US20110178026A1 - Ricin-like toxins for treatment of cancer - Google Patents

Ricin-like toxins for treatment of cancer Download PDF

Info

Publication number
US20110178026A1
US20110178026A1 US12/021,563 US2156308A US2011178026A1 US 20110178026 A1 US20110178026 A1 US 20110178026A1 US 2156308 A US2156308 A US 2156308A US 2011178026 A1 US2011178026 A1 US 2011178026A1
Authority
US
United States
Prior art keywords
nucleic acid
acid sequence
seq
chain
toxin
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US12/021,563
Inventor
Curtis Braun
Admir Purac
Thor Borgford
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
TWINSTRAND HOLDINGS Inc
Original Assignee
Twinstrand Therapeutics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Twinstrand Therapeutics Inc filed Critical Twinstrand Therapeutics Inc
Priority to US12/021,563 priority Critical patent/US20110178026A1/en
Assigned to TWINSTRAND THERAPEUTICS INC. reassignment TWINSTRAND THERAPEUTICS INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BRAUN, CURTIS, PURAC, ADMIR, BORGFORD, THOR
Assigned to TWINSTRAND HOLDINGS INC. reassignment TWINSTRAND HOLDINGS INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: TWINSTRAND THERAPEUTICS INC.
Publication of US20110178026A1 publication Critical patent/US20110178026A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/415Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from plants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the invention relates to proteins useful as therapeutics against unhealthy cells such as those which occur in inflammation and cancer.
  • the proteins contain A and B chains of a ricin-like toxin linked by a novel linker sequence that is specifically cleaved and activated by proteases specific to cancer.
  • cytotoxic proteins which may consist of one, two or several polypeptides or subunits. Those proteins having a single subunit may be loosely classified as Type I proteins. Many of the cytotoxins which have evolved two subunit structures are referred to as Type II proteins (Saelinger, C. B. in Trafficking of Bacterial Toxins (eds. Saelinger, C. B.) 1-13 (CRC Press Inc., Boca Raton, Fla., 1990). One subunit, the A chain, possesses the toxic activity whereas the second subunit, the B chain, binds cell surfaces and mediates entry of the toxin into a target cell. A subset of these toxins kill target cells by inhibiting protein biosynthesis.
  • bacterial toxins such as diphtheria toxin or Pseudomonas exotoxin inhibit protein synthesis by inactivating elongation factor 2.
  • Plant toxins such as ricin, abrin, and bacterial toxin Shiga toxin, inhibit protein synthesis by directly inactivating the ribosomes (Olsnes, S. & Phil, A. in Molecular action of toxins and viruses (eds. Cohen, P. & vanHeyningen, S.) 51-105 Elsevier Biomedical Press, Amsterdam, 1982).
  • Ricin derived from the seeds of Ricinus communis (castor oil plant), may be the most potent of the plant toxins. It is estimated that a single ricin A chain is able to inactivate ribosomes at a rate of 1500 ribosomes/minute. Consequently, a single molecule of ricin is enough to kill a cell (Olsnes, S. & Phil, A. in Molecular action of toxins and viruses (eds. Cohen, P. & vanHeyningen, S.) (Elsevier Biomedical Press, Amsterdam, 1982).
  • the ricin toxin is a glycosylated heterodimer consisting of A and B chains with molecular masses of 30,625 Da and 31,431 Da linked by a disulphide bond.
  • the A chain of ricin has an N-glycosidase activity and catalyzes the excision of a specific adenine residue from the 28S rRNA of eukaryotic ribosomes (Endo, Y. & Tsurugi, K. J., Biol. Chem. 262:8128 (1987)).
  • the B chain of ricin although not toxic in itself, promotes the toxicity of the A chain by binding to galactose residues on the surface of eukaryotic cells and stimulating receptor-mediated endocytosis of the toxin molecule (Simmons et al., Biol. Chem. 261:7912 (1986)).
  • the toxin molecule consisting of the A and B chains is internalized into the cell via clathrin-dependent or independent mechanisms, the greater reduction potential within the cell induces a release of the active A chain, eliciting its inhibitory effect on protein synthesis and its cytotoxicity (Emmanuel, F. et al., Anal. Biochem. 173: 134-141 (1988); Blum, J. S.
  • Protein toxins are initially produced in an inactive, precursor form.
  • Ricin is initially produced as a single polypeptide (preproricin) with an amino acid N-terminal presequence and 12 amino acid linker between the A and B chains.
  • the pre-sequence is removed during translocation of the ricin precursor into the endoplasmic reticulum (Lord, J. M., Eur. J. Biochem. 146:403-409 (1985) and Lord, J. M., Eur. J. Biochem. 146:411-416 (1985)).
  • the proricin is then translocated into specialized organelles called protein bodies where a plant protease cleaves the protein at a linker region between the A and B chains (Lord, J. M.
  • the ribosomes of the castor bean plant are themselves susceptible to inactivation by ricin A chain; however, as there is no cell surface galactose to permit B chain recognition the A chain cannot re-enter the cell.
  • the exact mechanism of A chain release and activation in target cell cytoplasm is not known (Lord, J. M. et al., FASAB journal 8:201-208 (1994)). However, it is known that for activation to take place the disulfide bond between the A and B chains must be reduced and, hence, the linkage between subunits broken.
  • Diphtheria toxin is produced by Corynebacterium diphtheriae as a 535 amino acid polypeptide with a molecular weight of approximately 58 kD (Greenfield, L. et al., Proc. Natl. Acad. Sci. USA 80:6853-6857 (1983); Pastan, I. et al., Annu. Rev. Biochem. 61:331-354 (1992); Collier, R. J. & Kandel, J., 1 . Biol. Chem. 246:1496-1503 (1971)). It is secreted as a single-chain polypeptide consisting of 2 functional domains.
  • the N-terminal domain contains the cytotoxic moiety whereas the C-terminal domain (B-chain) is responsible for binding to the cells and facilitates toxin endocytosis.
  • the mechanism of cytotoxicity for diphtheria toxin is based on ADP-ribosylation of EF-2 thereby blocking protein synthesis and producing cell death.
  • the 2 functional domains in diphtheria toxin are linked by an arginine-rich peptide sequence as well as a disulphide bond.
  • the arginine-rich peptide linker is cleaved by trypsin-like enzymes and the disulphide bond (Cys 186-201) is reduced.
  • the cytotoxic domain is subsequently translocated into the cytosol substantially as described above for ricin and elicits ribosomal inhibition and cytotoxicity.
  • Pseudomonas exotoxin is also a 66 kD single-chain toxin protein secreted by Pseudomonas aeruginosa with a similar mechanism of cytotoxicity to that of diphtheria toxin (Pastan, I. et al., Annu. Rev. Biochem. 61:331-354 (1992); Ogata, M. et al., J. Biol. Chem. 267:25396 25401 (1992); Vagil, M. L. et al., Infect. Immunol. 16:353-361 (1977)). Pseudomonas exotoxin consists of 3 conjoint functional domains.
  • the first domain I (amino acids 1-252) is responsible for cell binding and toxin endocytosis
  • a second domain II (amino acids 253-364) is responsible for toxin translocation from the endocytic vesicle to the cytosol
  • a third domain III (amino acids 400-613) is responsible for protein synthesis inhibition and cytotoxicity.
  • the liberation of the cytotoxic domain is effected by both proteolytic cleavage of a polypeptide sequence in the second domain (near Arg 279) and the reduction of the disulphide bond (Cys 265-287) in the endocytic vesicles.
  • the overall pathway to cytotoxicity is analogous to diphtheria toxin with the exception that the toxin translocation domain in Pseudomonas exotoxin is structurally distinct.
  • Class 2 ribosomal inhibitory proteins constitute other toxins possessing distinct functional domains for cytotoxicity and cell binding/toxin translocation which include abrin, modeccin, volkensin, (Sandvig, K. et al., Biochem. Soc. Trans. 21:707-711 (1993)) and mistle toe lectin (viscumin) (Olsnes, S. & Phil, A. in Molecular action of toxins and viruses (eds. Cohen, P. & vanHeyningen, S.) 51-105 Elsevier Biomedical Press, Amsterdam, 1982; Fodstad, et al. Canc. Res. 44: 862 (1984)).
  • Some toxins such as Shiga toxin and cholera toxin also have multiple polypeptide chains responsible for receptor binding and endocytosis.
  • the ricin gene has been cloned and sequenced, and the X-ray crystal structures of the A and B chains have been described (Rutenber, E. et al. Proteins 10:240-250 (1991); Weston et al., Mol. Bio. 244:410-422, 1994; Lamb and Lord, Eur. J. Biochem. 14:265 (1985); Halling, K. et al. Nucleic Acids Res. 13:8019 (1985)).
  • the genes for diptheria toxin and Pseudomonas exotoxin have been cloned and sequenced, and the 3-dimensional structures of the toxin proteins have been elucidated and described (Columblatti, M. et al., J.
  • Bacterial toxins such as Pseudomonas exotoxin and subunit A of diphtheria toxin; dual chain ribosomal inhibitory plant toxins such as ricin, and single chain ribosomal inhibitory proteins such as trichosanthin and pokeweed antiviral protein have been used for the elimination of HIV infected cells (Olson et al., AIDS Res. and Human Retroviruses 7:1025-1030 (1991)).
  • the high toxicity of these toxins for mammalian cells combined with a lack of specificity of action poses a major problem to the development of pharmaceuticals incorporating the toxins, such as immunotoxins.
  • An immunotoxin is a conjugate of a specific cell binding component, such as a monoclonal antibody or growth factor and the toxin in which the two protein components are covalently linked. Generally, the components are chemically coupled. However, the linkage may also be a peptide or disulfide bond.
  • the antibody directs the toxin to cell types presenting a specific antigen thereby providing a specificity of action not possible with the natural toxin.
  • Immunotoxins have been made both with the entire ricin molecule (i.e. both chains) and with the ricin A chain alone (Spooner et al., Mol. Immunol. 31:117-125, (1994)).
  • Immunotoxins made with the ricin dimer are more potent toxins than those made with only the A chain (IT-As).
  • IT-Rs The increased toxicity of IT-Rs is thought to be attributed to the dual role of the B chains in binding to the cell surface and in translocating the A chain to the cytosolic compartment of the target cell (Vitetta et al., Science 238:1098-1104 (1987); Vitetta & Thorpe, Seminars in Cell Biology 2:47-58 (1991)).
  • the presence of the B chain in these conjugates also promotes the entry of the immunotoxin into nontarget cells.
  • IT-As are more specific and safer to use than IT-Rs.
  • IT-Rs in the absence of the B chain the A chain has greatly reduced toxicity. Due to the reduced potency of IT-As as compared to IT-Rs, large doses of IT-As must be administered to patients. The large doses frequently cause immune responses and production of neutralizing antibodies in patients (Vitetta et al., Science 238:1098-1104 (1987)). IT-As and IT-Rs both suffer from reduced toxicity as the A chain is not released from the conjugate into the target cell cytoplasm.
  • a number of immunotoxins have been designed to recognize antigens on the surfaces of tumour cells and cells of the immune system (Pastan et al., Annals New York Academy of Sciences 758:345-353 (1995)).
  • a major problem with the use of such immunotoxins is that the antibody component is its only targeting mechanism and the target antigen is often found on non-target cells (Vitetta et al., Immunology Today 14:252-259 (1993)).
  • the preparation of a suitable specific cell binding component may be problematic.
  • antigens specific for the target cell may not be available and many potential target cells and infective organisms can alter their antigenic make up rapidly to avoid immune recognition.
  • the lack of specificity of the immunotoxins may severely limit their usefulness as therapeutics for the treatment of cancer and infectious diseases.
  • ⁇ -Hemolysin does not inhibit protein synthesis but is a heptameric transmembrane pore which acts as a channel to allow leakage of molecules up to 3 kD thereby disrupting the ionic balances of the living cell.
  • the ⁇ -hemolysin activation domain is likely located on the outside of the target cell (for activation by extracellular proteases).
  • the triggering mechanism in the disclosed hemolysin precursor does not involve the intracellular proteolytic cleavage of 2 functionally distinct domains.
  • the proteases used for the a-hemolysin activation are ubitquitiously secreted extracellular proteases and toxin activation would not be confined to activation in the vicinity of diseased cells. Such widespread activation of the toxin does not confer target specificity and limits the usefulness of said a-hemolysin toxin as therapeutics due to systemic toxicity.
  • cathepsin is a family of serine, cysteine or aspartic endopeptidases and exopeptidases which has been implicated to play a primary role in cancer metastasis (Schwartz, M. K., Clin. Chim. Acta 237:67-78 (1995); Spiess, E. et al., J. Histochem. Cytochem. 42:917-929 (1-994); Scarborough, P. E. et al., Protein Sci. 2:264276 (1993); Sloane, B. F. et al., Proc. Natl. Acad. Sci.
  • Matrix metalloproteinases are zinc-dependent proteinases consisting of collagenases, matrilysin, stromelysins, stromelysin-like MMPs, gelatinases, macrophage elastase, membrane-type MMPs (MT-MMPs) (Krane, S. M., Ann, N.Y. Acad. Sci. 732:1-10 (1994); Woessner, J. F., Ann, N.Y. Acad. Sci.
  • MMPs or matrixins zinc-dependent proteinases consisting of collagenases, matrilysin, stromelysins, stromelysin-like MMPs, gelatinases, macrophage elastase, membrane-type MMPs (MT-MMPs) (Krane, S. M., Ann, N.Y. Acad. Sci. 732:1-10 (1994); Woessner, J. F., Ann, N.Y. Ac
  • TIMPs tissue inhibitors of metalloproteinases
  • the expression of MMP genes is reported to be activated in inflammatory disorders (e.g. rheumatoid arthritis) and malignancy.
  • the present inventors have prepared novel recombinant toxic proteins which are specifically toxic to diseased cells but do not depend for their specificity of action on a specific cell binding component.
  • the recombinant proteins toxins have an A chain of a ricin-like toxin linked to a B chain by a synthetic linker sequence which may be cleaved specifically by a protease localised in cells or tissues affected by a specific disease to liberate the toxic A chain thereby selectively inhibiting or destroying the diseased cells or tissues (WO 98/49311 published Nov. 5, 1998 which is incorporated herein by reference).
  • the present invention relates to novel linker sequences that can be used to prepare recombinant toxic proteins having an A chain of a ricin-like toxin linked to a B chain by the linker sequence.
  • the novel linker sequences of the invention are illustrated in FIGS. 1-18 .
  • the present invention provides a purified and isolated nucleic acid encoding a linker sequence comprising: the nucleic acid sequence of pAP301 as shown in FIG. 1A ; the nucleic acid sequence of pAP302 as shown in FIG. 2A ; the nucleic acid sequence of pAP303 as shown in FIG. 3A ; the nucleic acid sequence of pAP304 as shown in FIG. 4A ; the nucleic acid sequence of pAP305 as shown in FIG. 5A ; the nucleic acid sequence of pAP308 as shown in FIG. 6A ; the nucleic acid sequence of pAP309 as shown in FIG. 7A ; the nucleic acid sequence of pAP313 as shown in FIG.
  • nucleic acid sequence of pAP314 as shown in FIG. 9A ; the nucleic acid sequence of pAP315 as shown in FIG. 10A ; the nucleic acid sequence of pAP316 as shown in FIG. 11A ; the nucleic acid sequence of pAP318 as shown in FIG. 12A ; the nucleic acid sequence of pAP320 as shown in FIG. 13A ; the nucleic acid sequence of pAP321 as shown in FIG. 14A ; the nucleic acid sequence of pAP322 as shown in FIG. 15A ; the nucleic acid sequence of pAP323 as shown in FIG. 16A ; the nucleic acid sequence of pAP324 as shown in FIG. 17A ; and the nucleic acid sequence of pAP325 as shown in FIG. 18A .
  • the present invention provides a purified and isolated nucleic acid encoding a recombinant toxic protein comprising (a) a nucleotide sequence encoding an A chain of a ricin-like toxin, (b) a nucleotide sequence encoding a B chain of a ricin-like toxin and (c) a heterologous linker amino acid sequence, linking the A and B chains.
  • the linker sequence is not a native linker sequence of a ricin-like toxin, but rather a synthetic heterologous linker sequence containing a cleavage recognition site for a specific protease.
  • the A and or the B chain may be those of ricin.
  • specific protease means a protease in any cell wherein there is expression of the protease at levels greater than those found in a corresponding healthy cell.
  • examples of a specific protease include MMPs, preferably MMP-2, MMP-9, MMP-14, and MT1-MMPs, and UPA, as well as others found in inflammatory cells and malignant cells.
  • An inflammatory cell includes any cell involved in the inflammation process having a specific protease.
  • the recombinant toxic proteins employing the novel linker sequences of the present invention may be used to treat various forms of cells having specific proteases such as inflammatory disorders including rheumatoid arthritis, atherosclerotic cells, Crohn's disease, central nervous system disease as well as in cancer including, but not limited to, T- and B-cell lymphoproliferative diseases, ovarian cancer, pancreatic cancer, head and neck cancer, squamous cell carcinoma, gastrointestinal cancer, breast cancer, prostate, cancer and non small cell lung cancer.
  • the cleavage recognition site of the linker is the cleavage recognition site for a cancer-associated protease.
  • the amino acid sequence of the linker comprises the sequence of PAP301 shown in FIG. 1C ; the sequence of PAP302 shown in FIG. 2C ; the sequence of PAP303 shown in FIG. 3C ; the sequence of PAP304 shown in FIG. 4C ; the sequence of PAP305 shown in FIG. 5C ; the sequence of PAP308 shown in FIG. 6C ; the sequence of PAP309 shown in FIG. 7C ; the sequence of PAP316 shown in FIG. 11C ; the sequence of PAP318 shown in FIG. 12C ; the sequence of PAP323 shown in FIG. 16C ; the sequence of PAP324 shown in FIG. 17C ; and the sequence of PAP325 shown in FIG.
  • nucleic acid sequences of the recombinant toxic proteins containing ricin A and B chains with each of the linker sequences are shown in FIGS. 1B , 2 B, 3 B, 4 B, 5 B, 6 B, 7 B, 8 B, 9 B, 10 B, 11 B, 12 B, 13 B, 14 B, 15 B, 16 B, 17 B and 18 B.
  • the present invention also provides a plasmid incorporating the nucleic acid of the invention.
  • the present invention provides a baculovirus transfer vector incorporating the nucleic acid of the invention.
  • the present invention provides a recombinant protein comprising an A chain of a ricin-like toxin, a B chain of a ricin-like toxin and a heterologous linker amino acid sequence, linking the A and B chains, wherein the linker sequence contains a cleavage recognition site for a specific protease.
  • the A and/or the B chain may be those of ricin.
  • the present invention provides a recombinant protein comprising an A chain of a ricin-like toxin, a B chain of a ricin-like toxin and a heterologous linker amino acid sequence, linking the A and B chains, wherein the linker sequence contains a cleavage recognition site for a inflammatory disease specific protease.
  • the A and/or the B chain may be those of ricin.
  • the cleavage recognition site is the cleavage recognition site for an inflammation based protease substantially as described above.
  • the inflammation is rheumatoid arthritis, atherosclerotic cells, Crohn's disease, or central nervous system disease.
  • the present invention provides a recombinant protein comprising an A chain of a ricin-like toxin, a B chain of a ricin-like toxin and a heterologous linker amino acid sequence, linking the A and B chains, wherein the linker sequence contains a cleavage recognition site for a cancer-specific protease.
  • the A and/or the B chain may be those of ricin.
  • the cleavage recognition site is the cleavage recognition site for a cancer protease substantially as described above.
  • the cancer is T-cell or B-cell lymphoproliferative disease, ovarian cancer, pancreatic cancer, head and neck cancer, squamous cell carcinoma, gastrointestinal cancer, breast cancer, prostate cancer, non small cell lung cancer.
  • the invention provides a pharmaceutical composition for treating a cell, such as an inflammatory cell or cancer cell, having a specific protease, comprising a recombinant protein of the invention and a pharmaceutically acceptable carrier, diluent or excipient.
  • the invention provides a method of inhibiting or destroying a cell having a specific protease, such as an inflammatory cell or a cancer cell, comprising the steps of preparing a recombinant protein of the invention having a heterologous linker sequence which contains a cleavage recognition site for the specific protease, and administering the recombinant protein to the cells.
  • the inflammatory state is rheumatoid arthritis, atherosclerotic cells, Crohn's disease, or central nervous system disease.
  • the cancer is T-cell or B-cell lymphoproliferative disease, ovarian cancer, pancreatic cancer, head and neck cancer, squamous cell carcinoma, gastrointestinal cancer, breast cancer, prostate cancer, non small cell lung cancer.
  • the present invention also relates to a method of treating a cell having a specific protease such as an inflammatory cell or a cancer cell, wherein the cells affected by the condition and which have a specific protease, are treated by administering an effective amount of one or more recombinant proteins of the invention to an animal in need thereof.
  • a specific protease such as an inflammatory cell or a cancer cell
  • a process for preparing a pharmaceutical for treating a cell having a specific protease, such as an inflammatory cell or a cancer cell, wherein cells affected by condition have a specific protease
  • the steps for preparing the pharmaceutical comprising the steps of preparing a purified and isolated nucleic acid having a nucleotide sequence encoding an A chain of a ricin-like toxin, a B chain of a ricin-like toxin and a heterologous linker amino acid sequence, linking the A and B chains, wherein the linker sequence contains a cleavage recognition site for the specific protease; introducing the nucleic acid into a host cell; expressing the nucleic acid in the host cell to obtain a recombinant protein comprising an A chain of a ricin-like toxin, a B chain of a ricin-like toxin and a heterologous linker amino acid sequence, linking the A and B chains wherein the linker sequence contains the
  • FIG. 1A shows the nucleotide sequence of the MMP-9 linker region of pAP301 (SEQ ID NOS:1-4);
  • FIG. 1B shows the nucleotide sequence of the pAP301 insert containing ricin and the MMP-9 linker (SEQ ID NO:5);
  • FIG. 1C shows the amino acid sequence of the PAP301 linker and the wild type ricin linker (SEQ ID NOS:6-7);
  • FIG. 2A shows the nucleotide sequence of the MMP-9 30 linker region of pAP302 (SEQ ID NOS:8-11);
  • FIG. 2B shows the nucleotide sequence of the pAP302 insert containing ricin and the MMP-9 linker (SEQ ID NO:12);
  • FIG. 2C shows the amino acid sequence of the PAP302 linker and the wild type ricin linker (SEQ ID NOS:13-14);
  • FIG. 3A shows the nucleotide sequence of the MMP-9 linker region of pAP303 (SEQ ID NOS:15-18);
  • FIG. 3B shows the nucleotide sequence of the pAP303 insert containing ricin and the MMP-9 linker (SEQ ID NO:19);
  • FIG. 3C shows the amino acid sequence of the PAP303 linker and the wild type ricin linker (SEQ ID NOS:20-21);
  • FIG. 4A shows the nucleotide sequence of the MMP-9 linker region of pAP304 (SEQ ID NOS:22-25);
  • FIG. 4B shows the nucleotide sequence of the pAP304 insert containing ricin and the MMP-9 linker (SEQ ID NO:26);
  • FIG. 4C shows the amino acid sequence of the PAP304 linker and the wild type ricin linker (SEQ ID NOS:27-28);
  • FIG. 5A shows the nucleotide sequence of the MMP-9 linker region of pAP305 (SEQ ID NOS:29-32);
  • FIG. 5B shows the nucleotide sequence of the pAP305 insert containing ricin and the MMP-9 linker (SEQ ID NO:33);
  • FIG. 5C shows the amino acid sequence of the PAP305 linker and the wild type ricin linker (SEQ ID NOS:34-35);
  • FIG. 6A shows the nucleotide sequence of the MMP-9 linker region of pAP308 (SEQ ID NOS:36-39);
  • FIG. 6B shows the nucleotide sequence of the pAP308 insert containing ricin and the MMP-9 linker (SEQ ID NO:40);
  • FIG. 6C shows the amino acid sequence of the pAP308 linker and the wild type ricin linker (SEQ ID NOS:41-42);
  • FIG. 7A shows the nucleotide sequence of the MMP-9 linker region of pAP309 (SEQ ID NOS:43-46);
  • FIG. 7B shows the nucleotide sequence of the pAP309 insert containing ricin and the MMP-9 linker (SEQ ID NO:47);
  • FIG. 7C shows the amino acid sequence of the PAP309 linker and the wild type ricin linker (SEQ ID NOS:48-49);
  • FIG. 8A shows the nucleotide sequence of the UPA linker region of pAP313 (SEQ ID NOS:50-53);
  • FIG. 8B shows the nucleotide sequence of the pAP313 insert containing ricin and the UPA linker (SEQ ID NO:54);
  • FIG. 8C shows the amino acid sequence of the PAP313 linker and the wild type ricin linker (SEQ ID NOS:55-56);
  • FIG. 9A shows the nucleotide sequence of the UPA linker region of pAP314 (SEQ ID NOS:57-60);
  • FIG. 9B shows the nucleotide sequence of the pAP314 insert containing ricin and the UPA linker (SEQ ID NO:61);
  • FIG. 9C shows the amino acid sequence of the PAP314 linker and the wild type ricin linker (SEQ ID NOS:62-63);
  • FIG. 10A shows the nucleotide sequence of the UPA linker region of pAP315 (SEQ ID NOS:64-67);
  • FIG. 10B shows the nucleotide sequence of the pAP315 insert containing ricin and the UPA linker (SEQ ID NO:68);
  • FIG. 10C shows the amino acid sequence of the PAP315 linker and the wild type ricin linker (SEQ ID NOS:69-70);
  • FIG. 11A shows the nucleotide sequence of the MMP-9 linker region of pAP316 (SEQ ID NOS:71-74);
  • FIG. 11B shows the nucleotide sequence of the pAP316 insert containing ricin and the MMP-9 linker (SEQ ID NO:75);
  • FIG. 11C shows the amino acid sequence of the PAP316 linker and the wild type ricin linker (SEQ ID NOS:76-77);
  • FIG. 12A shows the nucleotide sequence of the MMP-9 linker region of pAP318 (SEQ ID NOS:78-81);
  • FIG. 12B shows the nucleotide sequence of the pAP318 insert containing ricin and the MMP-9 linker (SEQ ID NO:82);
  • FIG. 12C shows the amino acid sequence of the PAP318 linker and the wild type ricin linker (SEQ ID NOS:83-84);
  • FIG. 13A shows the nucleotide sequence of the UPA linker region of pAP320 (SEQ ID NOS:85-88);
  • FIG. 13B shows the nucleotide sequence of the pAP320 insert containing ricin and the UPA linker (SEQ ID NO:89);
  • FIG. 13C shows the amino acid sequence of the PAP320 linker and the wild type ricin linker (SEQ ID NOS:90-91);
  • FIG. 14A shows the nucleotide sequence of the UPA linker region of pAP321 (SEQ ID NOS:92-95);
  • FIG. 14B shows the nucleotide sequence of the pAP321 insert containing ricin and the UPA linker (SEQ ID NO:96);
  • FIG. 14C shows the amino acid sequence of the PAP321 linker and the wild type ricin linker (SEQ ID NOS:97-98);
  • FIG. 15A shows the nucleotide sequence of the UPA linker region of pAP322 (SEQ ID NOS:99-102);
  • FIG. 15B shows the nucleotide sequence of the pAP322 insert containing ricin and the UPA linker (SEQ ID NO:103);
  • FIG. 15C shows the amino acid sequence of the PAP322 linker and the wild type ricin linker (SEQ ID NOS:104-105);
  • FIG. 16A shows the nucleotide sequence of the MMP-9 linker region of pAP323 (SEQ ID NOS:106-109);
  • FIG. 16B shows the nucleotide sequence of the pAP323 insert containing ricin and the MMP-9 linker (SEQ ID NO:110);
  • FIG. 16C shows the amino acid sequence of the PAP323 linker and the wild type ricin linker (SEQ ID NOS:111-112);
  • FIG. 17A shows the nucleotide sequence of the MMP-9 linker region of pAP324 (SEQ ID NOS:113-116);
  • FIG. 17B shows the nucleotide sequence of the pAP324 insert containing ricin and the MMP-9 linker (SEQ ID NO:117);
  • FIG. 17C shows the amino acid sequence of the PAP324 linker and the wild type ricin linker (SEQ ID NOS:118-119);
  • FIG. 18A shows the nucleotide sequence of the MMP-9 linker region of pAP325 (SEQ ID NOS:120-123);
  • FIG. 18B shows the nucleotide sequence of the pAP325 insert containing ricin and the MMP-9 linker (SEQ ID NO:124);
  • FIG. 18C shows the amino acid sequence of the PAP325 linker and the wild type ricin linker (SEQ ID NOS:125-126);
  • FIG. 19 shows the cleavage products of an MMP-9 digestion of PAP323, PAP324 and PAP325;
  • FIG. 20 is a graph showing the treatment of human tumour A431 with PAP304
  • FIG. 21 is a graph showing the treatment of human tumour A431 with PAP305.
  • FIG. 22 is a graph showing a significant delay in tumor growth in the murine tumor model.
  • the present invention relates to isolated and purified nucleic acid molecules encoding linker sequences.
  • the present invention also relates to isolated and purified nucleic acid molecules encoding a recombinant toxic protein comprising (a) a nucleotide sequence encoding an A chain of a ricin-like toxin, (b) a nucleotide sequence encoding a B chain of a ricin-like toxin and (c) a nucleotide sequence encoding a linker amino acid sequence of the invention, linking the A and B chains.
  • the heterologous linker sequence contains a cleavage recognition site for a specific protease.
  • isolated and purified refers to a nucleic acid substantially free of cellular material or culture medium when produced by recombinant DNA techniques, or chemical precursors, or other chemicals when chemically synthesized.
  • An “isolated and purified” nucleic acid is also substantially free of sequences which naturally flank the nucleic acid (i.e. sequences located at the 5′ and 3′ ends of the nucleic acid) from which the nucleic acid is derived.
  • nucleic acid is intended to include DNA and RNA and can be either double stranded or single stranded.
  • linker sequence refers to an internal amino acid sequence within the protein encoded by a nucleic acid molecule of the invention which contains residues linking the A and B chain of a ricin-like toxin so as to render the A chain incapable of exerting its toxic effect, for example catalytically inhibiting translation of an eukaryotic ribosome.
  • the linker sequences of the invention are heterologous to the A and B chain of a ricin-like toxin. By heterologous is meant that the linker sequence is not a sequence native to the A or B chain of a ricin-like toxin or precursor thereof.
  • the linker sequence may be of a similar length to the linker sequence of a ricin-like toxin and should not interfere with the role of the B chain in cell binding and transport into the cytoplasm.
  • the linker sequence is cleaved the A chain becomes active or toxic.
  • the nucleic acid molecule of the invention encoding a recombinant toxic protein is cloned by subjecting a preproricin cDNA clone to site-directed mutagenesis in order to generate a series of variants differing only in the sequence between the A and B chains (linker region).
  • Oligonucleotides, corresponding to the extreme 5′ and 3′ ends of the preproricin gene are synthesized and used to PCR amplify the gene.
  • oligonucleotide primers are designed to flank the start and stop codons of the preproricin open reading frame.
  • the preproricin cDNA is amplified using the upstream primer Ricin-99 or Ricin-109 and the downstream primer Ricin1729C with Vent DNA polymerase (New England Biolabs) using standard procedures (Sambrook et al., Molecular Cloning: A Laboratory Manual, Second Edition, (Cold Spring Harbor Laboratory Press, 1989)).
  • the purified PCR fragment encoding the preproricin cDNA is, then ligated into an Eco RV-digested pBluescript II SK plasmid (Stratagene), and is used to transform competent XL1-Blue cells (Stratagene).
  • the cloned PCR product containing the putative preproricin gene is confirmed by DNA sequencing of the entire cDNA clone.
  • the preproricin cDNA clone is subjected to site directed mutagenesis; in order to generate a series of variants differing only in the sequence between the A and B chains (linker region).
  • the wild-type preproricin linker region is replaced with the heterogenous linker sequences that are cleaved by the various specific proteases.
  • the linker regions of the variants encode a cleavage recognition sequence for a specific protease.
  • the mutagenesis and cloning strategies used to generate a specific protease-sensitive linker variant are summarized in WO 98149311 to the present inventor. Briefly, the first step involves a DNA amplification using a set of mutagenic primers in combination with the two flanking primers Ricin-109Eco and Ricin1729C PstI. Restriction digested PCR fragments are gel purified and then ligated with PVL1393 which has been digested with Eco RI and PstI. Ligation reactions are used to transform competent XLI-Blue cells (Stratagene). Recombinant clones are identified by restriction digests of plasmid miniprep, DNA and the mutant linker sequences are confirmed by DNA sequencing.
  • nucleotide sequences of the novel linker sequences of the invention are as follows: the nucleic acid sequence of pAP301 is shown in FIG. 1A ; the nucleic acid sequence of pAP302 is shown in FIG. 2A ; the nucleic acid sequence of pAP303 is shown in FIG. 3A ; the nucleic acid sequence of pAP304 is shown in FIG. 4A ; the nucleic acid sequence of pAP305 is shown in FIG. 5A ; the nucleic acid sequence of pAP308 is shown in FIG. 6A ; the nucleic acid sequence of pAP309 is shown in FIG. 7A ; the nucleic acid sequence of pAP313 is shown in FIG.
  • nucleic acid sequence of pAP314 is shown in FIG. 9A ; the nucleic acid sequence of pAP315 is shown in FIG. 10A ; the nucleic acid sequence of pAP316 is shown in FIG. 11A ; the nucleic acid sequence of pAP318 is shown in FIG. 12A ; the nucleic acid sequence of pAP320 is shown in FIG. 13A ; the nucleic acid sequence of pAP321 is shown in FIG. 14A ; the nucleic acid sequence of pAP322 is shown in FIG. 15A ; the nucleic acid sequence of pAP323 is shown in FIG. 16A ; the nucleic acid sequence of pAP324 is shown in FIG. 17A ; and the nucleic acid sequence of pAP325 is shown in FIG. 18A .
  • the nucleic acid molecule encoding a recombinant protein of the invention has sequences encoding an A chain of a ricin-like toxin, a B chain of a ricin-like toxin and a heterologous linker sequence containing a cleavage recognition site for a specific protease as described above.
  • the nucleotide sequences encoding the recombinant proteins of the invention are shown in FIGS. 1B-18B .
  • the nucleic acid may be expressed to provide a recombinant protein having an A chain of a ricin-like toxin, a B chain of a ricin-like toxin and a heterologous linker sequence containing a cleavage recognition site for a specific protease.
  • the nucleic acid molecule may comprise the A and/or B chain of ricin.
  • the ricin gene has been cloned and sequenced, and the X-ray crystal structures of the A and B chains are published (Rutenber, E., et al. Proteins 10:240-250 (1991); Weston et al., Mol. Biol. 244:410-422 (1994); Lamb and Lord, Eur. J. Biochem. 14:265 (1985); Halling, K., et al., Nucleic Acids Res. 13:8019 (1985)).
  • the invention includes nucleic acid molecules encoding truncations of A and B chains of ricin-like proteins and analogs and homologs of A and B chains of ricin-like proteins and truncations thereof (i.e., ricin-like proteins), as described herein. It will further be appreciated that variant forms of the nucleic acid molecules of the invention which arise by alternative splicing of an mRNA corresponding to a cDNA of the invention are encompassed by the invention.
  • Another aspect of the invention provides a nucleotide sequence which hybridizes under high stringency conditions to a nucleotide sequence encoding the A and/or B chains of a ricin-like protein.
  • Appropriate stringency conditions which promote DNA hybridization are known to those skilled in the art, or can be found in Current Protocols in Molecular Biology, John Wiley & Sons, N.Y. (1989), 6.3.1 6.3.6.
  • 6.0 ⁇ sodium chloride/sodium citrate (SSC) at about 45° C., followed by a wash of 2.0 ⁇ SSC at 50° C. may be employed.
  • the stringency may be selected based on the conditions used in the wash step.
  • the salt concentration in the wash step can be selected from a high stringency of about 0.2 ⁇ SSC at 50° C.
  • the temperature in the wash step can be at high stringency conditions, at about 65° C.
  • the nucleic acid molecule may comprise the A and/or B chain of a ricin-like toxin.
  • Methods for cloning ricin-like toxins are known in the art and are described, for example, in E.P. 466,222.
  • Sequences encoding ricin or ricin-like A and B chains may be obtained by selective amplification of a coding region, using sets of degenerative primers or probes for selectively amplifying the coding region in a genomic or cDNA library.
  • Appropriate primers may be selected from the nucleic acid sequence of A and B chains of ricin or ricin-like toxins.
  • oligonucleotide primers from the nucleotide sequences for use in PCR.
  • Suitable primers may be selected from the sequences encoding regions of ricin-like proteins which are highly conserved, as described for example in U.S. Pat. No. 5,101,025 and E.P. 466,222.
  • a nucleic acid can be amplified from cDNA or genomic DNA using these oligonucleotide primers and standard PCR amplification techniques.
  • the nucleic acid so amplified can be cloned into an appropriate vector and characterized by DNA sequence analysis.
  • cDNA may be prepared from mRNA, by isolating total cellular mRNA by a variety of techniques, for example, by using the guanidinium-thiocyanate extraction procedure of Chirgwin et al. ( Biochemistry 18, 5294-5299 (1979)).
  • cDNA is then synthesized from the mRNA using reverse transcriptase (for example, Moloney MLV reverse transcriptase available from Gibco/BRL, Bethesda, Md., or AMV reverse transcriptase available from Seikagaku America, Inc., St. Louis, Fla.).
  • reverse transcriptase for example, Moloney MLV reverse transcriptase available from Gibco/BRL, Bethesda, Md., or AMV reverse transcriptase available from Seikagaku America, Inc., St. Russia, Fla.
  • a sequence containing a cleavage recognition site for a specific protease may be selected based on the disease or condition which is to be targeted by the recombinant protein.
  • the cleavage recognition site may be selected from sequences known to encode a cleavage recognition site specific proteases of the disease or condition to be treated. Sequences encoding cleavage recognition sites may be identified by testing the expression product of the sequence for susceptibility to cleavage by the respective protease.
  • a polypeptide containing the suspected cleavage recognition site may be incubated with a specific protease and the amount of cleavage product determined (Dilannit, 1990, J. Biol. Chem. 285: 17345-17354 (1990)).
  • the specific protease may be prepared by methods known in the art and used to test suspected cleavage recognition sites.
  • the nucleic acid molecule of the invention may be prepared by site directed mutagenesis.
  • the cleavage site of a specific protease may be prepared by site directed mutagenesis of the homologous linker sequence of a proricin-like toxin. Procedures for cloning proricin-like genes, encoding a linker sequence are described in EP 466,222. Site directed mutagenesis may be accomplished by DNA amplification of mutagenic primers in combination with flanking primers.
  • the nucleic acid molecule of the invention may also encode a fusion protein.
  • a sequence encoding a heterologous linker sequence containing a cleavage recognition site for a specific protease may be cloned from a cDNA or genomic library or chemically synthesized based on the known sequence of such cleavage sites.
  • the heterologous linker sequence may then be fused in frame with the sequences encoding the A and B chains of the ricin-like toxin for expression as a fusion protein.
  • a nucleic acid molecule encoding a fusion protein may contain a sequence encoding an A chain and a B chain from the same ricin-like toxin or the encoded A and B chains may be from different toxins.
  • the A chain may be derived from ricin and the B chain may be derived from abrin.
  • a protein may also be prepared by chemical conjugation of the A and B chains and linker sequence using conventional coupling agents for covalent attachment.
  • RNA nucleic acid molecule of the invention which is RNA
  • a cDNA can be cloned downstream of a bacteriophage promoter, (e.g. a T7 promoter) in a vector, cDNA can be transcribed in vitro with T7 polymerase, and the resultant RNA can be isolated by standard techniques.
  • a bacteriophage promoter e.g. a T7 promoter
  • the amino acid sequence of the linker is selected from: the amino acid sequence of PAP301 as shown in FIG. 1C ; the amino acid sequence of PAP302 as shown in FIG. 2C ; the amino acid sequence of PAP303 as shown in FIG. 3C ; the amino acid sequence of PAP304 as shown in FIG. 4C ; the amino acid sequence of PAP305 as shown in FIG. 5C ; the amino acid sequence of PAP308 as shown in FIG. 6C ; the amino acid sequence of PAP309 as shown in FIG. 7C ; the amino acid sequence of PAP313 as shown in FIG. 8C ; the amino acid sequence of PAP314 as shown in FIG.
  • the present invention also provides recombinant proteins which incorporate the A and B chains of a ricin-like toxin linked by a heterologous linker sequence containing a cleavage recognition site for a specific protease as described above. It is an advantage of the recombinant proteins of the invention that they are non-toxic until the A chain is liberated from the B chain by specific cleavage of the linker by the target specific protease.
  • the recombinant protein may be used to specifically target for example, cancer cells in the absence of additional specific cell-binding components to target cancer cells. It is a further advantage that the specific protease cleaves the heterologous linker intracellularly thereby releasing the toxic A chain directly into the cytoplasm of the target cell. As a result, said cells are specifically targeted and normal cells are not directly exposed to the activated free A chain.
  • Ricin is a plant derived ribosome inhibiting protein which blocks protein synthesis in eukaryotic cells.
  • Ricin may be derived from the seeds of Ricinus communis (castor oil plant).
  • the ricin toxin is a glycosylated heterodimer with A and B chain molecular masses of 30,625 Da and 31,431 Da respectively.
  • the A chain of ricin has an N-glycosidase activity and catalyzes the excision of a specific adenine residue from the 28S rRNA of eukaryotic ribosomes (Endo, Y; & Tsurugi, K. J. Biol. Chem. 262:8128 (1987)).
  • the B chain of ricin although not toxic in itself, promotes the toxicity of the A chain by binding to galactose residues on the surface of eukaryotic cells and stimulating receptor-mediated endocytosis of the toxin molecule (Simmons et al., Biol. Chem. 261:7912 (1986)).
  • All protein toxins are initially produced in an inactive, precursor form.
  • Ricin is initially produced as a single polypeptide (preproricin) with a 35 amino acid N-terminal presequence and 12 amino acid linker between the A and B chains.
  • the pre-sequence is removed during translocation of the ricin precursor into the endoplasmic reticulum (Lord, J. M., Eur. J. Biochem. 146:403-409 (1985) and Lord, J. M., Eur. J. Biochem. 146:411-416 (1985)).
  • the proricin is then translocated into specialized organelles called protein bodies where a plant protease cleaves the protein at a linker region between the A and B chains (Lord, J.
  • Ricin-like proteins include, but are not limited to, bacterial, fungal and plant toxins which have A and B chains and inactivate ribosomes and inhibit protein synthesis.
  • the A chain is an active polypeptide subunit which is responsible for the pharmacologic effect of the toxin. In most cases the active component of the A chain is an enzyme.
  • the B chain is responsible for binding the toxin to the cell surface and is thought to facilitate entry of the A chain into the cell cytoplasm.
  • the A and B chains in the mature toxins are linked by disulfide bonds.
  • the toxins most similar in structure to ricin are plant toxins which have one A chain and one B chain. Examples of such toxins include abrin which may be isolated from the seeds of Abrus precatorius , modeccin, volkensin and viscumin.
  • Ricin-like bacterial proteins include diphtheria toxin, which is produced by Corynebacterium diphtheriae, Pseudomonas exotoxin and cholera toxin. It will be appreciated that the term ricin-like toxins is also intended to include the A chain of those toxins which have only an A chain.
  • the recombinant proteins of the invention could include the A chain of these toxins conjugated to, or expressed as, a recombinant protein with the B chain of another toxin. Examples of plant toxins having only an A chain include trichosanthin, MMC and pokeweed antiviral proteins, dianthin 30, dianthin 32, crotin II, curcin 11 and wheat germ inhibitor.
  • Examples of fungal toxins having only an A chain include alpha-sarcin, restrictocin, mitogillin, enomycin, phenomycin.
  • Examples of bacterial toxins having only an A chain include cytotoxin from Shigella dysenteriae and related Shiga-like toxins. Recombinant trichosanthin and the coding sequence thereof is disclosed in U.S. Pat. Nos. 5,101,025 and 5,128,460.
  • the recombinant protein of the invention may contain only that portion of the A chain which is necessary for exerting its cytotoxic effect.
  • the first 30 amino acids of the ricin A chain may be removed resulting in a truncated A chain which retains toxic activity.
  • the truncated ricin or ricin-like A chain may be prepared by expression of a truncated gene or by proteolytic degradation, for example with Nagarase (Funmatsu et al., Jap. J. Med. Sci. Biol. 23:264-267 (1970)).
  • the recombinant protein of the invention may contain only that portion of the B chain necessary for galactose recognition, cell binding and transport into the cell cytoplasm.
  • Truncated B chains are described for example in E.P. 145,111.
  • the A and B chains may be glycosylated or non-glycosylated.
  • Glycosylated A and B chains may be obtained by expression in the appropriate host cell capable of glycosylation.
  • Non-glycosylated chains may be obtained by expression in nonglycosylating host cells or by treatment to remove or destroy the carbohydrate moieties.
  • the proteins of the invention may be prepared using recombinant DNA methods. Accordingly, the nucleic acid molecules of the present invention may be incorporated in a known manner into an appropriate expression vector which ensures good expression of the protein.
  • Possible expression vectors include but are not limited to cosmids, plasmids, or modified viruses (e.g. replication defective retroviruses, adenoviruses and adeno-associated viruses), so long as the vector is compatible with the host cell used.
  • the expression vectors are “suitable for transformation of a host cell”, which means that the expression vectors contain a nucleic acid molecule of the invention and regulatory sequences selected on the basis of the host cells to be used for expression, which is operatively linked to the nucleic acid molecule. Operatively linked is intended to mean that the nucleic acid is linked to regulatory sequences in a manner which allows expression of the nucleic acid.
  • the invention therefore contemplates a recombinant expression vector of the invention containing a nucleic acid molecule of the invention, or a fragment thereof, and the necessary regulatory sequences for the transcription and translation of the inserted protein-sequence.
  • Suitable regulatory sequences may be derived from a variety of sources, including bacterial, fungal, viral, mammalian, or insect genes (For example, see the regulatory sequences described in Goeddel, Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, Calif. (1990). Selection of appropriate regulatory sequences is dependent on the host cell chosen as discussed below, and may be readily accomplished by one of ordinary skill in the art. Examples of such regulatory sequences include: a transcriptional promoter and enhancer or RNA polymerase binding sequence, a ribosomal binding sequence, including a translation initiation signal.
  • sequences such as an origin of replication, additional DNA restriction sites, enhancers, and sequences conferring inducibility of transcription may be incorporated into the expression vector. It will also be appreciated that the necessary regulatory sequences may be supplied by the native A and B chains and/or its flanking regions.
  • the recombinant expression vectors of the invention may also contain a selectable marker gene which facilitates the selection of host cells transformed or transfected with a recombinant molecule of the invention.
  • selectable marker genes are genes encoding a protein such as G418 and hygromycin which confer resistance to certain drugs, ⁇ -galactosidase, chloramphenicol acetyltransferase, firefly luciferase, or an immunoglobulin or portion thereof such as the Fc portion of an immunoglobulin preferably IgG.
  • selectable marker gene Transcription of the selectable marker gene is monitored by changes in the concentration of the selectable marker protein such as ⁇ -galactosidase, chloramphenicol acetyltransferase, or firefly luciferase. If the selectable marker gene encodes a protein conferring antibiotic resistance such as neomycin resistance transformant cells can be selected with G418. Cells that have incorporated the selectable marker gene will survive, while the other cells die. This makes it possible to visualize and assay for expression of recombinant expression vectors of the invention and in particular to determine the effect of a mutation on expression and phenotype. It will be appreciated that selectable markers can be introduced on a separate vector from the nucleic acid of interest.
  • the recombinant expression vectors may also contain genes which encode a fusion moiety which provides increased expression of the recombinant protein; increased solubility of the recombinant protein; and aid in the purification of the target recombinant protein by acting as a ligand in affinity purification.
  • a proteolytic cleavage site may be added to the target recombinant protein to allow separation of the recombinant protein from the fusion moiety subsequent to purification of the fusion protein.
  • Typical fusion expression vectors include pGEX (Amrad Corp., Melbourne, Australia), pMal (New England Biolabs, Beverly, Mass.) and pRIT5 (Pharmacia, Piscataway, N.J.) which fuse glutathione S-transferase (GST), maltose E binding protein, or protein A, respectively, to the recombinant protein.
  • GST glutathione S-transferase
  • maltose E binding protein or protein A, respectively, to the recombinant protein.
  • Recombinant expression vectors can be introduced into host cells to produce a transformed host cell.
  • the term “transformed host cell” is intended to include prokaryotic and eukaryotic cells which have been transformed or transfected with a recombinant expression vector of the invention.
  • the terms “transformed with”, “transfected with”, “transformation” and “transfection” are intended to encompass introduction of nucleic acid (e.g. a vector) into a cell by one of many possible techniques known in the art.
  • Prokaryotic cells can be transformed with nucleic acid by, for example, electroporation or calcium-chloride mediated transformation.
  • Nucleic acid can be introduced into mammalian cells via conventional techniques such as calcium phosphate or calcium chloride co-precipitation, DEAE-dextran mediated transfection, lipofectin, electroporation or microinjection. Suitable methods for transforming and transfecting host cells can be found in Sambrook et al. (Molecular Cloning: A Laboratory Manual, 2nd Edition, Cold Spring Harbor Laboratory press (1989)), and other laboratory textbooks.
  • Suitable host cells include a wide variety of prokaryotic and eukaryotic host cells.
  • the proteins of the invention may be expressed in bacterial cells such as E. coli , insect cells (using baculovirus), yeast cells or mammalian cells.
  • Other suitable host cells can be found in Goeddel, Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, Calif. (1991).
  • bacterial host cells suitable for carrying out the present invention include E. coli, B. subtilis, Salmonella typhimurium , and various species within the genus Pseudomonas, Streptomyces , and Staphylococcus , as well as many other bacterial species well known to one of ordinary skill in the art.
  • Suitable bacterial expression vectors preferably comprise a promoter which functions in the host cell, one or more selectable phenotypic markers, and a bacterial origin of replication.
  • Representative promoters include the ⁇ -lactamase (penicillinase) and lactose promoter system (see Chang et al., Nature 275:615 (1978)), the trp promoter (Nichols and Yanofsky, Meth in Enzymology 101:155, (1983) and the tac promoter (Russell et al., Gene 20:231, (1982)).
  • Representative selectable markers include various antibiotic resistance markers such as the kanamycin or ampicillin resistance genes.
  • Suitable expression vectors include but are not limited to bacteriophages such as lambda derivatives or plasmids such as pBR322 (Bolivar et al., Gene 2:9 S, (1977)), the pUC plasmids pUC18, pUC19, pUC118, pUC119 (see Messing, Meth in Enzymology 101:20-77, 1983 and Vieira and Messing, Gene 19:259-268 (1982)), and pNH8A, pNH16a, pNH18a, and Bluescript M13 (Stratagene, La Jolla, Calif.).
  • bacteriophages such as lambda derivatives or plasmids such as pBR322 (Bolivar et al., Gene 2:9 S, (1977))
  • the pUC plasmids pUC18, pUC19, pUC118, pUC119 see Messing, Meth in Enzymology 101:20-77, 1983
  • Typical fusion expression vectors which may be used are discussed above, e.g. pGEX (Amrad Corp., Melbourne, Australia), pMAL (New England Biolabs, Beverly, Mass.) and pRITS (Pharmacia, Piscataway, N.J.).
  • inducible non-fusion expression vectors include pTrc (Arnann et al., Gene 69:301-315 (1988)) and pET 11d (Studier et al., Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, Calif., 60-89 (1990)).
  • Yeast and fungi host cells suitable for carrying out the present invention include, but are not limited to Saccharomyces cerevisae , the genera Pichia or Kluyveromyces and various species of the genus Aspergillus .
  • yeast S. cerivisae examples include pYepSec1 (Baldari. et al., Embo J. 6:229-234 (1987)), pMFa (Kurjan and Herskowitz, Cell 30:933-943 (1982)), pJRY88 (Schultz et al., Gene 54:113-123 (1987)), and pYES2 (Invitrogen Corporation, San Diego, Calif.).
  • Mammalian cells suitable for carrying out the present invention include, among others: COS (e.g., ATCC No. CRL 1650 or 1651), BHK (e.g. ATCC No. CRL 6281), CHO (ATCC No. CCL 61), HeLa (e.g., ATCC No. CCL 2), 293 (ATCC No. 1573) and NS-1 cells.
  • Suitable expression vectors for directing expression in mammalian cells generally include a promoter (e.g., derived from viral material such as polyoma, Adenovirus 2, cytomegalovirus and Simian Virus 40), as well as other transcriptional and translational control sequences. Examples of mammalian expression vectors include pCDM8 (Seed, B., Nature 329:840 (1987)) and pMT2PC (Kaufman et al., EMBO J. 6:187-195 (1987)).
  • promoters, terminators, and methods for introducing expression vectors of an appropriate type into plant, avian, and insect cells may also be readily accomplished.
  • the proteins of the invention may be expressed from plant cells (see Sinkar et al., J. Biosci (Bangalore) 11:47-58 (1987), which reviews the use of Agrobacterium rhizogenes vectors; see also Zambryski et al., Genetic Engineering, Principles and Methods, Hollaender and Setlow (eds.), Vol. VI, pp. 253-278, Plenum Press, New York (1984), which describes the use of expression vectors for plant cells, including, among others, PAPS2022, PAPS2023, and PAPS2034)
  • Insect cells suitable for carrying out the present invention include cells and cell lines from Bombyx, Trichoplusia or Spodotera species.
  • Baculovirus vectors available for expression of proteins in cultured insect cells include the pAc series (Smith et al., Mol. Cell. Biol. 3:2156-2165 (1983)) and the pVL series (Lucklow, V. A., and Summers, M. D., Virology 170:31-39 (1989)).
  • Some baculovirus-insect cell expression systems suitable for expression of the recombinant proteins of the invention are described in PCT/US/02442.
  • the proteins of the invention may also be expressed in non-human transgenic animals such as, rats, rabbits, sheep and pigs (Hammer et al. Nature 315:680-683 (1985); Palmiter et al. Science 222:809-814 (1983); Brinster et al. Proc. Natl. Acad. Sci. USA 82:4438-4442 (1985); Palmiter and Brinster Cell 41:343-345 (1985) and U.S. Pat. No. 4,736,866).
  • non-human transgenic animals such as, rats, rabbits, sheep and pigs (Hammer et al. Nature 315:680-683 (1985); Palmiter et al. Science 222:809-814 (1983); Brinster et al. Proc. Natl. Acad. Sci. USA 82:4438-4442 (1985); Palmiter and Brinster Cell 41:343-345 (1985) and U.S. Pat. No. 4,736,866).
  • the proteins of the invention may also be prepared by chemical synthesis using techniques well known in the chemistry of proteins such as solid phase synthesis (Merrifield, J. Am. Chem. Assoc. 85:2149-2154 (1964)) or synthesis in homogenous solution (Houbenweyl, Methods of Organic Chemistry, ed. E. Wansch, Vol. 15 I and II, Thieme, Stuttgart (1987)).
  • the present invention also provides proteins comprising an A chain of a ricin-like toxin, a B chain of a ricin-like toxin and a heterologous linker amino acid sequence linking the A and B chains, wherein the linker sequence contains a cleavage recognition site for a specific protease.
  • a protein could be prepared other than by recombinant means, for example by chemical synthesis or by conjugation of A and B chains and a linker sequence isolated and purified from their natural plant, fungal or bacterial source.
  • Such A and B chains could be prepared having the glycosylation pattern of the native ricin-like toxin.
  • N-terminal or C-terminal fusion proteins comprising the protein of the invention conjugated with other molecules, such as proteins may be prepared by fusing, through recombinant techniques.
  • the resultant fusion proteins contain a protein of the invention fused to the selected protein or marker protein as described herein.
  • the recombinant protein of the invention may also be conjugated to other proteins by known techniques.
  • the proteins may be coupled using heterobifunctional thiol-containing linkers as described in WO 90/10457, N-succinimidyl-3-(2-pyridyldithio-proprionate) or N-succinimidyl-5 thioacetate.
  • proteins which may be used to prepare fusion proteins or conjugates include cell binding proteins such as immunoglobulins, hormones, growth factors, lectins, insulin, low density lipoprotein, glucagon, endorphins, transferrin, bombesin, asialoglycoprotein glutathione-S-transferase (GST), hemagglutinin (HA), and truncated myc.
  • cell binding proteins such as immunoglobulins, hormones, growth factors, lectins, insulin, low density lipoprotein, glucagon, endorphins, transferrin, bombesin, asialoglycoprotein glutathione-S-transferase (GST), hemagglutinin (HA), and truncated myc.
  • MMPs or matrixins matrix metalloproteinases
  • inflammatory disorders e.g. rheumatoid arthritis
  • malignancy e.g. rheumatoid arthritis
  • urokinase type plasminogen activator in inflammatory disorders such a rheumatoid arthritis
  • osteoarthritis Pap, G. et al., 2000
  • atherosclerotic cells infalkenberg, M., et al., 1998) Crohn's disease (Desreumaux P, et al.
  • the recombinant proteins of the invention may be used to specifically inhibit or destroy cells that contain a specific protease that can cleave the linker sequence of the recombinant protein. More particularly, the recombinant proteins of the invention may be used to specifically inhibit or destroy cancer cells that contain a protease that can cleave the linker sequence of the recombinant protein.
  • the recombinant proteins of the invention have specificity for cells that contain a specific protease, including those of inflammatory disorders and cancer cells, without the need for a cell binding component.
  • the ricin-like B chain of the recombinant proteins recognize galactose moieties on the cell surface and ensure that the protein is taken up by, for example, a cancer cell and released into the cytoplasm.
  • cleavage of the heterologous linker would not occur in the absence of the specific protease, and the A chain will remain inactive bound to the B chain.
  • the specific protease will cleave the cleavage recognition site in the linker thereby releasing the toxic A chain.
  • the present invention provides a method of inhibiting or destroying cells having a specific protease, for examples inflammatory cells or cancer cells, comprising contacting such cells with an effective amount of a recombinant protein or a nucleic acid molecule encoding a recombinant protein of the invention.
  • the present invention also provides a method of treating a cell having a specific protease, comprising administering an effective amount of a recombinant protein or a nucleic acid molecule encoding a recombinant protein of the invention to an animal in need thereof.
  • animal as used herein means any member of the animal kingdom including all mammals, birds, fish, reptiles and amphibians.
  • the animal to be treated is a mammal, more preferably a human.
  • the specificity of a recombinant protein of the invention may be tested by treating the protein with the specific protease which is thought to be specific for the cleavage recognition site of the linker and assaying for cleavage products.
  • specific proteases may be isolated from cancer cells, or they may be prepared recombinantly, for example following the procedures in Darket et al. (J. Biol. Chem. 254:2307-2312 (1988)).
  • the cleavage products may be identified for example based on size, antigenicity or activity.
  • the toxicity of the recombinant protein may be investigated by subjecting the cleavage products to an in vitro translation assay in cell lysates, for example using Brome Mosaic Virus mRNA as a template. Toxicity of the cleavage products may be determined using a ribosomal inactivation assay (Westby et al., Bioconjugate Chem. 3:377-382 (1992)). The effect of the cleavage products on protein synthesis may be measured in standardized assays of in vitro translation utilizing partially defined cell free systems composed for example of a reticulocyte lysate preparation as a source of ribosomes and various essential cofactors, such as mRNA template and amino acids.
  • the ability of the recombinant proteins of the invention to selectively inhibit or destroy cells having specific proteases may be readily tested in vitro using cell lines having the specific protease, such as cancer cell lines.
  • the selective inhibitory effect of the recombinant proteins of the invention may be determined, for example, by demonstrating the selective inhibition of cellular proliferation in cancer cells or infected cells.
  • Toxicity may also be measured based on cell viability, for example, the viability of cancer and normal cell cultures exposed to the recombinant protein may be compared. Cell viability may be assessed by known techniques, such as trypan blue exclusion assays.
  • a number of models may be used to test the cytotoxicity of recombinant proteins having a heterologous linker sequence containing a cleavage recognition site for a cancer associated matrix metalloprotease.
  • Thompson, E. W. et al. Breast Cancer Res. Treatment 31:357-370 (1994)
  • tumour cell-mediated proteolysis of extracellular matrix and tumour cell invasion of reconstituted basement membrane reconstituted basement membrane (collagen, laminin, fibronectin, Matrigel or gelatin).
  • Other applicable cancer cell models include cultured ovarian adenocarcinoma cells (Young, T. N. et al. Gynecol. Oncol.
  • specific cell binding components may optionally be conjugated to the proteins of the invention.
  • Such cell binding components may be expressed as fusion proteins with the proteins of the invention or the cell binding component may be physically or chemically coupled to the protein component.
  • suitable cell binding components include antibodies to cancer proteins.
  • Antibodies having specificity for a cell surface protein may be prepared by conventional methods.
  • a mammal e.g. a mouse, hamster, or rabbit
  • an immunogenic form of the peptide which elicits an antibody response in the mammal
  • Techniques for conferring immunogenicity on a peptide include conjugation to carriers or other techniques well known in the art.
  • the peptide can be administered in the presence of adjuvant.
  • the progress of immunization can be monitored by detection of antibody titers in plasma or serum. Standard ELISA or other immunoassay procedures can be used with the immunogen as antigen to assess the levels of antibodies.
  • antisera can be obtained and, if desired, polyclonal antibodies isolated from the sera.
  • antibody producing cells can be harvested from an immunized animal and fused with myeloma cells by standard somatic cell fusion procedures thus immortalizing these cells and yielding hybridoma cells.
  • myeloma cells can be harvested from an immunized animal and fused with myeloma cells by standard somatic cell fusion procedures thus immortalizing these cells and yielding hybridoma cells.
  • Such techniques are well known in the art, (e.g. the hybridoma technique originally developed by Kohler and Milstein ( Nature 256:495-497 (1975)) as well as other techniques such as the human B-cell hybridoma technique (Kozbor et al., Immunol.
  • Hybridoma cells can be screened immunochemically for production of antibodies specifically reactive with the peptide and the monoclonal antibodies can be isolated.
  • antibody as used herein is intended to include fragments thereof which also specifically react with a cell surface component.
  • Antibodies can be fragmented using conventional techniques and the fragments screened for utility in the same manner as described above. For example, F(ab′) 2 fragments can be generated by treating antibody with pepsin. The resulting F(ab′) 2 fragment can be treated to reduce disulfide bridges to produce Fab fragments.
  • Chimeric antibody derivatives i.e., antibody molecules that combine a non-human animal variable region and a human constant region are also contemplated within the scope of the invention.
  • Chimeric antibody molecules can include, for example, the antigen binding domain from an antibody of a mouse, rat, or other species, with human constant regions.
  • Conventional methods may be used to make chimeric antibodies containing the immunoglobulin variable region which recognizes a cell surface antigen (See, for example, Morrison et al., Proc. Natl. Acad. Sci. U.S.A. 81:6851 (1985); Takeda et al., Nature 314:452 (1985), Cabilly et al., U.S. Pat. No.
  • Monoclonal or chimeric antibodies specifically reactive against cell surface components can be further humanized by producing human constant region chimeras, in which parts of the variable regions, particularly the conserved framework regions of the antigen-binding domain, are of human origin and only the hypervariable regions are of non-human origin.
  • Such immunoglobulin molecules may be made by techniques known in the art, (e.g. Teng et al., Proc. Natl. Acad. Sci. U.S.A., 80:7308-7312 (1983); Kozbor et al., Immunology Today 4:7279 (1983); Olsson et al., Meth. Enzymol., 92:3-16 (1982), and PCT Publication WO92/06193 or EP 239,400). Humanized antibodies can also be commercially produced (Scotgen Limited, 2 Holly Road, Twickenham, Middlesex, Great Britain.)
  • Specific antibodies, or antibody fragments, reactive against cell surface components may also be generated by screening expression libraries encoding immunoglobulin genes, or portions thereof, expressed in bacteria with cell surface components.
  • complete Fab fragments, VH regions and FV regions can be expressed in bacteria using phage expression libraries (See for example Ward et al., Nature 341:544-546 (1989); Huse et al., Science 246:1275-1281 (1989); and McCafferty et al., Nature 348:552-554 (1990)).
  • phage expression libraries See for example Ward et al., Nature 341:544-546 (1989); Huse et al., Science 246:1275-1281 (1989); and McCafferty et al., Nature 348:552-554 (1990)).
  • SCID-hu mouse for example the model developed by Genpharm, can be used to produce antibodies, or fragments thereof.
  • the proteins and nucleic acids of the invention may be formulated into pharmaceutical compositions for administration to subjects in a biologically compatible form suitable for administration in vivo.
  • biologically compatible form suitable for administration in vivo is meant a form of the substance to be administered in which any toxic effects are outweighed by the therapeutic effects.
  • the substances may be administered to living organisms including humans, and animals.
  • Administration of a therapeutically active amount of the pharmaceutical compositions of the present invention is defined as an amount effective, at dosages and for periods of time necessary to achieve the desired result.
  • a therapeutically active amount of a substance may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of antibody to elicit a desired response in the individual. Dosage regime may be adjusted to provide the optimum therapeutic response. For example, several divided doses may be administered daily or the dose may be proportionally reduced as indicated by the exigencies of the therapeutic situation.
  • the present invention provides a pharmaceutical composition for treating cells having a specific protease comprising a recombinant protein or a nucleic acid encoding a recombinant protein of the invention and a pharmaceutically acceptable carrier, diluent or excipient.
  • the active substance may be administered in a convenient manner such as by injection (subcutaneous, intravenous, intramuscular, etc.), oral administration, inhalation, transdermal administration (such as topical cream or ointment, etc.), or suppository applications.
  • the active substance may be coated in a material to protect the compound from the action of enzymes, acids and other natural conditions which may inactivate the compound.
  • compositions described herein can be prepared by per se known methods for the preparation of pharmaceutically acceptable compositions which can be administered to subjects, such that an effective quantity of the active substance is combined in a mixture with a pharmaceutically acceptable vehicle.
  • Suitable vehicles are described, for example, in Remington's Pharmaceutical Sciences (Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa., USA 1985).
  • the compositions include, albeit not exclusively, solutions of the substances in association with one or more pharmaceutically acceptable vehicles or diluents, and contained in buffered solutions with a suitable pH and iso-osmotic with the physiological fluids.
  • compositions may be used in methods for treating animals, including mammals, preferably humans, with cancer. It is anticipated that the compositions will be particularly useful for treating patients with B-cell lymphoproliferative disease and melanoma.
  • the dosage and type of recombinant protein to be administered will depend on a variety of factors which may be readily monitored in human subjects. Such factors include the etiology and severity (grade and stage) of the neoplasia.
  • the preproricin gene was cloned from new foliage of the castor bean plant.
  • Total messenger RNA was isolated according to established procedures (Sambrook et al., Molecular Cloning: A Lab Manual (Cold Spring Harbour Press, Cold Spring Harbour, (1989)) and cDNA generated using reverse transcriptase.
  • Oligonucleotides corresponding to the extreme 5′ and 3′ ends of the preproricin gene were synthesized and used to PCR amplify the gene. Using the cDNA sequence for preproricin (Lamb et al., Eur. J. Biochem., 145:266-270, 1985), several oligonucleotide primers were designed to flank the start and stop codons of the preproricin open reading frame. The oligonucleotides were synthesized using an Applied Biosystems Model 392 DNA/RNA Synthesizer. First strand cDNA synthesis was primed using the oligonucleotide Ricin1729C.
  • the first strand cDNA synthesis reaction was used as template for DNA amplification by the polymerase chain reaction (PCR).
  • the preproricin cDNA was amplified using the upstream primer Ricin-99 (5′-CCGGGAGGAAATACTATTGTAAT-3′ (SEQ ID NO:128)) and the downstream primer Ricin1729C with Vent DNA polymerase (New England Biolabs) using standard procedures (Sambrook et al., Molecular Cloning: A Laboratory Manual, Second Edition, (Cold Spring Harbor Laboratory Press, 1989)).
  • Amplification was carried out in a Biometra thermal cycler (TRIO-Thermalcycler) using the following cycling parameters: denaturation 95° C. for 1 min., annealing 52° C.
  • the purified PCR fragment encoding the preproricin cDNA was then ligated (Sambrook et al., Molecular Cloning: A Laboratory Manual, Second Edition, (Cold Spring Harbor Laboratory Press, 1989)) into an Eco RV digested pBluescript II SK plasmid (Stratagene), and used to transform competent XL1-Blue cells (Stratagene). Positive clones were confirmed by restriction digestion of purified plasmid DNA. Plasmid DNA was extracted using a Qiaprep Spin Plasmid Miniprep Kit (Qiagen).
  • the cloned PCR product containing the putative preproricin gene (pAP144) was confirmed by DNA sequencing of the entire cDNA clone. Sequencing was performed using an Applied Biosystems 373A Automated DNA Sequencer, and confirmed by double-stranded dideoxy sequencing by the Sanger method using the Sequenase kit (USB) (see WO 98/49311).
  • pAP144 cut with EcoR1 was used as target for PCR pairs employing the Ricin109-Eco oligonucleotide (Ricin-109Eco primer: 5-GGAGGAATCCGGAGATGAAACCGGGAGGAAATACTATTGTAAT-3 (SEQ ID NO:129)) and a mutagenic primer for the 5′ half of the linker as well as the Ricin1729PstI primer (Ricin 1729-PstI: 5GTAGGCGCTGCAGATAACTTGCTGTCCTTTCAG-3 (SEQ ID NO:130)) and a mutagenic primer for the 3′ half of the linker.
  • the cycling conditions used for the PCRs were 98 degrees C. for 2 min.; 98° C.
  • PCR products were then digested by EcoRI and PstI respectively, electrophoresed on an agarose gel, and the bands purified by via glass wool spin columns. Triple ligations comprising the PCR product pairs (corresponding halves of the new linker) and pVL1393 vector digested with EcoR1 and PstI were carried out. Recombinant clones were identified by restriction digests of plasmid miniprep DNA and the altered linkers confirmed by DNA sequencing. Note that all altered linker variants were cloned directly into the pVL1393 vector.
  • Insect cells S. frugiperda (Sf9), and Trichoplusia ni (Tn368 and BTI-TN-581-4 (High Five)) were maintained on EX-CELL 405 medium JRH Biosciences) supplemented with 10% total calf serum (Summers et al., A Manual of Methods of Baculovirus Vectors and Insect Cell Culture Procedures , (Texas Agricultural Experiment Station, 1987)).
  • Recombinant viruses in the supernatants were then amplified by infecting Tn368 cells at a multiplicity of infection (moi) of 0.1, followed by collection of day 3 to 5 supernatants. A total of three rounds of amplification were performed for each recombinant following established procedures (Summers et al., A Manual of Methods of Baculovirus Vectors and Insect Cell Culture Procedures, (Texas Agricultural Experiment Station, 1987 and Gruenwald et al., Baculovirus Expression Vector System: Procedures and Methods Manual, 2nd Edition, (San Diego, Calif., 1993)).
  • Recombinant baculoviruses were used to infect 1 ⁇ 10 7 Tn368 or Sf9 cells at an moi of 9 in EX-CELL 405 media (JRH Biosciences) with 25 mM a-lactose in spinner flasks. Media supernatants containing mutant proricins were collected 3 or 4 days post-infection.
  • Protein samples were harvested three days post infection. The cells were removed by centrifuging the media at 8288 g for ten minutes using a GS3 (Sorvall) centrifuge rotor. The supernatant was further clarified by centrifuging at 25400 g using a SLA-1500 rotor (Sorvall) for 45 minutes. Protease inhibitor phenylmethylsulfonyl fluoride (Sigma) was slowly added to a final concentration of 1 mM. The samples were further prepared by adding a-lactose to a concentration of 20 mM (not including the previous lactose contained in the expression medium).
  • the samples were concentrated to 700 mL using a Prep/Scale-TFF Cartridge (2.5 ft, 10K regenerated cellulose (Millipore)) and a Masterflex pump. The samples were then dialysed for 2 days in 1 ⁇ Column Buffer (50 mM Tris, 100 mM NaCl, 0.02% NaN3, pH 7.5) using dialysis tubing (10 K MWCO, 32 mm flat width (Spectra/Por)). Subsequently, the samples were clarified by centrifuging at 25400 g using a SLA-1500 rotor (Sorvall) for 45 minutes.
  • a Prep/Scale-TFF Cartridge 2.5 ft, 10K regenerated cellulose (Millipore)
  • the samples were then dialysed for 2 days in 1 ⁇ Column Buffer (50 mM Tris, 100 mM NaCl, 0.02% NaN3, pH 7.5) using dialysis tubing (10 K MWCO, 32 mm flat width (Spec
  • the samples were degassed and applied at 4 degrees C. to a XK26/20 (Pharmacia) column (attached to a Pharmacia peristaltic pump, Pharmacia Single-path Monitor UV-1 Control and Optical Units, and Bromma LKB 2210 2-Channel Recorder) containing 20 mL a-Lactose Agarose Resin (Sigma).
  • the column was washed for 3 hours with 1 ⁇ Column buffer. Elution of proricin variant was performed by eluting with buffer (1 ⁇ Column buffer (0.1% NaN 3 ), 100 mM Lactose) until the baseline was again restored.
  • the samples were concentrated using an Amicon 8050 concentrator (Amicon) with a YM10 76 mm membrane, utilizing argon gas to pressurize the chamber.
  • the samples were further concentrated in Centricon 10 (Millipore) concentrators according to manufacturer's specifications.
  • the protein was applied to a SUPERDEX 75 (16/60) column and SUPERDEX 200 (16/60) column (Pharmacia) connected in series equilibrated with 100 mM Tris, 200 mM NaCl, pH 7.5 containing 100 mM lactose and 1.0% ⁇ -mercaptoethanol ( ⁇ ME).
  • the flow rate of the column was 0.15 mL/min and fractions were collected every 25 minutes.
  • the UV (280 nm) trace was used to determine the approximate location of the purified PAP-protein and thus determine the samples for Western analysis.
  • transfer buffer 48 mM Tris, 39 mM glycine, 0.0375% SDS, and 20% Methanol
  • PVDF Biorad membrane was presoaked for one minute in 100% methanol, rinsed in ddH 2 O and two minutes in transfer buffer.
  • Whatman paper was soaked briefly in transfer buffer. Five pieces of Whatman paper, membrane, gel, and another five pieces of Whatman paper were arranged on the bottom cathode (anode) of the Pharmacia Novablot transfer apparatus (Pharmacia). Transfer was for one hour at constant current (2 mA/cm 2 ).
  • Non-specifically bound primary antibody was removed by washing the blot for ten minutes with 1 ⁇ PBS containing 0.2% Tween 20. This was repeated four times. Secondary antibody donkey anti-rabbit (Amersham) was incubated with the blot under the same conditions as the primary antibody. Excess secondary antibody was washed as described above. Blots were developed with the ECL Western Blotting detection reagents according to the manufacturer's instructions. Blots were exposed to Medtec's Full Speed Blue Film (Medtec) or Amersham's ECL Hyperfilm (Amersham) for one second to five minutes. Film was developed in a KODAK Automatic Developer.
  • Immulon 2 plate VWR
  • 10 ⁇ g/ml of asialofetuin was coated with 100 ⁇ l per well of 10 ⁇ g/ml of asialofetuin and left overnight at 4° C.
  • the plate was washed with 3 ⁇ 300 ⁇ l per well with ddH 2 O using an automated plate washer (BioRad).
  • BioRad automated plate washer
  • the plate was blocked for one hour at 37° C. by adding 300 ⁇ L per well of PBS containing 1% ovalbumin.
  • the plate was washed again as above.
  • Proricin variant PAP-protein was added to the plate in various dilutions in 1 ⁇ Column Buffer, (50 mM Tris, 100 mM NaCl, pH 7.5).
  • a standard curve of RCA 60 (Sigma) from 1-10 ng was also included.
  • the plate was incubated for 1 h at 37° C. The plate was washed as above.
  • Anti-ricin monoclonal antibody (Sigma) was diluted 1:3000 in 1 ⁇ PBS containing 0.5% ovalburnin and 0.1% Tween-20, added at 100 ⁇ L per well and incubated for 1 h at 37° C. The plate was washed as above.
  • Donkey anti-rabbit polyclonal antibody was diluted 1:3000 in 1 ⁇ PBS containing 0.5% ovalburnin, 0.1% Tween-20, and added at 100 ⁇ L per well and incubated for 1 h at 37° C. The plate was given a final wash as described above.
  • Substrate was added to plate at 100 ⁇ L per well (1 mg/mL o-phenylenediamine (in H 2 O), 1 ⁇ L/mL H 2 O 2 ) and after development 25 ⁇ L of stop solution (20% H 2 SO 4 ) was added and the absorbance read (A490 nm-A630 nm) using a SPECTRA MAX 340 plate reader (Molecular Devices).
  • Ricin samples were prepared for reduction.
  • the required number of 0.5 mL tubes were labelled. (2 25 tubes for each sample, + and ⁇ aniline). To each of the sample tubes 20 ⁇ L of 1 ⁇ endo buffer was added, and 30 ⁇ L of buffer was added to the controls. To the sample tubes either 10 ⁇ L of 10 ng/ ⁇ L, Ricin or 10 ⁇ L of variant was added. Finally, 30 ⁇ L of rabbit reticulocyte lysate was added to all the tubes. The samples were incubated for 30 minutes at 30° C. using the thermal block. Samples were removed from the 0.5 mL tube and contents added into a 1.5 mL tube containing 1 mL of TRIZOL (Gibco). Samples were incubated for 15 minutes at room temperature.
  • RNA samples Pellets from the other samples (+aniline samples) were dissolved in 20 ⁇ L of DEPC treated ddH 2 O. An 80 ⁇ L aliquot of 1 M aniline (distilled) with 2.8 M acetic acid was added to these RNA samples and transferred to a fresh 0.5 mL tube. The samples were incubated in the dark for 3 minutes at 60° C. RNA was precipitated by adding 100 ⁇ l, of 95% ethanol and 5 ⁇ L of 3M sodium acetate, pH 5.2 to each tube and centrifuging at 12,000 g for 30 minutes at 4° C. Pellets were washed with 1 mL 70% ethanol and centrifuged again at 12,000 g for 5 minutes at 4° C. to precipitate RNA.
  • RNA ladder (BRL) (8 ⁇ L of ladder+8 ⁇ L of loading dye) was also included. Samples were incubated for 2 minutes at 70° C. on the thermal block. Electrophoresis was carried out on the samples using 1.2% agarose, 50% formamide gels in 0.1 ⁇ E buffer+0.2% SDS. The gel was run for 90 minutes at 75 volts. RNA was visualized by staining the gel in 1 ⁇ g/mL ethidium bromide in running buffer for 45 minutes. The gel was examined on a 302 nm UV box, photographed using the gel documentation system and saved to a computer disk.
  • Partially purified PAP-protein was applied to Superdex 75 and 200 (16/60) columns connected in series in order to remove the contaminating non-specifically processed PAP-protein. Eluted fractions were tested via Western analysis as described above and the fractions containing the most pure protein were pooled, concentrated and dialyzed against 1 ⁇ PBS buffer and then sterilized by filtration (Millipore). Final purified PAP-protein has less than 1% processed variant.
  • the purified PAP-protein was tested for susceptibility to cleavage by the particular protease and for activation of the A chain of the proricin variant, (inhibition of protein synthesis).
  • PAP-protein was incubated with and without protease for a specified time period and then electrophoresed and blotted. Cleaved PAP-protein will run as two 30 kDa proteins (B is slightly larger) under reducing (SDS-PAGE) conditions.
  • Unprocessed PAP-protein, which contains the linker region will migrate at 60 kDa.
  • Activation of protease treated PAP-protein is based on the method of May et al. ( EMBO Journal. 8 301-8, 1989).
  • Activation of ricin A chain upon cleavage of the intermediary linker results in catalytic depurination of the adenosine 4325 residue of 28S or 26S rRNA.
  • This depurination renders the molecule susceptible to amine-catalyzed hydrolysis by aniline of the phosphodiester bond on either side of the modification site.
  • the result is a diagnostic 390 base band.
  • reticulocyte ribosomes incubated with biochemically purified ricin A chain, released the characteristic RNA fragment upon aniline treatment of isolated rRNA (May, M. J.
  • the assay allows for the determination of activity of a ricin A chain which has been cleaved from the intact unit containing a particular variant linker sequence.
  • Affinity-purified proricin variant is treated with individual disease-specific proteases to confirm specific cleavage in the linker region.
  • Ricin-like toxin variants are eluted from the lactose-agarose matrix in protease digestion buffer (50 mM NaCl, 50 mM Na-acetate, pH 5.5, 1 mM dithiothreitol) containing 100 mM lactose.
  • Proricin substrate is then incubated at 37° C. for 60 minutes with a disease-specific protease.
  • the cleavage products consisting ricin A and B chains are identified using SDS/PAGE (Sambrook et al., Molecular Cloning: a Laboratory Manual, 2nd. ed., Cold Spring Harbor Press, 1989), followed by Western blot analysis using anti-ricin antibodies (Sigma).
  • FIG. 19 shows the cleavage products of an MMP-9 digestion of PAP323, PAP324 and PAP325.
  • Matrix metalloproteinases may be prepared substantially as described by Lark, M. W. et al. ( Proceedings of the 4 th International Conference of the Inflammation Research Association Abstract 145 (1988)) and Welch, A. R. et al. (Arch. Biochem. Biophys. 324:59-64 (1995)).
  • Urokinase plasminogen activator may be prepared substantially as described by Holmberg, L. et al. (Biochim Biophys Acta, 445:215-222, (1976)) and Someno, T. et al. (J Biochem 97:1493-1500 (1985)).
  • the Ricin and Ricin variants were sterile filtered using a 0.22 ⁇ m filter (Millipore). The concentration of the sterile samples were then quantified by A 280 and confirmed by BCA measurements (Pierce). For the variants digested with the MMP-9 protease in vitro, the digests were carried out as described in the digestion procedure for each protease. The digests were then diluted in the 1000 ng ⁇ ml ⁇ 1 dilution and sterile filtered.
  • Ricin and Ricin variants were serially diluted to the following concentrations: 1000 ng ⁇ ml ⁇ 1 , 100 ng ⁇ ml ⁇ 1 , 10 ng ⁇ ml ⁇ 1 , 1 ng ⁇ ml ⁇ 1 , 0.1 ng ⁇ ml ⁇ 1 , 0.01 ng ⁇ ml ⁇ 1 , 0.001 ng ⁇ ml ⁇ 1 with media containing 10% FBS and 1 ⁇ pen/strep.
  • the whole amount of media (and/or toxin) was removed from each well with a multichannel pipettor, and replaced with 100 ⁇ l of the substrate mixture (Promega Cell Titer 96 Aqueous Non-Radioactive Cell Proliferation Assay Kit).
  • the plates were incubated at 37° C. with 5% CO 2 for 2 to 4 hours, and subsequently read with a Spectramax 340 96 well plate reader at 490 nm.
  • the IC 50 values were calculated using the GRAFIT software program.
  • the protocol for the maximum tolerable dose (MTD) study involved three intravenous injections of variant, on days 1, 5 and 9, into the tail vein of either a Nude/SCID mouse. Three animals were used for each dose tested. The samples were diluted into saline solution containing 100 ⁇ g/mL Bovine Serum Albumin on the same day as the injection. Animals were observed for 14 days after dosing. Any surviving animals were euthanized after 14 days of study. The MTD value was defined as the highest dose of sample tested where all animals in the group survived. The results are presented in Table 5.
  • Tumour growth was monitored daily by measuring tumour dimensions with calipers.
  • the treatment initiation date was dependent on the rate of tumour growth.
  • the treatment groups are shown in Table 6.
  • Subcutaneous A431 tumours were established in SCID mice.
  • the tumours were treated with either PAP304 or PAP305 when the tumours reached 50 mm 3 on Days 1, 5 and 9.
  • the results shown in FIGS. 20 and 21 demonstrate that the linker decreases the toxicity of the variant (as compared with ricin) and the variants PAP304 and PAP305 are activated at or near the A431 (human epithelial carcinoma) solid tumour in mice.
  • a very exciting result is shown in FIG. 20 .
  • the variant PAP304 was able to slow down the growth of A431 solid tumour (17 day delay), without any signs of dose limiting toxicity (e.g., no weight loss or death).
  • MMP9 Variant Linker Size In Vivo ( ⁇ g/kg) PAP301 23 8 PAP302 16 40 PAP303 15 10 PAP304 8 150 PAP305 12 20 PAP308 12 30 PAP309 23 20 PAP316 23 20 PAP318 23 ⁇ 20 PAP323 21 15 PAP324 19 20 PAP325 17 20 (cf. Ricin - 1.6 ⁇ g/kg and PAP220 - 13 ⁇ g/kg)

Abstract

The present invention provides a protein having chain of a ricin-like toxin, a B chain of a ricin-like toxin and a novel heterologous linker amino acid sequence, linking the A and B chains. The linker sequence contains a cleavage recognition site for a specific protease such as those found in inflammatory cells and cancer cells. The invention also relates to a nucleic acid molecule encoding the protein and to expression vectors incorporating the nucleic acid molecule. Also provided is a method of inhibiting or destroying cells having a specific protease, such as cancer cells or inflammatory cells utilizing the nucleic acid molecules and proteins of the invention and pharmaceutical compositions for treating human inflammation and cancer.

Description

    RELATED APPLICATIONS
  • This application is a continuation of U.S. application Ser. No. 10/089,058 (now allowed) filed Sep. 19, 2002 which is a continuation of PCT/CA00/01162 filed Oct. 4, 2000 (which designated the U.S.) which claims the benefit of U.S. Provisional application No. 60/157,807 filed Oct. 4, 1999 (now abandoned). This application also claims benefit of U.S. Provisional application No. 60/197,409 filed Apr. 14, 2000 (now abandoned). All of the prior applications are incorporated herein in their entirety.
  • FIELD OF THE INVENTION
  • The invention relates to proteins useful as therapeutics against unhealthy cells such as those which occur in inflammation and cancer. The proteins contain A and B chains of a ricin-like toxin linked by a novel linker sequence that is specifically cleaved and activated by proteases specific to cancer.
  • BACKGROUND OF THE INVENTION
  • Bacteria and plants are known to produce cytotoxic proteins which may consist of one, two or several polypeptides or subunits. Those proteins having a single subunit may be loosely classified as Type I proteins. Many of the cytotoxins which have evolved two subunit structures are referred to as Type II proteins (Saelinger, C. B. in Trafficking of Bacterial Toxins (eds. Saelinger, C. B.) 1-13 (CRC Press Inc., Boca Raton, Fla., 1990). One subunit, the A chain, possesses the toxic activity whereas the second subunit, the B chain, binds cell surfaces and mediates entry of the toxin into a target cell. A subset of these toxins kill target cells by inhibiting protein biosynthesis. For example, bacterial toxins such as diphtheria toxin or Pseudomonas exotoxin inhibit protein synthesis by inactivating elongation factor 2. Plant toxins such as ricin, abrin, and bacterial toxin Shiga toxin, inhibit protein synthesis by directly inactivating the ribosomes (Olsnes, S. & Phil, A. in Molecular action of toxins and viruses (eds. Cohen, P. & vanHeyningen, S.) 51-105 Elsevier Biomedical Press, Amsterdam, 1982).
  • Ricin, derived from the seeds of Ricinus communis (castor oil plant), may be the most potent of the plant toxins. It is estimated that a single ricin A chain is able to inactivate ribosomes at a rate of 1500 ribosomes/minute. Consequently, a single molecule of ricin is enough to kill a cell (Olsnes, S. & Phil, A. in Molecular action of toxins and viruses (eds. Cohen, P. & vanHeyningen, S.) (Elsevier Biomedical Press, Amsterdam, 1982). The ricin toxin is a glycosylated heterodimer consisting of A and B chains with molecular masses of 30,625 Da and 31,431 Da linked by a disulphide bond. The A chain of ricin has an N-glycosidase activity and catalyzes the excision of a specific adenine residue from the 28S rRNA of eukaryotic ribosomes (Endo, Y. & Tsurugi, K. J., Biol. Chem. 262:8128 (1987)). The B chain of ricin, although not toxic in itself, promotes the toxicity of the A chain by binding to galactose residues on the surface of eukaryotic cells and stimulating receptor-mediated endocytosis of the toxin molecule (Simmons et al., Biol. Chem. 261:7912 (1986)). Once the toxin molecule consisting of the A and B chains is internalized into the cell via clathrin-dependent or independent mechanisms, the greater reduction potential within the cell induces a release of the active A chain, eliciting its inhibitory effect on protein synthesis and its cytotoxicity (Emmanuel, F. et al., Anal. Biochem. 173: 134-141 (1988); Blum, J. S. et al., J. Biol. Chem. 266: 22091-22095 (1991); Fiani, M. L. et al., Arch. Biochem. Biophys. 307: 225-230 (1993)). Empirical evidence suggests that activated toxin (e.g. ricin, shiga toxin and others) in the endosomes is transcytosed through the trans-Golgi network to the endoplasmic reticulum by retrograde transport before the A chain is translocated into the cytoplasm to elicit its action (Sandvig, K. & van Deurs, B., FEBS Lett. 346: 99-102 (1994).
  • Protein toxins are initially produced in an inactive, precursor form. Ricin is initially produced as a single polypeptide (preproricin) with an amino acid N-terminal presequence and 12 amino acid linker between the A and B chains. The pre-sequence is removed during translocation of the ricin precursor into the endoplasmic reticulum (Lord, J. M., Eur. J. Biochem. 146:403-409 (1985) and Lord, J. M., Eur. J. Biochem. 146:411-416 (1985)). The proricin is then translocated into specialized organelles called protein bodies where a plant protease cleaves the protein at a linker region between the A and B chains (Lord, J. M. et al., FASAB journal 8:201-208 (1994)). The two chains, however, remain covalently attached by an interchain disulfide bond (cysteine 259 in the A chain to cysteine 4 in the B chain) and mature disulfide linked ricin is stored in protein bodies inside the plant cells. The A chain is inactive in proricin (O'Hare, M. et al., FEBS Lett. 273:200-204 (1990)) and it is inactive in the disulfide-linked mature ricin (Richardson, P. T. et al., FEBS Lett. 255:15-20 (1989)). The ribosomes of the castor bean plant are themselves susceptible to inactivation by ricin A chain; however, as there is no cell surface galactose to permit B chain recognition the A chain cannot re-enter the cell. The exact mechanism of A chain release and activation in target cell cytoplasm is not known (Lord, J. M. et al., FASAB journal 8:201-208 (1994)). However, it is known that for activation to take place the disulfide bond between the A and B chains must be reduced and, hence, the linkage between subunits broken.
  • Diphtheria toxin is produced by Corynebacterium diphtheriae as a 535 amino acid polypeptide with a molecular weight of approximately 58 kD (Greenfield, L. et al., Proc. Natl. Acad. Sci. USA 80:6853-6857 (1983); Pastan, I. et al., Annu. Rev. Biochem. 61:331-354 (1992); Collier, R. J. & Kandel, J., 1. Biol. Chem. 246:1496-1503 (1971)). It is secreted as a single-chain polypeptide consisting of 2 functional domains. Similar to proricin, the N-terminal domain (A-chain) contains the cytotoxic moiety whereas the C-terminal domain (B-chain) is responsible for binding to the cells and facilitates toxin endocytosis. Conversely, the mechanism of cytotoxicity for diphtheria toxin is based on ADP-ribosylation of EF-2 thereby blocking protein synthesis and producing cell death. The 2 functional domains in diphtheria toxin are linked by an arginine-rich peptide sequence as well as a disulphide bond. Once the diphtheria toxin is internalized into the cell, the arginine-rich peptide linker is cleaved by trypsin-like enzymes and the disulphide bond (Cys 186-201) is reduced. The cytotoxic domain is subsequently translocated into the cytosol substantially as described above for ricin and elicits ribosomal inhibition and cytotoxicity.
  • Pseudomonas exotoxin is also a 66 kD single-chain toxin protein secreted by Pseudomonas aeruginosa with a similar mechanism of cytotoxicity to that of diphtheria toxin (Pastan, I. et al., Annu. Rev. Biochem. 61:331-354 (1992); Ogata, M. et al., J. Biol. Chem. 267:25396 25401 (1992); Vagil, M. L. et al., Infect. Immunol. 16:353-361 (1977)). Pseudomonas exotoxin consists of 3 conjoint functional domains. The first domain I (amino acids 1-252) is responsible for cell binding and toxin endocytosis, a second domain II (amino acids 253-364) is responsible for toxin translocation from the endocytic vesicle to the cytosol, and a third domain III (amino acids 400-613) is responsible for protein synthesis inhibition and cytotoxicity. After Pseudomonas exotoxin enters the cell, the liberation of the cytotoxic domain is effected by both proteolytic cleavage of a polypeptide sequence in the second domain (near Arg 279) and the reduction of the disulphide bond (Cys 265-287) in the endocytic vesicles. In essence, the overall pathway to cytotoxicity is analogous to diphtheria toxin with the exception that the toxin translocation domain in Pseudomonas exotoxin is structurally distinct.
  • Class 2 ribosomal inhibitory proteins (RIP-2) constitute other toxins possessing distinct functional domains for cytotoxicity and cell binding/toxin translocation which include abrin, modeccin, volkensin, (Sandvig, K. et al., Biochem. Soc. Trans. 21:707-711 (1993)) and mistle toe lectin (viscumin) (Olsnes, S. & Phil, A. in Molecular action of toxins and viruses (eds. Cohen, P. & vanHeyningen, S.) 51-105 Elsevier Biomedical Press, Amsterdam, 1982; Fodstad, et al. Canc. Res. 44: 862 (1984)). Some toxins such as Shiga toxin and cholera toxin also have multiple polypeptide chains responsible for receptor binding and endocytosis.
  • The ricin gene has been cloned and sequenced, and the X-ray crystal structures of the A and B chains have been described (Rutenber, E. et al. Proteins 10:240-250 (1991); Weston et al., Mol. Bio. 244:410-422, 1994; Lamb and Lord, Eur. J. Biochem. 14:265 (1985); Halling, K. et al. Nucleic Acids Res. 13:8019 (1985)). Similarly, the genes for diptheria toxin and Pseudomonas exotoxin have been cloned and sequenced, and the 3-dimensional structures of the toxin proteins have been elucidated and described (Columblatti, M. et al., J. Biol. Chem. 261:3030-3035 (1986); Allured, V. S. et al., Proc. Natl. Acad. Sci. USA 83:1320-1324 (1986); Gray, G. L. et al., Proc. Natl. Acad. Sci. USA 81:2645-2649 (1984); Greenfield, L. et al., Proc, Natl. Acad. Sci. USA 80:6853-6857 (1983); Collier, R. J. et al., J. Biol. Chem. 257:5283-5285 (1982)).
  • The potential of bacterial and plant toxins for inhibiting mammalian retroviruses, particularly acquired immunodeficiency syndrome (AIDS), has been investigated. Bacterial toxins such as Pseudomonas exotoxin and subunit A of diphtheria toxin; dual chain ribosomal inhibitory plant toxins such as ricin, and single chain ribosomal inhibitory proteins such as trichosanthin and pokeweed antiviral protein have been used for the elimination of HIV infected cells (Olson et al., AIDS Res. and Human Retroviruses 7:1025-1030 (1991)). The high toxicity of these toxins for mammalian cells, combined with a lack of specificity of action poses a major problem to the development of pharmaceuticals incorporating the toxins, such as immunotoxins.
  • Due to their extreme toxicity there has been much interest in making ricin-based immunotoxins as therapeutic agents for specifically destroying or inhibiting infected or tumourous cells or tissues (Vitetta et al., Science 238:1098-1104 (1987)). An immunotoxin is a conjugate of a specific cell binding component, such as a monoclonal antibody or growth factor and the toxin in which the two protein components are covalently linked. Generally, the components are chemically coupled. However, the linkage may also be a peptide or disulfide bond. The antibody directs the toxin to cell types presenting a specific antigen thereby providing a specificity of action not possible with the natural toxin. Immunotoxins have been made both with the entire ricin molecule (i.e. both chains) and with the ricin A chain alone (Spooner et al., Mol. Immunol. 31:117-125, (1994)).
  • Immunotoxins made with the ricin dimer (IT-Rs) are more potent toxins than those made with only the A chain (IT-As). The increased toxicity of IT-Rs is thought to be attributed to the dual role of the B chains in binding to the cell surface and in translocating the A chain to the cytosolic compartment of the target cell (Vitetta et al., Science 238:1098-1104 (1987); Vitetta & Thorpe, Seminars in Cell Biology 2:47-58 (1991)). However, the presence of the B chain in these conjugates also promotes the entry of the immunotoxin into nontarget cells. Even small amounts of B chain may override the specificity of the cell-binding component as the B chain will bind nonspecifically to galactose associated with N-linked carbohydrates, which is present on most cells. IT-As are more specific and safer to use than IT-Rs. However, in the absence of the B chain the A chain has greatly reduced toxicity. Due to the reduced potency of IT-As as compared to IT-Rs, large doses of IT-As must be administered to patients. The large doses frequently cause immune responses and production of neutralizing antibodies in patients (Vitetta et al., Science 238:1098-1104 (1987)). IT-As and IT-Rs both suffer from reduced toxicity as the A chain is not released from the conjugate into the target cell cytoplasm.
  • A number of immunotoxins have been designed to recognize antigens on the surfaces of tumour cells and cells of the immune system (Pastan et al., Annals New York Academy of Sciences 758:345-353 (1995)). A major problem with the use of such immunotoxins is that the antibody component is its only targeting mechanism and the target antigen is often found on non-target cells (Vitetta et al., Immunology Today 14:252-259 (1993)). Also, the preparation of a suitable specific cell binding component may be problematic. For example, antigens specific for the target cell may not be available and many potential target cells and infective organisms can alter their antigenic make up rapidly to avoid immune recognition. In view of the extreme toxicity of proteins such as ricin, the lack of specificity of the immunotoxins may severely limit their usefulness as therapeutics for the treatment of cancer and infectious diseases.
  • The insertion of intramolecular protease cleavage sites between the cytotoxic and cell-binding components of a toxin can mimic the way that the natural toxin is activated. European patent application no. 466,222 describes the use of maize-derived pro-proteins which can be converted into active form by cleavage with extracellular blood enzymes such as factor, Xa, thrombin or collagenase. Garred, 0. et al. (J. Biol. Chem. 270:10817-10821 (1995)) documented the use of a ubiquitous calcium-dependent serine protease, furin, to activate shiga toxin by cleavage of the trypsin-sensitive linkage between the cytotoxic A-chain and the pentamer of cell-binding B-units. Westby et al. (Bioconjugate Chem. 3:375-381 (1992)) documented fusion proteins which have a specific cell binding component and proricin with a protease sensitive cleavage site specific for factor Xa within the linker sequence. O'Hare et al. (FEBS Lett. 273:200-204 (1990)) also described a recombinant fusion protein of RTA and staphylococcal protein A joined by a trypsin-sensitive cleavage site. In view of the ubiquitous nature of the extracellular proteases utilized in these approaches, such artificial activation of the toxin precursor or immunotoxin does not confer a mechanism for intracellular toxin activation and the problems of target specificity and adverse immunological reactions to the cell-binding component of the immunotoxin remain.
  • In a variation of the approach of insertion of intramolecular protease cleavage sites on proteins which combine a binding chain and a toxic chain, Leppla, S. H. et al. (Bacterial Protein Toxins zbl.bakt.suppl. 24:431-442 (1994)) suggest the replacement of the native cleavage site of the protective antigen (PA) produced by Bacillus anthracis with a cleavage site that is recognized by cells that contain a particular protease. PA, recognizes, binds, and thereby assists in the internalization of lethal factor (U) and edema toxin (ET), also produced by Bacillus anthracis. However, this approach is wholly dependent on the availability of LF, or ET and PA all being localized to cells wherein the modified PA can be activated by the specific protease. It does not confer a mechanism for intracellular toxin activation and presents a problem of ensuring sufficient quantities of toxin for internalization in target cells.
  • The in vitro activation of a Staphylococcus-derived pore forming toxin, α-hemolysin by extracellular tumour-associated proteases has been documented (Panchel, R. G. et al., Nature Biotechnology 14:852-857 (1996)). Artificial activation of α-hemolysin in vitro by said proteases was reported but the actual activity and utility of α-hemolysin in the destruction of target cells were not demonstrated.
  • α-Hemolysin does not inhibit protein synthesis but is a heptameric transmembrane pore which acts as a channel to allow leakage of molecules up to 3 kD thereby disrupting the ionic balances of the living cell. The α-hemolysin activation domain is likely located on the outside of the target cell (for activation by extracellular proteases). The triggering mechanism in the disclosed hemolysin precursor does not involve the intracellular proteolytic cleavage of 2 functionally distinct domains. Also, the proteases used for the a-hemolysin activation are ubitquitiously secreted extracellular proteases and toxin activation would not be confined to activation in the vicinity of diseased cells. Such widespread activation of the toxin does not confer target specificity and limits the usefulness of said a-hemolysin toxin as therapeutics due to systemic toxicity.
  • A variety of proteases specifically associated with malignancy have been identified and described. For example, cathepsin is a family of serine, cysteine or aspartic endopeptidases and exopeptidases which has been implicated to play a primary role in cancer metastasis (Schwartz, M. K., Clin. Chim. Acta 237:67-78 (1995); Spiess, E. et al., J. Histochem. Cytochem. 42:917-929 (1-994); Scarborough, P. E. et al., Protein Sci. 2:264276 (1993); Sloane, B. F. et al., Proc. Natl. Acad. Sci. USA 83:2483-2487 (1986); Mikkelsen, T. et al., J. Neurosurge 83:285-290 (1995)). Matrix metalloproteinases (MMPs or matrixins) are zinc-dependent proteinases consisting of collagenases, matrilysin, stromelysins, stromelysin-like MMPs, gelatinases, macrophage elastase, membrane-type MMPs (MT-MMPs) (Krane, S. M., Ann, N.Y. Acad. Sci. 732:1-10 (1994); Woessner, J. F., Ann, N.Y. Acad. Sci. 732:11-21 (1994); Carvalho, K. et al., Biochem. Biophys. Res., Comm. 191:172-179 (1993); Nakano, A. et al. J. of Neurosurge, 83:298-307 (1995); Peng, K-W, et al. Human Gene Therapy, 8:729-738 (1997); More, D. H. et al. Gynaecologic oncology, 65:78-82 (1997), Ravanti, L., Kahari, V. Intl. J. Mol. Med. 6(4):391 (2000)). These proteases are involved in pathological matrix remodeling. Under normal physiological conditions, regulation of matrixin activity is effected at the level of gene expression. Enzymatic activity is also controlled stringently by tissue inhibitors of metalloproteinases (TIMPs) (Murphy, G. et al., Ann. N.Y. Acad. Sci., 732:31-41 (1994)). The expression of MMP genes is reported to be activated in inflammatory disorders (e.g. rheumatoid arthritis) and malignancy.
  • The present inventors have prepared novel recombinant toxic proteins which are specifically toxic to diseased cells but do not depend for their specificity of action on a specific cell binding component. The recombinant proteins toxins have an A chain of a ricin-like toxin linked to a B chain by a synthetic linker sequence which may be cleaved specifically by a protease localised in cells or tissues affected by a specific disease to liberate the toxic A chain thereby selectively inhibiting or destroying the diseased cells or tissues (WO 98/49311 published Nov. 5, 1998 which is incorporated herein by reference).
  • SUMMARY OF THE INVENTION
  • The present invention relates to novel linker sequences that can be used to prepare recombinant toxic proteins having an A chain of a ricin-like toxin linked to a B chain by the linker sequence. The novel linker sequences of the invention are illustrated in FIGS. 1-18.
  • In one aspect the present invention provides a purified and isolated nucleic acid encoding a linker sequence comprising: the nucleic acid sequence of pAP301 as shown in FIG. 1A; the nucleic acid sequence of pAP302 as shown in FIG. 2A; the nucleic acid sequence of pAP303 as shown in FIG. 3A; the nucleic acid sequence of pAP304 as shown in FIG. 4A; the nucleic acid sequence of pAP305 as shown in FIG. 5A; the nucleic acid sequence of pAP308 as shown in FIG. 6A; the nucleic acid sequence of pAP309 as shown in FIG. 7A; the nucleic acid sequence of pAP313 as shown in FIG. 8A; the nucleic acid sequence of pAP314 as shown in FIG. 9A; the nucleic acid sequence of pAP315 as shown in FIG. 10A; the nucleic acid sequence of pAP316 as shown in FIG. 11A; the nucleic acid sequence of pAP318 as shown in FIG. 12A; the nucleic acid sequence of pAP320 as shown in FIG. 13A; the nucleic acid sequence of pAP321 as shown in FIG. 14A; the nucleic acid sequence of pAP322 as shown in FIG. 15A; the nucleic acid sequence of pAP323 as shown in FIG. 16A; the nucleic acid sequence of pAP324 as shown in FIG. 17A; and the nucleic acid sequence of pAP325 as shown in FIG. 18A.
  • In another aspect, the present invention provides a purified and isolated nucleic acid encoding a recombinant toxic protein comprising (a) a nucleotide sequence encoding an A chain of a ricin-like toxin, (b) a nucleotide sequence encoding a B chain of a ricin-like toxin and (c) a heterologous linker amino acid sequence, linking the A and B chains. The linker sequence is not a native linker sequence of a ricin-like toxin, but rather a synthetic heterologous linker sequence containing a cleavage recognition site for a specific protease. The A and or the B chain may be those of ricin. As used herein “specific protease” means a protease in any cell wherein there is expression of the protease at levels greater than those found in a corresponding healthy cell. Examples of a specific protease include MMPs, preferably MMP-2, MMP-9, MMP-14, and MT1-MMPs, and UPA, as well as others found in inflammatory cells and malignant cells. An inflammatory cell includes any cell involved in the inflammation process having a specific protease.
  • The recombinant toxic proteins employing the novel linker sequences of the present invention may be used to treat various forms of cells having specific proteases such as inflammatory disorders including rheumatoid arthritis, atherosclerotic cells, Crohn's disease, central nervous system disease as well as in cancer including, but not limited to, T- and B-cell lymphoproliferative diseases, ovarian cancer, pancreatic cancer, head and neck cancer, squamous cell carcinoma, gastrointestinal cancer, breast cancer, prostate, cancer and non small cell lung cancer. In an embodiment, of the invention the cleavage recognition site of the linker is the cleavage recognition site for a cancer-associated protease.
  • In particular embodiments, the amino acid sequence of the linker comprises the sequence of PAP301 shown in FIG. 1C; the sequence of PAP302 shown in FIG. 2C; the sequence of PAP303 shown in FIG. 3C; the sequence of PAP304 shown in FIG. 4C; the sequence of PAP305 shown in FIG. 5C; the sequence of PAP308 shown in FIG. 6C; the sequence of PAP309 shown in FIG. 7C; the sequence of PAP316 shown in FIG. 11C; the sequence of PAP318 shown in FIG. 12C; the sequence of PAP323 shown in FIG. 16C; the sequence of PAP324 shown in FIG. 17C; and the sequence of PAP325 shown in FIG. 18C; all cleaved by MMP-9; the sequence of PAP313 shown in FIG. 8C; the sequence of PAP314 shown in FIG. 9C; the sequence of PAP315 shown in FIG. 10C; the sequence of PAP320 shown in FIG. 13C; the sequence of PAP321 shown in FIG. 14C; the sequence of PAP322 shown in FIG. 15C; all cleaved by urokinase-type plasminogen activator.
  • In a preferred embodiment, the nucleic acid sequences of the recombinant toxic proteins containing ricin A and B chains with each of the linker sequences are shown in FIGS. 1B, 2B, 3B, 4B, 5B, 6B, 7B, 8B, 9B, 10B, 11B, 12B, 13B, 14B, 15B, 16B, 17B and 18B.
  • The present invention also provides a plasmid incorporating the nucleic acid of the invention. In another embodiment, the present invention provides a baculovirus transfer vector incorporating the nucleic acid of the invention.
  • In an aspect, the present invention provides a recombinant protein comprising an A chain of a ricin-like toxin, a B chain of a ricin-like toxin and a heterologous linker amino acid sequence, linking the A and B chains, wherein the linker sequence contains a cleavage recognition site for a specific protease. The A and/or the B chain may be those of ricin.
  • In a further aspect, the present invention provides a recombinant protein comprising an A chain of a ricin-like toxin, a B chain of a ricin-like toxin and a heterologous linker amino acid sequence, linking the A and B chains, wherein the linker sequence contains a cleavage recognition site for a inflammatory disease specific protease. The A and/or the B chain may be those of ricin. In an embodiment, the cleavage recognition site is the cleavage recognition site for an inflammation based protease substantially as described above. In a particular embodiment the inflammation is rheumatoid arthritis, atherosclerotic cells, Crohn's disease, or central nervous system disease.
  • In a further aspect, the present invention provides a recombinant protein comprising an A chain of a ricin-like toxin, a B chain of a ricin-like toxin and a heterologous linker amino acid sequence, linking the A and B chains, wherein the linker sequence contains a cleavage recognition site for a cancer-specific protease. The A and/or the B chain may be those of ricin. In an embodiment, the cleavage recognition site is the cleavage recognition site for a cancer protease substantially as described above. In a particular embodiment, the cancer is T-cell or B-cell lymphoproliferative disease, ovarian cancer, pancreatic cancer, head and neck cancer, squamous cell carcinoma, gastrointestinal cancer, breast cancer, prostate cancer, non small cell lung cancer.
  • In a further aspect, the invention provides a pharmaceutical composition for treating a cell, such as an inflammatory cell or cancer cell, having a specific protease, comprising a recombinant protein of the invention and a pharmaceutically acceptable carrier, diluent or excipient.
  • In yet another aspect, the invention provides a method of inhibiting or destroying a cell having a specific protease, such as an inflammatory cell or a cancer cell, comprising the steps of preparing a recombinant protein of the invention having a heterologous linker sequence which contains a cleavage recognition site for the specific protease, and administering the recombinant protein to the cells. In an embodiment, the inflammatory state is rheumatoid arthritis, atherosclerotic cells, Crohn's disease, or central nervous system disease. In another embodiment, the cancer is T-cell or B-cell lymphoproliferative disease, ovarian cancer, pancreatic cancer, head and neck cancer, squamous cell carcinoma, gastrointestinal cancer, breast cancer, prostate cancer, non small cell lung cancer.
  • The present invention also relates to a method of treating a cell having a specific protease such as an inflammatory cell or a cancer cell, wherein the cells affected by the condition and which have a specific protease, are treated by administering an effective amount of one or more recombinant proteins of the invention to an animal in need thereof.
  • Still further, a process is provided for preparing a pharmaceutical for treating a cell having a specific protease, such as an inflammatory cell or a cancer cell, wherein cells affected by condition have a specific protease, the steps for preparing the pharmaceutical comprising the steps of preparing a purified and isolated nucleic acid having a nucleotide sequence encoding an A chain of a ricin-like toxin, a B chain of a ricin-like toxin and a heterologous linker amino acid sequence, linking the A and B chains, wherein the linker sequence contains a cleavage recognition site for the specific protease; introducing the nucleic acid into a host cell; expressing the nucleic acid in the host cell to obtain a recombinant protein comprising an A chain of a ricin-like toxin, a B chain of a ricin-like toxin and a heterologous linker amino acid sequence, linking the A and B chains wherein the linker sequence contains the cleavage recognition site for the specific protease; and suspending the protein in a pharmaceutically acceptable carrier, diluent or excipient.
  • Other features and advantages of the present invention will become apparent from the following detailed description. It should be understood, however, that the detailed description and the specific examples while indicating preferred embodiments of the invention are given by way of illustration only, since various changes and modifications within the spirit and scope of the invention will become apparent to those skilled in the art from this detailed description.
  • DESCRIPTION OF THE DRAWINGS
  • The invention will be better understood with reference to the drawings in which:
  • FIG. 1A shows the nucleotide sequence of the MMP-9 linker region of pAP301 (SEQ ID NOS:1-4);
  • FIG. 1B shows the nucleotide sequence of the pAP301 insert containing ricin and the MMP-9 linker (SEQ ID NO:5);
  • FIG. 1C shows the amino acid sequence of the PAP301 linker and the wild type ricin linker (SEQ ID NOS:6-7);
  • FIG. 2A shows the nucleotide sequence of the MMP-9 30 linker region of pAP302 (SEQ ID NOS:8-11);
  • FIG. 2B shows the nucleotide sequence of the pAP302 insert containing ricin and the MMP-9 linker (SEQ ID NO:12);
  • FIG. 2C shows the amino acid sequence of the PAP302 linker and the wild type ricin linker (SEQ ID NOS:13-14);
  • FIG. 3A shows the nucleotide sequence of the MMP-9 linker region of pAP303 (SEQ ID NOS:15-18);
  • FIG. 3B shows the nucleotide sequence of the pAP303 insert containing ricin and the MMP-9 linker (SEQ ID NO:19);
  • FIG. 3C shows the amino acid sequence of the PAP303 linker and the wild type ricin linker (SEQ ID NOS:20-21);
  • FIG. 4A shows the nucleotide sequence of the MMP-9 linker region of pAP304 (SEQ ID NOS:22-25);
  • FIG. 4B shows the nucleotide sequence of the pAP304 insert containing ricin and the MMP-9 linker (SEQ ID NO:26);
  • FIG. 4C shows the amino acid sequence of the PAP304 linker and the wild type ricin linker (SEQ ID NOS:27-28);
  • FIG. 5A shows the nucleotide sequence of the MMP-9 linker region of pAP305 (SEQ ID NOS:29-32);
  • FIG. 5B shows the nucleotide sequence of the pAP305 insert containing ricin and the MMP-9 linker (SEQ ID NO:33);
  • FIG. 5C shows the amino acid sequence of the PAP305 linker and the wild type ricin linker (SEQ ID NOS:34-35);
  • FIG. 6A shows the nucleotide sequence of the MMP-9 linker region of pAP308 (SEQ ID NOS:36-39);
  • FIG. 6B shows the nucleotide sequence of the pAP308 insert containing ricin and the MMP-9 linker (SEQ ID NO:40);
  • FIG. 6C shows the amino acid sequence of the pAP308 linker and the wild type ricin linker (SEQ ID NOS:41-42);
  • FIG. 7A shows the nucleotide sequence of the MMP-9 linker region of pAP309 (SEQ ID NOS:43-46);
  • FIG. 7B shows the nucleotide sequence of the pAP309 insert containing ricin and the MMP-9 linker (SEQ ID NO:47);
  • FIG. 7C shows the amino acid sequence of the PAP309 linker and the wild type ricin linker (SEQ ID NOS:48-49);
  • FIG. 8A shows the nucleotide sequence of the UPA linker region of pAP313 (SEQ ID NOS:50-53);
  • FIG. 8B shows the nucleotide sequence of the pAP313 insert containing ricin and the UPA linker (SEQ ID NO:54);
  • FIG. 8C shows the amino acid sequence of the PAP313 linker and the wild type ricin linker (SEQ ID NOS:55-56);
  • FIG. 9A shows the nucleotide sequence of the UPA linker region of pAP314 (SEQ ID NOS:57-60);
  • FIG. 9B shows the nucleotide sequence of the pAP314 insert containing ricin and the UPA linker (SEQ ID NO:61);
  • FIG. 9C shows the amino acid sequence of the PAP314 linker and the wild type ricin linker (SEQ ID NOS:62-63);
  • FIG. 10A shows the nucleotide sequence of the UPA linker region of pAP315 (SEQ ID NOS:64-67);
  • FIG. 10B shows the nucleotide sequence of the pAP315 insert containing ricin and the UPA linker (SEQ ID NO:68);
  • FIG. 10C shows the amino acid sequence of the PAP315 linker and the wild type ricin linker (SEQ ID NOS:69-70);
  • FIG. 11A shows the nucleotide sequence of the MMP-9 linker region of pAP316 (SEQ ID NOS:71-74);
  • FIG. 11B shows the nucleotide sequence of the pAP316 insert containing ricin and the MMP-9 linker (SEQ ID NO:75);
  • FIG. 11C shows the amino acid sequence of the PAP316 linker and the wild type ricin linker (SEQ ID NOS:76-77);
  • FIG. 12A shows the nucleotide sequence of the MMP-9 linker region of pAP318 (SEQ ID NOS:78-81);
  • FIG. 12B shows the nucleotide sequence of the pAP318 insert containing ricin and the MMP-9 linker (SEQ ID NO:82);
  • FIG. 12C shows the amino acid sequence of the PAP318 linker and the wild type ricin linker (SEQ ID NOS:83-84);
  • FIG. 13A shows the nucleotide sequence of the UPA linker region of pAP320 (SEQ ID NOS:85-88);
  • FIG. 13B shows the nucleotide sequence of the pAP320 insert containing ricin and the UPA linker (SEQ ID NO:89);
  • FIG. 13C shows the amino acid sequence of the PAP320 linker and the wild type ricin linker (SEQ ID NOS:90-91);
  • FIG. 14A shows the nucleotide sequence of the UPA linker region of pAP321 (SEQ ID NOS:92-95);
  • FIG. 14B shows the nucleotide sequence of the pAP321 insert containing ricin and the UPA linker (SEQ ID NO:96);
  • FIG. 14C shows the amino acid sequence of the PAP321 linker and the wild type ricin linker (SEQ ID NOS:97-98);
  • FIG. 15A shows the nucleotide sequence of the UPA linker region of pAP322 (SEQ ID NOS:99-102);
  • FIG. 15B shows the nucleotide sequence of the pAP322 insert containing ricin and the UPA linker (SEQ ID NO:103);
  • FIG. 15C shows the amino acid sequence of the PAP322 linker and the wild type ricin linker (SEQ ID NOS:104-105);
  • FIG. 16A shows the nucleotide sequence of the MMP-9 linker region of pAP323 (SEQ ID NOS:106-109);
  • FIG. 16B shows the nucleotide sequence of the pAP323 insert containing ricin and the MMP-9 linker (SEQ ID NO:110);
  • FIG. 16C shows the amino acid sequence of the PAP323 linker and the wild type ricin linker (SEQ ID NOS:111-112);
  • FIG. 17A shows the nucleotide sequence of the MMP-9 linker region of pAP324 (SEQ ID NOS:113-116);
  • FIG. 17B shows the nucleotide sequence of the pAP324 insert containing ricin and the MMP-9 linker (SEQ ID NO:117);
  • FIG. 17C shows the amino acid sequence of the PAP324 linker and the wild type ricin linker (SEQ ID NOS:118-119);
  • FIG. 18A shows the nucleotide sequence of the MMP-9 linker region of pAP325 (SEQ ID NOS:120-123);
  • FIG. 18B shows the nucleotide sequence of the pAP325 insert containing ricin and the MMP-9 linker (SEQ ID NO:124);
  • FIG. 18C shows the amino acid sequence of the PAP325 linker and the wild type ricin linker (SEQ ID NOS:125-126);
  • FIG. 19 shows the cleavage products of an MMP-9 digestion of PAP323, PAP324 and PAP325;
  • FIG. 20 is a graph showing the treatment of human tumour A431 with PAP304;
  • FIG. 21 is a graph showing the treatment of human tumour A431 with PAP305; and
  • FIG. 22 is a graph showing a significant delay in tumor growth in the murine tumor model.
  • DETAILED DESCRIPTION OF THE INVENTION 1. Nucleic Acid Molecules of the Invention
  • As mentioned above, the present invention relates to isolated and purified nucleic acid molecules encoding linker sequences. The present invention also relates to isolated and purified nucleic acid molecules encoding a recombinant toxic protein comprising (a) a nucleotide sequence encoding an A chain of a ricin-like toxin, (b) a nucleotide sequence encoding a B chain of a ricin-like toxin and (c) a nucleotide sequence encoding a linker amino acid sequence of the invention, linking the A and B chains. The heterologous linker sequence contains a cleavage recognition site for a specific protease.
  • The term “isolated and purified” as used herein refers to a nucleic acid substantially free of cellular material or culture medium when produced by recombinant DNA techniques, or chemical precursors, or other chemicals when chemically synthesized. An “isolated and purified” nucleic acid is also substantially free of sequences which naturally flank the nucleic acid (i.e. sequences located at the 5′ and 3′ ends of the nucleic acid) from which the nucleic acid is derived. The term “nucleic acid” is intended to include DNA and RNA and can be either double stranded or single stranded.
  • The term “linker sequence” as used herein refers to an internal amino acid sequence within the protein encoded by a nucleic acid molecule of the invention which contains residues linking the A and B chain of a ricin-like toxin so as to render the A chain incapable of exerting its toxic effect, for example catalytically inhibiting translation of an eukaryotic ribosome. The linker sequences of the invention are heterologous to the A and B chain of a ricin-like toxin. By heterologous is meant that the linker sequence is not a sequence native to the A or B chain of a ricin-like toxin or precursor thereof. However, preferably, the linker sequence may be of a similar length to the linker sequence of a ricin-like toxin and should not interfere with the role of the B chain in cell binding and transport into the cytoplasm. When the linker sequence is cleaved the A chain becomes active or toxic.
  • The nucleic acid molecule of the invention encoding a recombinant toxic protein is cloned by subjecting a preproricin cDNA clone to site-directed mutagenesis in order to generate a series of variants differing only in the sequence between the A and B chains (linker region). Oligonucleotides, corresponding to the extreme 5′ and 3′ ends of the preproricin gene are synthesized and used to PCR amplify the gene. Using the cDNA sequence for preproricin (Lamb et al., Eur. J. Biochem. 145:266-270 (1985)), several oligonucleotide primers are designed to flank the start and stop codons of the preproricin open reading frame.
  • The preproricin cDNA is amplified using the upstream primer Ricin-99 or Ricin-109 and the downstream primer Ricin1729C with Vent DNA polymerase (New England Biolabs) using standard procedures (Sambrook et al., Molecular Cloning: A Laboratory Manual, Second Edition, (Cold Spring Harbor Laboratory Press, 1989)). The purified PCR fragment encoding the preproricin cDNA is, then ligated into an Eco RV-digested pBluescript II SK plasmid (Stratagene), and is used to transform competent XL1-Blue cells (Stratagene). The cloned PCR product containing the putative preproricin gene is confirmed by DNA sequencing of the entire cDNA clone.
  • The preproricin cDNA clone is subjected to site directed mutagenesis; in order to generate a series of variants differing only in the sequence between the A and B chains (linker region). The wild-type preproricin linker region is replaced with the heterogenous linker sequences that are cleaved by the various specific proteases.
  • The linker regions of the variants encode a cleavage recognition sequence for a specific protease. The mutagenesis and cloning strategies used to generate a specific protease-sensitive linker variant are summarized in WO 98149311 to the present inventor. Briefly, the first step involves a DNA amplification using a set of mutagenic primers in combination with the two flanking primers Ricin-109Eco and Ricin1729C PstI. Restriction digested PCR fragments are gel purified and then ligated with PVL1393 which has been digested with Eco RI and PstI. Ligation reactions are used to transform competent XLI-Blue cells (Stratagene). Recombinant clones are identified by restriction digests of plasmid miniprep, DNA and the mutant linker sequences are confirmed by DNA sequencing.
  • The nucleotide sequences of the novel linker sequences of the invention are as follows: the nucleic acid sequence of pAP301 is shown in FIG. 1A; the nucleic acid sequence of pAP302 is shown in FIG. 2A; the nucleic acid sequence of pAP303 is shown in FIG. 3A; the nucleic acid sequence of pAP304 is shown in FIG. 4A; the nucleic acid sequence of pAP305 is shown in FIG. 5A; the nucleic acid sequence of pAP308 is shown in FIG. 6A; the nucleic acid sequence of pAP309 is shown in FIG. 7A; the nucleic acid sequence of pAP313 is shown in FIG. 8A; the nucleic acid sequence of pAP314 is shown in FIG. 9A; the nucleic acid sequence of pAP315 is shown in FIG. 10A; the nucleic acid sequence of pAP316 is shown in FIG. 11A; the nucleic acid sequence of pAP318 is shown in FIG. 12A; the nucleic acid sequence of pAP320 is shown in FIG. 13A; the nucleic acid sequence of pAP321 is shown in FIG. 14A; the nucleic acid sequence of pAP322 is shown in FIG. 15A; the nucleic acid sequence of pAP323 is shown in FIG. 16A; the nucleic acid sequence of pAP324 is shown in FIG. 17A; and the nucleic acid sequence of pAP325 is shown in FIG. 18A.
  • The nucleic acid molecule encoding a recombinant protein of the invention has sequences encoding an A chain of a ricin-like toxin, a B chain of a ricin-like toxin and a heterologous linker sequence containing a cleavage recognition site for a specific protease as described above. The nucleotide sequences encoding the recombinant proteins of the invention are shown in FIGS. 1B-18B. The nucleic acid may be expressed to provide a recombinant protein having an A chain of a ricin-like toxin, a B chain of a ricin-like toxin and a heterologous linker sequence containing a cleavage recognition site for a specific protease.
  • The nucleic acid molecule may comprise the A and/or B chain of ricin. The ricin gene has been cloned and sequenced, and the X-ray crystal structures of the A and B chains are published (Rutenber, E., et al. Proteins 10:240-250 (1991); Weston et al., Mol. Biol. 244:410-422 (1994); Lamb and Lord, Eur. J. Biochem. 14:265 (1985); Halling, K., et al., Nucleic Acids Res. 13:8019 (1985)). It will be appreciated that the invention includes nucleic acid molecules encoding truncations of A and B chains of ricin-like proteins and analogs and homologs of A and B chains of ricin-like proteins and truncations thereof (i.e., ricin-like proteins), as described herein. It will further be appreciated that variant forms of the nucleic acid molecules of the invention which arise by alternative splicing of an mRNA corresponding to a cDNA of the invention are encompassed by the invention.
  • Another aspect of the invention provides a nucleotide sequence which hybridizes under high stringency conditions to a nucleotide sequence encoding the A and/or B chains of a ricin-like protein. Appropriate stringency conditions which promote DNA hybridization are known to those skilled in the art, or can be found in Current Protocols in Molecular Biology, John Wiley & Sons, N.Y. (1989), 6.3.1 6.3.6. For example, 6.0× sodium chloride/sodium citrate (SSC) at about 45° C., followed by a wash of 2.0×SSC at 50° C. may be employed.
  • The stringency may be selected based on the conditions used in the wash step. By way of example, the salt concentration in the wash step can be selected from a high stringency of about 0.2×SSC at 50° C. In addition, the temperature in the wash step can be at high stringency conditions, at about 65° C.
  • The nucleic acid molecule may comprise the A and/or B chain of a ricin-like toxin. Methods for cloning ricin-like toxins are known in the art and are described, for example, in E.P. 466,222. Sequences encoding ricin or ricin-like A and B chains may be obtained by selective amplification of a coding region, using sets of degenerative primers or probes for selectively amplifying the coding region in a genomic or cDNA library. Appropriate primers may be selected from the nucleic acid sequence of A and B chains of ricin or ricin-like toxins. It is also possible to design synthetic oligonucleotide primers from the nucleotide sequences for use in PCR. Suitable primers may be selected from the sequences encoding regions of ricin-like proteins which are highly conserved, as described for example in U.S. Pat. No. 5,101,025 and E.P. 466,222.
  • A nucleic acid can be amplified from cDNA or genomic DNA using these oligonucleotide primers and standard PCR amplification techniques. The nucleic acid so amplified can be cloned into an appropriate vector and characterized by DNA sequence analysis. It will be appreciated that cDNA may be prepared from mRNA, by isolating total cellular mRNA by a variety of techniques, for example, by using the guanidinium-thiocyanate extraction procedure of Chirgwin et al. (Biochemistry 18, 5294-5299 (1979)). cDNA is then synthesized from the mRNA using reverse transcriptase (for example, Moloney MLV reverse transcriptase available from Gibco/BRL, Bethesda, Md., or AMV reverse transcriptase available from Seikagaku America, Inc., St. Petersburg, Fla.). It will be appreciated that the methods described above may be used to obtain the coding sequence from plants, bacteria or fungi, preferably plants, which produce known ricin-like proteins and also to screen for the presence of genes encoding as yet unknown ricin-like proteins.
  • A sequence containing a cleavage recognition site for a specific protease may be selected based on the disease or condition which is to be targeted by the recombinant protein. The cleavage recognition site may be selected from sequences known to encode a cleavage recognition site specific proteases of the disease or condition to be treated. Sequences encoding cleavage recognition sites may be identified by testing the expression product of the sequence for susceptibility to cleavage by the respective protease. A polypeptide containing the suspected cleavage recognition site may be incubated with a specific protease and the amount of cleavage product determined (Dilannit, 1990, J. Biol. Chem. 285: 17345-17354 (1990)). The specific protease may be prepared by methods known in the art and used to test suspected cleavage recognition sites.
  • The nucleic acid molecule of the invention may be prepared by site directed mutagenesis. For example, the cleavage site of a specific protease may be prepared by site directed mutagenesis of the homologous linker sequence of a proricin-like toxin. Procedures for cloning proricin-like genes, encoding a linker sequence are described in EP 466,222. Site directed mutagenesis may be accomplished by DNA amplification of mutagenic primers in combination with flanking primers.
  • The nucleic acid molecule of the invention may also encode a fusion protein. A sequence encoding a heterologous linker sequence containing a cleavage recognition site for a specific protease may be cloned from a cDNA or genomic library or chemically synthesized based on the known sequence of such cleavage sites. The heterologous linker sequence may then be fused in frame with the sequences encoding the A and B chains of the ricin-like toxin for expression as a fusion protein. It will be appreciated that a nucleic acid molecule encoding a fusion protein may contain a sequence encoding an A chain and a B chain from the same ricin-like toxin or the encoded A and B chains may be from different toxins. For example, the A chain may be derived from ricin and the B chain may be derived from abrin. A protein may also be prepared by chemical conjugation of the A and B chains and linker sequence using conventional coupling agents for covalent attachment.
  • An isolated and purified nucleic acid molecule of the invention which is RNA can be isolated by cloning a cDNA encoding an A and B chain and a linker into an appropriate vector which allows for transcription of the cDNA to produce an RNA molecule which encodes a protein of the invention. For example, a cDNA can be cloned downstream of a bacteriophage promoter, (e.g. a T7 promoter) in a vector, cDNA can be transcribed in vitro with T7 polymerase, and the resultant RNA can be isolated by standard techniques.
  • II. Novel Linkers and Recombinant Proteins of the Invention
  • As previously mentioned, the invention provides novel linker sequences. Preferably, the amino acid sequence of the linker is selected from: the amino acid sequence of PAP301 as shown in FIG. 1C; the amino acid sequence of PAP302 as shown in FIG. 2C; the amino acid sequence of PAP303 as shown in FIG. 3C; the amino acid sequence of PAP304 as shown in FIG. 4C; the amino acid sequence of PAP305 as shown in FIG. 5C; the amino acid sequence of PAP308 as shown in FIG. 6C; the amino acid sequence of PAP309 as shown in FIG. 7C; the amino acid sequence of PAP313 as shown in FIG. 8C; the amino acid sequence of PAP314 as shown in FIG. 9C; the amino acid sequence of PAP315 as shown in FIG. 10C; the amino acid sequence of PAP316 as shown in FIG. 11C; the amino acid sequence of PAP318 as shown in FIG. 12C; the amino acid sequence of PAP320 as shown in FIG. 13C; the amino acid sequence of PAP321 as shown in FIG. 14C; the amino acid sequence of PAP322 as shown in FIG. 15C; the amino acid sequence of PAP323 as shown in FIG. 16C; the amino acid sequence of PAP324 as shown in FIG. 17C; and the amino acid sequence of PAP325 as shown in FIG. 18C.
  • The present invention also provides recombinant proteins which incorporate the A and B chains of a ricin-like toxin linked by a heterologous linker sequence containing a cleavage recognition site for a specific protease as described above. It is an advantage of the recombinant proteins of the invention that they are non-toxic until the A chain is liberated from the B chain by specific cleavage of the linker by the target specific protease.
  • The recombinant protein may be used to specifically target for example, cancer cells in the absence of additional specific cell-binding components to target cancer cells. It is a further advantage that the specific protease cleaves the heterologous linker intracellularly thereby releasing the toxic A chain directly into the cytoplasm of the target cell. As a result, said cells are specifically targeted and normal cells are not directly exposed to the activated free A chain.
  • Ricin is a plant derived ribosome inhibiting protein which blocks protein synthesis in eukaryotic cells. Ricin may be derived from the seeds of Ricinus communis (castor oil plant). The ricin toxin is a glycosylated heterodimer with A and B chain molecular masses of 30,625 Da and 31,431 Da respectively. The A chain of ricin has an N-glycosidase activity and catalyzes the excision of a specific adenine residue from the 28S rRNA of eukaryotic ribosomes (Endo, Y; & Tsurugi, K. J. Biol. Chem. 262:8128 (1987)). The B chain of ricin, although not toxic in itself, promotes the toxicity of the A chain by binding to galactose residues on the surface of eukaryotic cells and stimulating receptor-mediated endocytosis of the toxin molecule (Simmons et al., Biol. Chem. 261:7912 (1986)).
  • All protein toxins are initially produced in an inactive, precursor form. Ricin is initially produced as a single polypeptide (preproricin) with a 35 amino acid N-terminal presequence and 12 amino acid linker between the A and B chains. The pre-sequence is removed during translocation of the ricin precursor into the endoplasmic reticulum (Lord, J. M., Eur. J. Biochem. 146:403-409 (1985) and Lord, J. M., Eur. J. Biochem. 146:411-416 (1985)). The proricin is then translocated into specialized organelles called protein bodies where a plant protease cleaves the protein at a linker region between the A and B chains (Lord, J. M. et al., FASAB journal 8:201-208 (1994)). The two chains, however, remain covalently attached by an interchain disulfide bond (cysteine 259 in the A chain to cysteine 4 in the B chain) and mature disulfide linked ricin is stored in protein bodies inside plant cells. The A chain is inactive in the proricin (O'Hare, M., et al., FEBS Lett. 273:200-204 (1990)) and it is inactive in the disulfide-linked mature ricin (Richardson, P. T. et al., FEBS Lett. 255:15-20 (1989)). The ribosomes of the castor bean plant are themselves susceptible to inactivation by ricin A chain; however, as there is no cell surface galactose to permit B chain recognition the A chain cannot re-enter the cell.
  • Ricin-like proteins include, but are not limited to, bacterial, fungal and plant toxins which have A and B chains and inactivate ribosomes and inhibit protein synthesis. The A chain is an active polypeptide subunit which is responsible for the pharmacologic effect of the toxin. In most cases the active component of the A chain is an enzyme. The B chain is responsible for binding the toxin to the cell surface and is thought to facilitate entry of the A chain into the cell cytoplasm. The A and B chains in the mature toxins are linked by disulfide bonds. The toxins most similar in structure to ricin are plant toxins which have one A chain and one B chain. Examples of such toxins include abrin which may be isolated from the seeds of Abrus precatorius, modeccin, volkensin and viscumin.
  • Ricin-like bacterial proteins include diphtheria toxin, which is produced by Corynebacterium diphtheriae, Pseudomonas exotoxin and cholera toxin. It will be appreciated that the term ricin-like toxins is also intended to include the A chain of those toxins which have only an A chain. The recombinant proteins of the invention could include the A chain of these toxins conjugated to, or expressed as, a recombinant protein with the B chain of another toxin. Examples of plant toxins having only an A chain include trichosanthin, MMC and pokeweed antiviral proteins, dianthin 30, dianthin 32, crotin II, curcin 11 and wheat germ inhibitor. Examples of fungal toxins having only an A chain include alpha-sarcin, restrictocin, mitogillin, enomycin, phenomycin. Examples of bacterial toxins having only an A chain include cytotoxin from Shigella dysenteriae and related Shiga-like toxins. Recombinant trichosanthin and the coding sequence thereof is disclosed in U.S. Pat. Nos. 5,101,025 and 5,128,460.
  • In addition to the entire A or B chains of a ricin-like toxin, it will be appreciated that the recombinant protein of the invention may contain only that portion of the A chain which is necessary for exerting its cytotoxic effect. For example, the first 30 amino acids of the ricin A chain may be removed resulting in a truncated A chain which retains toxic activity. The truncated ricin or ricin-like A chain may be prepared by expression of a truncated gene or by proteolytic degradation, for example with Nagarase (Funmatsu et al., Jap. J. Med. Sci. Biol. 23:264-267 (1970)). Similarly, the recombinant protein of the invention may contain only that portion of the B chain necessary for galactose recognition, cell binding and transport into the cell cytoplasm. Truncated B chains are described for example in E.P. 145,111. The A and B chains may be glycosylated or non-glycosylated. Glycosylated A and B chains may be obtained by expression in the appropriate host cell capable of glycosylation. Non-glycosylated chains may be obtained by expression in nonglycosylating host cells or by treatment to remove or destroy the carbohydrate moieties.
  • The proteins of the invention may be prepared using recombinant DNA methods. Accordingly, the nucleic acid molecules of the present invention may be incorporated in a known manner into an appropriate expression vector which ensures good expression of the protein. Possible expression vectors include but are not limited to cosmids, plasmids, or modified viruses (e.g. replication defective retroviruses, adenoviruses and adeno-associated viruses), so long as the vector is compatible with the host cell used. The expression vectors are “suitable for transformation of a host cell”, which means that the expression vectors contain a nucleic acid molecule of the invention and regulatory sequences selected on the basis of the host cells to be used for expression, which is operatively linked to the nucleic acid molecule. Operatively linked is intended to mean that the nucleic acid is linked to regulatory sequences in a manner which allows expression of the nucleic acid.
  • The invention therefore contemplates a recombinant expression vector of the invention containing a nucleic acid molecule of the invention, or a fragment thereof, and the necessary regulatory sequences for the transcription and translation of the inserted protein-sequence.
  • Suitable regulatory sequences may be derived from a variety of sources, including bacterial, fungal, viral, mammalian, or insect genes (For example, see the regulatory sequences described in Goeddel, Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, Calif. (1990). Selection of appropriate regulatory sequences is dependent on the host cell chosen as discussed below, and may be readily accomplished by one of ordinary skill in the art. Examples of such regulatory sequences include: a transcriptional promoter and enhancer or RNA polymerase binding sequence, a ribosomal binding sequence, including a translation initiation signal. Additionally, depending on the host cell chosen and the vector employed, other sequences, such as an origin of replication, additional DNA restriction sites, enhancers, and sequences conferring inducibility of transcription may be incorporated into the expression vector. It will also be appreciated that the necessary regulatory sequences may be supplied by the native A and B chains and/or its flanking regions.
  • The recombinant expression vectors of the invention may also contain a selectable marker gene which facilitates the selection of host cells transformed or transfected with a recombinant molecule of the invention. Examples of selectable marker genes are genes encoding a protein such as G418 and hygromycin which confer resistance to certain drugs, β-galactosidase, chloramphenicol acetyltransferase, firefly luciferase, or an immunoglobulin or portion thereof such as the Fc portion of an immunoglobulin preferably IgG. Transcription of the selectable marker gene is monitored by changes in the concentration of the selectable marker protein such as β-galactosidase, chloramphenicol acetyltransferase, or firefly luciferase. If the selectable marker gene encodes a protein conferring antibiotic resistance such as neomycin resistance transformant cells can be selected with G418. Cells that have incorporated the selectable marker gene will survive, while the other cells die. This makes it possible to visualize and assay for expression of recombinant expression vectors of the invention and in particular to determine the effect of a mutation on expression and phenotype. It will be appreciated that selectable markers can be introduced on a separate vector from the nucleic acid of interest.
  • The recombinant expression vectors may also contain genes which encode a fusion moiety which provides increased expression of the recombinant protein; increased solubility of the recombinant protein; and aid in the purification of the target recombinant protein by acting as a ligand in affinity purification. For example, a proteolytic cleavage site may be added to the target recombinant protein to allow separation of the recombinant protein from the fusion moiety subsequent to purification of the fusion protein. Typical fusion expression vectors include pGEX (Amrad Corp., Melbourne, Australia), pMal (New England Biolabs, Beverly, Mass.) and pRIT5 (Pharmacia, Piscataway, N.J.) which fuse glutathione S-transferase (GST), maltose E binding protein, or protein A, respectively, to the recombinant protein.
  • Recombinant expression vectors can be introduced into host cells to produce a transformed host cell. The term “transformed host cell” is intended to include prokaryotic and eukaryotic cells which have been transformed or transfected with a recombinant expression vector of the invention. The terms “transformed with”, “transfected with”, “transformation” and “transfection” are intended to encompass introduction of nucleic acid (e.g. a vector) into a cell by one of many possible techniques known in the art. Prokaryotic cells can be transformed with nucleic acid by, for example, electroporation or calcium-chloride mediated transformation. Nucleic acid can be introduced into mammalian cells via conventional techniques such as calcium phosphate or calcium chloride co-precipitation, DEAE-dextran mediated transfection, lipofectin, electroporation or microinjection. Suitable methods for transforming and transfecting host cells can be found in Sambrook et al. (Molecular Cloning: A Laboratory Manual, 2nd Edition, Cold Spring Harbor Laboratory press (1989)), and other laboratory textbooks.
  • Suitable host cells include a wide variety of prokaryotic and eukaryotic host cells. For example, the proteins of the invention may be expressed in bacterial cells such as E. coli, insect cells (using baculovirus), yeast cells or mammalian cells. Other suitable host cells can be found in Goeddel, Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, Calif. (1991).
  • More particularly, bacterial host cells suitable for carrying out the present invention include E. coli, B. subtilis, Salmonella typhimurium, and various species within the genus Pseudomonas, Streptomyces, and Staphylococcus, as well as many other bacterial species well known to one of ordinary skill in the art. Suitable bacterial expression vectors preferably comprise a promoter which functions in the host cell, one or more selectable phenotypic markers, and a bacterial origin of replication. Representative promoters include the β-lactamase (penicillinase) and lactose promoter system (see Chang et al., Nature 275:615 (1978)), the trp promoter (Nichols and Yanofsky, Meth in Enzymology 101:155, (1983) and the tac promoter (Russell et al., Gene 20:231, (1982)). Representative selectable markers include various antibiotic resistance markers such as the kanamycin or ampicillin resistance genes. Suitable expression vectors include but are not limited to bacteriophages such as lambda derivatives or plasmids such as pBR322 (Bolivar et al., Gene 2:9 S, (1977)), the pUC plasmids pUC18, pUC19, pUC118, pUC119 (see Messing, Meth in Enzymology 101:20-77, 1983 and Vieira and Messing, Gene 19:259-268 (1982)), and pNH8A, pNH16a, pNH18a, and Bluescript M13 (Stratagene, La Jolla, Calif.).
  • Typical fusion expression vectors which may be used are discussed above, e.g. pGEX (Amrad Corp., Melbourne, Australia), pMAL (New England Biolabs, Beverly, Mass.) and pRITS (Pharmacia, Piscataway, N.J.). Examples of inducible non-fusion expression vectors include pTrc (Arnann et al., Gene 69:301-315 (1988)) and pET 11d (Studier et al., Gene Expression Technology: Methods in Enzymology 185, Academic Press, San Diego, Calif., 60-89 (1990)).
  • Yeast and fungi host cells suitable for carrying out the present invention include, but are not limited to Saccharomyces cerevisae, the genera Pichia or Kluyveromyces and various species of the genus Aspergillus. Examples of vectors for expression in yeast S. cerivisae include pYepSec1 (Baldari. et al., Embo J. 6:229-234 (1987)), pMFa (Kurjan and Herskowitz, Cell 30:933-943 (1982)), pJRY88 (Schultz et al., Gene 54:113-123 (1987)), and pYES2 (Invitrogen Corporation, San Diego, Calif.). Protocols for the transformation of yeast and fungi are well known to those of ordinary skill in the art (see Hinnen et al., Proc. Natl. Acad. Sci. USA 75:1929 (1978); Itoh et al., J. Bacteriology 153:163 (1983), and Cullen et al. (Biol Technology 5:369 (1987)).
  • Mammalian cells suitable for carrying out the present invention include, among others: COS (e.g., ATCC No. CRL 1650 or 1651), BHK (e.g. ATCC No. CRL 6281), CHO (ATCC No. CCL 61), HeLa (e.g., ATCC No. CCL 2), 293 (ATCC No. 1573) and NS-1 cells. Suitable expression vectors for directing expression in mammalian cells generally include a promoter (e.g., derived from viral material such as polyoma, Adenovirus 2, cytomegalovirus and Simian Virus 40), as well as other transcriptional and translational control sequences. Examples of mammalian expression vectors include pCDM8 (Seed, B., Nature 329:840 (1987)) and pMT2PC (Kaufman et al., EMBO J. 6:187-195 (1987)).
  • Given the teachings provided herein, promoters, terminators, and methods for introducing expression vectors of an appropriate type into plant, avian, and insect cells may also be readily accomplished. For example, within one embodiment, the proteins of the invention may be expressed from plant cells (see Sinkar et al., J. Biosci (Bangalore) 11:47-58 (1987), which reviews the use of Agrobacterium rhizogenes vectors; see also Zambryski et al., Genetic Engineering, Principles and Methods, Hollaender and Setlow (eds.), Vol. VI, pp. 253-278, Plenum Press, New York (1984), which describes the use of expression vectors for plant cells, including, among others, PAPS2022, PAPS2023, and PAPS2034)
  • Insect cells suitable for carrying out the present invention include cells and cell lines from Bombyx, Trichoplusia or Spodotera species. Baculovirus vectors available for expression of proteins in cultured insect cells (SF 9 cells) include the pAc series (Smith et al., Mol. Cell. Biol. 3:2156-2165 (1983)) and the pVL series (Lucklow, V. A., and Summers, M. D., Virology 170:31-39 (1989)). Some baculovirus-insect cell expression systems suitable for expression of the recombinant proteins of the invention are described in PCT/US/02442.
  • Alternatively, the proteins of the invention may also be expressed in non-human transgenic animals such as, rats, rabbits, sheep and pigs (Hammer et al. Nature 315:680-683 (1985); Palmiter et al. Science 222:809-814 (1983); Brinster et al. Proc. Natl. Acad. Sci. USA 82:4438-4442 (1985); Palmiter and Brinster Cell 41:343-345 (1985) and U.S. Pat. No. 4,736,866).
  • The proteins of the invention may also be prepared by chemical synthesis using techniques well known in the chemistry of proteins such as solid phase synthesis (Merrifield, J. Am. Chem. Assoc. 85:2149-2154 (1964)) or synthesis in homogenous solution (Houbenweyl, Methods of Organic Chemistry, ed. E. Wansch, Vol. 15 I and II, Thieme, Stuttgart (1987)).
  • The present invention also provides proteins comprising an A chain of a ricin-like toxin, a B chain of a ricin-like toxin and a heterologous linker amino acid sequence linking the A and B chains, wherein the linker sequence contains a cleavage recognition site for a specific protease. Such a protein could be prepared other than by recombinant means, for example by chemical synthesis or by conjugation of A and B chains and a linker sequence isolated and purified from their natural plant, fungal or bacterial source. Such A and B chains could be prepared having the glycosylation pattern of the native ricin-like toxin.
  • N-terminal or C-terminal fusion proteins comprising the protein of the invention conjugated with other molecules, such as proteins may be prepared by fusing, through recombinant techniques. The resultant fusion proteins contain a protein of the invention fused to the selected protein or marker protein as described herein. The recombinant protein of the invention may also be conjugated to other proteins by known techniques. For example, the proteins may be coupled using heterobifunctional thiol-containing linkers as described in WO 90/10457, N-succinimidyl-3-(2-pyridyldithio-proprionate) or N-succinimidyl-5 thioacetate. Examples of proteins which may be used to prepare fusion proteins or conjugates include cell binding proteins such as immunoglobulins, hormones, growth factors, lectins, insulin, low density lipoprotein, glucagon, endorphins, transferrin, bombesin, asialoglycoprotein glutathione-S-transferase (GST), hemagglutinin (HA), and truncated myc.
  • III. Utility of the Nucleic Acid Molecules and Proteins of the Invention (a) Therapeutic Methods
  • As mentioned above, matrix metalloproteinases (MMPs or matrixins) are zinc-dependent proteinases and the expression of MMP genes is reported to be activated in inflammatory disorders (e.g. rheumatoid arthritis) and malignancy. In addition, there are reports of increased activation and expression of urokinase type plasminogen activator in inflammatory disorders such a rheumatoid arthritis (Slot, O., et al. 1999), osteoarthritis (Pap, G. et al., 2000), atherosclerotic cells (Falkenberg, M., et al., 1998) Crohn's disease (Desreumaux P, et al. 1999), central nervous system disease (Cuzner and Opdenakker, 1999) as well as in malignancy. Accordingly, the recombinant proteins of the invention may be used to specifically inhibit or destroy cells that contain a specific protease that can cleave the linker sequence of the recombinant protein. More particularly, the recombinant proteins of the invention may be used to specifically inhibit or destroy cancer cells that contain a protease that can cleave the linker sequence of the recombinant protein.
  • It is an advantage of the recombinant proteins of the invention that they have specificity for cells that contain a specific protease, including those of inflammatory disorders and cancer cells, without the need for a cell binding component. The ricin-like B chain of the recombinant proteins recognize galactose moieties on the cell surface and ensure that the protein is taken up by, for example, a cancer cell and released into the cytoplasm. When the protein is internalized into a normal cell, cleavage of the heterologous linker would not occur in the absence of the specific protease, and the A chain will remain inactive bound to the B chain. Conversely, when the protein is internalized into a cell having a specific protease, the specific protease will cleave the cleavage recognition site in the linker thereby releasing the toxic A chain.
  • Accordingly, the present invention provides a method of inhibiting or destroying cells having a specific protease, for examples inflammatory cells or cancer cells, comprising contacting such cells with an effective amount of a recombinant protein or a nucleic acid molecule encoding a recombinant protein of the invention. The present invention also provides a method of treating a cell having a specific protease, comprising administering an effective amount of a recombinant protein or a nucleic acid molecule encoding a recombinant protein of the invention to an animal in need thereof.
  • The term “effective amount” as used herein means an amount effective, at dosages and for periods of time necessary to achieve the desired result.
  • The term “animal” as used herein means any member of the animal kingdom including all mammals, birds, fish, reptiles and amphibians. Preferably, the animal to be treated is a mammal, more preferably a human.
  • The specificity of a recombinant protein of the invention may be tested by treating the protein with the specific protease which is thought to be specific for the cleavage recognition site of the linker and assaying for cleavage products. For example, specific proteases may be isolated from cancer cells, or they may be prepared recombinantly, for example following the procedures in Darket et al. (J. Biol. Chem. 254:2307-2312 (1988)). The cleavage products may be identified for example based on size, antigenicity or activity. The toxicity of the recombinant protein may be investigated by subjecting the cleavage products to an in vitro translation assay in cell lysates, for example using Brome Mosaic Virus mRNA as a template. Toxicity of the cleavage products may be determined using a ribosomal inactivation assay (Westby et al., Bioconjugate Chem. 3:377-382 (1992)). The effect of the cleavage products on protein synthesis may be measured in standardized assays of in vitro translation utilizing partially defined cell free systems composed for example of a reticulocyte lysate preparation as a source of ribosomes and various essential cofactors, such as mRNA template and amino acids. Use of radiolabelled amino acids in the mixture allows quantitation of incorporation of free amino acid precursors into trichloroacetic acid precipitable proteins. Rabbit reticulocyte lysates may be conveniently used (O'Hare, FEBS Lett. 273:200-204 (1990)).
  • The ability of the recombinant proteins of the invention to selectively inhibit or destroy cells having specific proteases may be readily tested in vitro using cell lines having the specific protease, such as cancer cell lines. The selective inhibitory effect of the recombinant proteins of the invention may be determined, for example, by demonstrating the selective inhibition of cellular proliferation in cancer cells or infected cells.
  • Toxicity may also be measured based on cell viability, for example, the viability of cancer and normal cell cultures exposed to the recombinant protein may be compared. Cell viability may be assessed by known techniques, such as trypan blue exclusion assays.
  • In another example, a number of models may be used to test the cytotoxicity of recombinant proteins having a heterologous linker sequence containing a cleavage recognition site for a cancer associated matrix metalloprotease. Thompson, E. W. et al. (Breast Cancer Res. Treatment 31:357-370 (1994)) has described a model for the determination of invasiveness of human breast cancer cells in vitro by measuring tumour cell-mediated proteolysis of extracellular matrix and tumour cell invasion of reconstituted basement membrane (collagen, laminin, fibronectin, Matrigel or gelatin). Other applicable cancer cell models include cultured ovarian adenocarcinoma cells (Young, T. N. et al. Gynecol. Oncol. 62:89-99 (1996); Moore, D. H. et al. Gynecol. Oncol. 65:78-82 (1997)), human follicular thyroid cancer cells (Demeure, M. J. et al., World J. Surg. 16:770-776 (1992)), human melanoma (A-2058) and fibrosarcoma (HT-1080) cell lines (Mackay, A. R. et al. Lab. Invest. 70:781 783 (1994)), and lung squamous (HS-24) and adenocarcinoma (SB-3) cell lines (Spiess, E. et al. J. Histochem. Cytochem. 42:917-929 (1994)). An in vivo test system involving the implantation of tumours and measurement of tumour growth and metastasis in athymic nude mice has also been described (Thompson, E. W. et al., Breast Cancer Res. Treatment 31:357-370 (1994); Shi, Y. E. et al., Cancer Res. 53:1409-1415 (1993)).
  • Although the primary specificity of the proteins of the invention for cells having a specific protease is mediated by the specific cleavage of the cleavage recognition site of the linker, it will be appreciated that specific cell binding components may optionally be conjugated to the proteins of the invention. Such cell binding components may be expressed as fusion proteins with the proteins of the invention or the cell binding component may be physically or chemically coupled to the protein component. Examples of suitable cell binding components include antibodies to cancer proteins.
  • Antibodies having specificity for a cell surface protein may be prepared by conventional methods. A mammal, (e.g. a mouse, hamster, or rabbit) can be immunized with an immunogenic form of the peptide which elicits an antibody response in the mammal Techniques for conferring immunogenicity on a peptide include conjugation to carriers or other techniques well known in the art. For example, the peptide can be administered in the presence of adjuvant. The progress of immunization can be monitored by detection of antibody titers in plasma or serum. Standard ELISA or other immunoassay procedures can be used with the immunogen as antigen to assess the levels of antibodies. Following immunization, antisera can be obtained and, if desired, polyclonal antibodies isolated from the sera.
  • To produce monoclonal antibodies, antibody producing cells (lymphocytes) can be harvested from an immunized animal and fused with myeloma cells by standard somatic cell fusion procedures thus immortalizing these cells and yielding hybridoma cells. Such techniques are well known in the art, (e.g. the hybridoma technique originally developed by Kohler and Milstein (Nature 256:495-497 (1975)) as well as other techniques such as the human B-cell hybridoma technique (Kozbor et al., Immunol. Today 4:72 (1983)), the EBV-hybridoma technique to produce human monoclonal antibodies (Cole et al., Methods Enzymol, 121:140-67 (1986)), and screening of combinatorial antibody libraries (Huse et al., Science 246:1275 (1989)). Hybridoma cells can be screened immunochemically for production of antibodies specifically reactive with the peptide and the monoclonal antibodies can be isolated.
  • The term “antibody” as used herein is intended to include fragments thereof which also specifically react with a cell surface component. Antibodies can be fragmented using conventional techniques and the fragments screened for utility in the same manner as described above. For example, F(ab′)2 fragments can be generated by treating antibody with pepsin. The resulting F(ab′)2 fragment can be treated to reduce disulfide bridges to produce Fab fragments.
  • Chimeric antibody derivatives, i.e., antibody molecules that combine a non-human animal variable region and a human constant region are also contemplated within the scope of the invention. Chimeric antibody molecules can include, for example, the antigen binding domain from an antibody of a mouse, rat, or other species, with human constant regions. Conventional methods may be used to make chimeric antibodies containing the immunoglobulin variable region which recognizes a cell surface antigen (See, for example, Morrison et al., Proc. Natl. Acad. Sci. U.S.A. 81:6851 (1985); Takeda et al., Nature 314:452 (1985), Cabilly et al., U.S. Pat. No. 4,816,567; Boss et al., U.S. Pat. No. 4,816,397; Tanaguchi et al., E.P. Patent No. 171,496; European Patent No. 173,494; United Kingdom Patent No. GB 2177096B). It is expected that chimeric antibodies would be less immunogenic in a human subject than the corresponding non-chimeric antibody.
  • Monoclonal or chimeric antibodies specifically reactive against cell surface components can be further humanized by producing human constant region chimeras, in which parts of the variable regions, particularly the conserved framework regions of the antigen-binding domain, are of human origin and only the hypervariable regions are of non-human origin. Such immunoglobulin molecules may be made by techniques known in the art, (e.g. Teng et al., Proc. Natl. Acad. Sci. U.S.A., 80:7308-7312 (1983); Kozbor et al., Immunology Today 4:7279 (1983); Olsson et al., Meth. Enzymol., 92:3-16 (1982), and PCT Publication WO92/06193 or EP 239,400). Humanized antibodies can also be commercially produced (Scotgen Limited, 2 Holly Road, Twickenham, Middlesex, Great Britain.)
  • Specific antibodies, or antibody fragments, reactive against cell surface components may also be generated by screening expression libraries encoding immunoglobulin genes, or portions thereof, expressed in bacteria with cell surface components. For example, complete Fab fragments, VH regions and FV regions can be expressed in bacteria using phage expression libraries (See for example Ward et al., Nature 341:544-546 (1989); Huse et al., Science 246:1275-1281 (1989); and McCafferty et al., Nature 348:552-554 (1990)). Alternatively, a SCID-hu mouse, for example the model developed by Genpharm, can be used to produce antibodies, or fragments thereof.
  • (b) Pharmaceutical Compositions
  • The proteins and nucleic acids of the invention may be formulated into pharmaceutical compositions for administration to subjects in a biologically compatible form suitable for administration in vivo. By “biologically compatible form suitable for administration in vivo” is meant a form of the substance to be administered in which any toxic effects are outweighed by the therapeutic effects. The substances may be administered to living organisms including humans, and animals. Administration of a therapeutically active amount of the pharmaceutical compositions of the present invention is defined as an amount effective, at dosages and for periods of time necessary to achieve the desired result. For example, a therapeutically active amount of a substance may vary according to factors such as the disease state, age, sex, and weight of the individual, and the ability of antibody to elicit a desired response in the individual. Dosage regime may be adjusted to provide the optimum therapeutic response. For example, several divided doses may be administered daily or the dose may be proportionally reduced as indicated by the exigencies of the therapeutic situation.
  • Accordingly, the present invention provides a pharmaceutical composition for treating cells having a specific protease comprising a recombinant protein or a nucleic acid encoding a recombinant protein of the invention and a pharmaceutically acceptable carrier, diluent or excipient.
  • The active substance may be administered in a convenient manner such as by injection (subcutaneous, intravenous, intramuscular, etc.), oral administration, inhalation, transdermal administration (such as topical cream or ointment, etc.), or suppository applications. Depending on the route of administration, the active substance may be coated in a material to protect the compound from the action of enzymes, acids and other natural conditions which may inactivate the compound.
  • The compositions described herein can be prepared by per se known methods for the preparation of pharmaceutically acceptable compositions which can be administered to subjects, such that an effective quantity of the active substance is combined in a mixture with a pharmaceutically acceptable vehicle. Suitable vehicles are described, for example, in Remington's Pharmaceutical Sciences (Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa., USA 1985). On this basis, the compositions include, albeit not exclusively, solutions of the substances in association with one or more pharmaceutically acceptable vehicles or diluents, and contained in buffered solutions with a suitable pH and iso-osmotic with the physiological fluids.
  • The pharmaceutical compositions may be used in methods for treating animals, including mammals, preferably humans, with cancer. It is anticipated that the compositions will be particularly useful for treating patients with B-cell lymphoproliferative disease and melanoma. The dosage and type of recombinant protein to be administered will depend on a variety of factors which may be readily monitored in human subjects. Such factors include the etiology and severity (grade and stage) of the neoplasia.
  • The following non-limiting examples are illustrative of the present invention:
  • EXAMPLES Example 1 Cloning and Expression of Proricin Variants Activated by Disease Specific Proteases Isolation of Total RNA
  • The preproricin gene was cloned from new foliage of the castor bean plant. Total messenger RNA was isolated according to established procedures (Sambrook et al., Molecular Cloning: A Lab Manual (Cold Spring Harbour Press, Cold Spring Harbour, (1989)) and cDNA generated using reverse transcriptase.
  • cDNA Synthesis
  • Oligonucleotides, corresponding to the extreme 5′ and 3′ ends of the preproricin gene were synthesized and used to PCR amplify the gene. Using the cDNA sequence for preproricin (Lamb et al., Eur. J. Biochem., 145:266-270, 1985), several oligonucleotide primers were designed to flank the start and stop codons of the preproricin open reading frame. The oligonucleotides were synthesized using an Applied Biosystems Model 392 DNA/RNA Synthesizer. First strand cDNA synthesis was primed using the oligonucleotide Ricin1729C. Three micrograms of total RNA was used as a template for oligo Ricin1729C (5′-ATAACTTGCTGCTCCTTTCA-3′ (SEQ ID NO:127)) primed synthesis of cDNA using Superscript II Reverse Transcriptase (BRL) following the manufacturer's protocol.
  • DNA Amplification and Cloning
  • The first strand cDNA synthesis reaction was used as template for DNA amplification by the polymerase chain reaction (PCR). The preproricin cDNA was amplified using the upstream primer Ricin-99 (5′-CCGGGAGGAAATACTATTGTAAT-3′ (SEQ ID NO:128)) and the downstream primer Ricin1729C with Vent DNA polymerase (New England Biolabs) using standard procedures (Sambrook et al., Molecular Cloning: A Laboratory Manual, Second Edition, (Cold Spring Harbor Laboratory Press, 1989)). Amplification was carried out in a Biometra thermal cycler (TRIO-Thermalcycler) using the following cycling parameters: denaturation 95° C. for 1 min., annealing 52° C. for 1 min., and extension 72° C. for 2 min., (33 cycles), followed by a final extension cycle at 72° C. for 10 min. The 1846 by amplified product was fractionated on an agarose gel (Sambrook et al., Molecular Cloning: A Laboratory Manual, Second Edition, (Cold Spring Harbor Laboratory Press, 1989), and the DNA purified from the gel slice using Qiaex resin (Qiagen) following the manufacturer's protocol. The purified PCR fragment encoding the preproricin cDNA was then ligated (Sambrook et al., Molecular Cloning: A Laboratory Manual, Second Edition, (Cold Spring Harbor Laboratory Press, 1989)) into an Eco RV digested pBluescript II SK plasmid (Stratagene), and used to transform competent XL1-Blue cells (Stratagene). Positive clones were confirmed by restriction digestion of purified plasmid DNA. Plasmid DNA was extracted using a Qiaprep Spin Plasmid Miniprep Kit (Qiagen).
  • DNA Sequencing
  • The cloned PCR product containing the putative preproricin gene (pAP144) was confirmed by DNA sequencing of the entire cDNA clone. Sequencing was performed using an Applied Biosystems 373A Automated DNA Sequencer, and confirmed by double-stranded dideoxy sequencing by the Sanger method using the Sequenase kit (USB) (see WO 98/49311).
  • Production and Cloning of Linker Variants
  • pAP144 cut with EcoR1 was used as target for PCR pairs employing the Ricin109-Eco oligonucleotide (Ricin-109Eco primer: 5-GGAGGAATCCGGAGATGAAACCGGGAGGAAATACTATTGTAAT-3 (SEQ ID NO:129)) and a mutagenic primer for the 5′ half of the linker as well as the Ricin1729PstI primer (Ricin 1729-PstI: 5GTAGGCGCTGCAGATAACTTGCTGTCCTTTCAG-3 (SEQ ID NO:130)) and a mutagenic primer for the 3′ half of the linker. The cycling conditions used for the PCRs were 98 degrees C. for 2 min.; 98° C. 1 min., 52° C. 1 min., 72° C. 1 min. 15 sec. (30 cycles); 72 degrees C. 10 min.; 4 degrees C. soak. The PCR products were then digested by EcoRI and PstI respectively, electrophoresed on an agarose gel, and the bands purified by via glass wool spin columns. Triple ligations comprising the PCR product pairs (corresponding halves of the new linker) and pVL1393 vector digested with EcoR1 and PstI were carried out. Recombinant clones were identified by restriction digests of plasmid miniprep DNA and the altered linkers confirmed by DNA sequencing. Note that all altered linker variants were cloned directly into the pVL1393 vector.
  • Isolation of Recombinant Baculoviruses
  • Insect cells S. frugiperda (Sf9), and Trichoplusia ni (Tn368 and BTI-TN-581-4 (High Five)) were maintained on EX-CELL 405 medium JRH Biosciences) supplemented with 10% total calf serum (Summers et al., A Manual of Methods of Baculovirus Vectors and Insect Cell Culture Procedures, (Texas Agricultural Experiment Station, 1987)). Two micrograms of recombinant pVL1393 DNA was co-transfected with 0.5 microgram of BaculoGold AcNPV DNA (Pharmingen) into 2×106 Tn368 insect cells following the manufacturer's protocol (Gruenwald et al., Baculovirus Expression Vector System: Procedures and Methods Manual, 2nd Edition, (San Diego, Calif., 1993)). On day 5 post-transfection, media were centrifuged and the supernatants tested in limiting dilution assays with Tn368 cells (Summers et al., A Manual of Methods of Baculovirus Vectors and Insect Cell Culture Procedures, (Texas Agricultural Experiment Station, 1987)). Recombinant viruses in the supernatants were then amplified by infecting Tn368 cells at a multiplicity of infection (moi) of 0.1, followed by collection of day 3 to 5 supernatants. A total of three rounds of amplification were performed for each recombinant following established procedures (Summers et al., A Manual of Methods of Baculovirus Vectors and Insect Cell Culture Procedures, (Texas Agricultural Experiment Station, 1987 and Gruenwald et al., Baculovirus Expression Vector System: Procedures and Methods Manual, 2nd Edition, (San Diego, Calif., 1993)).
  • Expression of Mutant Proricin
  • Recombinant baculoviruses were used to infect 1×107 Tn368 or Sf9 cells at an moi of 9 in EX-CELL 405 media (JRH Biosciences) with 25 mM a-lactose in spinner flasks. Media supernatants containing mutant proricins were collected 3 or 4 days post-infection.
  • Example 2 Harvesting and Affinity Column Purification of Pro-Ricin Variants
  • Protein samples were harvested three days post infection. The cells were removed by centrifuging the media at 8288 g for ten minutes using a GS3 (Sorvall) centrifuge rotor. The supernatant was further clarified by centrifuging at 25400 g using a SLA-1500 rotor (Sorvall) for 45 minutes. Protease inhibitor phenylmethylsulfonyl fluoride (Sigma) was slowly added to a final concentration of 1 mM. The samples were further prepared by adding a-lactose to a concentration of 20 mM (not including the previous lactose contained in the expression medium). The samples were concentrated to 700 mL using a Prep/Scale-TFF Cartridge (2.5 ft, 10K regenerated cellulose (Millipore)) and a Masterflex pump. The samples were then dialysed for 2 days in 1× Column Buffer (50 mM Tris, 100 mM NaCl, 0.02% NaN3, pH 7.5) using dialysis tubing (10 K MWCO, 32 mm flat width (Spectra/Por)). Subsequently, the samples were clarified by centrifuging at 25400 g using a SLA-1500 rotor (Sorvall) for 45 minutes.
  • Following centrifugation, the samples were degassed and applied at 4 degrees C. to a XK26/20 (Pharmacia) column (attached to a Pharmacia peristaltic pump, Pharmacia Single-path Monitor UV-1 Control and Optical Units, and Bromma LKB 2210 2-Channel Recorder) containing 20 mL a-Lactose Agarose Resin (Sigma). The column was washed for 3 hours with 1× Column buffer. Elution of proricin variant was performed by eluting with buffer (1× Column buffer (0.1% NaN3), 100 mM Lactose) until the baseline was again restored. The samples were concentrated using an Amicon 8050 concentrator (Amicon) with a YM10 76 mm membrane, utilizing argon gas to pressurize the chamber. The samples were further concentrated in Centricon 10 (Millipore) concentrators according to manufacturer's specifications.
  • Purification of Variant PAP-Protein by Gel Filtration Chromatography
  • In order to purify variant from processed material produced during fermentation, the protein was applied to a SUPERDEX 75 (16/60) column and SUPERDEX 200 (16/60) column (Pharmacia) connected in series equilibrated with 100 mM Tris, 200 mM NaCl, pH 7.5 containing 100 mM lactose and 1.0% β-mercaptoethanol (βME). The flow rate of the column was 0.15 mL/min and fractions were collected every 25 minutes. The UV (280 nm) trace was used to determine the approximate location of the purified PAP-protein and thus determine the samples for Western analysis.
  • Western Analysis of Column Fractions
  • Fractions eluted from the SUPERDEX columns (Pharmacia) were analyzed for purity using standard Western blotting techniques. An aliquot of 10 μL from each fraction was boiled in 1× sample buffer (62.6 mM Tris-Cl, pH 6.8, 4.4% 13ME, 2% sodium dodecyl sulfate (SDS), 5% glycerol (all from Sigma) and 0.002% bromophenol blue (Biorad)) for five minutes. Denatured samples were loaded on 12% Tris-Glycine Gels (Biorad) along with 50 ng of RCA60 (Sigma) and 5 μL of kaleidoscope prestained standards (Biorad). Electrophoresis was carried out for ninety minutes at 100V in 25 mM Tris-Cl, pH 8.3, 0.1% SDS, and 192 mM glycine using the BioRad Mini Protean II cells (Biorad).
  • Following electrophoresis gels were equilibrated in transfer buffer (48 mM Tris, 39 mM glycine, 0.0375% SDS, and 20% Methanol) for a few minutes. PVDF Biorad membrane was presoaked for one minute in 100% methanol, rinsed in ddH2O and two minutes in transfer buffer. Whatman paper was soaked briefly in transfer buffer. Five pieces of Whatman paper, membrane, gel, and another five pieces of Whatman paper were arranged on the bottom cathode (anode) of the Pharmacia Novablot transfer apparatus (Pharmacia). Transfer was for one hour at constant current (2 mA/cm2).
  • Transfer was confirmed by checking for the appearance of the prestained standards on the membrane. Non-specific sites on the membrane were blocked by incubating the blot for thirty minutes in 1× Phosphate Buffered Saline (1×PBS; 137 mM NaCl, 2.7 mM KCl, 8 mM Na2HPO4,1.5 mM KH2PO4, pH 7.4) with 5% skim milk powder (Carnation). Primary antibody rabbit anti-ricin, (Sigma) was diluted 1:3000 in 1×PBS containing 0.1% Tween 20 (Sigma) and 2.5% skim milk and incubated with blot for forty five minutes on a orbital shaker (VWR). Non-specifically bound primary antibody was removed by washing the blot for ten minutes with 1×PBS containing 0.2% Tween 20. This was repeated four times. Secondary antibody donkey anti-rabbit (Amersham) was incubated with the blot under the same conditions as the primary antibody. Excess secondary antibody was washed as described above. Blots were developed with the ECL Western Blotting detection reagents according to the manufacturer's instructions. Blots were exposed to Medtec's Full Speed Blue Film (Medtec) or Amersham's ECL Hyperfilm (Amersham) for one second to five minutes. Film was developed in a KODAK Automatic Developer.
  • Determination of Lectin Binding Ability of Pro-Ricin Variant
  • An Immulon 2 plate (VWR) was coated with 100 μl per well of 10 μg/ml of asialofetuin and left overnight at 4° C. The plate was washed with 3×300 μl per well with ddH2O using an automated plate washer (BioRad). The plate was blocked for one hour at 37° C. by adding 300 μL per well of PBS containing 1% ovalbumin. The plate was washed again as above. Proricin variant PAP-protein was added to the plate in various dilutions in 1× Column Buffer, (50 mM Tris, 100 mM NaCl, pH 7.5). A standard curve of RCA60 (Sigma) from 1-10 ng was also included. The plate was incubated for 1 h at 37° C. The plate was washed as above. Anti-ricin monoclonal antibody (Sigma) was diluted 1:3000 in 1×PBS containing 0.5% ovalburnin and 0.1% Tween-20, added at 100 μL per well and incubated for 1 h at 37° C. The plate was washed as above. Donkey anti-rabbit polyclonal antibody was diluted 1:3000 in 1×PBS containing 0.5% ovalburnin, 0.1% Tween-20, and added at 100 μL per well and incubated for 1 h at 37° C. The plate was given a final wash as described above. Substrate was added to plate at 100 μL per well (1 mg/mL o-phenylenediamine (in H2O), 1 μL/mL H2O2) and after development 25 μL of stop solution (20% H2SO4) was added and the absorbance read (A490 nm-A630 nm) using a SPECTRA MAX 340 plate reader (Molecular Devices).
  • Determination of PAP-Protein Activity Using the Rabbit Reticulocyte Assay
  • Ricin samples were prepared for reduction.
    • A) RCA60=3,500 ng/μL of RCA60+997 μL 1× Endo buffer (25 mM Tris, 25 mM KCl, 5 mM MGCl2, pH 7.6)
      • Reduction=95 μL of 10 ng/μL+5 μL β-mercaptoethanol
    • B) Ricin variants
      • Reduction=40 μL variant+2 μL β-mercaptoethanol
      • The ricin standard and the variants were incubated for 30 minutes at room temperature.
    Ricin—Rabbit Reticulocyte Lysate Reaction
  • The required number of 0.5 mL tubes were labelled. (2 25 tubes for each sample, + and −aniline). To each of the sample tubes 20 μL of 1× endo buffer was added, and 30 μL of buffer was added to the controls. To the sample tubes either 10 μL of 10 ng/μL, Ricin or 10 μL of variant was added. Finally, 30 μL of rabbit reticulocyte lysate was added to all the tubes. The samples were incubated for 30 minutes at 30° C. using the thermal block. Samples were removed from the 0.5 mL tube and contents added into a 1.5 mL tube containing 1 mL of TRIZOL (Gibco). Samples were incubated for 15 minutes at room temperature. After the incubation, 200 μL of chloroform was added, and the sample was vortexed and spun at 12,000 g for 15 minutes at 4° C. The top aqueous layer from the samples was removed and contents added to a 1 mL tube containing 500 μL of isopropanol. Samples were incubated for 15 minutes at room temperature and then centrifuged at 12,000 for 15 minutes at 4° C. Supernatant was removed and the pellets were washed with 1 mL of 70% ethanol. Centrifugation at 12,000 g for 5 minutes at 4° C. pelleted the RNA. All but approximately 20 μL of the supernatant was removed and the RNA pellet was allowed to air dry. Pellets from the other samples (+aniline samples) were dissolved in 20 μL of DEPC treated ddH2O. An 80 μL aliquot of 1 M aniline (distilled) with 2.8 M acetic acid was added to these RNA samples and transferred to a fresh 0.5 mL tube. The samples were incubated in the dark for 3 minutes at 60° C. RNA was precipitated by adding 100 μl, of 95% ethanol and 5 μL of 3M sodium acetate, pH 5.2 to each tube and centrifuging at 12,000 g for 30 minutes at 4° C. Pellets were washed with 1 mL 70% ethanol and centrifuged again at 12,000 g for 5 minutes at 4° C. to precipitate RNA. The supernatant was removed and air dried. These pellets were dissolved in 10 μL of 0.1× E buffer. To all samples, 10 μL of formamide loading dye was added. The RNA ladder (BRL) (8 μL of ladder+8 μL of loading dye) was also included. Samples were incubated for 2 minutes at 70° C. on the thermal block. Electrophoresis was carried out on the samples using 1.2% agarose, 50% formamide gels in 0.1× E buffer+0.2% SDS. The gel was run for 90 minutes at 75 volts. RNA was visualized by staining the gel in 1 μg/mL ethidium bromide in running buffer for 45 minutes. The gel was examined on a 302 nm UV box, photographed using the gel documentation system and saved to a computer disk.
  • Results: Protein Expression Yields
  • Aliquots were taken at each stop of the harvesting/purification and tested. Yields of functional ricin variant were determined by ELISA. Typical results of an 3400 mL prep of infected T ni cells are given below.
  • Aliquot μg PAP 304
    Before concentration and dialysis 14,472
    after concentration and dialysis 13,611
    alpha-Lactose agarose column flow through 418
    alpha-Lactose agarose column elution 8,682
  • Yield: 8,682/14,472=60% Purification of PAP-Protein and Western Analysis of Column Fractions
  • Partially purified PAP-protein was applied to Superdex 75 and 200 (16/60) columns connected in series in order to remove the contaminating non-specifically processed PAP-protein. Eluted fractions were tested via Western analysis as described above and the fractions containing the most pure protein were pooled, concentrated and dialyzed against 1×PBS buffer and then sterilized by filtration (Millipore). Final purified PAP-protein has less than 1% processed variant.
  • The purified PAP-protein was tested for susceptibility to cleavage by the particular protease and for activation of the A chain of the proricin variant, (inhibition of protein synthesis). Typically, PAP-protein was incubated with and without protease for a specified time period and then electrophoresed and blotted. Cleaved PAP-protein will run as two 30 kDa proteins (B is slightly larger) under reducing (SDS-PAGE) conditions. Unprocessed PAP-protein, which contains the linker region, will migrate at 60 kDa.
  • Activation of PAP-Protein Variant with Specific Protease
  • Activation of protease treated PAP-protein is based on the method of May et al. (EMBO Journal. 8 301-8, 1989). Activation of ricin A chain upon cleavage of the intermediary linker results in catalytic depurination of the adenosine 4325 residue of 28S or 26S rRNA. This depurination renders the molecule susceptible to amine-catalyzed hydrolysis by aniline of the phosphodiester bond on either side of the modification site. The result is a diagnostic 390 base band. As such, reticulocyte ribosomes incubated with biochemically purified ricin A chain, released the characteristic RNA fragment upon aniline treatment of isolated rRNA (May, M. J. et al. Embo. Journal, 8:301-308 at 302-303 (1989)). It is on this basis that the assay allows for the determination of activity of a ricin A chain which has been cleaved from the intact unit containing a particular variant linker sequence.
  • Example 3 In Vitro Protease Digestion of Proricin Variants:
  • Affinity-purified proricin variant is treated with individual disease-specific proteases to confirm specific cleavage in the linker region. Ricin-like toxin variants are eluted from the lactose-agarose matrix in protease digestion buffer (50 mM NaCl, 50 mM Na-acetate, pH 5.5, 1 mM dithiothreitol) containing 100 mM lactose. Proricin substrate is then incubated at 37° C. for 60 minutes with a disease-specific protease. The cleavage products consisting ricin A and B chains are identified using SDS/PAGE (Sambrook et al., Molecular Cloning: a Laboratory Manual, 2nd. ed., Cold Spring Harbor Press, 1989), followed by Western blot analysis using anti-ricin antibodies (Sigma). FIG. 19 shows the cleavage products of an MMP-9 digestion of PAP323, PAP324 and PAP325.
  • Matrix metalloproteinases may be prepared substantially as described by Lark, M. W. et al. (Proceedings of the 4th International Conference of the Inflammation Research Association Abstract 145 (1988)) and Welch, A. R. et al. (Arch. Biochem. Biophys. 324:59-64 (1995)).
  • Urokinase plasminogen activator may be prepared substantially as described by Holmberg, L. et al. (Biochim Biophys Acta, 445:215-222, (1976)) and Someno, T. et al. (J Biochem 97:1493-1500 (1985)).
  • Example 4 Cytotoxicity of Ricin and Ricin Variants on Cell Lines Cell Lines COS-I (African Green Monkey Kidney Cells)
  • This is an SV40 transformed cell line which was prepared from established simian cells CV-1. (Reference: Gluzman, Y. (1975) Cell, 23, 175-182)(ATCC CRL 1650).
  • HT-1080 Human Fibrosarcoma
  • (ATCC CCL 121) This cell line was shown to produce active MMP-9 in tissue culture. (References: Moore et al. (1997) Gynecologic Oncology 65, 83-88.)
  • Cell Preparation
  • After washing with 1×PBS (0.137 M NaCl, 2.68 mM KCl, 8.10 mM Na2HP04, 1.47 mM KH2PO4), cells in log phase growth were removed from plates with 1× trypsin/EDTA (Gibco/BRL). The cells were centrifuged at 1100 rpm for 3 min, resuspended in Dulbecco's Modified Eagle Medium containing 10% FBS and 1× pen/strep, and then counted using a haemocytometer. They were adjusted to a concentration of 5×104 cells·ml−1. One hundred microliters per well of cells was added to wells 2B-2G through to wells 9B-9G of a Falcon 96 well tissue culture plate. A separate 96 well tissue culture plate was used for each sample of Ricin or Ricin variant. The plates were incubated at 37° C. with 5% CO2 for 24 hours.
  • Toxin Preparation
  • The Ricin and Ricin variants were sterile filtered using a 0.22 μm filter (Millipore). The concentration of the sterile samples were then quantified by A280 and confirmed by BCA measurements (Pierce). For the variants digested with the MMP-9 protease in vitro, the digests were carried out as described in the digestion procedure for each protease. The digests were then diluted in the 1000 ng·ml−1 dilution and sterile filtered. Ricin and Ricin variants were serially diluted to the following concentrations: 1000 ng·ml−1, 100 ng·ml−1, 10 ng·ml−1, 1 ng·ml−1, 0.1 ng·ml−1, 0.01 ng·ml−1, 0.001 ng·ml−1 with media containing 10% FBS and 1× pen/strep.
  • Application of Toxin or Variants to Plates
  • Columns 2 to 9 were labeled: control, 1000 ng·ml−1, 100 ng·ml−1, 10 ng·ml−1, 1 ng·ml−1, 0.1 ng·ml−1, 0.01 ng·ml−1, 0.001 ng·ml−1 consecutively. The media was removed from all the sample wells with a multichannel pipettor. For each plate of variant and toxin, 50 μl of media was added to wells 2B to 2G as the control, and 50 μl of each sample dilution was added to the corresponding columns. The plates were incubated for one hour at 37° C. with 5% CO2, then washed once and replaced with media, then incubated for 48 hours at 37° C. with 5% CO2.
  • Sample Application
  • The whole amount of media (and/or toxin) was removed from each well with a multichannel pipettor, and replaced with 100 μl of the substrate mixture (Promega Cell Titer 96 Aqueous Non-Radioactive Cell Proliferation Assay Kit). The plates were incubated at 37° C. with 5% CO2 for 2 to 4 hours, and subsequently read with a Spectramax 340 96 well plate reader at 490 nm. The IC50 values were calculated using the GRAFIT software program.
  • Results
  • The results of the cytotoxicity assay are shown in Tables 1 to 4. In almost all cases the novel variants show preferential activation in the tumour cell line HT-1080 (human fibrosarcoma) as compared with the non-tumourogenic cell line COS-1 (immortalized cell line form the kidney of an African green monkey).
  • Example 5 Maximum Tolerable Dose Data
  • The protocol for the maximum tolerable dose (MTD) study involved three intravenous injections of variant, on days 1, 5 and 9, into the tail vein of either a Nude/SCID mouse. Three animals were used for each dose tested. The samples were diluted into saline solution containing 100 μg/mL Bovine Serum Albumin on the same day as the injection. Animals were observed for 14 days after dosing. Any surviving animals were euthanized after 14 days of study. The MTD value was defined as the highest dose of sample tested where all animals in the group survived. The results are presented in Table 5.
  • These results demonstrate that linkers of the invention in proricin variants decrease the toxicity of the recombinant proteins.
  • Example 6
  • In vivo Studies
  • (a) Protocol for A431 Animal Model Studies
  • Tumour growth was monitored daily by measuring tumour dimensions with calipers. The treatment initiation date was dependent on the rate of tumour growth. Four groups (4 mice per group) of mice develop tumours of the desired size (50 mm3-100 mm3) Such mice are weighed and treatment initiated. This treatment initiation date is considered as day 1, and the mice were given a bolus intravenous injection of variant on this day. Injections were administered through the lateral tail vein. The treatment groups are shown in Table 6.
  • All samples and buffer were made up in saline solution containing 100 μg/mL Bovine Serum Albumin.
  • (b) In Vivo Efficacy Studies
  • Subcutaneous A431 tumours were established in SCID mice. The tumours were treated with either PAP304 or PAP305 when the tumours reached 50 mm3 on Days 1, 5 and 9. The results shown in FIGS. 20 and 21 demonstrate that the linker decreases the toxicity of the variant (as compared with ricin) and the variants PAP304 and PAP305 are activated at or near the A431 (human epithelial carcinoma) solid tumour in mice. A very exciting result is shown in FIG. 20. In this study, the variant PAP304 was able to slow down the growth of A431 solid tumour (17 day delay), without any signs of dose limiting toxicity (e.g., no weight loss or death).
  • (c) Protocol and Efficacy for Testing PAP304 against P388 Murine Leukemia Tumour Model
  • Mice were grouped according to body weight. Animals (n=4) were inoculated (Day=0) with 1×106 cells implanted subcutaneously in the flank of the BDF-1 mouse in a volume of 50 μL with a 28 g needle. P388 murine leukemia cells from the ATCC tumor repository were maintained as an ascitic fluid in the BDF-1 mouse which were passaged to new mice weekly. The cells used for experiment were used within passage 3-20. For the experiment, cells were rinsed with Hanks Balanced Salt Solution, counted on a heamocytometer and diluted with HBSS to a concentration of 20×106 cells/ml. PAP304 was injected intravenously on days 3, 6 and 9 after tumour injection. The results are shown in FIG. 22. A significant delay in tumor growth in the murine tumor model.
  • Having illustrated and described the principles of the invention in a preferred embodiment, it should be appreciated to those skilled in the art that the invention can be modified in arrangement and detail without departure from such principles. We claim all modifications coming within the scope of the following claims.
  • All publications, patents and patent applications referred to herein are incorporated by reference in their entirety to the same extent as if each individual publication, patent or patent application was specifically and individually indicated to be incorporated by reference in its entirety.
  • FULL CITATIONS FOR CERTAIN REFERENCES REFERRED TO IN THE SPECIFICATION
    • Bever Jr., C. T., Panitch, H. S., and Johnson, K. P. (1994) Neurology 44(4), 745-8. increased cathepsin B activity in peripheral blood mononuclear 5 cells of multiple sclerosis patients.
    • Cohen, P., Graves, H. C., Peehl, D. M., Kamarei, M., Giudice, L. C., and Rosenfeld, R. G. (1992) journal of Clinal Endocrinology and Metabolism 75(4), 1046-53. Prostate-specific antigen (PSA) is an insulin-like growth factor binding protein-3 protease found in seminal plasma.
    • Conover, C. A. and De Leon, D. D. (1994) J. Biol. Chem. 269(10), 7076-80. Acid activated insulin-like growth factor-binding protein-3 proteolysis in normal and transformed cells. Role of cathepsin D.
    • Cuzner, M. L., Opdenakker, G. Plasminogen activators and matrix metalloproteases, mediators of extracellular proteolysis in inflammatory demyelination of the central nervous system. J. Neuroimmunol 94(1-2):1-14 (1999).
    • Desreumaux, P., Huet, G., Zerimech, F., Gambiez, L., Balduyck, M., Baron, P., Degand, P., Cortot, A., Colombel, J. F., Janin, A. Acute inflammatory intestinal vascular lesions and in situ abnormalities of the plasminogen activation system in Crohn's disease. Eur. J. Gastroenterol Hepatol, 11(10): 1113-9 (1999).
    • Hansen, G., Schuster, A., Zubrod, C., and Wahn, V. (1995) Resp 62(3), 117-24. Alpha 1-proteinase inhibitor abrogates proteolytic and secretagogue activity of cystic fibrosis sputum.
    • Muller, H. L., Oh, Y., Gargosky, S. E., Lehrnbecher, T., Hintz, R. L., and Rosenfeld, R. G. (1993) journal of Clinical Endocrinology and Metabolism 77(5), 1113-9. Concentrations of insulin-like growth factor (IGF)-binding protein-3 (IGFBP-3), IGF, and 1GFBP-3 protease activity in cerebrospinal fluid of children with leukemia, central nervous system tumor, or meningitis.
    • Pap, G., Eberhardt, R., Rocken, C., Nebelung, W., Neumann, H. W., Roessner, A. Expression of stromelysin and urokinase type plasminogen activator protein in resection specimens and biopsies at different stages of osteoarthritis of the knee. Pathol Res Pract 196(4):219-26 (2000).
    • Slot, O., Brunner, N., Locht, H., Oxholm, P., Stephens, R. W., Soluble urokinase plasminogen activator receptor in plasma of patients with inflammatory rheumatic disorders: increased concentrations in rheumatoid arthritis. Ann Rheum Dis, 58(8):488-92 (1999).
    1) Cytotoxicity of Selected Variants
  • TABLE 1
    Selected Variants against COS-1 Cells - Target Protease MMP-9
    Ricin PAP220 PAP301 PAP302 PAP303 PAP304 PAP305 PAP308
    Linker Length 23 23 16 15 8 12 12
    (residues)
    Reduction in 1X 23X 24X 118X 63X 1220X 145X 89X
    toxicity relative
    to Ricin
  • TABLE 2
    Selected Variants against HT1080 Cells - Target Protease MMP-9
    Ricin PAP220 PAP301 PAP302 PAP303 PAP304 PAP305 PAP308
    Linker Length 23 23 16 15 8 12 12
    (residues)
    Reduction in 1X 4X 5X 24X 12X 137X 38X 21X
    toxicity relative
    to Ricin

    2) Cytotoxicity Data from Selected Variants
  • TABLE 3
    Selected Variants against COS-1 cells
    MMP9 Variants
    Ricin PAP316 PAP318 PAP323 PAP324 PAP325
    Linker Length 23 23 21 19 17
    (residues)
    Reduction in 1X 39X 100X 65X 67X 82X
    toxicity relative
    to Ricin
    UPA Variants
    Ricin PAP313 PAP314 PAP315 PAP320 PAP321 PAP322
    Linker Length 7 15 14 13 11 9
    (residues)
    Reduction in 1X 110X 52X 75X 55X 1283X 82X
    toxicity relative
    to Ricin
  • TABLE 4
    Selected Variants against HT1080 Cells
    MMP9 Variants
    Ricin PAP316 PAP318 PAP323 PAP324 PAP325
    Linker Length 23 23 21 19 17
    (residues)
    Reduction in 1X 13X 51X 15X 14X 20X
    toxicity relative
    to Ricin
    UPA Variants
    Ricin PAP313 PAP314 PAP315 PAP320 PAP321 PAP322
    Linker Length 7 15 14 13 11 9
    (residues)
    Reduction in 1X 43X 27X 18X 14X 367X 51X
    toxicity relative
    to Ricin
  • TABLE 5
    Maximum Tolerable Dose of MMP9 Variants
    MMP9 Variant Linker Size In Vivo (μg/kg)
    PAP301 23 8
    PAP302 16 40
    PAP303 15 10
    PAP304 8 150
    PAP305 12 20
    PAP308 12 30
    PAP309 23 20
    PAP316 23 20
    PAP318 23 <20
    PAP323 21 15
    PAP324 19 20
    PAP325 17 20
    (cf. Ricin - 1.6 μg/kg and PAP220 - 13 μg/kg)
  • TABLE 6
    Drug Dose Treatment
    Group Sample (μg/kg) (days)
    1 Control - Buffer 0 1, 5, and 9
    2 PAP304 75 1, 5, and 9
    3 PAP304 100 1, 5, and 9
    4 PAP304 150 1, 5, and 9

Claims (43)

1-42. (canceled)
43. A purified and isolated nucleic acid molecule comprising (a) a nucleotide sequence encoding an A chain of a ricin-like toxin, (b) a nucleotide sequence encoding a B chain of a ricin-like toxin and (c) a nucleotide sequence encoding a heterologous linker amino acid sequence linking the A and B chains, the heterologous linker sequence containing a cleavage recognition site for a specific protease.
44. A nucleic acid molecule of claim 43 wherein the specific protease is an MMP or UPA.
45. A nucleic acid molecule according to claim 43 wherein the protease is associated with a cancer cell.
46. A nucleic acid molecule according to claim 45 wherein the cancer cell is one found in T- and B-cell lymphoproliferative diseases, ovarian cancer, pancreatic cancer, head and neck cancer, squamous cell carcinoma, gastrointestinal cancer, breast cancer, prostate, cancer or non small cell lung cancer.
47. A nucleic acid molecule according to claim 43 wherein the protease is associated with an inflammatory cell.
48. A nucleic acid molecule according to claim 47 wherein the cell is one found in rheumatoid arthritis, atherosclerotic cells, Crohn's disease, or central nervous system disease.
49. A nucleic acid molecule of claim 43 wherein the A chain is ricin A chain, abrin toxin A chain, diphtheria toxin A chain, Domain III of Pseudomonas exotoxin, volkensin toxin A chain, cholera toxin A chain, modeccin toxin A chain, viscumin toxin A chain or shiga toxin A chain.
50. A nucleic acid molecule of claim 43 wherein the B chain is ricin B chain, abrin toxin B chain, diphtheria toxin B chain, Domain I/II of Pseudomonas exotoxin, volkensin toxin B chain, cholera toxin B chain, modeccin toxin B chain, viscumin toxin B chain or shiga toxin B chain.
51. A nucleic acid molecule according to claim 43 having a nucleic acid sequence selected from the group consisting of the nucleic acid sequence of pAP301 as shown in FIG. 1B (SEQ ID NO:5); the nucleic acid sequence of pAP302 as shown in FIG. 2B (SEQ ID NO:12); the nucleic acid sequence of pAP303 as shown in FIG. 3B (SEQ ID NO:19); the nucleic acid sequence of pAP304 as shown in FIG. 4B (SEQ ID NO:26); the nucleic acid sequence of pAP305 as shown in FIG. 5B (SEQ ID NO:33); the nucleic acid 5 sequence of pAP308 as shown in FIG. 6B (SEQ ID NO:40); the nucleic acid sequence of pAP309 as shown in FIG. 7B (SEQ ID NO:47); the nucleic acid sequence of pAP313 as shown in FIG. 8B (SEQ ID NO:54); the nucleic acid sequence of pAP314 as shown in FIG. 9B (SEQ ID NO:61); the nucleic acid sequence of pAP315 as shown in FIG. 10B (SEQ ID NO:68); the nucleic acid sequence of pAP316 as shown in FIG. 11B (SEQ ID NO:75); the nucleic acid sequence of pAP318 as shown in FIG. 12B (SEQ ID NO:82); the nucleic acid sequence of pAP320 as shown in FIG. 13B (SEQ ID NO:89); the nucleic acid sequence of pAP321 as shown in FIG. 14B (SEQ ID NO:96); the nucleic acid sequence of pAP322 as shown in FIG. 15B (SEQ ID NO:103); the nucleic acid sequence of pAP323 as shown in FIG. 16B (SEQ ID NO:110); the nucleic acid sequence of pAP324 as shown in FIG. 17B (SEQ ID NO:117); and the nucleic acid sequence of pAP325 as shown in FIG. 18B (SEQ ID NO:124).
52. A nucleic acid molecule according to claim 43 wherein the nucleotide sequence of the linker is selected from the group consisting of: the nucleic acid sequence of pAP301 as shown in FIG. 1A (SEQ ID NO:4); the nucleic acid sequence of pAP302 as shown in FIG. 2A (SEQ ID NO:11); the nucleic acid sequence of pAP303 as shown in FIG. 3A (SEQ ID NO:18); the nucleic acid sequence of pAP304 as shown in FIG. 4A (SEQ ID NO:25); the nucleic acid sequence of pAP305 as shown in FIG. 5A (SEQ ID NO:32); the nucleic acid sequence of pAP308 as shown in FIG. 6A (SEQ ID NO:39); the nucleic acid sequence of pAP309 as shown in FIG. 7A (SEQ ID NO:46); the nucleic acid sequence of pAP313 as shown in FIG. 8A (SEQ ID NO:53); the nucleic acid sequence of pAP314 as shown in FIG. 9A (SEQ ID NO:60); the nucleic acid sequence of pAP315 as shown in FIG. 10A (SEQ ID NO:67); the nucleic acid sequence of pAP316 as shown in FIG. 11A (SEQ ID NO:74); the nucleic acid sequence of pAP318 as shown in FIG. 12A (SEQ ID NO:81); the nucleic acid sequence of pAP320 as shown in FIG. 13A (SEQ ID NO:88); the nucleic acid sequence of pAP321 as shown in FIG. 14A (SEQ ID NO:95); the nucleic acid sequence of pAP322 as shown in FIG. 15A (SEQ ID NO:102); the nucleic acid sequence of pAP323 as shown in FIG. 16A (SEQ ID NO:109); the nucleic acid sequence of pAP324 as shown in FIG. 17A (SEQ ID NO:116); and the nucleic acid sequence of pAP325 as shown in FIG. 18A (SEQ ID NO:123).
53. A plasmid incorporating the nucleic acid molecule of claim 43.
54. A baculovirus transfer vector incorporating the nucleic acid molecule according to claim 43.
55. A recombinant protein comprising an A chain of a ricin-like toxin, a B chain of a ricin-like toxin and a heterologous linker amino acid sequence, linking the A and B chains, wherein the linker sequence contains a cleavage recognition site for a specific protease.
56. A recombinant protein of claim 55 wherein the specific protease is an MMP or UPA.
57. A protein according to claim 55 wherein the protease is associated with a cancer cell.
58. A protein according to claim 57 wherein the cancer cell is one found in T- and B cell lymphoproliferative diseases, ovarian cancer, pancreatic cancer, head and neck cancer, squamous cell carcinoma, gastrointestinal cancer, breast cancer, prostate, cancer or non small cell lung cancer.
59. A protein according to claim 55 wherein the protease is associated with an inflammatory cell.
60. A protein according to claim 59 wherein the cell is one found in rheumatoid arthritis, atherosclerotic cells, Crohn's disease, or central nervous system disease.
61. A recombinant protein of claim 55 wherein the A chain is ricin A chain, abrin toxin A chain, diphtheria toxin A chain, Domain III of Pseudomonas exotoxin, volkensin toxin A chain, cholera toxin A chain, modeccin toxin A chain, viscumin toxin A chain, or shiga toxin A chain.
62. A recombinant protein of claim 55 wherein the B chain is ricin B chain, abrin toxin B chain, diphtheria toxin B chain, Domain I/II of Pseudomonas exotoxin, volkensin toxin B chain, cholera toxin B chain, modeccin toxin B chain, viscumin toxin B chain, or shiga toxin B chain.
63. A recombinant protein of claim 55 wherein the linker amino acid sequence is selected from the group consisting of: the amino acid sequence of PAP301 as shown in FIG. 1C (SEQ ID NO:7); the amino acid sequence of PAP309 as shown in FIG. 7C (SEQ ID NO:49); the amino acid sequence of PAP314 as shown in FIG. 9C (SEQ ID NO:63); the amino acid sequence of PAP315 as shown in FIG. 10C (SEQ ID NO:70); the amino acid sequence of PAP318 as shown in FIG. 12C (SEQ ID NO:84); the amino acid sequence of PAP320 as shown in FIG. 13C (SEQ ID NO:91); the amino acid sequence of PAP321 as shown in FIG. 14C (SEQ ID NO:98); and the amino acid sequence of PAP322 as shown in FIG. 15C (SEQ ID NO:105.
64. A method of inhibiting or destroying cells having a specific protease comprising the steps of:
(a) preparing a purified and isolated nucleic acid having a nucleotide sequence encoding an A chain of a ricin-like toxin, a B chain of a ricin-like toxin, and a heterologous linker amino acid sequence, linking the A and B chains, wherein the linker sequence contains a cleavage recognition site for the protease;
(b) introducing the nucleic acid into a host cell and expressing the nucleic acid in the host cell to obtain a recombinant protein comprising an A chain of a ricin-like toxin, a B chain of a ricin-like toxin and a linker amino acid sequence;
(c) suspending the protein in a pharmaceutically acceptable carrier, diluent or excipient, and
(d) contacting the cells with the recombinant protein.
65. A method according to claim 64 wherein the protease is an MMP or UPA.
66. A method according to claim 64 wherein the protease is associated with a cancer cell.
67. A method according to claim 66 wherein the cancer cell is one found in T- and B cell lymphoproliferative diseases, ovarian cancer, pancreatic cancer, head and neck cancer, squamous cell carcinoma, gastrointestinal cancer, breast cancer, prostate, cancer or non small cell lung cancer.
68. A method according to claim 64 wherein the protease is associated with an inflammatory cell.
69. A method according to claim 68 wherein the cell is one found in rheumatoid arthritis, atherosclerotic cells, Crohn's disease, or central nervous system disease.
70. A method of inhibiting or destroying cells having a specific protease comprising contacting the cells with an effective amount a recombinant protein according to claim 55.
71. A method of treating a cell having a specific protease comprising administering an effective amount of a recombinant protein according to claim 55 to an animal in need thereof.
72. A method of treating a cell having a specific protease comprising administering an effective amount of a nucleic acid molecule according to claim 43 to an animal in need thereof.
73. A process for preparing a pharmaceutical for treating a cell having a specific protease comprising the steps of:
(a) preparing a purified and isolated nucleic acid having a nucleotide sequence encoding an A chain of a ricin-like toxin, a B chain of a ricin-like toxin, and a heterologous linker amino acid sequence, linking the A and B chains, wherein the linker sequence contains a cleavage recognition site for a specific protease;
(b) introducing the nucleic acid into a host cell and expressing the nucleic acid in the host cell to obtain a recombinant protein comprising an A chain of a ricin-like toxin, a B chain of a ricin-like toxin and a linker amino acid sequence;
(c) suspending the protein in a pharmaceutically acceptable carrier, diluent or excipient.
74. A process according to claim 73 wherein the protease is an MMP or UPA.
75. A process according to claim 73 wherein the protease is associated with a cancer cell.
76. A process according to claim 75 wherein the cancer cell is one found in T- and B cell lymphoproliferative diseases, ovarian cancer, pancreatic cancer, head and neck cancer, squamous cell carcinoma, gastrointestinal cancer, breast cancer, prostate, cancer or non small cell lung cancer.
77. A process according to claim 73 wherein the protease is associated with an inflammatory cell.
78. A process according to claim 77 wherein the cell is one found in rheumatoid arthritis, atherosclerotic cells, Crohn's disease, or central nervous system disease.
79. A pharmaceutical composition for treating cancer comprising a recombinant protein of claim 55 and a pharmaceutically acceptable carrier, diluent or excipient.
80. A pharmaceutical composition for treating inflammation comprising a recombinant protein of claim 55 and a pharmaceutically acceptable carrier, diluent or excipient.
81. A pharmaceutical composition for treating a cell having a specific protease comprising a nucleic acid molecule of claim 43 and a pharmaceutically acceptable carrier, diluent or excipient.
82. A pharmaceutical composition for treating a cell having a specific protease comprising an amino acid molecule of claim 43 and a pharmaceutically acceptable carrier, diluent or excipient.
83. A purified and isolated nucleic acid molecule having a nucleic acid sequence selected from the group consisting of: the nucleic acid sequence of pAP301 as shown in FIG. 1A (SEQ ID NO:4); the nucleic acid sequence of pAP302 as shown in FIG. 2A (SEQ ID NO:11); the nucleic acid sequence of pAP303 as shown in FIG. 3A (SEQ ID NO:18); the nucleic acid sequence of pAP304 as shown in FIG. 4A (SEQ ID NO:25); the nucleic acid sequence of pAP305 as shown in FIG. 5A (SEQ ID NO:32); the nucleic acid sequence of pAP308 as shown in FIG. 6A (SEQ ID NO:39); the nucleic acid sequence of pAP309 as shown in FIG. 7A (SEQ ID NO:46); the nucleic acid sequence of pAP313 as shown in FIG. 8A (SEQ ID NO:53); the nucleic acid sequence of pAP314 as shown in FIG. 9A (SEQ ID NO:60); the nucleic acid sequence of pAP315 as shown in FIG. 10A (SEQ ID NO:67); the nucleic acid sequence of pAP316 as shown in FIG. 11A (SEQ ID NO:74); the nucleic acid sequence of pAP318 as shown in FIG. 12A (SEQ ID NO:81); the nucleic acid sequence of pAP320 as shown in FIG. 13A (SEQ ID NO:88); the nucleic acid sequence of pAP321 as shown in FIG. 14A (SEQ ID NO:95); the nucleic acid sequence of pAP322 as shown in FIG. 15A (SEQ ID NO:102); the nucleic acid sequence of pAP323 as shown in FIG. 16A (SEQ ID NO:109); the nucleic acid sequence of pAP324 as shown in FIG. 17A (SEQ ID NO:116); and the nucleic acid sequence of pAP325 as shown in FIG. 18A (SEQ ID NO:123).
84. A linker protein having an amino acid sequence selected from the group consisting of: the amino acid sequence of PAP301 as shown in FIG. 1C (SEQ ID NO:7); the amino acid sequence of PAP309 as shown in FIG. 7C (SEQ ID NO:49); the amino acid sequence of PAP314 as shown in FIG. 9C (SEQ ID NO:63); the amino acid sequence of PAP315 as shown in FIG. 10C (SEQ ID NO:70); the amino acid sequence of PAP318 as shown in FIG. 12C (SEQ ID NO:84); the amino acid sequence of PAP320 as shown in FIG. 13C (SEQ ID NO:91); the amino acid sequence of PAP321 as shown in FIG. 14C (SEQ ID NO:98); and the amino acid sequence of PAP322 as shown in FIG. 15C (SEQ ID NO:105).
US12/021,563 1999-10-04 2008-01-29 Ricin-like toxins for treatment of cancer Abandoned US20110178026A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US12/021,563 US20110178026A1 (en) 1999-10-04 2008-01-29 Ricin-like toxins for treatment of cancer

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US15780799P 1999-10-04 1999-10-04
US19740900P 2000-04-14 2000-04-14
PCT/CA2000/001162 WO2001025267A2 (en) 1999-10-04 2000-10-04 Improved ricin-like toxins for treatment of cancer
US10/089,058 US7060789B1 (en) 1999-10-04 2000-10-04 Ricin-like toxins for treatment of cancer
US11/414,227 US7348008B2 (en) 1999-10-04 2006-06-09 Ricin-like toxins for treatment of cancer
US12/021,563 US20110178026A1 (en) 1999-10-04 2008-01-29 Ricin-like toxins for treatment of cancer

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/414,227 Continuation US7348008B2 (en) 1999-10-04 2006-06-09 Ricin-like toxins for treatment of cancer

Publications (1)

Publication Number Publication Date
US20110178026A1 true US20110178026A1 (en) 2011-07-21

Family

ID=26854503

Family Applications (3)

Application Number Title Priority Date Filing Date
US10/089,058 Expired - Lifetime US7060789B1 (en) 1997-04-30 2000-10-04 Ricin-like toxins for treatment of cancer
US11/414,227 Expired - Fee Related US7348008B2 (en) 1999-10-04 2006-06-09 Ricin-like toxins for treatment of cancer
US12/021,563 Abandoned US20110178026A1 (en) 1999-10-04 2008-01-29 Ricin-like toxins for treatment of cancer

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US10/089,058 Expired - Lifetime US7060789B1 (en) 1997-04-30 2000-10-04 Ricin-like toxins for treatment of cancer
US11/414,227 Expired - Fee Related US7348008B2 (en) 1999-10-04 2006-06-09 Ricin-like toxins for treatment of cancer

Country Status (4)

Country Link
US (3) US7060789B1 (en)
AU (1) AU7636800A (en)
CA (1) CA2386237A1 (en)
WO (1) WO2001025267A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014099488A1 (en) * 2012-12-19 2014-06-26 Stanley Mo Personalized search library based on continual concept correlation

Families Citing this family (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8921521B2 (en) 2003-06-18 2014-12-30 Ocera Therapeutics, Inc. Macrocyclic modulators of the Ghrelin receptor
JP2007530019A (en) * 2004-03-24 2007-11-01 ツインストランド セラピューティックス インコーポレイテッド Glycosylation variants of ricin-like proteins
WO2007050105A2 (en) * 2004-12-08 2007-05-03 Smiths Detection Inc. Oligonucleotide sequences for the detection of ricin gene and toxin
EP2047199A2 (en) 2006-07-20 2009-04-15 Modine Manufacturing Company Flat tube, flat tube heat exchanger, and method of manufacturing same
CA2691539C (en) * 2007-06-21 2014-08-26 Angelica Therapeutics, Inc. Modified toxins
EP2268297A4 (en) * 2008-02-29 2011-11-16 Angelica Therapeutics Inc Modified toxins
US10059750B2 (en) 2013-03-15 2018-08-28 Angelica Therapeutics, Inc. Modified toxins
EP4098743A1 (en) * 2015-05-29 2022-12-07 CureVac AG Method for adding cap structures to rna using immobilized enzymes

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4869903A (en) * 1987-05-29 1989-09-26 Genelabs Incorporated Method of selectively inhibiting HIV
US5101025A (en) * 1989-04-04 1992-03-31 Genelabs Incorporated Recombinant trichosanthin and coding sequence
US5128468A (en) * 1989-11-23 1992-07-07 FIS--Fabbrica Italiana Sintetici S.p.A. Process for preparing 2-aryl-3-hydroxy-cis-2,3-dihydro-1,5-benzothiazepin-4(5H)-ones and their derivatives
US5820866A (en) * 1994-03-04 1998-10-13 National Jewish Center For Immunology And Respiratory Medicine Product and process for T cell regulation

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2521657B2 (en) 1983-07-15 1996-08-07 インペリアル・ケミカル・インダストリーズ・ピーエルシー Recombinant DNA molecule and transformed host cell containing the same
JPH02502287A (en) 1987-11-24 1990-07-26 ジェネティックス・インスティテュート・インコーポレイテッド Improved ricin molecules and ricin toxin conjugates
US5128460A (en) 1989-04-04 1992-07-07 Genelabs Incorporated Recombinant trichosanthin and coding sequence
US5248606A (en) 1990-06-11 1993-09-28 Dowelanco Dna encoding inactive precursor and active forms of maize ribosome inactivating protein
US5677274A (en) 1993-02-12 1997-10-14 The Government Of The United States As Represented By The Secretary Of The Department Of Health And Human Services Anthrax toxin fusion proteins and related methods
US6333303B1 (en) * 1996-04-30 2001-12-25 Twinstrand Therapeutics Inc. Antiviral ricin-like proteins
AU7023798A (en) 1997-04-30 1998-11-24 De Novo Enzyme Corporation Ricin-like toxin variants for treatment of cancer, viral or parasitic infections
US6803358B1 (en) * 2000-04-14 2004-10-12 Twinstrand Therapeutics Inc. Ricin-like toxin variants for treatment of cancer, viral or parasitic infections
US6531125B1 (en) * 1999-03-02 2003-03-11 Twinstrand Therapeutics Inc. Antiviral ricin-like proteins

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4869903A (en) * 1987-05-29 1989-09-26 Genelabs Incorporated Method of selectively inhibiting HIV
US5101025A (en) * 1989-04-04 1992-03-31 Genelabs Incorporated Recombinant trichosanthin and coding sequence
US5128468A (en) * 1989-11-23 1992-07-07 FIS--Fabbrica Italiana Sintetici S.p.A. Process for preparing 2-aryl-3-hydroxy-cis-2,3-dihydro-1,5-benzothiazepin-4(5H)-ones and their derivatives
US5820866A (en) * 1994-03-04 1998-10-13 National Jewish Center For Immunology And Respiratory Medicine Product and process for T cell regulation

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014099488A1 (en) * 2012-12-19 2014-06-26 Stanley Mo Personalized search library based on continual concept correlation
US9582572B2 (en) 2012-12-19 2017-02-28 Intel Corporation Personalized search library based on continual concept correlation

Also Published As

Publication number Publication date
AU7636800A (en) 2001-05-10
WO2001025267A3 (en) 2002-03-28
US20070036815A1 (en) 2007-02-15
WO2001025267A2 (en) 2001-04-12
CA2386237A1 (en) 2001-04-12
US7348008B2 (en) 2008-03-25
US7060789B1 (en) 2006-06-13

Similar Documents

Publication Publication Date Title
US20110144004A1 (en) Ricin-like toxin variants for treatment of cancer, viral or parasitic infections
US7348008B2 (en) Ricin-like toxins for treatment of cancer
DK2197903T3 (en) Deletions in Domain II from Pseudomonas exotoxin A that reduce non-specific TOXICITY
JP6169561B2 (en) Recombinant immunotoxins targeting mesothelin
Shapira et al. An immunoconjugate of anti-CD24 and Pseudomonas exotoxin selectively kills human colorectal tumors in mice
CA2668017A1 (en) Improved conjugates
JPH09509043A (en) A novel ribosome-inactivating protein isolated from the plant Bryonia dioica
CA2315880A1 (en) Streptococcal heat shock proteins of the hsp60 family
EP0909322B1 (en) Antiviral ricin-like proteins
US6803358B1 (en) Ricin-like toxin variants for treatment of cancer, viral or parasitic infections
US7247715B2 (en) Ricin-like toxin variants for treatment of cancer, viral or parasitic infections
US20090304817A1 (en) Glycosylation Variants of Ricin-Like Proteins

Legal Events

Date Code Title Description
AS Assignment

Owner name: TWINSTRAND THERAPEUTICS INC., BRITISH COLUMBIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:BRAUN, CURTIS;PURAC, ADMIR;BORGFORD, THOR;SIGNING DATES FROM 20030307 TO 20030703;REEL/FRAME:020453/0627

AS Assignment

Owner name: TWINSTRAND HOLDINGS INC., CANADA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:TWINSTRAND THERAPEUTICS INC.;REEL/FRAME:023015/0658

Effective date: 20090723

STCB Information on status: application discontinuation

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