acid sequence SEQ. No. 104 of the structure described above was used as a template to generate its coding DNA sequence SEQ. No. 121. A plasmid containing the coding sequence of DNA was generated and overexpression of the fusion protein was d out in accordance with the general procedures described above. Overexpression was performed according to the general procedure A, using E. coli Tuner (DE3) strain from n. The n was separated by electrophoresis in accordance with the general procedure described above. The protein was expressed without histidine tag.
Example 48 Fusion protein of SEQ. No. 105 The protein of SEQ. No. 105 is a fusion protein having the length of 215 amino acids and the mass of 24.4 kDa, wherein domain (a) is the sequence of TRAIL 121-281, and domain (b) of the effector peptide is the 25-amino acids melittin peptide (SEQ. No. 132), ed at the inus of domain (a).
Additionally, between domain (a) and domain (b) there are sequentially incorpo- rated steric linker (GGGGSGGGG), metalloprotease cleavage site (PLGLAG), urokinase cleavage site (RWR) and steric linker (66). Additionally, to the C- terminus of domain (b) is attached a polyarginine transporting domain (RRRRRRRR), forming C-terminal fragment of entire uct.
Thus, the structure of the fusion protein of the invention is as follows: (TRAIL121-281)-L|NKER10-MMP-UROKIN-LINKER1-(SEQ.No.132)-TRANS3 The amino acid sequence and the DNA encoding sequence comprising codons optimized for expression in E. coli are, respectively SEQ. No. 105 and SEQ. No. 122 as shown in the attached Sequence Listing.
The amino acid sequence SEQ. No. 105 of the structure described above was used as a template to te its coding DNA sequence SEQ. No. 122. A plasmid 7373 containing the coding sequence of DNA was generated and overexpression of the fusion protein was carried out in accordance with the general procedures described above. pression was performed according to the general procedure A, using E. coli Tuner (DE3) strain from Novagen. The protein was separated by ophoresis in accordance with the l procedure described above.
The protein was expressed without histidine tag.
Example 49 Fusion n of SEQ. No. 106 The protein of SEQ. No. 106 is a fusion protein having the length of 203 amino acids and the mass of 23.3 kDa, wherein domain (a) is the sequence of TRAIL 121-281, and domain (b) of the effector peptide is the 15-amino acids synthetic lytic peptide (SEQ. No. 35), attached at the C-terminus of domain (a).
Additionally, between domain (a) and domain (b) there are sequentially incorpo- rated steric linker (GGGGSGGGG), metalloprotease cleavage site (PLGLAG), urokinase cleavage site (RWR) and ginine transporting domain (RRRRRRRR) Thus, the ure of the fusion protein of the invention is as follows: 121-281)-L|NKER10-MMP-UROKIN-TRANS3-(SEQ.No.35) The amino acid sequence and the DNA encoding sequence comprising codons optimized for expression in E. coli are, respectively SEQ. No. 106 and SEQ. No. 123 as shown in the attached Sequence Listing.
The amino acid sequence SEQ. No. 106 of the structure described above was used as a template to generate its coding DNA sequence SEQ. No. 123. A plasmid containing the coding sequence of DNA was generated and overexpression of the fusion protein was carried out in ance with the general procedures described above. Overexpression was performed according to the general procedure A, using E. coli Tuner (0E3) strain from Novagen. The protein was separated by electrophoresis in accordance with the l procedure described above. The protein was expressed without histidine tag. e 50 Fusion protein of SEQ. No. 107 The protein of SEQ. No. 107 is a fusion protein having the length of 208 amino acids and the mass of 23.7 kDa, wherein domain (a) is the sequence of TRAIL 121-281, and domain (b) of the effector peptide is the 15-amino acids synthetic lytic peptide (SEQ. No. 35), attached at the C-terminus of domain (a). 7474 Additionally, between domain (a) and domain (b) there are sequentially incorpo- rated steric linker (GGGGSGGGG), metalloprotease cleavage site (PLGLAG), urokinase cleavage site (RWR), transporting domain (YGRKKRRQRRR) and steric linker (66).
Thus, the structure of the fusion protein of the invention is as follows: (TRAIL121-281)-L|NKER10-MMP-UROKlN-TRANS6-LINKER1-(SEQ.No.35) The amino acid sequence and the DNA encoding sequence comprising codons optimized for sion in E. coli are, respectively SEQ. No. 107 and SEQ. No. 124 as shown in the attached Sequence Listing.
The amino acid sequence SEQ. No. 107 of the structure described above was used as a template to generate its coding DNA sequence SEQ. No. 124. A plasmid containing the coding sequence of DNA was generated and overexpression of the fusion protein was carried out in accordance with the general ures described above. Overexpression was med according to the general procedure A, using E. coli Tuner (DE3) strain from Novagen. The protein was separated by electrophoresis in accordance with the general ure described above. The protein was expressed without histidine tag. e 51. Examination of anti-tumor activity of the fusion proteins Examination of anti-tumor activity of the fusion proteins was carried out in vitro in a cytotoxicity assay on tumor cell lines and in vivo in mice. For comparison purposes, rhTRAIL114-281 protein and placebo were used. 1. Measurement of ar ism: determination of secondary structures composition of the obtained proteins Quality of the preparations of fusion proteins in terms of their structures was determined by ar ism for the fusion proteins of Ex. 23, Ex. 11, and Ex. 13.
Circular dichroism is used for determination of secondary structures and conformation of proteins. CD method uses optical activity of the protein structures, manifested in rotating the plane of polarization of light and the appearance of elliptical polarization. CD spectrum of proteins in far ultraviolet (UV) es precise data on the conformation of the main polypeptide chain.
Dialysis Samples of the protein to be analysed, after formulation into a buffer ting of 50 mM Tris-HCl pH 8.0, 100 mM NaCl, 10% glycerol, 0.1 mM ZnClz, 80 mM 7575 saccharose, 5mM DTT, were dialysed in dialysis bags (Sigma-Aldrich) with cut-off 12 kDa. is was performed against 100 fold excess (v/v) of buffer with respect to protein preparations, with stirring for several hours at 4°C. After dialysis was completed, each preparation was centrifuged (25 000 rpm., 10 min., 4°C) and atants were collected. Protein concentration in the samples thus obtained was determined by Bradford method.
Measurement of circular dichroism for proteins in the concentration range of 7 mg/ml was performed on Jasco J-710 opolarimeter, in a quartz e with optical way 0.2 mm or 1 mm. The measurement was performed under the flow of en at 7 l/min, which d to perform the measure- ment in the wavelength range from 195 to 250 nm. Parameters of the measure- ment: spectral tion of - 1 nm; half width of the light beam 1 nm; sensitivi- ty 20 mdeg, the averaging time for one wavelength - 8 s, scan speed 10 nm/min. ed spectra were analyzed numerically in the range of 193-250 nm using CDPro software. Points for which the voltage at the photomultiplier exceeded 700 V were omitted, due to too low signal to noise ratio in this wavelength range.
The data ed served for calculations of particular secondary structures content in the analyzed proteins with use of CDPro software (Table 1).
Table 1. Content of secondary structures in the analyzed proteins..
(Exp-gal)NRM D —WA ———-— * value obtained on the basis of crystalline structure 1D4V ** values obtained on the basis of crystalline structures 1|KQ, 1R4Q, 1ABR, 3PX8 The control molecule (rhTRAIL114-281) shows CD spectrum characteristic for the proteins with predominantly type [Es-sheet structures (sharply outlined ellipticity minimum at the wavelength of 220 nm). This confirms the calculation of secon- dary ure components, suggesting a marginal number of a-helix elements.
The result obtained is also consistent with the data from the crystal structure of hTRAIL protein, and is characteristic for fusion protein of the invention of Ex. 7676 23, wherein beta elements constitute 42% of their structure. For proteins of Ex. 11 and Ex. 13 higher alpha-helix content was observed (additional minimum of the spectrum at ngth 208 nm). This is due to the presence in constructs of KLAKLAK motifs which have strong amphipathic character and form alpha- helical- like structures. Unfortunately, due to low stability of proteins from Ex. 23, Ex. 11 and Ex. 13 in the buffer for CD measurements and low concentrations of analyzed preparations their spectra are characterized by a high noise level and with low tion. Therefore, they may not fully t the actual situation, and only suggest the result. 2. Tests on cell lines in vitro Cell lines Cell lines were obtained from ATCC and CLS, and then propagated and deposited in the ’s Laboratory of Biology Cell Line Bank. During the experiment, cells were routinely checked for the presence of Mycoplasma by PCR que using the kit Venor®GeM Mycoplasma PCR Detection Kit (Minerva Biolabs, Berlin, Germany). The cultures were maintained at standard conditions: 37°C, 5% C02 (in the case of DMEM - 10% C02), and 85% relative humidity. ular cell lines were cultured in appropriate media as recommended by ATCC.
Table 2. Adherent cells number of Cell line Cancer type Medium cells per well (thousands) Cil$C2€5 human colorectal RPMI + 10% FBS + penicillin + cancer streptomycm #CCL-222 '1ng McCoy’s + 10% FBS + penicillin + - cancer streptomycm # CCL-2 cancer streptomycm # HTB-81 :TCC RPMI + 10% FBS + penicillin + - cancer streptomycm # CRL-1435 cancer streptomycm MD‘XQACBC'M DMEM + 10% FBS + penicillin + - cancer streptomycin # HTB-Z6 MDA-MB-435s DMEM + 10% FBS + penicillin + - ATCC# HTB-129 cancer streptomycm 7777 UM-UC-3 human bladder MEM + 10% FBS + penic1llin +. . .
ATCC 3.5 cancer streptomycin # CLR-1749 $3.210 human bladder DMEM + 10% FBS + llin + cancer streptomycm #CRL-21 69 Sm)? human colorectal DMEM + 10% FBS + penicillin + cancer streptomycm #CCL-227 BXPC-3 human pancreatic RPMI + 10% FBS + penicillin + ATCC 4'5 c0”cer stre tomp Vcin #CRL-1687 ’213923 human n McCoy’s + 10% FBS + penicillin + cancer streptomycm # HTB-77 NIH:A(_)I_\éCCAR-3 human ovarian RPMI + 20% FBS + 0,01 mg/ml cancer insulina + penic1llin + streptomycm #HTB-161 l-IEDCGCZ human liver MEM + 10% FBS + penicillin + ma streptomycm # HB-8065 Human embrional MEM + 10% FBS + penicillin + ATCC 4 k'd ll . 1 ney ce 5 5 rep omycmt t # CLR-1573 ACHN human kidney. MEM + 10% FBS + penic1llin +. . .
ATCC 4 cancer streptomycm #CCL-222 CAKI 1 human kidney. McCoy s + 10% FBS + penic1llin +, . . .
ATCC 3'5 cancer stre tomp ycin #HTB-46 CAKI 2 human kidney. McCoy s + 10% FBS + penic1llin +, . . .
ATCC 3'5 cancer stre tomp ycin # HTB-47 NCIATHcf’gAR human small cell RPMI + 10% FBS + penicillin + lung cancer streptomycm 1351 HT144 human melanoma McCoy s + 10% FBS + pemc1llin +, . . .
ATCC 7 cells # HTB-63 streptomycm NCI-H46O human lung RPMI + 10% FBS + penicillin + ATCC 2'5 cancer stre tomp ycin #HTB-177 A549 human lung RPMI + 10% FBS + pemCillin +. . .
ATCC 2'5 cancer stre tomp ycin # CCL-185 NEECSCA human uterine s + 10% FBS + penicillin + sarcoma streptomycm # CRL-1976 MES-SA/Dx5 multidrug- MCCOY 5 +5236 5?; :fiemaum +, . . .
ATCC resistant human 4 p y #CRL-1977 uterine sarcoma 7878 MES-SA/MX2 Waymouth’s MB 752/1 + McCoy’s human uterine ATCC (1 : 1) + 10% FBS + penicillin + #CRL-2274 streptomycin # HTB-58 cancer streptomycin HCT-116 ATCC human colorectal McCoy’s + 10% FBS + penicillin + # CC L-247 cancer streptomycin DMEM:F12 + 5% horse plasma + 0.5 MCF10A ATCC mammary pg/ml ortisone + 10 pg/ml # CRL-10317 epithelial cells insuline + 20 ng/ml growth factor Panc-1 CLS human atic DMEM + 10% FBS + llin + 330228 cancer streptomycin Paxfii‘n human pancreatic RPMI + 10% FBS + penicillin + cancer streptomycm # CRL-2549 PLC/PRF/5 CLS human liver DMEM + 10% FBS + penicillin + 330315 hepatoma streptomycin LNCaP human prostate RPMI + 10% FBS + penic1llin +. . .
ATCC cancer streptomycm # CRL- 1740 SK-Hep-1 human liver RPMI + 10% FBS + penicillin + CLS300334 hepatoma omycin MEM + 13:15»; A498 human kidney p ypceirr1nc1llin + CLS 300113 cancer MEM + flaw; [3521mm + HT1080 ATCC Human p y 3 #CCL-121 fibrosarcoma HUV-EC-C human umbilical M199 + 20% FBS + penicillin + 0,05 ATCC vein endothelial mg/ml ECGS + 0,1 mg/ml heparine 8.5 # 30 cells + llin + streptomycin Table 3. Nonadherent cells: number of Cell line Cancer type Medium cells per well t(housands) NCI- H69 human small cell RPMI + 10% FBS + penicillin + ATCC # HTB- 119 lung cancer streptomycin ATCC #CRL-2570 streptomycin ATCC # CCL-240 streptomycin ATCC # CCL- 119 streptomycin M'I'I' cytotoxicity test MIT assay is a colorimetric assay used to measure proliferation, viability and cytotoxicity of cells. It consists in decomposition of a yellow tetrazolium salt M'I'I' (4,5-dimethylthiazolyl)-2,5-diphenyltetrazolium bromide) to the water- 7979 insoluble purple dye formazan by mitochondrial enzyme succinate-tetrazolium ase 1. MIT reduction occurs only in living cells. Data analysis consists in determining |C50 concentration of the protein (in ng/ml), at which the 50% reduction in the number of cells occurs in the population treated compared to control cells. Results were ed using GraphPad Prism 5.0 software. The test was performed according to the literature ptions , JE, (1998). Cell Biology, a Laboratory Handbook, second edition, Academic Press, San Diego; Yang, Y., Koh, LW, Tsai, JH., (2004); Involvement of viral and chemical factors with oral cancer in Taiwan, Jpn J Clin Oncol, 34 (4), 176-183).
Cell culture medium was diluted to a defined density (104 - 105 cells per 100 pl).
Then 100 pl of appropriately d cell suspension was applied to a 96-well plate in triplicates. Thus prepared cells were ted for 24 h at 37°C in 5% or % C02, depending on the medium used, and then to the cells (in 100 pl of medium) r 100 pl of the medium containing various concentrations of tested ns were added. After incubation of the cells with tested proteins over the period of next 72 hours, which is equivalent to 3-4 times of cell division, the medium with the test protein was added with 20 ml of MIT working solution [5 mg/ml], and incubation was continued for 3 h at 37°C in 5% C02.
Then the medium with M'I'I' solution was removed, and formazan crystals were dissolved by adding 100 pl of DMSO. After stirring, the absorbance was measured at 570 nm (reference filter 690 nm).
EZ4U cytotoxicity test EZ4U (Biomedica) test was used for testing cytotoxic activity of the proteins in nonadherent cell lines. The test is a modification of the MIT method, n formazan formed in the ion of tetrazolium salt is water-soluble. Cell viability study was carried out after continuous r incubation of the cells with protein (seven concentrations of n, each in triplicates). On this basis |C50 values were determined (as an average of two independent experiments) using the GraphPad Prism 5 software. Control cells were incubated with the 3O solvent only.
The results of in vitro cytotoxicity tests are summarized as |C50 values (ng/ ml), which corresponds to the protein concentration at which the cytotoxic effect of fusion proteins is observed at the level of 50% with respect to control cells treated only with solvent. Each experiment represents the average value of at least two independent experiments performed in triplicates. As a criterion of lack of activity of protein preparations the IC50 limit of 2000 ng/ml was adopted.
Fusion proteins with an |C5o value above 2000 were considered inactive. 8080 Cells selected for this test ed tumor cell lines that are naturally resistant to TRAIL protein (the ion of natural resistance to TRAIL: IC50 for TRAIL protein > 2000), as well as tumor cell lines sensitive to TRAIL protein and resistant to doxorubicin line MES-SA/DXS as a cancer line resistant to conventional anticancer medicaments.
Undifferentiated HUVEC cell line was used as a healthy l cell line for assessment of the effect/toxicity of the fusion proteins in non-cancer cells.
The results obtained confirm the possibility of overcoming the resistance of the cell lines to TRAIL by administration of certain fusion proteins of the invention to cells naturally resistant to TRAIL. When fusion proteins of the invention were administered to the cells sensitive to TRAIL, in some cases a clear and strong potentiation of the potency of action was ed, which was manifested in reduced IC50 values of the fusion protein compared with |C5o for the TRAIL alone.
Furthermore, cytotoxic activity of the fusion protein of the invention in the cells resistant to classical anti-cancer medicament doxorubicin was obtained, and in some cases it was stronger than ty of TRAIL alone.
The IC50 values above 2000 obtained for the non-cancer cell lines show the ab- sence of toxic effects associated with the use of ns of the invention for healthy cells, which tes potential low ic toxicity of the protein. ination of xic activity of selected n preparations against extended panel of tumor cell lines Table 4 presents the results of the tests of cytotoxic activity in vitro for selected fusion proteins of the invention against a broad panel of tumor cells from different organs, corresponding to the broad range of most common cancers.
The experimental results are presented as a mean value i standard deviation (SD). All calculations and graphs were prepared using the GraphPad Prism 5.0 software.
Obtained |C50 values confirm high cytotoxic activity of fusion proteins and thus their potential utility in the treatment of cancer. 8181 28%: S 223335 c0552: US 3 8.5335 2583 msoscscou some ho IIII'IIIIIE 3.55% an 8x835 338. IIIIIII IIIIIIIIIIIII IIIIIEI Illlllllllagé 8282 28%: “mm: Mm c0552: 223335 3 85835 2.583 no.5: cou 3.55% HxBotG .mv IIIIIIIIIIIII 338. 8383 28%: “mm: mm 223335 c0552: US 3 85835 2583 cou some 3.55% %§%%%% u._x33>u .3 5305 338. IIIIIIIIIIIH; 8484 :ER: “mm”: vw ama c0552: U8 3 8.5.835 2.583 msoscscou 8.53 3.55% %%%%%% 8x883 .uv £30; 338. Eéégéél 8585 26%: “mm: Lm>o .nw 223335 c0552: US 3 85835 2583 cou some 3.55% I%%%%% 8x833 .3 5305 338. 8686 3. Antitumor effectiveness of fusion ns in vivo on xenografts Antitumor activity of protein preparations was tested in a mouse model of human colon cancer Colo 205 and HCT-116, human lung cancer A549 and NCI- H460-Luc, human prostate cancer PC-3, human pancreas cancer Panc-1 and MIA PaCa-Z, human liver cancer PCL/PRF/S and HepGZ, and human multidrug resistant MES e sarcoma 5.
Cells The cells of human lung cancer A549 and NCI-H460-Luc2 and human prostate cancer PC3 were maintained in RPMI1640 medium (HyClone, Logan, UT, USA) supplemented with 10% fetal calf serum and 2 mM glutamine.
The cells of human colon cancer Colo 205 were maintained in RPM|1640 medium (HyClone, Logan, UT, USA) (optionally mixed in the ratio of 1:1 with Opto-MEM (Invitrogen, Cat. No. 22600-134) supplemented with 10% fetal calf serum and 2 mM glutamine.
The cells of human pancreas cancer PANC-1, human liver cancer PLC/PRF/5, as cancer MIA PaCa-Z and human colon cancer SW-620 were maintained in DMEM medium (HyClone, Logan, UT, USA) supplemented with 10% fetal calf serum and 2 mM glutamine.
The cells of human colon cancer 6 were maintained in McCoy’s medium (HyClone, Logan, UT, USA) supplemented with 10% fetal calf serum and 2 mM glutamine.
The cells of multidrug resistant human uterine sarcoma MES-SA.Dx5 were maintained in McCoy’s medium (HyClone, Logan, UT, USA) supplemented with % fetal calf serum and 2 mM glutamine, and 1 uM doxorubicin hydrochloride (Sigma, Cat. No. D1515-10MG). Three days before the cells tation, the cells were ed in medium without doxorubicin.
The cells of human liver cancer HepGZ were maintained in MEM medium ne, Logan, UT, USA) supplemented with 10% fetal calf serum and 2 mM glutamine. On the day of mice grafting, the cells were detached from the support by washing the cells with trypsin (Invitrogen), then the cells were centrifuged at 1300 rpm, 4°C, 8 min., suspended in HBSS buffer (Hanks medium). 8787 On the day of mice grafting, the cells were detached from the t by washing the cells with trypsin (Invitrogen), then the cells were centrifuged at 1300 rpm, 4°C, 8 min., suspended in HBSS buffer (Hanks medium).
Mice Examination of antitumor activity of proteins of the invention was conducted on 4-6 week-old (lung cancer model) foxn1(nu)/J) mice or 9-10 week old (prostate cancer model) obtained from Centrum Medycyny Doswiadczalnej in Bialystok, 4-5 week-old female Crl:SHO-Prkdc$ddHrh mice obtained from Charles River Germany. Mice were kept under specific pathogen-free conditions with free access to food and demineralised water (ad libitum). All experiments on animals were carried in ance with the guidelines: ”Interdisciplinary Principles and Guidelines for the Use of Animals in Research, Marketing and Education" issued by the New York Academy of Sciences' Ad Hoc Committee on Animal ch and were approved by the IV Local Ethics Committee on Animal Experimentation in Warsaw (No. 9).
The course and evaluation of the experiments Tumor size was measured using electronic er, tumor volume was calculated using the formula: (a2 x b)/2, where a = shorter diagonal of the tumor (mm) and b = longer diagonal of the tumor (mm). tion of tumor growth was calculated using the formula: TGI [%] (Tumor growth inhibition) = (WT/WC) x 100 - 100% wherein WT is the average tumor volume in the treatment group, and WC is the average tumor volume in the control group.
The experimental results are presented as a mean value i rd deviation (SD). All calculations and graphs were prepared using the program GraphPad Prism 5.0.
Lung cancer model c Experiment A.
On day 0 be.Cg-foxn1(nu)/J mice were d subcutaneously (s.c.) in the right side with 5x106 of A549 cells suspended in 0.1 ml of the mixture HBSS:Matrigel 421 using syringe with a needle 0.5 x 25 mm (Bogmark). On the 19 day of the experiment mice were randomized to obtain the average size of tumors in the group of ~75 mm3 and assigned to treatment groups. The treatment groups were stered with the preparations of fusion protein of the invention of Ex. 11a (15 mg/kg), and rhTRAIL114-281 (20 mg/kg) as a 8888 comparison and water for injections as a control. The preparations were admi- nistered intravenously (i.v) 6 times once daily every second day. On the 35 day of the experiment mice were sacrificed by disruption of the spinal cord.
The experimental results are shown on Fig. 1 and Fig. 2 as a diagram of chan- ges of the tumor volume and tumor growth inhibition (%TG|) as the percentage of control in mice treated with fusion protein of the invention of Ex. 11"11 and comparatively with rhTRAIL114-281.
The experimental results presented in Fig. 1 and 2 show that stration of the fusion n of the invention of Ex.11a caused A549 tumor growth inhibi- tion, with TGI 28% relative to the control on 35 day of experiment. For rhTRAIL114-281 used as the ative reference, a slight inhibitory effect on tumor cell growth was obtained relative to the control, with TGI at the level of 0%. Thus, fusion proteins of the invention exerted much er effect compared to TRAIL alone.
Experiment B.On day 0 Crl:SHO-Prkdc5CidHrhr mice were grafted subcutaneously (s.c.) in the right side with 7x106 of A549 cells suspended in 0.1 ml of the mixture HBSS:Matrigel 3:1 using syringe with a needle 0.5 x 25 mm (Bogmark).
On the 17 day of the experiment mice were ized to obtain the e size of tumors in the group of ~100-120 mm3 and assigned to treatment groups.
The treatment groups were administered with the preparations of fusion protein of the invention of Ex. 11a (40 mg/kg), and rhTRAIL114-281 (20 mg/kg) as a comparison against formulation buffer (19 mM NaHzPO4, 81 mM NazHPO4, 50 mM NaCl, 5 mM glutathione, 0.1 mM ZnClz, 10% glycerol, pH 7.4) as a control. The preparations were administered intravenously (i.v) 6 times once daily every second day. On the 34 day of the experiment mice were iced by disruption of the spinal cord.
The experimental results are shown on Fig. 3 and Fig. 4 as a diagram of chan- ges of the tumor volume and tumor growth inhibition (%TG|) as the tage of control in mice treated with fusion n of the invention of Ex.11a and 3O comparatively rhTRAIL114-281.
The experimental results presented in Fig. 3 and 4 show that administration of the fusion protein of the invention of Ex.11a caused A549 tumor growth inhibition, with TGI 45% relative to the control on 34 day of experiment. For rhTRAIL114-281 used as the comparative reference, a slight inhibitory effect on tumor cell growth was obtained relative to the control, with TGI at the level of 8989 21,8%. Thus, fusion protein of the invention exerted much stronger effect compared to TRAIL alone.
Experiment C.
On day 0 Crl:SHO-PrkchddHrhr mice were grafted subcutaneously (s.c.) in the right side with 5x106 of NCI-H460-Luc2 cells suspended in 0.1 ml of HBSS buffer using syringe with a needle 0.5 x 25 mm (Bogmark). On the 11 day of the experiment mice were randomized to obtain the average size of tumors in the group of ~100-120 mm3 and assigned to treatment groups. The treatment groups were administered with the preparations of fusion protein of the invention of Ex.11a (40 mg/kg and 30 mg/kg), and rhTRAIL114-281 (20 mg/kg) as a comparison against formulation buffer (19 mM NaHzPO4, 81 mM NazHPO4, 50 mM NaCl, 5 mM glutathione, 0.1 mM ZnClz, 10% glycerol, pH 7.4) as a control. The preparations were stered intravenously (i.v) 6 times once daily every second day (in case of fusion protein of the invention of Ex.11a first administration at a dose 40 mg/kg and subsequent at 30 mg/kg. On the 29 day of the experiment mice were sacrificed by tion of the spinal cord.
The experimental results are shown on Fig. 5 and Fig. 6 as a diagram of changes of the tumor volume and tumor growth inhibition (%TG|) as the percentage of control in mice treated with fusion protein of the invention of Ex. 11a and comparatively with L114-281.
The mental s presented in Fig. 5 and 6 show that administration of the fusion protein of the invention of Ex.11a caused tumor 60-Luc2 growth inhibition, with TGI 93% relative to the control on 29 day of experi- ment. For rhTRAIL114-281 used as the comparative nce, an inhibitory effect on tumor cell growth was obtained relative to the control, with TGI at the level of 76%. Thus, fusion protein of the invention exerted much stronger effect compared to TRAIL alone.
The tested fusion protein did not cause significant side effects sted by a decrease in body weight of mice (i.e. less than 10% of the baseline body 3O weight). This shows low ic toxicity of the protein of the invention.
Prostate cancer model On day 0 be.Cg-foxn1(nu)/J mice were grafted subcutaneously (s.c.) in the right side with 5x106 of PC3 cells suspended in 0.18 ml of HBSS buffer and 0.02 ml of Matrigel using syringe with a needle 0.5 x 25 mm (Bogmark). On the 29 day of the ment mice were randomized to obtain the average size of tumors in the group of ~90 mm3 and assigned to treatment groups. The 9090 treatment groups were administered with the preparations of fusion protein of the invention of Ex. 11a (15 mg/kg) and 0.9% NaCl as a control. The prepa- rations were administered intravenously (i.v) once daily for 6 days. On the 60 day of the experiment mice were sacrificed by disruption of the spinal cord.
The mental results are shown on Fig. 7 and Fig. 8 as a diagram of chan- ges of the tumor volume and tumor growth inhibition (%TG|) as the percentage of control in mice treated with fusion protein of the ion of Ex. 11a.
The experimental results presented in Fig. 7 and 8 show that administration of the fusion protein of the invention of Ex.11a caused PC3 tumor growth inhibition, with TGI 33% relative to the control on the 60 day of experiment.
The tested fusion protein did not cause significant side effects manifested by a decrease in body weight of mice (i.e. less than 10% of the baseline body weight). This shows low systemic toxicity of the n of the invention.
Pancreas cancer model Experiment A on PANC-1 cells On day 0 Crl:SHO-Prkdc5ddHrhr mice were grafted subcutaneously (s.c.) in the right side with 5x106 of PANC-1 cells suspended in 0.1 ml of the mixture HBSS:Matrigel 3:1 using syringe with a needle 0.5 x 25 mm (Bogmark). On the 31 day of the experiment mice were ized to obtain the average size of tumors in the group of ~110 mm3 and assigned to treatment groups. The treatment groups were administered with the preparations of fusion protein of the invention of Ex. 11a (40 mg/kg), and rhTRAIL114-281 (20 mg/kg) as a comparison against formulation buffer (19 mM NaHzPO4, 81 mM NazHPO4, 50 mM NaCl, 5 mM glutathione, 0.1 mM ZnClz, 10% ol, pH 7.4) as a control. The preparations were administered intravenously (i.v) 6 times once daily every second day. On the 42 day of the experiment mice were sacrificed by tion of the spinal cord.
The experimental s are shown on Fig. 9 and Fig. 10 as a diagram of chan- ges of the tumor volume and tumor growth tion (%TG|) as the percentage of control in mice treated with fusion n of the invention of Ex.11a and comparatively with rhTRAIL114-281.
The experimental results presented in Fig. 9 and 10 show that administration of the fusion protein of the invention of Ex. 113 caused PANC-1 tumor growth inhibition, with TGI 32.6% relative to the control on 42 day of experiment. For rhTRAIL114-281 used as the ative reference, a slight inhibitory effect on 9191 tumor cell growth was obtained relative to the control, with TGI at the level of 26%. Thus, fusion protein of the invention exerted much stronger effect compared to TRAIL alone.
The tested fusion protein did not cause significant side effects manifested by a decrease in body weight of mice (i.e. less than 10% of the baseline body weight). This shows low systemic toxicity of the protein of the invention.
Experiment B on MIA PaCa-Z cells On day 0 O-Prkdc5€idHrhr mice were grafted subcutaneously (s.c.) in the right flank region with 5x106 of MIA PaCa-Z cells suspended in 0.1 ml of 3:1 mixture HBSS bufferzMatrigel using e with a 0.5 x 25 mm needle (Bogmark). When tumors reached the size of 60-398 mm3 (day 20), mice were randomized to obtain the e size of tumors in the group of ~ 170 mm3 and assigned to treatment groups. The treatment groups were administered with the preparations of fusion protein of the invention of Ex. 16b (50 mg/kg) and rhTRAIL114-281 (50 mg/kg) as a comparison and reference compound gemcy- tabine (Gemzar, Eli Lilly) (50 mg/kg). The ations were administered intravenously (i.v.) six times every second day, gemcytabine was applied intraperitoneally (i.p.) in the same schedule. The control group received formulation buffer.
When a therapeutic group d the average tumor size of ~ 1000 mm3, mice were sacrificed by the cervical dislocation.
The experimental results obtained in mice Crl:SHO-Prkdc$CidHrhr burdened with MIA PaCa-Z pancreatic oma treated with fusion protein of the ion of Ex. 16b and comparatively with rhTRAIL114-281 and gemcytabine are shown in Fig. 19 as a diagram of changes of the tumor volume and in Figure 20 which shows tumor growth inhibition (%TGI) as a percentage of control.
The s of the experiment presented in Figures 19 and 20 show that administration of the fusion protein of the invention of Ex. 16b caused MIA PaCa-Z tumor growth inhibition, with TGI 93% relative to the control on 61th 3O day of the ment. For rhTRAIL114-281 and gemcytabine as comparative references, a slight inhibitory effect on tumor cell growth was obtained relative to the control, with TGI at the level of 68% and 42,6%, respectively.
Thus, fusion ns of the invention exerted much stronger effect compared to TRAIL alone and standard chemotherapy.
Colon cancer model 9292 Experiment A on HCT116 cellsOn day 0 Crl:SHO-PrkchCidHrhr mice were grafted subcutaneously (s.c.) in the right side with 5x106 of HCT116 cells suspended in 0.1 ml of HBSS buffer using syringe with a needle 0.5 x 25 mm (Bogmark). On the18 day of the experiment mice were randomized to obtain the average size of tumors in the group of ~400mm3 and assigned to treatment groups. The treatment groups were administered with the preparations of fusion protein of the invention of Ex. 118 (35 mg/kg), and L114-281 (20 mg/kg) as a comparison against formulation buffer (5 mM NaHzPO4, 95 mM NazHPO4, 200 mM NaCl, 5 mM glutathione, 0.1 mM ZnClz, 10% glycerol, 80 mM saccharose, pH 8.0) as a control. The ations were administered intravenously (i.v) 6 times once daily every second day. On the 32 day of the experiment mice were sacrificed by disruption of the spinal cord.
The experimental results are shown on Fig. 11 and Fig. 12 as a diagram of changes of the tumor volume and tumor growth inhibition (%TG|) as a tage of control in mice treated with fusion protein of the ion of Ex. 11a and atively with rhTRAIL114-281.
The experimental s presented in Fig. 11 and 12 show that administration of the fusion protein of the invention of Ex.11a caused tumor HCT116 growth inhibition, with TGI 33.5% relative to the control on 32 day of experiment. For rhTRAIL114-281 used as the comparative reference, a slight inhibitory effect on tumor cell growth was obtained relative to the control, with TGI at the level of .6%. Thus, fusion protein of the invention exerted much stronger effect compared to TRAIL alone.
The tested fusion protein did not cause icant side s manifested by a decrease in body weight of mice (i.e. less than 10% of the baseline body weight). This shows low systemic toxicity of the protein of the invention.
Experiment A1 on HCT116 cells On day 0 Crl:SHO-Prkdc5€idHrhr mice were grafted subcutaneously (s.c.) in the right flank region with 5x106 of HCT116 cells suspended in 0.1 ml of the 3:1 3O mixture HBSS bufferzMatrigel using syringe with a 0.5 x 25 mm needle (Bogmark). When tumors reached the size of 71 -432 mm3 (day 13), mice were ized to obtain the average size of tumors in the group of ~ 180 mm3 and assigned to treatment groups. The treatment groups were administered with the preparations of fusion protein of the invention of Ex. 16b (90 mg/kg) and rhTRAIL114-281 (65 mg/kg) as a comparison. The preparations were adminis- 9393 tered intravenously (i.v.) six times every second day. The control group received formulation .
When an mental group reached the average tumor size of ~ 1000 mm3, mice were sacrificed by cervical dislocation.
The experimental results obtained in mice Crl:SHO-PrkdcscjdHrhr burdened with HCT116 colon cancer treated with fusion proteins of the invention of Ex. 16b and comparatively with rhTRAIL114-281 are shown in Fig. 21 as a diagram of changes of the tumor volume and in Figure 22 which shows tumor growth inhibition (%TG|) as a percentage of control.
The results of experiments presented in Figures 21 and 22 show that administration of the fusion protein of the invention of Ex. 16b caused HCT116 tumor growth inhibition, with TGI 65.8% relative to the control on 24th day of the experiment. For rhTRAIL114-281 used as the comparative reference, a slight inhibitory effect on tumor cell growth was obtained relative to the control, with TGI at the level of 37.9%. Thus, fusion proteins of the invention exerted much er effect compared to TRAIL alone.
Experiment B on SW620 cells On day 0 Crl:SHO-PrkchddHrhr mice were grafted aneously (s.c.) in the right side with 5x106 of SW620 cells suspended in 0.1 ml of the mixture HBSS:Matrigel 3:1 using syringe with a needle 0.5 x 25 mm (Bogmark). On the 17 day of the experiment mice were randomized to obtain the average size of tumors in the group of ~320 mm3 and assigned to treatment groups. The treatment groups were administered with the preparation of fusion protein of the invention of Ex. 168 (20 mg/kg) and rhTRAIL114-281 (30 mg/kg) as a comparison against formulation buffer (19 mM 4, 81 mM NazHPO4, 50 mM NaCl, 5 mM hione, 0.1 mM ZnClz, 10% glycerol, pH 7.4) as a control. The ations were administered intravenously (i.v) 6 times once daily every second day. On the 31 day of the experiment mice were sacrificed by disruption of the spinal cord. 3O The experimental results in mice Crl:SHO-Prkdc$ddHrhr burdened with SW620 d with fusion protein of the invention of Ex.16a and comparatively with rhTRAIL114-281 are shown on Fig. 13 and Fig. 14 as a diagram of changes of the tumor volume and tumor growth inhibition (%TG|) as a percentage of control.
The experimental results ted in Fig. 13 and 14 show that stration of the fusion protein of the invention of Ex.16a caused tumor SW620 growth inhibition, with TGI equal to 25% comparing to control on the 31 day of 9494 experiment. For rhTRAIL114-281 used as the ative reference, no inhibitory effect on tumor cell growth was obtained relative to the control, with TGI at the level of -9%. Thus, fusion proteins of the invention exerted much stronger effect compared to TRAIL alone.
The tested fusion protein did not cause significant side effects manifested by a decrease in body weight of mice (i.e. less than 10% of the baseline body ). This shows low systemic toxicity of the protein of the invention.
Experiment C on Col0205 cellsOn day 0 Crl:SHO-Prkdc$ddHrhr mice were grafted subcutaneously (s.c.) in the right side with 5x106 of Col0205 cells ded in 0.1 ml of H355 buffer using syringe with a needle 0.5 x 25 mm (Bogmark). On the 13 day of the experiment mice were randomized to obtain the average size of tumors in the group of ~115 mm3 and ed to treatment groups.
The treatment groups were administered with the preparations of fusion protein of the invention of Ex.16a (30 mg/kg), and rhTRAIL114-281 (30 mg/kg) as a comparison against formulation buffer (19 mM NaHzPO4, 81 mM NazHPO4, 50 mM NaCl, 5 mM glutathione, 0.1 mM ZnClz, 10% glycerol, pH 7.4) as a control. The preparations were administered enously (i.v) 6 times once daily every second day. On the 33 day of the experiment mice were sacrificed by disruption of spinal cord.
The experimental results are shown on Fig. 15 and Fig. 16 as a diagram of changes of the tumor volume and tumor growth inhibition (%TG|) as the percentage of control.
The experimental s presented in Fig. 15 and 16 show that administration of the fusion protein of the invention of Ex.16a caused Col0205 tumor growth inhibition, with TGI equal to 80% ve to the control on 33 day of experiment. For rhTRAIL114-281 used as the comparative reference, a slight inhibitory effect on tumor cell growth was obtained relative to the control, with TGI at the level of 18.8%. Thus, fusion ns of the invention exerted much stronger effect compared to TRAIL alone.
The tested fusion proteins did not cause icant side effects manifested by a decrease in body weight of mice (i.e. less than 10% of the baseline body weight). This shows low systemic toxicity of the protein of the invention.
Multidrug resistant uterine sarcoma model On day 0 Crl:SHO-Prkdc5ddHrhr mice were grafted subcutaneously (s.c.) in the right side with 7x106 of MES-SA/Dx5 cells suspended in 0.1 ml of the e atrigel 10:1 using syringe with a needle 0.5 x 25 mm (Bogmark). On the 19 day of the experiment mice were randomized to obtain the average size of 9595 tumors in the group of ~180 mm3 and assigned to treatment groups. The treatment groups were administered with the preparation of fusion protein of the invention of Ex.11a(30 mg/kg) and rhTRAIL114-281 (10 mg/kg) as a comparison against formulation buffer (19 mM 4, 81 mM NazHPO4, 50 mM NaCl, 5 mM glutathione, 0,1 mM ZnClz, 10% glycerol, pH 7.4) as a l.
Preparations were administered intravenously (i.v.) 6 times once daily every second day. On the 35 day of the experiment mice were sacrificed by disruption of spinal cord.
The experimental results are shown on Fig. 17 and Fig. 18 as a diagram of changes of the tumor volume and tumor growth inhibition (%TG|) as a percentage of control.
The experimental results presented in Fig. 17 and 18 show that stration of the fusion protein of the ion of Ex.11a caused MES-SA/Dx5 tumor growth inhibition, with TGI equal to 81% relative to the control on 35 day of experiment. For rhTRAIL114-281 used as the comparative reference, a slight inhibitory effect on tumor cell growth was obtained ve to the control, with TGI at the level of 29%. Thus, fusion protein of the invention exerted much stronger effect compared to TRAIL alone.
The tested fusion protein did not cause significant side effects manifested by a decrease in body weight of mice (i.e. less than 10% of the baseline body weight). This shows low systemic toxicity of the protein of the invention.
Liver cancer model Experiment A on HepGZ cells On day 0 Crl:SHO-Prkdc5CidHrhr mice were d subcutaneously (s.c.) in the right flank region with 7x106 of HepGZ cells suspended in 0.1 ml of the 3:1 mixture HBSS bufferzMatrigel using syringe with a 0.5 x 25 mm needle (Bogmark). When tumors reached the size of 64-529 mm3 (day 25), mice were randomized to obtain the e size of tumors in the group of ~ 230 mm3 and assigned to treatment groups. The treatment groups were administered with the preparations of fusion protein of the invention of Ex. 16b (80 mg/kg) and rhTRAIL114-281 (50 mg/kg) as a comparison and reference nd 5-FU (5- Fluorouracil, Sigma-Aldrich) (20 mg/kg). The ations were administered intravenously (i.v.) six times every second day, 5-FU was applied intraperitoneally (i.p.). The control group received formulation buffer.
When the therapeutic group reached the average tumor size of ~ 1000 mm3, mice were iced by cervical dislocation. 9696 The experimental results obtained are shown in Fig. 23 as a diagram of changes of the tumor volume and in Figure 24 which shows tumor growth inhibition (%TG|) as a percentage of control.
The results of the experiment presented in Figures 23 and 24 show that administration of the fusion protein of the invention of Ex. 16b caused HepGZ tumor growth inhibition, with TGI 94.6% relative to the control on 42th day of the experiment. For rhTRAIL114-281 as the comparative reference, a slight inhibitory effect on tumor cell growth was obtained relative to the control, with TGI at the level of 23.2%. Reference compound, 5-FU, didn’t show any efficacy against HepGZ . Thus, fusion protein of the invention exerted much stronger effect compared to TRAIL alone and standard chemotherapy.
Experiment B on PLC/PRF/5 cells On day 0 Crl:SHO-Prkdc5°idHr“’ mice were grafted subcutaneously (s.c.) in the right flank region with 7x106 of PLC/PRF/5 cells suspended in 0.1 ml of the 3:1 mixture HBSS zMatrigel using e with a 0.5 x 25 mm needle (Bogmark). When tumors reached the size of 72-536 mm3 (day 29), mice were randomized to obtain the e size of tumors in the group of ~ 205 mm3 and assigned to treatment groups. The treatment groups were administered with the preparations of fusion protein of the ion of Ex. 16b (50 mg/kg) and rhTRAIL114-281 (50 mg/kg) as a comparison and reference compound 5-FU (5- Fluorouracil, Sigma-Aldrich) (30 mg/kg). The preparations were administered intravenously (i.v.) six times every second day, except 5-FU, which was applied intraperitoneally (i.p.) in the schedule (q1dx5)x2. The l group received formulation buffer.
When an experimental group reached the e tumor size of ~ 1000 mm3, mice were iced by cervical dislocation.
The experimental results obtained are shown in Fig. 25 as a diagram of changes of the tumor volume and in Figure 26 which shows tumor growth inhibition (%TG|) as a percentage of control.
The results of the experiment presented in Figures 25 and 26 show that administration of the fusion protein of the invention of Ex. 16b caused F/5 tumor growth inhibition, with TGI 53% relative to the control on 43th day of the experiment. For rhTRAIL114-281 and 5-FU as comparative references, a slight inhibitory effect on tumor cell growth was obtained relative to the l, with TGI at the level of 25.2% and 32.2%, respectively.
Thus, fusion protein of the invention exerted much stronger effect compared to TRAIL alone and standard chemotherapy.

Claims (24)

We claim:
1. A fusion protein comprising: domain (a) which is a functional fragment of the sequence of soluble hTRAIL protein, which fragment begins with an amino acid in the range from hTRAIL95 to hTRAIL121, inclusive, and ends with the amino acid at the position hTRAIL281, or a g of said functional fragment having at least 70% sequence identity, preferably 85% identity, and at least one domain (b) which is the sequence of a cytolytic effector peptide with an amphipathic alpha-helix conformation forming pores in the cell membrane, wherein the sequence of the domain (b) is attached at the C-terminus and/or N- terminus of domain (a).
2. The fusion protein according to claim 1, wherein domain (a) is selected from the group ting of hTRAIL95-281, hTRAIL114-281, 115-281, 1 16-281, hTRAIL1 19-281, and hTRAIL1 21-281.
3. The fusion protein according to claim 1 to 2, in which domain (b) is ed from the group consisting of: - piLosuLin-1 of SEQ. No. 36, - piLosulin-5 of SEQ. No. 37, - 14-amino acids tic Lytic peptide of SEQ. No. 41, - 27-amino acids e FFhCAP18 of SEQ. No. 43, - BAMP-28 peptide of SEQ. No. 44, - analogue of isoform C of lytic peptide from Entamoeba histolytica of SEQ. No. 45, - analogue of isoform A of lytic peptide from Entamoeba histolytica SEQ. No. 46, - analogue of isoform B of Lytic peptide from Entamoeba histolytica of SEQ. No. 47, - fragment of HA2 domain of influenza virus haemagglutinin of SEQ. No. - active fragment of human perforin of SEQ. No. 54, - parasporin-2 z us thuringensis of SEQ. No. 55, - fusion n comprising synthetic lytic peptide with KLLK motif and a peptide being antagonist of PDGF receptor of SEQ. No. 125, - pleurocidin analogue of SEQ. No. 126, - pleurocidin analogue of SEQ. No. 127, and - tic lytic peptide of SEQ. No. 128.
4. The fusion protein according to any of the claims 1 to 3, which between domain (a) and domain (b) or between domains (b) contains domain (C) containing protease cleavage site, selected from a sequence recognized by metatloprotease MMP, a sequence recognized by urokinase uPA, and sequence recognized by furin and a sequence recognized by native furin.
5. The fusion protein according to claim 4, in which a ce recognized by metalloprotease MMP is Pro Leu Gly Leu Ala Gly, a sequence recognized by urokinase uPA is Arg Val Vat Arg, a sequence recognized by furin is Arg Lys Lys Arg, and a sequence ized by native furin is Arg His Arg Gin Pro Arg Gly Trp Glu Gin Leu or His Arg Gin Pro Arg Gly Trp Glu Gin.
6. The fusion n according to claim 4 or 5, in which domain (c) is a combination of sequence recognized by metalloprotease MMP and a sequence recognized by ase uPA located next to each other.
7. The fusion protein according to any of preceding claims, in which effector peptide of domain (b) is additionally connected with transporting domain (d), selected from the group consisting of: (dl) polyhistidine sequence orting through the cell membrane comprising 6, 7, 8, 9, 10 or 11 His residues, and (d2) polyarginine sequence transporting through a cell membrane, consisting of 6, 7, 8, 9, 10 or 11 Arg residues, (0) PD4 transporting sequence (protein transduction domain 4) Tyr Ala Arg Ala Ala Ala Arg Gin Ala Arg Ala, (d4) a transporting sequence consisting of transferrin receptor g sequence Thr His Arg Pro Pro Met Trp Ser Pro Val Trp Pro, and (6) PD5 transporting ce (protein transduction domain 5, TAT protein) Tyr Gly Arg Lys Lys Arg Arg Gin Arg Arg Arg, and combinations thereof.
8. The fusion protein according to claim 7, wherein sequence (d) is located at the C-terminus or N-terminus of the effector peptide domain (b).
9. The fusion protein according to claim 7, wherein transporting domain (d) is located n domain (b) and domain (C), or between domain (a) and domain (c), or between two domains (c).
10. The fusion n according to claim 7, wherein sequence (d) is located at the C-terminus of the fusion protein.
11. The fusion protein according to any one of claims 4 to 10, which between two (c) domains contains domain (d) which is a linker for ment of PEG molecule, selected from Ala Ser Gly Cys Gly Pro Glu Gly and Ala Ser Gly Cys Gly Pro Glu.
12. The fusion protein according to any one of claims 4 to 11, which additionally comprises a flexible steric linker between domains (a), (b) and/or (c).
13. The fusion protein according to claim 12, n the steric linker is selected from Gly Gly, Gly Gly Gly, Gly Ser Gly, Gly Gly Gly Gly Ser, Gly Gly Gly Gly Gly Ser, Gly Gly Ser Gly Gly, Gly Gly Gly Ser Gly Gly Gly, Gly Gly Gly Gly Ser Gly, Gly Gly Gly Ser GLy GLy Gly Gly Gly Ser, Gly Gly GLy Gly Ser Gly Gly Gly Gly, GLy Ser GLy Gly Gly Ser Gly Gly GLy, Cys Ala Ala Cys Ala Ala Ala Cys, Cys Ala Ala Ala Cys Ala Ala Cys, Ser GLy Gly, single glycine residue GLy, and single cysteine residue Cys, and combinations thereof.
14. The fusion protein according to claim 1, having the amino acid sequence selected from the group consisting of SEQ. No. 4; SEQ. No. 5; SEQ. No. 6; SEQ. No. 11; SEQ. No. 12; SEQ. No. 13; SEQ. No. 14; SEQ. No. 15; SEQ. No. 16; SEQ. No. 18; SEQ. No. 19; SEQ. No. 20; SEQ. No. 21; SEQ. No. 22; SEQ. No. 23; SEQ. No. 30; SEQ. No. 31; SEQ. No. 32, SEQ. No. 91; SEQ. No. 92; SEQ. No. 93; SEQ. No. 94; SEQ. No. 95; SEQ. No. 96; SEQ. No. 97, SEQ. No. 98; and SEQ. No. 102.
15. The fusion n according to any one of the preceding claims, which is a recombinant protein.
16. A polynucleotide, coding the fusion protein as defined in any one of claims 1 to 14.
17. The polynucleotide according to claim 16, optimized for genetic expression in E. coil.
18. The polynucleotide according to claim 17, selected from the group consisting of SEQ. No. 60; SEQ. No. 61; SEQ. No. 62; SEQ. No. 67; SEQ. No. 68; SEQ. No. 69; SEQ. No. 70; SEQ. No. 71; SEQ. No. 72; SEQ. No. 74; SEQ. No. 75; SEQ. No. 76; SEQ. No. 77; SEQ. No. 78; SEQ. No. 79; SEQ. No. 86; SEQ. No. 87; SEQ. No. 88; SEQ. No. 108; SEQ. No. 109; SEQ. No. 110; SEQ. No. 111; SEQ. No. 112; SEQ. No. 113; SEQ. No. 114, SEQ. No. 115; SEQ. No. 119.
19. An expression vector comprising polynucleotide according to any one of claims 16 to 18.
20. A host cell comprising the expression vector as defined in claim 19, wherein said host is not within a human body.
21. The host cell according to claim 20, which is an E. coil cell.
22. A pharmaceutical composition comprising as an active ingredient the fusion protein as d in any one of claims 1 to 15, in ation with a pharmaceutically acceptable carrier.
23. The ceutical composition according to claim 22 in a form for parenteral administration.
24. The fusion protein as defined in any one of claims 1 to 15 for use in the treatment of neoplastic diseases in mammals, including humans.
NZ627445A 2011-12-28 2012-12-22 Anticancer fusion protein NZ627445B2 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
PL397595A PL223487B1 (en) 2011-12-28 2011-12-28 Anti-tumor fusion protein
PLP.397595 2011-12-28
PCT/IB2012/057657 WO2013098755A2 (en) 2011-12-28 2012-12-22 Anticancer fusion protein

Publications (2)

Publication Number Publication Date
NZ627445A NZ627445A (en) 2016-01-29
NZ627445B2 true NZ627445B2 (en) 2016-05-03

Family

ID=

Similar Documents

Publication Publication Date Title
AU2012360086B2 (en) Anticancer fusion protein
KR101861872B1 (en) Anticancer fusion protein
US20150044162A1 (en) Anticancer fusion protein
US9175059B2 (en) Anticancer fusion protein comprising trail and a growth factor receptor inhibitor
US20140031283A1 (en) Anticancer fusion protein
NZ627445B2 (en) Anticancer fusion protein
NZ617353B2 (en) Anticancer fusion protein