WO2002066506A2 - Use of casb7439 (ash2) in the immunotherapy of breast cancer - Google Patents

Use of casb7439 (ash2) in the immunotherapy of breast cancer Download PDF

Info

Publication number
WO2002066506A2
WO2002066506A2 PCT/EP2002/001649 EP0201649W WO02066506A2 WO 2002066506 A2 WO2002066506 A2 WO 2002066506A2 EP 0201649 W EP0201649 W EP 0201649W WO 02066506 A2 WO02066506 A2 WO 02066506A2
Authority
WO
WIPO (PCT)
Prior art keywords
seq
polypeptide
polynucleotide
sequence
casb7439
Prior art date
Application number
PCT/EP2002/001649
Other languages
French (fr)
Other versions
WO2002066506A3 (en
Inventor
Carlota Vinals Y De Bassols
Jean-Pol Cassart
Original Assignee
Glaxosmithkline Biologicals S.A.
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 Glaxosmithkline Biologicals S.A. filed Critical Glaxosmithkline Biologicals S.A.
Priority to AU2002249221A priority Critical patent/AU2002249221A1/en
Publication of WO2002066506A2 publication Critical patent/WO2002066506A2/en
Publication of WO2002066506A3 publication Critical patent/WO2002066506A3/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • C07K14/4701Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals not used
    • C07K14/4748Tumour specific antigens; Tumour rejection antigen precursors [TRAP], e.g. MAGE
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies

Definitions

  • the present invention relates methods for the utilisation of polynucleotides, herein referred to as CASB7439 polynucleotides, and of polypeptides encoded thereby (referred to herein as CASB7439 polypeptides) for the prophylaxis and treatment of breast cancer.
  • the invention relates to diagnostic assays for detecting breast cancer associated with inappropriate CASB7439 polypeptide activity or levels and for detecting breast tumour cells expressing CASB7439 polypeptide.
  • Polypeptides and polynucleotides disclosed herein are believed to be important immunogens for specific prophylactic or therapeutic immunisation against breast tumours, because they are specifically expressed or highly over-expressed in breast tumours compared to normal cells and can thus be targeted by antigen-specific immune mechanisms leading to the destruction of the tumour cell. They can also be used to diagnose the occurrence of tumour cells. Furthermore, their inappropriate expression in certain circumstances can cause an induction of autoimmune, inappropriate immune responses, which could be corrected through appropriate vaccination using the same polypeptides or polynucleotides. hi this respect the most important biological activities to our purpose are the antigenic and immunogenic activities of the polypeptide disclosed herein.
  • a polypeptide for use according to the present invention may also exhibit at least one other biological activity of a CASB7439 polypeptide, which could qualify it as a target for therapeutic or prophylactic intervention different from that linked to the immune response.
  • the present invention relates to the use of CASB7439 polypeptides in the prophylaxis, treatment or diagnosis of breast cancer.
  • Such peptides include isolated polypeptides comprising an amino acid sequence which has at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95% identity, most preferably at least 97-99% identity, to that of SEQ ID NO:2, SEQ ID NO:3 or SEQ ID NO:8 over the entire length of SEQ ID NO:2, SEQ ID NO:3 or SEQ ID NO 8.
  • Such polypeptides include those comprising the amino acid of SEQ LD NO:2, SEQ ID NO:3 or SEQ ID NO: 8.
  • a most preferred polypeptide for use according to the present invention is that of SEQ LD NO:2.
  • polypeptides are recombinantly produced.
  • polypeptides according to the invention are purified, and are substantially free of any other proteins or contaminating host-originating material.
  • peptides that find utility in the present invention include isolated polypeptides encoded by a polynucleotide comprising the sequence contained in SEQ ID NO: 1 or SEQ ID NO:4.
  • the invention also contemplates the use of an immunogenic fragment of a CASB7439 polypeptide, that is a contiguous portion of the CASB7439 polypeptide which has the same or substantially similar immunogenic properties to the polypeptide comprising the amino acid sequence of SEQ ID NO:2 or SEQ ID NO:3 or SEQ ID NO:8. That is to say, the fragment (if necessary when coupled to a carrier) is capable of raising an immune response which recognises the CASB7439 polypeptide.
  • an immunogenic fragment may include, for example, the CASB7439 polypeptide lacking an N-terminal leader sequence, a transmembrane domain or a C-terminal anchor domain.
  • the immunogenic fragment of CASB7439 comprises substantially all of the extracellular domain of a polypeptide which has at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95% identity, most preferably at least 97-99% identity, to that of SEQ ID NO:2 or SEQ ID NO:3 or SEQ ID NO:8 over the entire length of SEQ ID NO:2 or SEQ ID NO: 3 or SEQ ID NO:8.
  • an immunogenic fragment according to the invention comprises at least one epitope.
  • Peptide fragments incorporating an epitope of CASB7439 typically will comprise at least 7, preferably 9 or 10 contiguous amino acids from SEQ LO NO:2 or SEQ ID NO:3 or SEQ ID NO:8.
  • Preferred epitopes are shown in SEQ LD NO: 16 to SEQ LO NO:33.
  • Peptides that incorporate these epitopes are preferably contemplated in the present invention.
  • Mimotopes which have the same characteristics as these epitopes, and immunogens comprising such mimotopes which generate an immune response which cross-react with an epitope in the context of the CASB7439 molecule, are also used according to the present invention.
  • the present invention includes the use of isolated peptides encompassing these epitopes themselves, and any mimotope thereof in the treatment of breast cancer.
  • mimotope is defined as an entity which is sufficiently similar to the native CASB7439 epitope so as to be capable of being recognised by antibodies which recognise the native molecule; (Gheysen, H.M., et al., 1986, Synthetic peptides as antigens. Wiley, Chichester, Ciba foundation symposium 119, pl30-149; Gheysen, H.M., 1986, Molecular
  • Immunology, 23,7, 709-715 are capable of raising antibodies, when coupled to a suitable carrier, which antibodies cross-react with the native molecule.
  • Peptide mimotopes of the above-identified epitopes may be designed for a particular purpose by addition, deletion or substitution of elected amino acids.
  • the peptides according to the present invention may be modified for the purposes of ease of conjugation to a carrier, preferably a protein carrier.
  • a carrier preferably a protein carrier.
  • peptides conjugated to a protein carrier to include a hydrophobic terminus distal from the conjugated terminus of the peptide, such that the free unconjugated end of the peptide remains associated with the surface of the carrier protein.
  • the peptides may be altered to have an N-terminal cysteine and a C- terminal hydrophobic amidated tail.
  • the addition or substitution of a D-stereoisomer form of one or more of the amino acids may be performed to create a beneficial derivative, for example to enhance stability of the peptide.
  • modified peptides could be a wholly or partly non-peptide mimotope wherein the constituent residues are not necessarily confined to the 20 naturally occurring amino acids.
  • these may be cyclised by techniques known in the art to constrain the peptide into a conformation that closely resembles its shape when the peptide sequence is in the context of the whole molecule.
  • a preferred method of cyclising a peptide comprises the addition of a pair of cysteine residues to allow the formation of a disulphide bridge.
  • mimotopes or immunogens for use in the present invention may be larger than the above-identified epitopes, and as such may comprise the sequences disclosed herein. Accordingly, the mimotopes disclosed in the present invention may consist of addition of N and/or C terminal extensions of a number of other natural residues at one or both ends.
  • the peptide mimotopes may also be retro sequences of the natural sequences, in that the sequence orientation is reversed; or alternatively the sequences maybe entirely or at least in part comprised of D-stereo isomer amino acids (inverso sequences).
  • the peptide sequences may be retro-inverso in character, in that the sequence orientation is reversed and the amino acids are of the D-stereoisomer form.
  • retro or retro-inverso peptides have the advantage of being non-self, and as such may overcome problems of self-tolerance in the immune system.
  • peptide mimotopes may be identified using antibodies which are capable themselves of binding to the epitopes of the present invention using techniques such as phage display technology (EP 0 552267 Bl).
  • This technique generates a large number of peptide sequences which mimic the structure of the native peptides and are, therefore, capable of binding to anti-native peptide antibodies, but may not necessarily themselves share significant sequence homology to the native peptide.
  • This approach may have significant advantages by allowing the possibility of identifying a peptide with enhanced immunogenic properties, or may overcome any potential self-antigen tolerance problems which may be associated with the use of the native peptide sequence. Additionally this technique allows the identification of a recognition pattern for each native-peptide in terms of its shared chemical properties amongst recognised mimotope sequences.
  • the covalent coupling of the peptide to the immunogenic carrier can be carried out in a manner well known in the art.
  • a carbodiimide, glutaraldehyde or (N-[ ⁇ - maleimidobutyryloxy] succinimide ester utilising common commercially available heterobifunctional linkers such as CDAP and SPDP (using manufacturers instructions).
  • the immunogen can easily be isolated and purified by means of a dialysis method, a gel filtration 5. method, a fractionation method etc.
  • the present invention further provides for the use of a CASB7439 polypeptide as defined hereabove, which is chemically conjugated to a carrier protein.
  • the types of carriers used in the immunogens of the present invention will be 0 readily known to the man skilled in the art.
  • the function of the carrier is to provide cytokine help in order to help induce an immune response against the peptide.
  • a non-exhaustive list of carriers which may be used in the present invention include: Keyhole limpet Haemocyanin (KLH), serum albumins such as bovine serum albumin (BSA), inactivated bacterial toxins such as tetanus or 5 diptheria toxins (TT and DT), or recombinant fragments thereof (for example, Domain 1 of Fragment C of TT, or the translocation domain of DT), or the purified protein derivative of tuberculin (PPD).
  • KLH Keyhole limpet Haemocyanin
  • BSA bovine serum albumin
  • TT and DT inactivated bacterial toxins
  • TT and DT inactivated bacterial toxins
  • recombinant fragments thereof for example, Domain 1 of
  • the mimotopes or epitopes may be directly conjugated to liposome carriers, which may additionally comprise immunogens capable of providing T-cell help.
  • liposome carriers which may additionally comprise immunogens capable of providing T-cell help.
  • the ratio of 0 mimotopes to carrier is in the order of 1 : 1 to 20: 1 , and preferably each carrier should carry between 3-15 peptides.
  • a preferred carrier is Protein D from Haemophilus influenzae (EP 0 594 610 B1).
  • Protein D is an IgD-binding protein from Haemophilus influenzae and has been patented by Forsgren (WO 91/18926, granted EP 0 594 610 Bl).
  • fragments of protein D for example Protein D l/3 rd (comprising the N-terminal 100-110 amino acids of protein D (GB 9717953.5)).
  • Another prefened method of presenting the peptides for use in the present invention is in the context of a recombinant fusion molecule.
  • EP 0 421 635 B describes the use of chimaeric hepadnavirus core antigen particles to present foreign peptide sequences in a virus-like particle.
  • immunogens for use in the present invention may comprise peptides presented in chimaeric particles consisting of hepatitis B core antigen.
  • the recombinant fusion proteins may comprise the mimotopes of the present invention and a carrier protein, such as NSl of the influenza virus, or a fragment thereof such as the first 1-80 or 1-81 amino acids of the NSl protein, which is preferred.
  • Preferred fusion proteins according to this invention are sequences set forth in SEQ LD NO: 7 and in SEQ ID NO: 14.
  • the nucleic acid which encodes said immunogen can also be used in the present invention.
  • Preferred nucleotidic sequences are given in SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO: 13, SEQ ID NO:15 and SEQ ID NO:34.
  • Peptides used in the present invention can be readily synthesised by solid phase procedures well known in the art. Suitable syntheses may be performed by utilising "T-boc” or "F-moc” procedures. Cyclic peptides can be synthesised by the solid phase procedure employing the well-known "F-moc” procedure and polyamide resin in the fully automated apparatus. Alternatively, those skilled in the art will know the necessary laboratory procedures to perform the process manually. Techniques and procedures for solid phase synthesis are described in 'Solid Phase Peptide Synthesis: A Practical Approach' by E. Atherton and R..C. Sheppard, published by IRL at Oxford University Press (1989).
  • the peptides may be produced by recombinant methods, including expressing nucleic acid molecules encoding the mimotopes in a bacterial or mammalian cell line, followed by purification of the expressed mimotope.
  • Techniques for recombinant expression of peptides and proteins are known in the art, and are described in Maniatis, T., Fritsch, E.F. and Sambrook et al., Molecular cloning, a laboratory manual, 2nd Ed.; Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York (1989).
  • polypeptides or immunogenic fragment utilised in the invention may be in the form of the "mature" protein or may be a part of a larger protein such as a precursor or a fusion protein. It is often advantageous to include an additional amino acid sequence which contains secretory or leader sequences, pro-sequences, sequences which aid in purification such as multiple histidine residues, or an additional sequence for stability during recombinant production. Furthermore, addition of exogenous polypeptide or lipid tail or polynucleotide sequences to increase the immunogenic potential of the final molecule is also considered.
  • another aspect of the invention concerns the use of genetically engineered soluble fusion proteins comprising a polypeptide of the present invention, or a fragment thereof, and various portions of the constant regions of heavy or light chains of immunoglobulins of various subclasses (IgG, IgM, IgA, IgE).
  • immunoglobulin is the constant part of the heavy chain of human IgG, particularly IgGl, where fusion takes place at the hinge region.
  • the Fc part can be removed simply by incorporation of a cleavage sequence which can be cleaved with blood clotting factor Xa.
  • this invention relates to the use of these fusion proteins for drug screening, diagnosis and therapy. Examples of fusion protein technology can be found in International Patent Application Nos. WO94/29458 and WO94/22914.
  • the proteins for use according to the present invention may be chemically conjugated, or expressed as recombinant fusion proteins allowing increased levels to be produced in an expression system as compared to non-fused protein.
  • the fusion partner may assist in providing T helper epitopes (immunological fusion partner), preferably T helper epitopes recognised by humans, or assist in expressing the protein (expression enhancer) at higher yields than the native recombinant protein.
  • the fusion partner will be both an immunological fusion partner and expression enhancing partner.
  • Fusion partners include protein D from Haemophilus influenza B and the non- structural protein from influenzae virus, NSl (hemagglutinin), or a fragment thereof having retained T helper epitopes.
  • NSl hemagglutinin
  • T helper epitopes C.hackett, D. Horowitz, M. Wysocka & S. Dillon, 1992, J. Gen. Nirology, 73, 1339-1343.
  • ⁇ S1 is the immunological fusion partner it has the additional advantage in that it allows higher expression yields to be achieved.
  • SEQ ID NO:7 An example of such ⁇ S1-CASB7439 fusion protein is given in SEQ ID NO:7, and is encoded by the polynucleotide sequences given in SEQ ID NO:5 or in SEQ LD NO:6, being respectively the native (unmodified) cDNA sequence or codon-optimised (for E.coli expression) sequence.
  • the polypeptide sequence of SEQ ID NO:7 and polynucleotide sequences of SEQ ID NO:5 and SEQ ID NO:6 are preferred for the use according to the present invention.
  • Another immunological fusion partner is the protein known as LYTA.
  • the C terminal portion of the molecule is used.
  • Lyta is derived from Streptococcus pneumoniae which synthesise an N-acetyl-L-alanine amidase, amidase LYTA, (coded by the lytA gene ⁇ Gene, 43 (1986) page 265-272 ⁇ an autolysin that specifically degrades certain bonds in the peptidoglycan backbone.
  • the C- terminal domain of the LYTA protein is responsible for the affinity to the choline or to some choline analogues such as DEAE. This property has been exploited for the development of E.coli C-LYTA expressing plasmids useful for expression of fusion proteins.
  • the present invention also includes the use of xenogeneic forms (also termed ortholog forms) of the aforementioned polypeptides, said xenogeneic forms referring to an antigen having substantial sequence identity to the human antigen (also termed autologous antigen) which serves as a reference antigen but which is derived from a different non-human species.
  • the substantial identity refers to concordance of an amino acid sequence with another amino acid sequence or of a polynucleotide sequence with another polynucleotide sequence when such sequence are arranged in a best fit alignment in any of a number of sequence alignment proteins known in the art.
  • the xenogeneic CASB7439 polypeptide will be a CASB7439 polypeptide which is xenogeneic with respect to human CASB7439, in other words which is isolated from a species other than human.
  • the polypeptide is isolated from mouse, rat, pig, or rhesus monkey, most preferably from mouse or rat.
  • the present invention also provides a method of inducing an immune response against human CASB7439 having an amino acid sequence as set forth in SEQ ID NO:9 and SEQ ID NO:10 in a human, comprising administering to the subject an effective dosage of a composition comprising a xenogeneic form of said human CASB7439 as described herein.
  • a preferred embodiment is a method of inducing an immune response against human CASB7439 using the xenogeneic CASB7439 isolated from mouse, rat, pig or rhesus monkey.
  • Another preferred method of inducing an immune response according to the present invention is using an antigen composition including a live viral expression system which expresses said xenogeneic antigen.
  • the isolated xenogeneic CASB7439 polypeptide will generally share substantial sequence similarity, and include isolated polypeptides comprising an amino acid sequence which has at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95% identity, most preferably at least 97-99% identity, to that of SEQ ID NO:9 and SEQ ID NO: 10 over the entire length of SEQ ID NO:9 and SEQ ID NO: 10, respectively.
  • the xenogeneic polypeptide will comprise an immunogenic fragment of the polypeptide of SEQ ID NO:9 and SEQ ID NO:10 in which the immunogenic activity of the immunogenic fragment is substantially the same as the polypeptide of SEQ ID NO:9 and SEQ ID NO: 10.
  • the xenogeneic CASB7439 polypeptide can be a fragment of at least about 20 consecutive amino acids, preferably about 30, more preferably about 50, yet more preferably about 100, most preferably about 150 contiguous amino acids selected from the amino acid sequences as shown in SEQ ID NO:9 and SEQ LD NO: 10.
  • xenogeneic CASB7439 fragments will retain some functional property, preferably an immunological activity, of the larger molecule set forth in SEQ ID NO:9 and SEQ LD NO: 10, and are useful in the methods described herein (e.g. in pharmaceutical and vaccine compositions, in diagnostics, etc.).
  • the fragments will be able to generate an immune response against the human counterpart, such as the generation of cross-reactive antibodies which react with the autologous human form of CASB7439 as set forth in SEQ LD NO:2.
  • the xenogeneic polypeptide of the invention may be part of a larger fusion, comprising the xenogeneic CASB7439 polypeptide or fragment thereof and a heterologous protein or part of a protein acting as a fusion partner as described hereabove.
  • the present invention also contemplates the use of variants of the aforementioned polypeptides, that is polypeptides that vary from the referents by conservative amino acid substitutions, whereby a residue is substituted by another with like characteristics.
  • Typical such substitutions are among Ala, Nal, Leu and De; among Ser and Thr; among the acidic residues Asp and Glu; among Asn and Gin; and among the basic residues Lys and Arg; or aromatic residues Phe and Tyr.
  • Particularly preferred are variants in which several, 5-10, 1-5, 1-3, 1-2 or 1 amino acids are substituted, deleted, or added in any combination.
  • Polypeptides for use in the present invention can be prepared in any suitable manner.
  • Such polypeptides include isolated naturally occurring polypeptides, recombinantly produced polypeptides, synthetically produced polypeptides, or polypeptides produced by a combination of these methods. Means for preparing such polypeptides are well understood in the art.
  • the present invention relates to the use of CASB7439 polynucleotides in the treatment or diagnosis of breast cancer.
  • polynucleotides include isolated polynucleotides comprising a nucleotide sequence encoding a polypeptide which has at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95% identity, to the amino acid sequence of SEQ ID ⁇ O:2 or SEQ ID NO:3 or SEQ ID NO:8, over the entire length of SEQ ID NO:2 or SEQ ID NO:3 or SEQ ID NO:8.
  • polypeptides which have at least 97% identity are highly preferred, whilst those with at least 98-99% identity are more highly preferred, and those with at least 99% identity are most highly preferred.
  • Such polynucleotides include SEQ ID NO: 1 , or include a polynucleotide comprising the nucleotide sequence contained in SEQ ID NO:l encoding the polypeptide of SEQ ID NO:2 or comprising the nucleotide contained in SEQ ID NO:4 encoding the polypeptide of SEQ ID NO:3 or SEQ ID NO:8.
  • polynucleotides which have at least 97% identity are highly preferred, whilst those with at least 98-99% identity are more highly preferred, and those with at least 99% identity are most highly preferred.
  • polynucleotides include isolated polynucleotides comprising a nucleotide sequence which has at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95% identity, to SEQ ID NO: 1 or SEQ ID NO:4 or to the coding sequence of SEQ ID NO: 1 or SEQ ID NO:4 over the entire length of SEQ ID NO: 1 or SEQ ID NO:4 or over the entire length of the coding sequence of SEQ TD NO:l or SEQ ED NO:4 respectively.
  • polynucleotides which have at least 97% identity are highly preferred, whilst those with at least 98-99% identity are more highly preferred, and those with at least 99% identity are most highly preferred.
  • Such polynucleotides include a polynucleotide comprising the polynucleotide of SEQ ID NO:l or SEQ ID NO:4 as well as the polynucleotide of SEQ ID NO:l or SEQ ID NO:4 or the coding region of SEQ LD NO: 1 or SEQ ID NO:4.
  • Said polynucleotide can be inserted in a suitable plasmid or recombinant microrganism vector and used for immunisation (see for example Wolff et. al, Science 247:1465-1468 (1990); Corr et. al., J. Exp. Med. 184:1555-1560 (1996); Doe et. al., Proc. Natl. Acad. Sci. 93:8578-8583 (1996)).
  • the present invention also provides a nucleic acid encoding the aforementioned xenogeneic proteins of the present invention and their use in medicine.
  • the xenogeneic CASB7439 polynucleotide for use in pharmaceutical compositions has the sequence set forth in SEQ ID NO: 11 and SEQ ID NO: 12.
  • the isolated xenogeneic CASB7439 polynucleotides according to the invention may be single-stranded (coding or antisense) or double-stranded, and maybe DNA (genomic, cDNA or synthetic) or RNA molecules. Additional coding or non-coding sequences may, but need not, be present within a polynucleotide of the present invention.
  • the present invention provides polynucleotide variants having substantial identity to the sequences disclosed herein in SEQ LD NO:l 1 and SEQ ID NO: 12 for example those comprising at least 70% sequence identity, preferably at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% or higher, sequence identity compared to a polynucleotide sequence of this invention using the methods described herein, (e.g., BLAST analysis using standard parameters).
  • the isolated xenogeneic polynucleotide of the invention will comprise a nucleotide sequence encoding a polypeptide that has at least 90%, preferably 95% and above, identity to the amino acid sequence of SEQ TD NO:l 1 and SEQ ED NO: 12, over the entire length of SEQ ED NO: 11 and SEQ LD NO: 12, respectively; or a nucleotide sequence complementary to said isolated polynucleotide.
  • the invention also contemplates the use of polynucleotides which are complementary to all the above described polynucleotides.
  • the invention also provides for the use of a fragment of a CASB7439 polynucleotide which when administered to a subject has the same immunogenic properties as the polynucleotide of SEQ LD NO:l or SEQ ID NO:4.
  • the invention also provides for the use of a polynucleotide encoding an immunological fragment of a CASB7439 polypeptide as hereinbefore defined. Also contemplated are the use of such fragments that have a level of immunogenic activity of at least about 50%, preferably at least about 70% and more preferably at least about 90% of the level of immunogenic activity of a polypeptide sequence encoded by a polynucleotide sequence set forth in SEQ LD NO: 1 or SEQ ED NO:4.
  • Polypeptide fragments for use according to the invention preferably comprise at least about 5, 10, 15, 20, 25, 50, or 100 contiguous amino acids, or more, including all intermediate lengths, of a polypeptide composition set forth herein, such as those set forth in SEQ ED NO:2 or SEQ ID NO:3 or SEQ ED NO:8, or those encoded by a polynucleotide sequence set forth in a sequence of SEQ ID NO:l or SEQ ED NO:4.
  • the nucleotide sequence of SEQ ID NO:l is a cDNA sequence which comprises a polypeptide encoding sequence (nucleotide 545 to 1126) encoding a polypeptide of 193 amino acids, the polypeptide of SEQ ED NO:2.
  • the nucleotide sequence encoding the polypeptide of SEQ ED NO:2 may be identical to the polypeptide encoding sequence contained in SEQ ED NO: 1 or it may be a sequence other than the one contained in SEQ ED NO: 1, which, as a result of the redundancy (degeneracy) of the genetic code, also encodes the polypeptide of SEQ ED NO:2.
  • the polypeptide of the SEQ ED NO:2 is structurally related to other proteins of the achaete scute family, and is also named "human Achaete Scute homologue 2" (HASH2) (accession number NP_005161 and AAB86993).
  • HASH2 human Achaete Scute homologue 2
  • HASH2 Human Achaete Scute homologue 2
  • ASCL2 Achaete Scute complex like 2
  • Human ASCL2 is expressed in the extravillus trophoblasts of the developing placenta only, and maps on chromosome 1 lpl5 close to IGF2 and H19.
  • the mouse achaete-scute homolog-2 gene (MASH2) encodes a transcription factor playing a role in the development of the trophoblast.
  • the Mash2 gene is paternally imprinted in the mouse, and the lack of human ASCL2 expression in non-malignant hydatidiform (androgenetic) moles indicates that human Ascl2 is also imprinted in man.
  • Ascl2 genes are members of the basic helix-loop-helix (BHLH) family of transcription factors. They activate transcription by binding to the E box (5 - CANNTG-3'). Dimerization with other BHLH proteins is required for efficient DNA binding. They are involved in the determination of the neuronal precursors in the peripheral nervous system and the central nervous system in drosophila melanogaster, and probably in mammals as well.
  • BHLH basic helix-loop-helix
  • the complementary strand of the nucleotide sequence of SEQ ED NO:l is the polynucleotide sequence of SEQ ED NO:4. This strand also comprises two other polypeptide encoding sequences. He first polypeptide encoding sequence (nucleotide 1184 to 399 of SEQ ED:1, nucleotide 608 to 1393 of SEQ LD NO:4) encodes a polypeptide of 262 amino acids, the polypeptide of SEQ ED NO:3.
  • the second polypeptide encoding sequence (nucleotide 840 to 262 of SEQ LD NO:l, nucleotide 952 to 1530 of SEQ ED NO:4) encodes a polypeptide of 193 amino acids, the polypeptide of SEQ ED NO:8.
  • the nucleotide sequence encoding the polypeptides of SEQ ID NO:3 and 8 may be identical to the polypeptides encoding sequence contained in SEQ ED NO: 1 or 4 or it may be a sequence other than the one contained in SEQ ID NO: 1 or 4, which, as a result of the redundancy (degeneracy) of the genetic code, also encodes the polypeptides of SEQ ID NO:3 and 8.
  • polypeptide of the SEQ ED NO:3 is structurally related to other proteins of the splicing coactivator protein family, having homology and/or structural similarity with homo sapiens splicing coactivator subunit srm300 (genbank accession AAF21439).
  • the polypeptide of SEQ ID NO:8 is not related to any known protein.
  • Preferred polypeptides and polynucleotides for use in the present invention are expected to have, ter alia, similar biological functions/properties to their homologous polypeptides and polynucleotides. Furthermore, preferred polypeptides, immunological fragments and polynucleotides of the present invention have at least one activity of either SEQ ED NO: 1, SEQ ED NO:2, SEQ ED NO:3, SEQ ED NO:4 or SEQ ED NO:8 as appropriate.
  • Polynucleotides for use in the invention may be obtained, using standard cloning and screening techniques, from a cDNA library derived from mRNA in cells of human tumour tissue (breast for example), (for example Sambrook et al., Molecular Cloning: A Laboratory Manual, 2 nd Ed., Cold Spring harbor Laboratory Press, Cold Spring harbor, N.Y. (1989)).
  • Polynucleotides of the invention can also be obtained from natural sources such as genomic DNA libraries or can be synthesised using well known and commercially available techniques.
  • the polynucleotide may include the coding sequence for the mature polypeptide, by itself; or the coding sequence for the mature polypeptide in reading frame with other coding sequences, such as those encoding a leader or secretory sequence, a pre-, or pro- or prepro- protein sequence, or other fusion peptide portions.
  • a marker sequence which facilitates purification of the fused polypeptide can be encoded.
  • the marker sequence is a hexa-histidine peptide, as provided in the pQE vector (Qiagen, hie.) and described in Gentz et al, Proc Natl Acad Sci USA (1989) 86:821-824, or is an HA tag.
  • the polynucleotide may also contain non-coding 5' and 3' sequences, such as transcribed, non-translated sequences, splicing and polyadenylation signals, ribosome binding sites and sequences that stabilise mRNA.
  • polypeptide variants which comprise the amino acid sequence of SEQ ID NO:2, SEQ ED NO: 3 or SEQ LD NO:8 and in which several, for instance from 5 to 10, 1 to 5, 1 to 3, 1 to 2 or 1, amino acid residues are substituted, deleted or added, in any combination.
  • Polynucleotides which are identical or sufficiently identical to a nucleotide sequence contained in SEQ LD NO: 1 or SEQ ID NO:4, may be used as hybridisation probes for cDNA and genomic DNA or as primers for a nucleic acid amplification (PCR) reaction, to isolate full-length cDNAs and genomic clones encoding polypeptides according to the present invention and to isolate cDNA and genomic clones of other genes (including genes encoding paralogs from human sources and orthologs and paralogs from species other than human) that have a high sequence similarity to SEQ ED NO: 1 or SEQ LD NO:4.
  • PCR nucleic acid amplification
  • these nucleotide sequences are 70% identical, preferably 80% identical, more preferably 90% identical, most preferably 95% identical to that of the referent.
  • the probes or primers will generally comprise at least 15 nucleotides, preferably, at least 30 nucleotides and may have at least 50 nucleotides. Particularly preferred probes will have between 30 and 50 nucleotides. Particularly preferred primers wiU have between 20 and 25 nucleotides.
  • polypeptides or polynucleotides derived from sequences from homologous animal origin could be used as immunogens to obtain a cross-reactive immune response to the human gene.
  • a polynucleotide encoding a polypeptide for use in of the present invention may be obtained by a process which comprises the steps of screening an appropriate library under stringent hybridisation conditions with a labelled probe having the sequence of SEQ ED NO: 1 or SEQ ED NO:4 or a fragment thereof; and isolating full-length cDNA and genomic clones containing said polynucleotide sequence.
  • Such hybridisation techniques are well known to the skilled artisan.
  • Preferred stringent hybridisation conditions include overnight incubation at 42°C in a solution comprising: 50% formamide, 5xSSC (150mM NaCI, 15mM trisodium citrate), 50 mM sodium phosphate (pH7.6), 5x Denhardt's solution, 10 % dextran sulfate, and 20 microgram/ml denatured, sheared salmon sperm DNA; followed by washing the filters in O.lx SSC at about 65°C.
  • the present invention also includes the use of polynucleotides obtainable by screening an appropriate library under stringent hybridisation conditions with a labelled probe having the sequence of SEQ ED NO:l or SEQ ED NO:4 or a fragment thereof.
  • an isolated cDNA sequence will be incomplete, in that the region coding for the polypeptide is short at the 5' end of the cDNA.
  • PCR Nucleic acid amplification
  • Recombinant polypeptides for use in the present invention may be prepared by processes well known in the art from genetically engineered host cells comprising expression systems. Accordingly, in a further aspect, the present invention relates to an expression system which comprises a polynucleotide, to host cells which are genetically engineered with such expression systems and to the production of polypeptides of the invention by recombinant techniques. Cell-free translation systems can also be employed to produce such proteins using RNAs derived from the DNA constructs of the present invention.
  • host cells can be genetically engineered to incorporate expression systems or portions thereof for polynucleotides of the present invention.
  • Introduction of polynucleotides into host cells can be effected by methods described in many standard laboratory manuals, such as Davis et al., Basic Methods in Molecular Biology (1986) and Sambrook et al, Molecular Cloning: A Laboratory Manual, 2nd Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY. (1989).
  • Preferred such methods include, for instance, calcium phosphate transfection, DEAE-dextran mediated transfection, transvection, microinjection, cationic lipid-mediated transfection, electroporation, transduction, scrape loading, ballistic introduction or infection.
  • the nucleic acids transfected into the host cells can be genetically engineered to comprise the optimisation of codon usage involving the replacement of at least one non-preferred or less preferred codon in a given sequence encoding a CASB7439 protein or fusion or fragment or variant thereof, by a preferred codon encoding the same amino acid.
  • the optimisation will be carried out to fit the preferred codons or more prefened codons for a given expression host, in particular for non-mammalian cell expression such as yeast or bacterial expression.
  • Particularly contemplated is a CASB7439 sequence optimised for E.coli expression.
  • At least one codon, and more prefereably all the codons of the CASB7439 protein or fragment or variant thereof can be changed to fit at best the host usage, especially the E.coli usage, that is, the one (or ones) that is the most prevalent.
  • a codon-optimised CASB7439 polynucleotide or polypeptide sequence Particularly preferred optimised CASB7439 sequences are given in SEQ ID NO: 6 coding for the polypeptide sequence set out in SEQ LD NO:7, and SEQ ED NO: 15 or SEQ ED NO:34 coding for the polypeptide sequence setu out in SEQ ED NO: 14.
  • Vectors comprising such DNA, hosts transformed thereby and the truncated or hybrid proteins themselves, expressed as described hereinbelow all form part of the invention.
  • the proteins are coexpressed with thioredoxin in trans (TIT).
  • TIT thioredoxin in trans
  • Coexpression of thioredoxin in trans versus in cis is preferred to keep antigen free of thioredoxin without the need for protease.
  • Thioredoxin coexpression eases the solubilisation of the proteins of the invention.
  • Thioredoxin coexpression has also a significant impact on protein purification yield, on purified-protein solubility and quality.
  • bacterial cells such as Streptococci, Staphylococci, E. coli, Streptomyces and Bacillus subtilis cells
  • fungal cells such as yeast cells and Aspergillus ceUs
  • insect cells such as Drosophila S2 and Spodoptera Sf9 cells
  • animal cells such as CHO, COS, HeLa, C127, 3T3, BHK, HEK 293 and Bowes melanoma cells
  • plant cells such as CHO, COS, HeLa, C127, 3T3, BHK, HEK 293 and Bowes melanoma cells.
  • expression systems can be used, for instance, chromosomal, episomal and virus-derived systems, e.g., vectors derived from bacterial plasmids, from bacteriophage, from transposons, from yeast episomes, from insertion elements, from yeast chromosomal elements, from viruses such as baculoviruses, papova viruses, such as SV40, vaccinia viruses, adenoviruses, fowl pox viruses, pseudorabies viruses and retroviruses, and vectors derived from combinations thereof, such as those derived from plasmid and bacteriophage genetic elements, such as cosmids and phagemids.
  • the expression systems may contain control regions that regulate as well as engender expression.
  • any system or vector which is able to maintain, propagate or express a polynucleotide to produce a polypeptide in a host may be used.
  • the appropriate nucleotide sequence may be inserted into an expression system by any of a variety of well-known and routine techniques, such as, for example, those set forth in Sambrook et al, Molecular Cloning, A Laboratory Manual (supra).
  • Appropriate secretion signals may be incorporated into the desired polypeptide to allow secretion of the translated protein into the lumen of the endoplasmic reticulum, the periplasmic space or the extracellular environment. These signals may be endogenous to the polypeptide or they may be heterologous signals.
  • the expression system may also be a recombinant live microorganism, such as a virus or bacterium.
  • the gene of interest can be inserted into the genome of a live recombinant virus or bacterium. Inoculation and in vivo infection with this live vector will lead to in vivo expression of the antigen and induction of immune responses.
  • polynucleotides encoding immunogenic polypeptides for use according to the present invention are introduced into suitable mammalian host cells for expression using any of a number of known viral-based systems.
  • refroviruses provide a convenient and effective platform for gene delivery systems.
  • a selected nucleotide sequence encoding a polypeptide for use in the present invention can be inserted into a vector and packaged in retroviral particles using techniques known in the art. The recombinant virus can then be isolated and delivered to a subject.
  • retroviral systems have been described (e.g., U.S. Pat. No.
  • adenovirus-based systems have also been described. Unlike retroviruses which integrate into the host genome, adenoviruses persist extrachromosomally thus mimmising the risks associated with insertional mutagenesis (Haj-Ahmad and Graham (1986) J. Virol. 57:267-274; Bett et al. (1993) J. Virol. 67:5911-5921; Mittereder et al. (1994) Human Gene Therapy 5:717-729; Seth et al. (1994) J. Virol. 68:933-940; Barr et al. (1994) Gene Therapy 1 :51-58; Berkner, K. L. (1988) BioTechniques 6:616-629; and Rich et al.
  • AAV vectors can be readily constructed using • techniques well known in the art. See, e.g., U.S. Pat. Nos. 5,173,414 and 5,139,941; International Publication Nos. WO 92/01070 and WO 93/03769; Lebkowski et al. (1988) Molec. Cell. Biol. 8:3988-3996; Vincent et al. (1990) Vaccines 90 (Cold Spring Harbor Laboratory Press); Carter, B. J. (1992) Current Opinion in Biotechnology 3:533-539; Muzyczka, N. (1992) Current Topics in Microbiol. and Immunol. 158:97-129; Kotin, R. M. (1994) Human Gene Therapy 5:793-801; Shelling and Smith (1994) Gene Therapy 1:165-169; and Zhou et al.
  • Additional viral vectors useful for delivering the nucleic acid molecules encoding polypeptides for use in the present invention by gene transfer include those derived from the pox family of viruses, such as vaccinia virus and avian poxvirus.
  • vaccinia virus recombinants expressing the molecules of interest can be constructed as follows. The DNA encoding a polypeptide is first inserted into an appropriate vector so that it is adjacent to a vaccinia promoter and flanking vaccinia DNA sequences, such as the sequence encoding thymidine kinase (TK). This vector is then used to transfect cells which are simultaneously infected with vaccinia.
  • TK thymidine kinase
  • Homologous recombination serves to insert the vaccinia promoter plus the gene encoding the polypeptide of interest into the viral genome.
  • the resulting TK.sup.(-) recombinant can be selected by culturing the cells in the presence of 5-bromodeoxyuridine and picking viral plaques resistant thereto.
  • a vaccinia-based infection/transfection system can be conveniently used to provide for inducible, transient expression or coexpression of one or more polypeptides described herein in host cells of an organism.
  • cells are first infected in vitro with a vaccinia virus recombinant that encodes the bacteriophage T7 RNA polymerase.
  • This polymerase displays extraordinar specificity in that it only transcribes templates bearing T7 promoters.
  • cells are transfected with the polynucleotide or polynucleotides of interest, driven by a T7 promoter.
  • the polymerase expressed in the cytoplasm from the vaccinia virus recombinant transcribes the transfected DNA into RNA which is then translated into polypeptide by the host translational machinery.
  • the method provides for high level, transient, cytoplasmic production of large quantities of RNA and its translation products. See, e.g., Elroy-Stein and Moss, Proc. Natl. Acad. Sci. USA (1990) 87:6743-6747; Fuerst et al. Proc. Natl. Acad. Sci. USA (1986) 83:8122-8126.
  • avipoxviruses such as the fowlpox and canarypox viruses
  • canarypox viruses can also be used to deliver the coding sequences of interest.
  • Recombinant avipox viruses expressing immunogens from mammalian pathogens, are known to confer protective immunity when administered to non-avian species.
  • the use of an Avipox vector is particularly desirable in human and other mammalian species since members of the Avipox genus can only productively replicate in susceptible avian species and therefore are not infective in mammalian cells.
  • Methods for producing recombinant Avipoxviruses are known in the art and employ genetic recombination, as described above with respect to the production of vaccinia viruses. See, e.g., WO 91/12882; WO 89/03429; and WO 92/03545.
  • alphavirus vectors can also be used for delivery of polynucleotide compositions for use in the present invention, such as those vectors described in U.S. Patent Nos. 5,843,723; 6,015,686; 6,008,035 and 6,015,694.
  • Certain vectors based on Venezuelan Equine Encephalitis (NEE) can also be used, illustrative examples of which can be found in U.S. Patent ⁇ os. 5,505,947 and 5,643,576.
  • molecular conjugate vectors such as the adenovirus chimeric vectors described in Michael et al. J. Biol. Chem. (1993) 268:6866-6869 and Wagner et al. Proc. ⁇ atl. Acad. Sci. USA (1992) 89:6099-6103, can also be used for gene delivery under the invention.
  • the recombinant live microorganisms described above can be virulent, or attenuated in various ways in order to obtain live vaccines. Such live vaccines also form part of the invention.
  • a polynucleotide may be integrated into the genome of a target cell. This integration may be in the specific location and orientation via homologous recombination (gene replacement) or it may be integrated in a random, non-specific location (gene augmentation).
  • the polynucleotide may be stably maintained in the cell as a separate, episomal segment of DNA. Such polynucleotide segments or "episomes" encode sequences sufficient to permit maintenance and replication independent of or in synchronization with the host cell cycle. The manner in which the expression construct is delivered to a cell and where in the cell the polynucleotide remains is dependent on the type of expression construct employed.
  • a polynucleotide is administered/delivered as "naked" DNA, for example as described in Ul er et al., Science 259:1145-1149, 1993 and reviewed by Cohen, Science 259:1691-1692, 1993.
  • the uptake of naked DNA may be increased by coating the DNA onto biodegradable beads, which are efficiently transported into the cells.
  • a composition of the present invention can be delivered via a particle bombardment approach, many of which have been described.
  • gas-driven particle acceleration can be achieved with devices such as those manufactured by Powderject Pharmaceuticals PLC (Oxford, UK) and Powderject Vaccines Inc. (Madison, WI), some examples of which are described in U.S. Patent Nos. 5,846,796; 6,010,478; 5,865,796; 5,584,807; and EP Patent No. 0500 799.
  • This approach offers a needle-free delivery approach wherein a dry powder formulation of microscopic particles, such as polynucleotide or polypeptide particles, are accelerated to high speed within a helium gas jet generated by a hand held device, propelling the particles into a target tissue of interest.
  • microscopic particles such as polynucleotide or polypeptide particles
  • compositions of the present invention include those provided by Bioject, Inc. (Portland, OR), some examples of which are described in U.S. Patent Nos. 4,790,824; 5,064,413; 5,312,335; 5,383,851; 5,399,163; 5,520,639 and 5,993,412.
  • Polypeptides for use in the present invention can be recovered and purified from recombinant cell cultures by well-known methods including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and lectin chromatography. Most preferably, ion metal affinity chromatography (EMAC) is employed for purification.
  • EMAC ion metal affinity chromatography
  • Well known techniques for refolding proteins may be employed to regenerate active conformation when the polypeptide is denatured during intracellular synthesis, isolation and or purification.
  • Another important aspect of the invention relates to a method for inducing , reinforcing or modulating an immunological response in a mammal which comprises inoculating the mammal with a fragment or the entire polypeptide or polynucleotide of the invention, adequate to produce antibody and or T cell immune response for prophylaxis or for therapeutic treatment of breast cancer.
  • Yet another aspect of the invention relates to a method of inducing, re-inforcing or modulating immunological response in a mammal which comprises, delivering a polypeptide as contemplated in the present invention via a vector or cell directing expression of the polynucleotide and coding for the polypeptide in vivo in order to induce such an immunological response to produce immune responses for prophylaxis or treatment of said mammal from breast cancer.
  • a further aspect of the invention relates to the use of an immunological/vaccine formulation (composition) which, when introduced into a mammalian host suffering from or susceptible to breast cancer, induces, re-inforces or modulates an immunological response in that mammal to a polypeptide according to the present invention wherein the composition comprises a polypeptide or polynucleotide of the invention or an immunological fragment thereof as herein before defined.
  • the vaccine formulation for use in breast cancer indication may further comprise a suitable carrier. Since a polypeptide may be broken down in the stomach, it is preferably administered parenterally (for instance, subcutaneous, intramuscular, intravenous, or intradermal injection).
  • Formulations suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents or thickening agents.
  • the formulations may be presented in unit-dose or multi-dose containers, for example, sealed ampoules and vials and may be stored in a freeze-dried condition requiring only the addition of the sterile liquid carrier immediately prior to use.
  • a further aspect of the invention relates to the in vitro induction of immune responses to a fragment or the entire polypeptide or polynucleotide disclosed herein or a molecule comprising said polypeptide or polynucleotide, using cells from the immune system of a mammal, and reinfusing these activated immune cells of the mammal for the treatment of breast cancer.
  • Activation of the cells from the immune system is achieved by in vitro incubation with the entire polypeptide or polynucleotide according to the present invention or a molecule comprising said polypeptide or polynucleotide in the presence or absence of various immunomodulator molecules.
  • a further aspect of the invention relates to the immunisation of a mammal suffering from or susceptible to breast cancer by administration of antigen presenting cells modified by in vitro loading with part or the entire polypeptide disclosed herein or a molecule comprising said polypeptide and administered in vivo in an immunogenic way.
  • antigen presenting cells can be transfected in vitro with a vector containing a fragment or the entire polynucleotide according to the present invention or a molecule comprising said polynucleotide, such as to express the corresponding polypeptide, and administered in vivo in an immunogenic way.
  • the pharmaceutical compositions described herein will comprise one or more immunostimulants in addition to the immunogenic polynucleotide, polypeptide, antibody, T-cell and/or antigen presenting cell (APC) compositions of this invention.
  • An immunostimulant refers to essentially any substance that enhances or potentiates an immune response (antibody and/or cell-mediated) to an exogenous antigen.
  • One preferred type of immunostimulant comprises an adjuvant.
  • Many adjuvants contain a substance designed to protect the antigen from rapid catabolism, such as aluminum hydroxide or mineral oil, and a stimulator of immune responses, such as lipid A, Bortadella pertussis or Mycobacterium tuberculosis derived proteins.
  • adjuvants are commercially available as, for example, Freund's Incomplete Adjuvant and Complete Adjuvant (Difco Laboratories, Detroit, MI); Merck Adjuvant 65 (Merck and Company, Inc., Rahway, NJ); AS-2 (SmithKline Beecham, Philadelphia, PA); aluminum salts such as aluminum hydroxide gel (alum) or aluminum phosphate; salts of calcium, iron or zinc; an insoluble suspension of acylated tyrosine; acylated sugars; cationically or anionically derivatized polysaccharides; polyphosphazenes; biodegradable microspheres; monophosphoryl lipid A and quil A.
  • Cytokines such as GM-CSF, interleukin-2, - 7, -12, and other like growth factors, may also be used as adjuvants.
  • the adjuvant composition is preferably one that induces an immune response predominantly of the Thl type.
  • High levels of Thl -type cytokines e.g., IFN- ⁇ , TNF ⁇ , IL-2 and IL-12
  • high levels of Th2-type cytokines e.g., IL-4, IL-5, IL-6 and IL-10
  • a patient will support an immune response that includes Thl- and Th2-type responses.
  • Thl -type cytokines will increase to a greater extent than the level of Th2-type cytokines.
  • the levels of these cytokines may be readily assessed using standard assays. For a review of the families of cytokines, see Mosmann and Coffman, Ann. Rev. Immunol. 7:145- 173, 1989.
  • Certain preferred adjuvants for eliciting a predominantly Thl -type response include, for example, a combination of monophosphoryl lipid A, preferably 3-de- O-acylated monophosphoryl lipid A, together with an aluminum salt.
  • MPL ® adjuvants are available from Corixa Corporation (Seattle, WA; see, for example, US Patent Nos. 4,436,727; 4,877,611; 4,866,034 and 4,912,094).
  • CpG-containing oligonucleotides in which the CpG dinucleotide is unrnethylated also induce a predominantly Thl response.
  • Such oligonucleotides are well known and are described, for example, in WO 96/02555, WO 99/33488 and U.S. Patent Nos.
  • Another preferred adjuvant comprises a saponin, such as Quil A, or derivatives thereof, including QS21 and QS7 (Aquila Biopharmaceuticals Inc., Framingham, MA); Escin; Digitonin; or Gypsophila or Chenopodium quinoa saponins .
  • Other preferred formulations include more than one saponin in the adjuvant combinations of the present invention, for example combinations of at least two of the following group comprising QS21, QS7, Quil A, ⁇ -escin, or digitonin.
  • the saponin formulations may be combined with vaccine vehicles composed of chitosan or other polycationic polymers, polylactide and polylactide- co-glycolide particles, poly-N-acetyl glucosamine-based polymer matrix, particles composed of polysaccharides or chemically modified polysaccharides, liposomes and lipid-based particles, particles composed of glycerol monoesters, etc.
  • vaccine vehicles composed of chitosan or other polycationic polymers, polylactide and polylactide- co-glycolide particles, poly-N-acetyl glucosamine-based polymer matrix, particles composed of polysaccharides or chemically modified polysaccharides, liposomes and lipid-based particles, particles composed of glycerol monoesters, etc.
  • the saponins may also be formulated in the presence of cholesterol to form particulate structures such as liposomes or ISCOMs.
  • the saponins may be formulated together with a polyoxyethylene ether or ester, in either a non- particulate solution or suspension, or in a particulate structure such as a paucilamelar liposome or ISCOM.
  • the saponins may also be formulated with excipients such as Carbopol R to increase viscosity, or may be formulated in a dry powder form with a powder excipient such as lactose.
  • the adjuvant system includes the combination of a monophosphoryl lipid A and a saponin derivative, such as the combination of QS21 and 3D-MPL ® adjuvant, as described in WO 94/00153, or a less reactogenic composition where the QS21 is quenched with cholesterol, as described in WO 96/33739.
  • a monophosphoryl lipid A and a saponin derivative such as the combination of QS21 and 3D-MPL ® adjuvant, as described in WO 94/00153
  • a less reactogenic composition where the QS21 is quenched with cholesterol as described in WO 96/33739.
  • Other preferred formulations comprise an oil-in-water emulsion and tocopherol.
  • Another particularly preferred adjuvant formulation employing QS21 , 3D-MPL ® adjuvant and tocopherol in an oil-in-water emulsion is described in WO 95/17210.
  • Another enhanced adjuvant system involves the combination of a CpG-containing oligonucleotide and a saponin derivative particularly the combination of CpG and QS21 as disclosed in WO 00/09159.
  • the formulation additionally comprises an oil in water emulsion and tocopherol.
  • Additional illustrative adjuvants for use in the pharmaceutical compositions of the invention include Montanide ISA 720 (Seppic, France), SAF (Chiron, California, United States), ISCOMS (CSL), MF-59 (Chiron), the SBAS series of adjuvants (e.g., SBAS-2 or SBAS-4, available from SmithKline Beecham, Rixensart, Belgium), Detox (Enhanzyn ® ) (Corixa, Hamilton, MT), RC-529 (Corixa, Hamilton, MT) and other aminoalkyl glucosaminide 4-phosphates (AGPs), such as those described in pending U.S. Patent Application Serial Nos. 08/853,826 and 09/074,720, the disclosures of which are incorporated herein by reference in their entireties, and polyoxyethylene ether adjuvants such as those described in WO 99/52549A1.
  • n 1-50
  • A is a bond or -C(O)-
  • R is Ci- 50 alkyl or Phenyl C ⁇ - 50 alkyl.
  • One embodiment of the present invention consists of a vaccine formulation for use in the treatment or prophylaxis of breast cancer comprising a polyoxyethylene ether of general formula (I), wherein n is between 1 and 50, preferably 4-24, most preferably 9; the R component is C 1 -50, preferably C 4 -C 20 alkyl and most preferably C 12 alkyl, and A is a bond.
  • the concentration of the polyoxyethylene ethers should be in the range 0.1-20%, preferably from 0.1-10%, and most preferably in the range 0.1-1%.
  • Preferred polyoxyethylene ethers are selected from the following group: polyoxyethylene-9-lauryl ether, polyoxyethylene-9- steoryl ether, polyoxyethylene-8-steoryl ether, polyoxyethylene-4-lauryl ether, polyoxyethylene-35-lauryl ether, and polyoxyethylene-23-lauryl ether.
  • Polyoxyethylene ethers such as polyoxyethylene lauryl ether are described in the Merck index (12 edition: entry 7717). These adjuvant molecules are described in WO 99/52549.
  • polyoxyethylene ether according to the general formula (I) above may, if desired, be combined with another adjuvant.
  • a prefened adjuvant combination is preferably with CpG as described in the pending UK patent application GB 9820956.2.
  • a carrier is also present in the vaccine composition for use according to the invention.
  • the carrier may be an oil in water emulsion, or an aluminium salt, such as aluminium phosphate or aluminium hydroxide.
  • a prefened oil-in-water emulsion comprises a metabolisible oil, such as squalene, alpha tocopherol and Tween 80.
  • a metabolisible oil such as squalene, alpha tocopherol and Tween 80.
  • the antigens in the vaccine composition according to the invention are combined with QS21 and 3D-MPL in such an emulsion.
  • the oil in water emulsion may contain span 85 and/or lecithin and/or tricaprylin.
  • QS21 and 3D-MPL will be present in a vaccine in the range of l ⁇ g - 200 ⁇ g, such as 10-100 ⁇ g, preferably lO ⁇ g - 50 ⁇ g per dose.
  • the oil in water will comprise from 2 to 10% squalene, from 2 to 10% alpha tocopherol and from 0.3 to 3% tween 80.
  • the ratio of squalene: alpha tocopherol is equal to or less than 1 as this provides a more stable emulsion.
  • Span 85 may also be present at a level of 1 %. In some cases it may be advantageous that the vaccines of the present invention will further contain a stabiliser.
  • Non-toxic oil in water emulsions preferably contain a non-toxic oil, e.g. squalane or squalene, an emulsifier, e.g. Tween 80, in an aqueous carrier.
  • a non-toxic oil e.g. squalane or squalene
  • an emulsifier e.g. Tween 80
  • the aqueous carrier may be, for example, phosphate buffered saline.
  • the present invention also provides the use of a polyvalent vaccine composition in the treatment or prophylaxis of breast cancer comprising a vaccine formulation of the invention in combination with other antigens, useful for treating breast cancers, .
  • a polyvalent vaccine composition may include a TH-1 inducing adjuvant as hereinbefore described.
  • An immunogenic composition for use according to the invention is delivered to a host via antigen presenting cells (APCs), such as dendritic cells, macrophages, B cells, monocytes and other cells that may be engineered to be efficient APCs.
  • APCs antigen presenting cells
  • Such cells may, but need not, be genetically modified to increase the capacity for presenting the antigen, to improve activation and or maintenance of the T cell response, to have anti-tumour effects per se and/or to be immunologically compatible with the receiver (i.e., matched HLA haplotype).
  • APCs may generally be isolated from any of a variety of biological fluids and organs, including tumour and peritumoural tissues, and may be autologous, allogeneic, syngeneic or xenogeneic cells.
  • Dendritic cells are highly potent APCs (Banchereau and Steinman, Nature 392:245-251, 1998) and have been shown to be effective as a physiological adjuvant for eliciting prophylactic or therapeutic antitumour immunity (see Timmerman and Levy, Ann. Rev. Med. 50:507-529, 1999). Ln general, dendritic cells may be identified based on their typical shape (stellate in situ, with marked cytoplasmic processes (dendrites) visible in vitro), their ability to take up, process and present antigens with high efficiency and their ability to activate naive T cell responses.
  • Dendritic cells may, of course, be engineered to express specific cell-surface receptors or ligands that are not commonly found on dendritic cells in vivo or ex vivo, and such modified dendritic cells are contemplated by the present invention.
  • secreted vesicles antigen-loaded dendritic cells called exosomes
  • exosomes antigen-loaded dendritic cells
  • Dendritic cells and progenitors may be obtained from peripheral blood, bone marrow, tumour-infiltrating cells, peritumoural tissues-infiltrating cells, lymph nodes, spleen, skin, umbilical cord blood or any other suitable tissue or fluid.
  • dendritic cells may be differentiated ex vivo by adding a combination of cytokines such as GM-CSF, IL-4, IL-13 and/or TNF ⁇ to cultures of monocytes harvested from peripheral blood.
  • CD34 positive cells harvested from peripheral blood, umbilical cord blood or bone manow may be differentiated into dendritic cells by adding to the culture medium combinations of GM-CSF, EL-3, TNF ⁇ , CD40 ligand, LPS, flt3 ligand and/or other compound(s) that induce differentiation, maturation and proliferation of dendritic cells.
  • Dendritic cells are conveniently categorized as “immature” and “mature” cells, which allows a simple way to discriminate between two well characterized phenotypes. However, this nomenclature should not be construed to exclude all possible intermediate stages of differentiation. Immature dendritic cells are characterized as APC with a high capacity for antigen uptake and processing, which corcelates with the high expression of Fc ⁇ receptor and mannose receptor.
  • APCs may generally be transfected with a polynucleotide of the invention (or portion or other variant thereof) such that the encoded polypeptide, or an immunogenic portion thereof, is expressed on the cell surface. Such transfection may take place ex vivo, and a pharmaceutical composition comprising such transfected cells may then be used for therapeutic purposes, as described herein.
  • a gene delivery vehicle that targets a dendritic or other antigen presenting cell may be administered to a patient, resulting in transfection that occurs in vivo.
  • In vivo and ex vivo transfection of dendritic cells may generally be performed using any methods known in the art, such as those described in WO 97/24447, or the gene gun approach described by Mahvi et al., Immunology and cell Biology 75:456-460, 1997.
  • Antigen loading of dendritic cells may be achieved by incubating dendritic cells or progenitor cells with the tumour polypeptide, DNA (naked or within a plasmid vector) or RNA; or with antigen-expressing recombinant bacterium or viruses (e.g., vaccinia, fowlpox, adenovirus or lentivirus vectors).
  • the polypeptide Prior to loading, the polypeptide may be covalently conjugated to an immunological partner that provides T cell help (e.g., a carrier molecule).
  • a dendritic cell may be pulsed with a non- conjugated immunological partner, separately or in the presence of the polypeptide.
  • compositions for use according to the present invention, the type of carrier will typically vary depending on the mode of administration.
  • the compositions may be formulated for any appropriate manner of administration, including for example, topical, oral, nasal, mucosal, intravenous, intracranial, intraperitoneal, subcutaneous and intramuscular administration.
  • Carriers for use within such pharmaceutical compositions are biocompatible, and may also be biodegradable.
  • the formulation preferably provides a relatively constant level of active component release. En other embodiments, however, a more rapid rate of release immediately upon administration may be desired.
  • the formulation of such compositions is well within the level of ordinary skill in the art using known techniques.
  • Illustrative carriers useful in this regard include microparticles of poly(lactide-co-glycolide), polyacrylate, latex, starch, cellulose, dextran and the like.
  • illustrative delayed-release carriers include supramolecular biovectors, which comprise a non- liquid hydrophilic core (e.g., a cross-linked polysaccharide or oligosaccharide) and, optionally, an external layer comprising an amphiphihc compound, such as a phospholipid (see e.g., U.S. Patent No. 5,151,254 and PCT applications WO 94/20078, WO/94/23701 and WO 96/06638).
  • the amount of active compound contained within a sustained release formulation depends upon the site of implantation, the rate and expected duration of release and the nature of the condition to be treated or prevented.
  • biodegradable microspheres e.g., polylactate polyglycolate
  • Suitable biodegradable microspheres are disclosed, for example, in U.S. Patent Nos. 4,897,268; 5,075,109; 5,928,647; 5,811,128; 5,820,883; 5,853,763;
  • hepatitis B core protein carrier systems such as described in WO/99 40934, and references cited therein, will also be useful for many applications.
  • Another illustrative carrier/delivery system employs a carrier comprising particulate-protein complexes, such as those described in U.S. Patent No. 5,928,647, which are capable of inducing a class I- restricted cytotoxic T lymphocyte responses in a host.
  • compositions according to the invention will often further comprise one or more buffers (e.g., neutral buffered saline or phosphate buffered saline), carbohydrates (e.g., glucose, mannose, sucrose or dextrans), mannitol, proteins, polypeptides or amino acids such as glycine, antioxidants, bacteriostats, chelating agents such as EDTA or glutathione, adjuvants (e.g., aluminum hydroxide), solutes that render the formulation isotonic, hypotonic or weakly hypertonic with the blood of a recipient, suspending agents, thickening agents and/or preservatives.
  • buffers e.g., neutral buffered saline or phosphate buffered saline
  • carbohydrates e.g., glucose, mannose, sucrose or dextrans
  • mannitol proteins
  • proteins polypeptides or amino acids
  • proteins e.glycine
  • antioxidants e.g., antioxidants
  • compositions described herein may be presented in unit-dose or multi-dose containers, such as sealed ampoules or vials. Such containers are typically sealed in such a way to preserve the sterility and stability of the formulation until use.
  • formulations may be stored as suspensions, solutions or emulsions in oily or aqueous vehicles.
  • a pharmaceutical composition may be stored in a freeze-dried condition requiring only the addition of a sterile liquid carrier immediately prior to use.
  • compositions disclosed herein may be delivered via oral administration to an animal.
  • these compositions may be formulated with an inert diluent or with an assimilable edible carrier, or they may be enclosed in hard- or soft-shell gelatin capsule, or they may be compressed into tablets, or they may be incorporated directly with the food of the diet.
  • the active compounds may even be incorporated with excipients and used in the form of ingestible tablets, buccal tables, troches, capsules, elixirs, suspensions, syrups, wafers, and the like (see, for example, Mathiowitz et ⁇ l., Nature 1997 Mar 27;386(6623):410-4; Hwang et ⁇ l, CritRev Ther Drug Carrier Syst 1998;15(3):243-84; U. S. Patent 5,641,515; U. S. Patent 5,580,579 an U. S. Patent 5,792,451).
  • Tablets, troches, pills, capsules and the like may also contain any of a variety of additional components, for example, a binder, such as gum tragacanth, acacia, cornstarch, or gelatin; excipients, such as dicalcium phosphate; a disintegrating agent, such as com starch, potato starch, alginic acid and the like; a lubricant, such as magnesium stearate; and a sweetening agent, such as sucrose, lactose or saccharin maybe added or a flavoring agent, such as peppermint, oil of wintergreen, or cheny flavoring.
  • a binder such as gum tragacanth, acacia, cornstarch, or gelatin
  • excipients such as dicalcium phosphate
  • a disintegrating agent such as com starch, potato starch, alginic acid and the like
  • a lubricant such as magnesium stearate
  • a sweetening agent such as sucrose, lac
  • any material used in preparing any dosage unit form should be pharmaceutically pure and substantially non-toxic in the amounts employed.
  • the active compounds may be incorporated into sustained- release preparation and formulations.
  • these formulations will contain at least about 0.1% of the active compound or more, although the percentage of the active ingredient(s) may, of course, be varied and may conveniently be between about 1 or 2% and about 60% or 70% or more of the weight or volume of the total formulation.
  • the amount of active compound(s) in each therapeutically useful composition may be prepared is such a way that a suitable dosage will be obtained in any given unit dose of the compound.
  • Factors such as solubility, bioavailability, biological half- life, route of administration, product shelf life, as well as other pharmacological considerations will be contemplated by one skilled in the art of preparing such pharmaceutical formulations, and as such, a variety of dosages and treatment regimens may be desirable.
  • compositions according to the present invention may alternatively be incorporated with one or more excipients in the form of a mouthwash, dentifrice, buccal tablet, oral spray, or sublingual orally-administered formulation.
  • the active ingredient may be incorporated into an oral solution such as one containing sodium borate, glycerin and potassium bicarbonate, or dispersed in a dentifrice, or added in a therapeutically-effective amount to a composition that may include water, binders, abrasives, flavoring agents, foaming agents, and humectants.
  • the compositions may be fashioned into a tablet or solution form that may be placed under the tongue or otherwise dissolved in the mouth.
  • solutions of the active compounds as free base or pharmacologically acceptable salts may be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose.
  • Dispersions may also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations generally will contain a preservative to prevent the growth of microorganisms.
  • Illustrative pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions (for example, see U. S. Patent 5,466,468).
  • the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and/or vegetable oils.
  • polyol e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like
  • suitable mixtures thereof e.g., vegetable oils
  • vegetable oils e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like
  • suitable mixtures thereof e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like
  • vegetable oils e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like
  • Proper fluidity may be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion
  • isotonic agents for example, sugars or sodium chloride.
  • Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • aqueous solutions for parenteral administration in an aqueous solution, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose.
  • a sterile aqueous medium that can be employed will be known to those of skill in the art in light of the present disclosure.
  • one dosage maybe dissolved in 1 ml of isotonic NaCI solution and either added to 1000 ml of hypodermoclysis fluid or injected at the proposed site of infusion, (see for example, "Remington's Pharmaceutical Sciences" 15th Edition, pages 1035-1038 and 1570-1580).
  • preparations will of course preferably meet sterility, pyrogenicity, and the general safety and purity standards as required by FDA Office of Biologies standards.
  • compositions disclosed herein may be formulated in a neutral or salt form.
  • Illustrative pharmaceutically-acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like. Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective.
  • the carriers can further comprise any and all solvents, dispersion media, vehicles, coatings, diluents, antibacterial and antifungal agents, isotonic and absorption delaying agents, buffers, carrier solutions, suspensions, colloids, and the like.
  • solvents dispersion media, vehicles, coatings, diluents, antibacterial and antifungal agents, isotonic and absorption delaying agents, buffers, carrier solutions, suspensions, colloids, and the like.
  • the use of such media and agents for pharmaceutical active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions.
  • pharmaceutically-acceptable refers to molecular entities and compositions that do not produce an allergic or similar untoward reaction when administered to a human.
  • the pharmaceutical compositions may be delivered by intranasal sprays, inhalation, and/or other aerosol delivery vehicles.
  • Methods for delivering genes, nucleic acids, and peptide compositions directly to the lungs via nasal aerosol sprays has been described, e.g., inU. S. Patent 5,756,353 and U. S. Patent 5,804,212.
  • the delivery of drugs using intranasal microparticle resins (Takenaga et al, J Controlled Release 1998 Mar 2;52(l-2):81-7) and lysophosphatidyl-glycerol compounds (U. S. Patent 5,725,871) are also well- known in the pharmaceutical arts.
  • illustrative transmucosal drug delivery in the form of a polytetrafluoroetheylene support matrix is described in U. S. Patent 5,780,045.
  • liposomes, nanocapsules, microparticles, lipid particles, vesicles, and the like are used for the introduction of the compositions of the present invention into suitable host cells/organisms.
  • the compositions of the present invention may be formulated for delivery either encapsulated in a lipid particle, a liposome, a vesicle, a nanosphere, or a nanoparticle or the like.
  • the compositions disclosed herein can be bound, either covalently or non-covalently, to the surface of such carrier vehicles.
  • liposome and liposome-like preparations as potential drag carriers is generally known to those of skill in the art (see for example, Lasic, Trends Biotechnol 1998 Jul; 16(7): 307-21; Takakura, Nippon Rinsho 1998 Mar;56(3):691-5; Chandran et al, Indian J Exp Biol. 1997 Aug;35(8):801-9; Margalit, Crit Rev Ther Drug Carrier Syst. 1995;12(2-3):233-61; U.S. Patent 5,567,434; U.S. Patent 5,552,157; U.S. Patent 5,565,213; U.S. Patent 5,738,868 and U.S. Patent 5,795,587, each specifically incorporated herein by reference in its entirety).
  • Liposomes have been used successfully with a number of cell types that are normally difficult to transfect by other procedures, including T cell suspensions, primary hepatocyte cultures and PC 12 cells (Renneisen et al, J Biol Chem. 1990 Sep 25;265(27): 16337-42; Muller et al, DNA Cell Biol. 1990 Apr;9(3):221-9).
  • liposomes are free of the DNA length constraints that are typical of viral- based delivery systems. Liposomes have been used effectively to introduce genes, various drugs, radiotherapeutic agents, enzymes, viruses, transcription factors, allosteric effectors and the like, into a variety of cultured cell lines and animals.
  • liposomes does not appear to be associated with autoimmune responses or unacceptable toxicity after systemic delivery.
  • liposomes are formed from phospholipids that are dispersed in an aqueous medium and spontaneously form multilamellar concentric bilayer vesicles (also termed multilamellar vesicles (MLVs).
  • MLVs multilamellar vesicles
  • the invention provides the use of pharmaceutically-acceptable nanocapsule formulations of the compositions disclosed herein.
  • Nanocapsules can generally entrap compounds in a stable and reproducible way (see, for example, Quintanar-Guenero et al., Drug Dev End Pharm. 1998 Dec;24(12): 1113-28).
  • ultrafine particles sized around 0.1 ⁇ m
  • Such particles can be made as described, for example, by Couvreur et al, Crit Rev Ther Drug Carrier Syst.
  • This invention also relates to the use of polynucleotides, in the form of primers derived from the polynucleotides of the present invention, and of polypeptides, in the form of antibodies or reagents specific for the polypeptide of the present invention, as diagnostic reagents in the context of breast cancer.
  • the identification of genetic or biochemical markers in blood or tissues that will enable the detection of very early changes along the carcinogenesis pathway will help in determining the best treatment for the patient.
  • Surrogate tumour markers such as polynucleotide expression
  • the identification of expression levels of the polynucleotides of the invention will be useful in both the staging of the cancerous disorder and grading the nature of the cancerous tissue.
  • the staging process monitors the advancement of the cancer of the breast and is determined on the presence or absence of malignant tissue in the areas biopsied.
  • the polynucleotides of the invention can help to perfect the staging process by identifying markers for the aggressivity of said cancer, for example the presence in different areas of the body.
  • the grading of the cancer describes how closely a tumour resembles normal tissue of its same type and is assessed by its cell morphology and other markers of differentiation.
  • the polynucleotides of the invention can be useful in determining the tumour grade as they can help in the determination of the differentiation status of the cells of a tumour.
  • the diagnostic assays offer a process for diagnosing or determining a susceptibility to breast cancer, through diagnosis by methods comprising determining from a sample derived from a subject an abnormally decreased or increased level of polypeptide or mRNA.
  • This method of diagnosis is known as differential expression.
  • the expression of a particular gene is compared between a diseased tissue and a normal tissue.
  • a difference between the polynucleotide-related gene, mRNA, or protein in the two tissues is compared, for example in molecular weight, amino acid or nucleotide sequence, or relative abundance, indicates a change in the gene, or a gene which regulates it, in the tissue of the human that was suspected of being diseased.
  • RNA level Decreased or increased expression can be measured at the RNA level.
  • PolyA RNA is first isolated from the two tissues and the detection of mRNA encoded by a gene conesponding to a differentially expressed polynucleotide according to the invention can be performed by, for example, in situ hybridisation in tissue sections, reverse trascriptase-PCR, using Northern blots containing poly A+ mRNA, or any other direct or inderect RNA detection method.
  • An increased or decreased expression of a given RNA in a diseased tissue compared to a normal tissue suggests that the transcript and/or the expressed protein has a role in the disease.
  • detection of a higher or lower level of mRNA conesponding to SEQ TD NO: 1 or SEQ ED NO:4 relative to normal level is indicative of the presence of cancer in the patient.
  • mRNA expression levels in a sample can be determined by generation of a library of expressed sequence tags (ESTs) from the sample.
  • ESTs expressed sequence tags
  • the relative representation of ESTs in the library can be used to assess the relative representation of the gene transcript in the starting sample.
  • the EST analysis of the test can then be compared to the EST analysis of a reference sample to determine the relative expression levels of the polynucleotide of interest.
  • Other mRNA analyses can be carried out using serial analysis of gene expression (SAGE) methodology (Velculescu et. Al. Science (1995) 270:484) , differential display methodology (For example, US 5,776,683) or hybridisation analysis which relies on the specificity of nucleotide interactions.
  • the comparison could be made at the protein level.
  • the protein sizes in the two tissues may be compared using antibodies to detect polypeptides in Western blots of protein extracts from the two tissues. Expression levels and subcellular localization may also be detected immtmologically using antibodies to the conesponding protein. Further assay techniques that can be used to determine levels of a protein, such as a polypeptide of the present invention, in a sample derived from a host are well-known to those of skill in the art. A raised or decreased level of polypeptide expression in the diseased tissue compared with the same protein expression level in the normal tissue indicates that the expressed protein may be involved in the disease.
  • the diagnosis can be determined by detection of gene product expression levels encoded by at least one sequence set forth in SEQ ID NO: 1 or SEQ ED NO:4.
  • a comparison of the mRNA or protein levels in a diseased versus normal tissue may also be used to follow the progression or remission of a disease.
  • polynucleotide sequences in a sample can be assayed using polynucleotide anays. These can be used to examine differential expression of genes and to determine gene function.
  • anays of the polynucleotide sequences SEQ ED NO: 1 or SEQ ED NO:4 can be used to determine if any of the polynucleotides are differentially expressed between a normal and cancer cell.
  • an anay of oligonucleotides probes comprising the SEQ TD NO: 1 or SEQ ED NO:4 nucleotide sequence or fragments thereof can be constructed to conduct efficient screening of e.g., genetic mutations.
  • Anay technology methods are well known and have general applicability and can be used to address a variety of questions in molecular genetics including gene expression, genetic linkage, and genetic variability (see for example: M.Chee et al., Science, Vol 274, pp 610-613 (1996)).
  • Diagnosis includes determination of a subject's susceptibility to breast cancer disease, determination as to whether a subject presently has said disease, and also the prognosis of a subject affected by said disease.
  • the present invention further relates to a diagnostic kit for performing a diagnostic assay in a patient suffering from or susceptible to breast cancer disease which comprises:
  • a polynucleotide of the present invention preferably the nucleotide sequence of SEQ ID NO: 1 or SEQ ED NO:4, or a fragment thereof ;
  • polypeptide of the present invention preferably the polypeptide of SEQ TD NO: 2, SEQ ED NO:3 or SEQ ED NO:8, or a fragment thereof; or
  • an antibody to a polypeptide of the present invention preferably to the polypeptide of SEQ ID NO: 2, SEQ ED NO:3 or SEQ ED NO:8.
  • the nucleotide sequences for use in the present invention are also valuable for chromosomal localisation.
  • the sequence is specifically targeted to, and can hybridize with, a particular location on an individual human chromosome.
  • the mapping of relevant sequences to chromosomes according to the present invention is an important first step in conelating those sequences with gene associated disease. Once a sequence has been mapped to a precise chromosomal location, the physical position of the sequence on the chromosome can be conelated with genetic map data. Such data are found in, for example, V. McKusick, Mendelian Inheritance in Man (available on-line through Johns Hopkins University Welch Medical Library). The relationship between genes and diseases that have been mapped to the same chromosomal region are then identified through linkage analysis (coinheritance of physically adjacent genes). The differences in the cDNA or genomic sequence between affected and unaffected individuals can also be determined.
  • polypeptides for use in the invention or their fragments or analogs thereof, or cells expressing them can also be used as immunogens to produce antibodies immunospecific for polypeptides of the present invention.
  • immunospecific means that the antibodies have substantially greater affinity for the polypeptides disclosed herein than their affinity for other related polypeptides in the prior art.
  • the invention provides for the use of an antibody immunospecific for a polypeptide according to the invention or an immunological fragment thereof as hereinbefore defined.
  • the antibody is a monoclonal antibody
  • Antibodies generated against the polypeptides disclosed herein may be obtained by administering the polypeptides or epitope-bearing fragments, analogs or cells to an animal, preferably a non-human animal, using routine protocols.
  • any technique which provides antibodies produced by continuous cell line cultures can be used. Examples include the hybridoma technique (Kohler, G. and Milstein, C, Nature (1975) 256:495-497), the trioma technique, the human B-cell hybridoma technique (Kozbor et al, Immunology Today (1983) 4:72) and the EBV-hybridoma technique (Cole et al, Monoclonal Antibodies and Cancer Therapy, 77-96, Alan R. Liss, Inc., 1985).
  • the above-described antibodies may be employed to isolate or to identify clones expressing the polypeptide or to purify the polypeptides by affinity chromatography.
  • the antibody of the invention may also be employed to prevent or treat breast cancer in a patient.
  • Another aspect of the invention relates to a method for inducing or modulating an immunological response in a mammal which comprises inoculating the mammal with a polypeptide of the present invention, adequate to produce antibody and/or T cell immune response to protect or ameliorate the symptoms or progression of the breast cancer.
  • Yet another aspect of the invention relates to a method of inducing or modulating immunological response in a mammal which comprises, delivering a polypeptide of the present invention via a vector directing expression of the polynucleotide and coding for the polypeptide in vivo in order to induce such an immunological response to produce antibody to protect said animal from breast cancer.
  • the present invention therefore provides a method of treating breast cancer, related to either a presence of, an excess of, or an under- expression of, CASB7439 polypeptide activity.
  • the present invention further provides for a method of screening compounds to identify those which stimulate or which inhibit the function of the CASB7439 polypeptide.
  • agonists or antagonists may be employed for therapeutic and prophylactic purposes for breast cancer as hereinbefore mentioned.
  • Compounds may be identified from a variety of sources, for example, cells, ceU-free preparations, chemical libraries, and natural product mixtures.
  • Such agonists, antagonists or inhibitors so-identified may be natural or modified substrates, ligands, receptors, enzymes, etc., as the case may be, of the polypeptide; or may be structural or functional mimetics thereof (see Coligan et al, Current Protocols in Immunology l(2):Chapter 5 (1991)).
  • the invention provides a method for screening to identify compounds which stimulate or which inhibit the function of the polypeptides according to the invention which comprises a method selected from the group consisting of:
  • polypeptides according to the may be used to identify membrane bound or soluble receptors, if any, through standard receptor binding techniques known in the art. Well known screening methods may also be used to identify agonists and antagonists of the polypeptides disclosed herein which compete with the binding of the polypeptide of the invention to its receptors, if any.
  • the present invention relates to a screening kit for identifying agonists, antagonists, ligands, receptors, substrates, enzymes, etc. for polypeptides disclosed herein that are used for therapeutic and prophylactic purposes for breast cancer; or compounds which decrease or enhance the production of such polypeptides, which comprises:
  • polypeptide of the present invention (c) a cell membrane expressing a polypeptide of the present invention; or (d) antibody to a polypeptide of the present invention; which polypeptide is preferably that of SEQ ED NO: 2, SEQ ED NO:3 or SEQ ID NO:8.
  • a polypeptide according to the present invention may also be used in a method for the structure-based design of an agonist, antagonist or inhibitor of the polypeptide, by: (a) determining in the first instance the three-dimensional structure of the polypeptide; (b) deducing the three-dimensional structure for the likely reactive or binding site(s) of an agonist, antagonist or inhibitor;
  • Gene therapy may also be employed to effect the endogenous production of CASB7439 polypeptide by the relevant cells in the subject.
  • Gene therapy see Chapter 20, Gene Therapy and other Molecular Genetic-based Therapeutic Approaches, (and references cited therein) in Human Molecular Genetics, T Strachan and A P Read, BIOS Scientific Publishers Ltd (1996).
  • Vaccine preparation is generally described in Pharmaceutical Biotechnology, Vol.61 Vaccine Design - the subunit and adjuvant approach, edited by Powell and
  • Encapsulation within liposomes is described, for example, by Fullerton, U.S.
  • Patent 4,235,877 Conjugation of proteins to macromolecules is disclosed, for example, by Likhite, U.S. Patent 4,372,945 and by Armor et al, U.S. Patent
  • each vaccine dose is selected as an amount which induces an immunoprotective response without significant, adverse side effects in typical vaccinees. Such amount will vary depending upon which specific immunogen is employed. Generally, it is expected that each dose will comprise l-1000 ⁇ g of protein, preferably 2- 1 OO ⁇ g, most preferably 4-40 ⁇ g. An optimal amount for a particular vaccine can be ascertained by standard studies involving observation of antibody titres and other responses in subjects. Following an initial vaccination, subjects may receive a boost in about 4 weeks.
  • Isolated means altered “by the hand of man” from the natural state. If an "isolated” composition or substance occurs in nature, it has been changed or removed from its original environment, or both.
  • a polynucleotide or a polypeptide naturally present in a living animal is not “isolated,” but the same polynucleotide or polypeptide separated from the coexisting materials of its natural state is “isolated”, as the term is employed herein.
  • Polynucleotide generally refers to any polyribonucleotide or polydeoxribonucleotide, which may be unmodified RNA or DNA or modified RNA or DNA including single and double stranded regions.
  • Variant refers to a polynucleotide or polypeptide that differs from a reference polynucleotide or polypeptide, but retains essential properties.
  • a typical variant of a polynucleotide differs in nucleotide sequence from another, reference polynucleotide. Changes in the nucleotide sequence of the variant may or may not alter the amino acid sequence of a polypeptide encoded by the reference polynucleotide. Nucleotide changes may result in amino acid substitutions, additions, deletions, fusions and truncations in the polypeptide encoded by the reference sequence, as discussed below.
  • a typical variant of a polypeptide differs in amino acid sequence from another, reference polypeptide.
  • a variant and reference polypeptide may differ in amino acid sequence by one or more substitutions, additions, deletions in any combination.
  • a substituted or inserted amino acid residue may or may not be one encoded by the genetic code.
  • a variant of a polynucleotide or polypeptide may be a naturally occurring such as an allelic variant, or it may be a variant that is not known to occur naturally. Non-naturally occurring variants of polynucleotides and polypeptides may be made by mutagenesis techniques or by direct synthesis.
  • Identity is a relationship between two or more polypeptide sequences or two or more polynucleotide sequences, as determined by comparing the sequences.
  • identity also means the degree of sequence relatedness between polypeptide or polynucleotide sequences, as the case may be, as determined by the match between strings of such sequences.
  • Prefened methods to determine identity are designed to give the largest match between the sequences tested. Methods to determine identity and similarity are codified in publicly available computer programs. Prefened computer program methods to determine identity and similarity between two sequences include, but are not limited to, the GCG program package (Devereux, J., et al., Nucleic Acids Research 12(1): 387 (1984)), BLASTP, BLASTN, and FASTA (Atschul, S.F. et al., J. Molec. Biol.
  • the BLAST X program is publicly available from NCBI and other sources (BLAST Manual, Altschul, S., et al, NCBI NLM NEH Bethesda, MD 20894; Altschul, S., et al, J. Mol. Biol. 215: 403-410 (1990).
  • the well known Smith Waterman algorithm may also be used to determine identity.
  • the prefened algorithm used is FASTA.
  • the prefened parameters for polypeptide or polynuleotide sequence comparison using this algorithm include the following:
  • Prefened parameters for polypeptide sequence comparison with other methods include the following:
  • Gap Length Penalty 4 A program useful with these parameters is publicly available as the "gap" program from Genetics Computer Group, Madison WI. The aforementioned parameters are the default parameters for polypeptide comparisons (along with no penalty for end gaps).
  • a program useful with these parameters is publicly available as the "gap" program from Genetics Computer Group, Madison WI.
  • the aforementioned parameters are the default parameters for polynucleotide comparisons.
  • a polynucleotide sequence of the present invention may be identical to the reference sequence of SEQ TD NO: 1 or SEQ ED NO:4, that is be 100% identical, or it may include up to a certain integer number of nucleotide alterations as compared to the reference sequence.
  • Such alterations are selected from the group consisting of at least one nucleotide deletion, substitution, including transition and transversion, or insertion, and wherein said alterations may occur at the 5' or 3' terminal positions of the reference nucleotide sequence or anywhere between those terminal positions, interspersed either individually among the nucleotides in the reference sequence or in one or more contiguous groups within the reference sequence.
  • the number of nucleotide alterations is determined by multiplying the total number of nucleotides in SEQ TD NO:l or SEQ ED NO:4 by the numerical percent of the respective percent identity(divided by 100) and subtracting that product from said total number of nucleotides in SEQ TD NO:l or SEQ LD NO:4, or: ⁇ x n - (xn » y), wherein n n is the number of nucleotide alterations, x n is the total number of nucleotides in SEQ TD NO:l or SEQ ED NO:4, and y is, for instance, 0.70 for 70%, 0.80 for 80%, 0.85 for 85%, 0.90 for 90%, 0.95 for 95%,etc, and wherein any non-integer product of x n and y is rounded down to the nearest integer prior to subtracting it from x n .
  • Alterations of a polynucleotide sequence encoding the polypeptide of SEQ TD NO:2 or SEQ ED NO:3 or SEQ ED NO:8 may create nonsense, missense or frameshift mutations in this coding sequence and thereby alter the polypeptide encoded by the polynucleotide following such alterations.
  • a polypeptide sequence of the present invention may be identical to the reference sequence of SEQ ID NO:2 or SEQ ED NO:3 or SEQ ED NO:8, that is be 100% identical, or it may include up to a certain integer number of amino acid alterations as compared to the reference sequence such that the % identity is less than 100%.
  • Such alterations are selected from the group consisting of at least one amino acid deletion, substitution, including conservative and non-conservative substitution, or insertion, and wherein said alterations may occur at the amino- or carboxy-terminal positions of the reference polypeptide sequence or anywhere between those terminal positions, interspersed either individually among the amino acids in the reference sequence or in one or more contiguous groups within the reference sequence.
  • the number of amino acid alterations for a given % identity is determined by multiplying the total number of amino acids in SEQ ED NO:2 or SEQ ED NO:3 or SEQ ED NO:8 by the numerical percent of the respective percent identity(divided by 100) and then subtracting that product from said total number of amino acids in SEQ ED NO:2 or SEQ ED NO:3 or SEQ ED NO:8, or: n a ⁇ x a - (x a » y), wherein n a is the number of amino acid alterations, x a is the total number of amino acids in SEQ TD NO:2 or SEQ ED NO:3 or SEQ ED NO:8, and y is, for instance 0.70 for 70%, 0.80 for 80%, 0.85 for 85% etc., and wherein any non- integer product of x a and y is rounded down to the nearest integer prior to subtracting it from x a .
  • “Homolog” is a generic term used in the art to indicate a polynucleotide or polypeptide sequence possessing a high degree of sequence relatedness to a subject sequence. Such relatedness may be quntified by determining the degree of identity and or similarity between the sequences being compared as hereinbefore described. Falling within this generic term are the terms “ortholog”, meaning a polynucleotide or polypeptide that is the functional equivalent of a polynucleotide or polypeptide in another species and "paralog” meaning a functionally similar sequence when considered within the same species.
  • the invention will be further illustrated with reference to the following, non-limiting, examples.
  • Real-time RT-PCR (U. Gibson. 1996. Genome Research: 6,996) is used to compare mRNA transcript abundance of the candidate antigen in a panel of tumour and matched normal tissues of breast from multiple patient samples. Ln addition, mRNA levels of the candidate antigen in different normal and tumour cell lines of breast, in a panel of normal tissues, and finally in breast tumours grown in vivo in SCED mice are also evaluated by this approach. This analysis is important to establish the breast tumour specificity of CASB7439 expression, which is an important criterion a good antigen candidate must fulfil.
  • cDNA was derived from prostate, ovarian, colon and breast tumour cell lines as well as breast tumours, normal breast and other normal tissues.
  • Human tumour explants from breast cancer patients ( ⁇ lmm 3 ) are implanted subcutaneously in SCID mice.
  • Time-release hormone pellets ( ⁇ -estradiol) are co-implanted subcutaneously.
  • tumours reach -5% of the total mouse body weight they are excised and a portion ( ⁇ lmm3) is passaged into another set of SCED mice. This process can be repeated indefinitely.
  • SCED-derived tumour specimens are designated by the passage number followed by 'p'.
  • RNA is extracted from snap frozen biopsie or cell lines using TriPure reagent (Boehringer). Poly-A+ mRNA is purified from total RNA after DNAase treatment using oligo-dT magnetic beads (Dynal). Quantification of the mRNA is performed by spectrofluorimetry (VersaFluor, BioRad) using Sybrll dye (Molecular Probes).
  • Primers for real-time PCR amplification are designed with the Perkin-Elmer Primer Express software using default options for TaqMan amplification conditions.
  • Real time PCR was performed with CASB7439 specific probes and the expression levels in the tissues determined : 2 distinct TaqMan primer pairs are designed (one pair in CASB7439 3' UTR region and one pair in CASB7439 ORF region) and used in subsequent real-time RT-PCR analysis.
  • Real-time reactions are assembled according to standard PCR protocols using 2 ng of reverse transcribed mRNA (Expand RT, Roche) for each reaction. Sybrl dye (Molecular Probes) is added at a final dilution of 1/75000 for real-time detection. Amplification (40 cycles) and real-time detection is performed in a Perkin-Elmer Biosystems PE7700 system using conventional instrument settings. Ct values are calculated using the PE7700 Sequence Detector software, and results are standardised with respect to Actin.
  • Figure 1 demonstrates while almost all colon cell lines expressed CASB7439 so did the breast tumour cell lines MCF-7, MDA-MB415, BT474 and T47D. These results on a panel of cell lines suggest that, in addition to colon cancer, CASB7439 transcript is also surprisingly over-expressed in breast tumour cell lines, as compared to other normal tissues. Expression is also seen in pooled breast tumour tissue, SCED-derived breast tumour 9987 (both passages) and 9077 (late passage only), as shown in figures 2 and 3. Primers derived from the CASB7439 ORF as well as the 3' end yielded similar profiles.
  • Figure 4 confirms breast tumour specificity of CASB7439 expression on an extended panel of breast tumours and normal tissues. Taken together, these real-time RT-PCR results cleary indicates CASB7439 is over-expressed in an majority of breast tumours, as compared to normal tissues.
  • Example 2 Example 2
  • Total bacterial DNA is extracted from 100 ⁇ l over-night cultures. Bacteria are lysed with guanidium isothiocyanate and the bacterial DNA is affinity purified using magnetic glass (Boehringer). Plasmid inserts are recovered from the bacterial DNA by Advantage PCR amplification (Clontech). The PCR products are dotted onto two nylon membranes to produce high density cDNA anays using the Biomek 96 HDRT tool (Beekman). The spotted cDNA is covalently linked to the membrane by UV irradiation. The first membrane is hybridised with a mixed cDNA probe prepared from the tumour of a single patient. The second membrane is hybridised with an equivalent amount of mixed cDNA probe prepared from normal breast of the same patient.
  • the probe cDNA is prepared by PCR amplification as described above and is labelled using the AlkPhos Direct System (Amersham). Hybridisation conditions and stringency washes are as described in the AlkPhos Direct kit. Hybridized probe is detected by chemiluminescence. Hybridisation intensities for each cDNA fragment on both blots are measured by film densitometry or direct measurement (BioRad Fluor-S Max). The ratio of the tumour to normal hybridisation intensities (T/N) is calculated for each gene to evaluate the degree of over-expression in the tumour. Genes which are significantly over-expressed in breast tumours are followed-up. Significance is arbitrarily defined as one standard deviation of the T/N frequency distribution.
  • RNA from multiple patient donors >18
  • DNA anays are hybridised with mixed cDNA probes from normal tissues other than breast (see list above) to determine the level of expression of the candidate gene in these tissues.
  • DNA micro-anays are used to examine mRNA expression profiles of large collections of genes in multiple samples. This information is used to complement the data obtained by real-time PCR and provides an independent measure of gene expression levels in tumours and normal tissues.
  • Examples of cunent technologies for production of DNA micro-arrays include 1) The Affymetrix "GeneChip” anays in which oligonucleotides are synthetized on the surface of the chip by solid phase chemical synthesis using a photolithographic process 2) DNA spotting technology in which small volumes of a DNA solution are robotically deposited and then immobilized onto the surface of a solid phase (e.g. glass).
  • the chips are hybridized with cDNA or cRNA which has been extracted from the tissue of interest (e.g. normal tissue, tumour etc%) and labeled with radioactivity or with a fluorescent reporter molecule.
  • the labeled material is hybridized to the chip and the amount of probe bound to each sequence on the chip is determined using a specialized scanner.
  • the experiment can be set-up with a single fluorescent reporter (or radioactivity) or, alternatively, can be performed using two fluorescent reporters. Ln this latter case, each of the two samples is labeled with one of the reporter molecules. The two labeled samples are then hybridized competitively to the sequences on the DNA chip. The ratio of the two fluorescent signals is determined for each sequence on the chip. This ratio is used to calculate the relative abundance of the transcript in the two samples. Detailed protocols are available from a number of sources including "DNA Microanays: A practical approach. Schena M. Oxford University Press 1999" and the World Wide Web
  • Northern blots are produced according to standard protocols using 1 ⁇ g of poly A+ mRNA. Radioactive probes are prepared using the Ready-to-Go system (Pharmacia).
  • Expression in microbial hosts, or alternatively in vitro transcription/translation, is used to produce the antigen of the invention for vaccine purposes and to produce protein fragments or whole protein for rapid purification and generation of antibodies needed for characterization of the naturally expressed protein by western blot, immunohistochemistry or for follow-up of purification.
  • Recombinant proteins may be expressed in two microbial hosts, E. coli and in yeast (such as Saccharomyces cerevisiae or Pichia pastoris). This allows the selection of the expression system with the best features for this particular antigen production. In general, the recombinant antigen will be expressed in E. coli and the reagent protein expressed in yeast.
  • the expression strategy first involves the design of the primary structure of the recombinant antigen.
  • an expression fusion partner EFP
  • EFP expression fusion partner
  • an immune fusion partner EFP
  • an affinity fusion partner AFP
  • polynucleotide sequence of the coding region of the antigen can also be modified to optimise its expression. Using a codon usage that is adapted to the chosen recombinant expression host can dramatically increase antigen expression yields.
  • a comparative evaluation of the different versions of the expressed antigen will allow the selection of the most promising candidate that is to be used for further purification and immunological evaluation.
  • Construct 1 Full length wild type CASB7439 cDNA in fusion with the N terminal fragment (the first 80 amino acids) of Influenza protein NSl coding cDNA as EFP and with a histidine tail coding cDNA as an AFP (SEQ ED NO:5).
  • the encoded fusion protein sequence is SEQ ED NO:7.
  • the CASB7439 protein design is shown in Figure 5.
  • Contruct 2 Full length mutated CASB7439 cDNA in fusion with the N terminal fragment of Influenza protein NS 1 coding cDNA as EFP and with a histidine tail coding cDNA as an AFP (SEQ ED NO:6).
  • This construct was designed in order to comprise the first 50 codons of native CASB7439 cDNA replaced by codons specific of the E. coli codon usage, to enhance expression potential of CASB7439 in its E. coli host. Only CASB7439 codons that are known to be problematic in E coli host by the man skilled in the art were replaced by an E. Coli codon. For each CASB7439 problematic codon, the E coli codon is chosen to encode for the same aminoacid and to have a low GC content.
  • the encoded fusion protein sequence is SEQ ID NO:7.
  • Contruct 3 Full length wild type CASB7439 cDNA in fusion with a ll amino acids T7*Tag ® (Novagen, Madison, WI) coding cDNA as EFP and with a histidine tail coding cDNA as an AFP (SEQ ED NO: 13).
  • the encoded fusion protein is SEQ ED NO: 14.
  • Construct 4 Full length mutated CASB7439 cDNA in fusion with a 11 amino acids T7»Tag ® (Novagen, Madison, WI) coding cDNA as EFP and with a histidine tail coding cDNA as an AFP (SEQ ID NO:34).
  • the first 50 codons of native CASB7439 cDNA replaced by codons specific of the E. coli codon usage. The same substitutions as those performed in construct 2 have been carried out in contract 4.
  • the encoded fusion protein sequence is SEQ ED NO: 14.
  • Construct 5 Full length mutated CASB7439 cDNA is fusion with a l l amino acids T7»Tag ® (Novagen, Madison, WTjcoding cDNA as EFP and a histine tail coding cDNA as AFP (SEQ ED NO: 15). This construct also has the first 50 codons of native CASB7439 cDNA replaced by codons specific of the E. coli codon usage. All CASB7439 codons are modified in this construct to have specific E Coli codons and to respect E. Coli codon frequency distribution.
  • the encoded fusion protein sequence is SEQ TD NO: 14.
  • Plamid pMGl is a derivative of pMG27N.
  • Plasmid pMG27N is a pBR322-derived expression vector which contains the P L promoter, an N utilization site (to relieve transcriptional polarity in the presence of N protein) and the ell ribisome binding site including the ell translation initiation codon incorporated in an Nde I restriction site (Gross et al. 1985, Mol.&Cell.Biol. 5:1015).
  • Plasmid pMGl has been constructed by inserting the 81 first codons of the NSl coding region from influenza strain A/PR/8/34 cleaved from plasmid pASl ⁇ H/801 (Young et al. 1983. Proc.Natl.Acad.Sci.USA 80:6105) by BamH I and Nco I into pMG27N digested by BamH I and Sac I. A synthetic DNA linker was introduced between the Nco I and the Sac I sites.
  • a schematic represention of pMGl construction is shown in Figure 6. Construct 3 to 5 are cloned in a commercial pET24b(+) plamid from Novagen (Madison, WI).
  • Constructs 1 to 5 are transformed in the AR58 E. coli strain, which is a cryptic ⁇ lysogen derived from N99 that is gal E::Tn 10,A-8(chlD-pgl),A-Hl(cro-chlA),N i' , and cI857 (Proc.Natl.Acad.Sci.USA vol82, pp.88-92, January 1985 Biochemistry).
  • the recombinant product is characterized by the evaluation of the level of expression and the prediction of further solubility of the protein by analysis of the behavior in the crude extract. After growth on appropriate culture medium and induction of the recombinant protein expression, total extracts are analyzed by SDS-PAGE. The recombinant proteins are visualised in stained gels and identified by Western blot analysis using specific antibodies.
  • the cell extract was prepared as follows: cells were resuspended in PBS buffer, disrupted (French press, 3 times), and centrifuged 30 min at 14000t. More than 90% of the protein was found in the supernatant of the cell extract.
  • the purification scheme follows a classical approach based on the presence of an His affinity tail in the recombinant protein.
  • the disrupted cells are filtered and the acellular extracts loaded onto an Ion Metal Affinity Chromatography (EMAC; Ni 'H TA from Qiagen) that will specifically retain the recombinant protein.
  • EMAC Ion Metal Affinity Chromatography
  • the retained proteins are eluted by 0-500 mM imidazole gradient (possibly in presence of a detergent) in a phosphate buffer.
  • the supernatant from the harvested culture was denatured in 6M urea,100mM NaH 2 PO 4 , lOmM Tris, pH 8, and loaded on a chromatographic column EMAC Qiagen NTA Ni "1-1" under the following conditions :
  • the eluted protein in 500mM imidazole + 6M urea is dialysed under the following conditions :
  • the unpurified cell extracts is run on a 12.5% SDS PAGE, and subsequently stained with Coomassie blue.
  • a Western blot is also performed using a monoclonal antibody directed against the 80 aminoacids N terminal fragment of the NSl protein in fusion with CASB7439 construct 1.
  • the resulting gels (figures 7 and 8) show that the protein is expressed and visible in the cell extract supernatant.
  • the final purified material is freezed and stored.
  • the protein content was quantified using a Lowry protein assay (0,9 mg/1.2 ml).
  • the purity was assessed by a 12.5% PAGE SDS stained with Coomassie blue (figure 9), and the presence of the recombinant protein was checked by Western blot, using a monoclonal antibody directed against the 80 aminoacids N terminal fragment of the NSl protein in fusion with CASB7439 construct 1 (figure 10).
  • the immunogenicity of the antigen of the present invention has been verified by immunising rabbits and mice using various means of immunisation. Indeed, immunisation with CASB7439 forms, either peptide or recombinant protein could induce humoral immune response with the generation of specific antibodies against CASB7439 and/or could induce a CASB7439 specific cellular immune response. Additionally, in vivo delivery of CASB7439 protein using for instance, naked DNA in an appropriate vector encoding CASB7439 or fragments of CASB7439, CASB7439 gene delivered by a viral vector encoding CASB7439 or fragments of CASB7439, is equally appropriate to demonstrate CASB7439 immunogenicity.
  • plasmid expression vector for CASB7439 was constructed through the recombination of ⁇ DOR207/CASB7439 with the destination vector pJB 16 (pVRl012-Dest) by the LR reaction of the Gateway Cloning Technology.
  • HEK293T cells were transfected in 6-well plate with pJB76 DNA (0.25 or 1 ⁇ g), or empty vector (pJB16; lug).
  • Recombinant adenoviruses are effective vectors for gene-based vaccination because they are capable of eliciting humoral and cellular immune responses against the encoded antigen. Recombining CASB7439 by such an approach is recommended for further immunological validations. Recombinant adenovirus for CASB7439 (AdC7439) was constructed using a Lac/GW-1 expression cassette under control of the CMV promoter.
  • FIG. 13 A schematic representation of the recombinant expression vector is shown in Figure 13.
  • 2 x 10 5 human fibroblasts from donor D93 were infected by the recombinant adenoviras for CASB7439 (AdC7439) in 6-well plate at various MOL Cells were harvested at 48 hours post-infection and cell lysates were analyzed by Western blot for expression of CASB7439 using rabbit anti-NSl- CASB7439-His polyclonal antibody (1:500).
  • a band at the size expected for CASB7439 is observed at low level in fibroblasts infected at an MOI of 200 and 300 and at high level at MOI of 400 or 500. No expression is seen in uninfected fibroblasts.
  • Small amounts of relatively purified protein can be used to generate immunological tools in order to a) detect the expression by immunohistochemistry in normal or cancer tissue sections; b) detect the expression, and to follow the protein during the purification process (ELISA/ Western Blot); or c) characterise/ quantify the purified protein (ELISA).
  • 96 well microplates (maxisorb Nunc) are coated with 5 ⁇ g of protein overnight at 4°C. After lhour saturation at 37°C with PBS NCS 1%, serial dilution of the rabbit sera is added for 1H 30 at 37°C (starting at 1/10). After 3 washings in PBS Tween, anti rabbit biotinylated anti serum (Amersham ) is added (1/5000). Plates are washed and peroxydase coupled streptavidin (1/5000) is added for 30 min at 37°C. After washing, 50 ⁇ l TMB (BioRad) is added for 7 min and the reaction then stopped with H2SO4 0.2M. The OD can be measured at 450 nm and midpoint dilutions calculated by SoftmaxPro.
  • Rabbits are immunised with two synthetic peptides designed from CASB7439 aminoacid sequence SEQ ED NO:2. Chosen peptide are CASB7439 aminoacid positions 1 to 14 (SB595) and 157 to 172 (SB600). Synthetic peptides are then conjugated to a canier protein (KLH). Conjugates are formulated with Freund's adjuvant, and two rabbits are immunised with each of the conjugates. Four weeks after the second immunisation and four weeks after the third immunisation, a blood sample is taken and analysed.
  • KLH canier protein
  • Anti-CASB7439 antibody titers are estimated in the serum by ELISA following standard protocols as described above. 6.2.3 Monoclonal antibodies: Immunisation
  • mice are immunized 3 times at 3 week intervals with 5 ⁇ g of purified protein. Bleedings are performed 14 days post II and 1 week post 3. The sera are tested by Elisa on purified protein used as coated antigen. Based on these results (midpoint dilution >10000) one mouse is selected for fusion.
  • Fusion/ HATselection Spleen cells are fused with the SP2/0 myeloma according to a standard protocol using PEG 40% and DMSO 5%. Cells are then seeded in 96 well plates 2.5 xlO 4 - 10 5 cells/well and resistant clones will be selected in HAT medium. The supernatant of these hybridomas will be tested for their content in specific antibodies and when positive, will be submitted to 2 cycles of limited dilution. After 2 rounds of screening, 3 hybridomas will be chosen for ascitis production.
  • immuno staining is performed on normal or cancer tissue sections, in order to determine : 0 the level of expression of the antigen of the invention in cancer relative to normal tissue or
  • tissue sample is mounted on a cork disk in OCT compound and rapidly frozen in isopentane previously super cooled in liquid nitrogen (- 160°C). The block will then be conserved at -70°C until use. 7- 1 O ⁇ m sections will be realised in a cryostat chamber (-20, -30°C).
  • Tissue sections are dried for 5 min at room Temperature (RT), fixed in acetone for lOmin at RT, dried again, and saturated with PBS 0.5% BSA 5% serum. After 30 min at RT either a direct or indirect staining is performed using antigen specific antibodies. A direct staining leads to a better specificity but a less intense staining whilst an indirect staining leads to a more intense but less specific staining.
  • the immunological relevance of the antigen of the invention can be assessed by in vitro priming of human T cells. All T cell lymphocyte lines and dendritic cells are derived from PBMCs (peripheral blood mononuclear cells) of healthy donors (prefened HLA-A2 subtype). An HLA-A2.1/Kb transgenic mouse model is also used for screening of HLA-A2.1 peptides.
  • Newly discovered antigen-specific CD8 + T cell lines are raised and maintained by weekly in vitro stimulation.
  • the lytic activity and the ⁇ -EFN production of the CD8 + lines in response to the antigen or antigen derived-peptides is tested using standard procedures.
  • transgenic mice are immunized with adjuvanted HLA-A2 peptides, those unable to induce a CD8 + response (as defined by an efficient lysis of peptide- pulsed autologous spleen cells) will be further analyzed in the human system.
  • Human dendritic cells (cultured according to Romani et al.) will be pulsed with peptides and used to stimulate CD8 + -sorted T cells (by Facs).
  • the CD8 + lines will be first tested on peptide-pulsed autologous BLCL (EBV-B transformed cell lines). To verify the proper in vivo processing of the peptide, the CD8 + lines will be tested on cDNA-transfected tumour cells (HLA-A2 transfected LnCaP, Skov3 or CAMA tumour cells).
  • CD8 + T cell lines will be primed and stimulated with either gene-gun transfected dendritic cells, retrovirally transduced B7.1 -transfected fibroblasts, recombinant pox virus or adenoviras infected dendritic cells.
  • Virus infected cells are very efficient to present antigenic peptides since the antigen is expressed at high level but can only be used once to avoid the over-growth of viral T cells lines.
  • CD8 + lines are tested on cDNA-transfected tumour cells as indicated above. Peptide specificity and identity is determined to confirm the immunological validation.
  • CD4 + T-cell immune response can also be assessed.
  • Generation of specific CD4 + T-cells is made using dendritic cells loaded with recombinant purified protein or peptides to stimulate the T-cells.
  • HLA Class I binding peptide sequences are predicted either by the Parker's algorithm (Parker, K. C, M. A. Bednarek, and J. E. Coligan. 1994. Scheme for ranking potential HLA-A2 binding peptides based on independent binding of individual peptide side-chains. J. Immunol. 152:163 and http:/ ⁇ imas.dcrt.r-ih.gov/molbio/hla_bind/) or the Rammensee method (Rammensee, Friede, Stevanovic, MHC ligands and peptide motifs: 1st listing, Immunogenetics 41, 178-228, 1995 ; Rammensee, Bachmann, Stevanovic: MHC ligands and peptide motifs.
  • HLA-A2.1/Kb transgenic mice model (Nitiello et al.).
  • the HLA Class II binding peptide sequences are predicted using the Tepitope algorithm, with a score cut-offset to 6 (Sturniolo, Hammer at al., Nature Biotechnology. 1999. 17;555-561).
  • HLA-DRB1*1106 nonamers

Abstract

The use of CASB7439 polypeptides and polynucleotides, and variants, immunogenic fragments and fusions thereof, and of immunogenic compositions thereof, in the prophylaxis, treatment and diagnosis of breast tumours is disclosed. Methods of diagnosing the presence of breast cancer or a susceptility to breast cancer in a subject related to the expression or activity of a CASB7439 polynucleotide or polypeptide are also disclosed.

Description

Novel Use
The present invention relates methods for the utilisation of polynucleotides, herein referred to as CASB7439 polynucleotides, and of polypeptides encoded thereby (referred to herein as CASB7439 polypeptides) for the prophylaxis and treatment of breast cancer. In a further aspect, the invention relates to diagnostic assays for detecting breast cancer associated with inappropriate CASB7439 polypeptide activity or levels and for detecting breast tumour cells expressing CASB7439 polypeptide.
Polypeptides and polynucleotides disclosed herein are believed to be important immunogens for specific prophylactic or therapeutic immunisation against breast tumours, because they are specifically expressed or highly over-expressed in breast tumours compared to normal cells and can thus be targeted by antigen-specific immune mechanisms leading to the destruction of the tumour cell. They can also be used to diagnose the occurrence of tumour cells. Furthermore, their inappropriate expression in certain circumstances can cause an induction of autoimmune, inappropriate immune responses, which could be corrected through appropriate vaccination using the same polypeptides or polynucleotides. hi this respect the most important biological activities to our purpose are the antigenic and immunogenic activities of the polypeptide disclosed herein. A polypeptide for use according to the present invention may also exhibit at least one other biological activity of a CASB7439 polypeptide, which could qualify it as a target for therapeutic or prophylactic intervention different from that linked to the immune response.
In a first aspect, the present invention relates to the use of CASB7439 polypeptides in the prophylaxis, treatment or diagnosis of breast cancer. Such peptides include isolated polypeptides comprising an amino acid sequence which has at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95% identity, most preferably at least 97-99% identity, to that of SEQ ID NO:2, SEQ ID NO:3 or SEQ ID NO:8 over the entire length of SEQ ID NO:2, SEQ ID NO:3 or SEQ ID NO 8. Such polypeptides include those comprising the amino acid of SEQ LD NO:2, SEQ ID NO:3 or SEQ ID NO: 8. A most preferred polypeptide for use according to the present invention is that of SEQ LD NO:2.
Preferably the aforementioned polypeptides are recombinantly produced. Most preferably the polypeptides according to the invention are purified, and are substantially free of any other proteins or contaminating host-originating material.
Other peptides that find utility in the present invention include isolated polypeptides encoded by a polynucleotide comprising the sequence contained in SEQ ID NO: 1 or SEQ ID NO:4.
The invention also contemplates the use of an immunogenic fragment of a CASB7439 polypeptide, that is a contiguous portion of the CASB7439 polypeptide which has the same or substantially similar immunogenic properties to the polypeptide comprising the amino acid sequence of SEQ ID NO:2 or SEQ ID NO:3 or SEQ ID NO:8. That is to say, the fragment (if necessary when coupled to a carrier) is capable of raising an immune response which recognises the CASB7439 polypeptide. Such an immunogenic fragment may include, for example, the CASB7439 polypeptide lacking an N-terminal leader sequence, a transmembrane domain or a C-terminal anchor domain. In a preferred aspect the immunogenic fragment of CASB7439 according to the invention comprises substantially all of the extracellular domain of a polypeptide which has at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95% identity, most preferably at least 97-99% identity, to that of SEQ ID NO:2 or SEQ ID NO:3 or SEQ ID NO:8 over the entire length of SEQ ID NO:2 or SEQ ID NO: 3 or SEQ ID NO:8. Preferably an immunogenic fragment according to the invention comprises at least one epitope.
Peptide fragments incorporating an epitope of CASB7439 typically will comprise at least 7, preferably 9 or 10 contiguous amino acids from SEQ LO NO:2 or SEQ ID NO:3 or SEQ ID NO:8. Preferred epitopes are shown in SEQ LD NO: 16 to SEQ LO NO:33. Peptides that incorporate these epitopes are preferably contemplated in the present invention. Mimotopes which have the same characteristics as these epitopes, and immunogens comprising such mimotopes which generate an immune response which cross-react with an epitope in the context of the CASB7439 molecule, are also used according to the present invention.
The present invention, therefore, includes the use of isolated peptides encompassing these epitopes themselves, and any mimotope thereof in the treatment of breast cancer. The meaning of mimotope is defined as an entity which is sufficiently similar to the native CASB7439 epitope so as to be capable of being recognised by antibodies which recognise the native molecule; (Gheysen, H.M., et al., 1986, Synthetic peptides as antigens. Wiley, Chichester, Ciba foundation symposium 119, pl30-149; Gheysen, H.M., 1986, Molecular
Immunology, 23,7, 709-715); or are capable of raising antibodies, when coupled to a suitable carrier, which antibodies cross-react with the native molecule.
Peptide mimotopes of the above-identified epitopes may be designed for a particular purpose by addition, deletion or substitution of elected amino acids. Thus, the peptides according to the present invention may be modified for the purposes of ease of conjugation to a carrier, preferably a protein carrier. For example, it may be desirable for some chemical conjugation methods to include a terminal cysteine to the epitope. In addition it may be desirable for peptides conjugated to a protein carrier to include a hydrophobic terminus distal from the conjugated terminus of the peptide, such that the free unconjugated end of the peptide remains associated with the surface of the carrier protein. This reduces the conformational degrees of freedom of the peptide, and thus increases the probability that the peptide is presented in a conformation that most closely resembles that of the peptide as found in the context of the whole molecule. For example, the peptides may be altered to have an N-terminal cysteine and a C- terminal hydrophobic amidated tail. Alternatively, the addition or substitution of a D-stereoisomer form of one or more of the amino acids may be performed to create a beneficial derivative, for example to enhance stability of the peptide. Those skilled in the art will realise that such modified peptides, or mimotopes, could be a wholly or partly non-peptide mimotope wherein the constituent residues are not necessarily confined to the 20 naturally occurring amino acids. In addition, these may be cyclised by techniques known in the art to constrain the peptide into a conformation that closely resembles its shape when the peptide sequence is in the context of the whole molecule. A preferred method of cyclising a peptide comprises the addition of a pair of cysteine residues to allow the formation of a disulphide bridge.
Further, those skilled in the art will realise that mimotopes or immunogens for use in the present invention may be larger than the above-identified epitopes, and as such may comprise the sequences disclosed herein. Accordingly, the mimotopes disclosed in the present invention may consist of addition of N and/or C terminal extensions of a number of other natural residues at one or both ends. The peptide mimotopes may also be retro sequences of the natural sequences, in that the sequence orientation is reversed; or alternatively the sequences maybe entirely or at least in part comprised of D-stereo isomer amino acids (inverso sequences). Also, the peptide sequences may be retro-inverso in character, in that the sequence orientation is reversed and the amino acids are of the D-stereoisomer form. Such retro or retro-inverso peptides have the advantage of being non-self, and as such may overcome problems of self-tolerance in the immune system.
Alternatively, peptide mimotopes may be identified using antibodies which are capable themselves of binding to the epitopes of the present invention using techniques such as phage display technology (EP 0 552267 Bl). This technique, generates a large number of peptide sequences which mimic the structure of the native peptides and are, therefore, capable of binding to anti-native peptide antibodies, but may not necessarily themselves share significant sequence homology to the native peptide. This approach may have significant advantages by allowing the possibility of identifying a peptide with enhanced immunogenic properties, or may overcome any potential self-antigen tolerance problems which may be associated with the use of the native peptide sequence. Additionally this technique allows the identification of a recognition pattern for each native-peptide in terms of its shared chemical properties amongst recognised mimotope sequences.
The covalent coupling of the peptide to the immunogenic carrier can be carried out in a manner well known in the art. Thus, for example, for direct covalent coupling it is possible to utilise a carbodiimide, glutaraldehyde or (N-[γ- maleimidobutyryloxy] succinimide ester, utilising common commercially available heterobifunctional linkers such as CDAP and SPDP (using manufacturers instructions). After the coupling reaction, the immunogen can easily be isolated and purified by means of a dialysis method, a gel filtration 5. method, a fractionation method etc. Accordingly, the present invention further provides for the use of a CASB7439 polypeptide as defined hereabove, which is chemically conjugated to a carrier protein.
The types of carriers used in the immunogens of the present invention will be 0 readily known to the man skilled in the art. The function of the carrier is to provide cytokine help in order to help induce an immune response against the peptide. A non-exhaustive list of carriers which may be used in the present invention include: Keyhole limpet Haemocyanin (KLH), serum albumins such as bovine serum albumin (BSA), inactivated bacterial toxins such as tetanus or 5 diptheria toxins (TT and DT), or recombinant fragments thereof (for example, Domain 1 of Fragment C of TT, or the translocation domain of DT), or the purified protein derivative of tuberculin (PPD). Alternatively the mimotopes or epitopes may be directly conjugated to liposome carriers, which may additionally comprise immunogens capable of providing T-cell help. Preferably the ratio of 0 mimotopes to carrier is in the order of 1 : 1 to 20: 1 , and preferably each carrier should carry between 3-15 peptides.
In an embodiment of the invention a preferred carrier is Protein D from Haemophilus influenzae (EP 0 594 610 B1). Protein D is an IgD-binding protein from Haemophilus influenzae and has been patented by Forsgren (WO 91/18926, granted EP 0 594 610 Bl). In some circumstances, for example in recombinant immunogen expression systems it may be desirable to use fragments of protein D, for example Protein D l/3rd (comprising the N-terminal 100-110 amino acids of protein D (GB 9717953.5)).
Another prefened method of presenting the peptides for use in the present invention is in the context of a recombinant fusion molecule. For example, EP 0 421 635 B describes the use of chimaeric hepadnavirus core antigen particles to present foreign peptide sequences in a virus-like particle. As such, immunogens for use in the present invention may comprise peptides presented in chimaeric particles consisting of hepatitis B core antigen. Additionally, the recombinant fusion proteins may comprise the mimotopes of the present invention and a carrier protein, such as NSl of the influenza virus, or a fragment thereof such as the first 1-80 or 1-81 amino acids of the NSl protein, which is preferred. Preferred fusion proteins according to this invention are sequences set forth in SEQ LD NO: 7 and in SEQ ID NO: 14. For any recombinantly expressed protein which is used according to the present invention, the nucleic acid which encodes said immunogen can also be used in the present invention. Preferred nucleotidic sequences are given in SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO: 13, SEQ ID NO:15 and SEQ ID NO:34.
Peptides used in the present invention can be readily synthesised by solid phase procedures well known in the art. Suitable syntheses may be performed by utilising "T-boc" or "F-moc" procedures. Cyclic peptides can be synthesised by the solid phase procedure employing the well-known "F-moc" procedure and polyamide resin in the fully automated apparatus. Alternatively, those skilled in the art will know the necessary laboratory procedures to perform the process manually. Techniques and procedures for solid phase synthesis are described in 'Solid Phase Peptide Synthesis: A Practical Approach' by E. Atherton and R..C. Sheppard, published by IRL at Oxford University Press (1989). Alternatively, the peptides may be produced by recombinant methods, including expressing nucleic acid molecules encoding the mimotopes in a bacterial or mammalian cell line, followed by purification of the expressed mimotope. Techniques for recombinant expression of peptides and proteins are known in the art, and are described in Maniatis, T., Fritsch, E.F. and Sambrook et al., Molecular cloning, a laboratory manual, 2nd Ed.; Cold Spring Harbor Laboratory Press, Cold Spring Harbor, New York (1989).
The polypeptides or immunogenic fragment utilised in the invention may be in the form of the "mature" protein or may be a part of a larger protein such as a precursor or a fusion protein. It is often advantageous to include an additional amino acid sequence which contains secretory or leader sequences, pro-sequences, sequences which aid in purification such as multiple histidine residues, or an additional sequence for stability during recombinant production. Furthermore, addition of exogenous polypeptide or lipid tail or polynucleotide sequences to increase the immunogenic potential of the final molecule is also considered. Accordingly, another aspect of the invention concerns the use of genetically engineered soluble fusion proteins comprising a polypeptide of the present invention, or a fragment thereof, and various portions of the constant regions of heavy or light chains of immunoglobulins of various subclasses (IgG, IgM, IgA, IgE). Preferred as an immunoglobulin is the constant part of the heavy chain of human IgG, particularly IgGl, where fusion takes place at the hinge region. In a particular embodiment, the Fc part can be removed simply by incorporation of a cleavage sequence which can be cleaved with blood clotting factor Xa. Furthermore, this invention relates to the use of these fusion proteins for drug screening, diagnosis and therapy. Examples of fusion protein technology can be found in International Patent Application Nos. WO94/29458 and WO94/22914.
The proteins for use according to the present invention may be chemically conjugated, or expressed as recombinant fusion proteins allowing increased levels to be produced in an expression system as compared to non-fused protein. The fusion partner may assist in providing T helper epitopes (immunological fusion partner), preferably T helper epitopes recognised by humans, or assist in expressing the protein (expression enhancer) at higher yields than the native recombinant protein. Preferably the fusion partner will be both an immunological fusion partner and expression enhancing partner.
Fusion partners include protein D from Haemophilus influenza B and the non- structural protein from influenzae virus, NSl (hemagglutinin), or a fragment thereof having retained T helper epitopes. Typically the N-terminal 80 or 81 amino acids are utilised, although different fragments may be used provided they include T helper epitopes (C. Hackett, D. Horowitz, M. Wysocka & S. Dillon, 1992, J. Gen. Nirology, 73, 1339-1343). When ΝS1 is the immunological fusion partner it has the additional advantage in that it allows higher expression yields to be achieved. An example of such ΝS1-CASB7439 fusion protein is given in SEQ ID NO:7, and is encoded by the polynucleotide sequences given in SEQ ID NO:5 or in SEQ LD NO:6, being respectively the native (unmodified) cDNA sequence or codon-optimised (for E.coli expression) sequence. The polypeptide sequence of SEQ ID NO:7 and polynucleotide sequences of SEQ ID NO:5 and SEQ ID NO:6 are preferred for the use according to the present invention. Another immunological fusion partner is the protein known as LYTA. Preferably the C terminal portion of the molecule is used. Lyta is derived from Streptococcus pneumoniae which synthesise an N-acetyl-L-alanine amidase, amidase LYTA, (coded by the lytA gene {Gene, 43 (1986) page 265-272} an autolysin that specifically degrades certain bonds in the peptidoglycan backbone. The C- terminal domain of the LYTA protein is responsible for the affinity to the choline or to some choline analogues such as DEAE. This property has been exploited for the development of E.coli C-LYTA expressing plasmids useful for expression of fusion proteins. Purification of hybrid proteins containing the C-LYTA fragment at its amino terminus has been described {Biotechnology: 10, (1992) page 795- 798} . It is possible to use the repeat portion of the Lyta molecule found in the C terminal end starting at residue 178, for example residues 188 - 305.
The present invention also includes the use of xenogeneic forms (also termed ortholog forms) of the aforementioned polypeptides, said xenogeneic forms referring to an antigen having substantial sequence identity to the human antigen (also termed autologous antigen) which serves as a reference antigen but which is derived from a different non-human species. In this context the substantial identity refers to concordance of an amino acid sequence with another amino acid sequence or of a polynucleotide sequence with another polynucleotide sequence when such sequence are arranged in a best fit alignment in any of a number of sequence alignment proteins known in the art. By substantial identity is meant at least 70-95%, and preferably at least 85-95%, most preferably at least 90%-95%, sequence identity between the compared sequences. Therefore according to the invention the xenogeneic CASB7439 polypeptide will be a CASB7439 polypeptide which is xenogeneic with respect to human CASB7439, in other words which is isolated from a species other than human. In a preferred embodiment, the polypeptide is isolated from mouse, rat, pig, or rhesus monkey, most preferably from mouse or rat. Accordingly the present invention also provides a method of inducing an immune response against human CASB7439 having an amino acid sequence as set forth in SEQ ID NO:9 and SEQ ID NO:10 in a human, comprising administering to the subject an effective dosage of a composition comprising a xenogeneic form of said human CASB7439 as described herein. A preferred embodiment is a method of inducing an immune response against human CASB7439 using the xenogeneic CASB7439 isolated from mouse, rat, pig or rhesus monkey. Another preferred method of inducing an immune response according to the present invention is using an antigen composition including a live viral expression system which expresses said xenogeneic antigen.
The isolated xenogeneic CASB7439 polypeptide will generally share substantial sequence similarity, and include isolated polypeptides comprising an amino acid sequence which has at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95% identity, most preferably at least 97-99% identity, to that of SEQ ID NO:9 and SEQ ID NO: 10 over the entire length of SEQ ID NO:9 and SEQ ID NO: 10, respectively.
Accordingly the xenogeneic polypeptide will comprise an immunogenic fragment of the polypeptide of SEQ ID NO:9 and SEQ ID NO:10 in which the immunogenic activity of the immunogenic fragment is substantially the same as the polypeptide of SEQ ID NO:9 and SEQ ID NO: 10. In addition the xenogeneic CASB7439 polypeptide can be a fragment of at least about 20 consecutive amino acids, preferably about 30, more preferably about 50, yet more preferably about 100, most preferably about 150 contiguous amino acids selected from the amino acid sequences as shown in SEQ ID NO:9 and SEQ LD NO: 10. More particularly xenogeneic CASB7439 fragments will retain some functional property, preferably an immunological activity, of the larger molecule set forth in SEQ ID NO:9 and SEQ LD NO: 10, and are useful in the methods described herein (e.g. in pharmaceutical and vaccine compositions, in diagnostics, etc.). In particular the fragments will be able to generate an immune response against the human counterpart, such as the generation of cross-reactive antibodies which react with the autologous human form of CASB7439 as set forth in SEQ LD NO:2. In a specific embodiment, the xenogeneic polypeptide of the invention may be part of a larger fusion, comprising the xenogeneic CASB7439 polypeptide or fragment thereof and a heterologous protein or part of a protein acting as a fusion partner as described hereabove. The present invention also contemplates the use of variants of the aforementioned polypeptides, that is polypeptides that vary from the referents by conservative amino acid substitutions, whereby a residue is substituted by another with like characteristics. Typical such substitutions are among Ala, Nal, Leu and De; among Ser and Thr; among the acidic residues Asp and Glu; among Asn and Gin; and among the basic residues Lys and Arg; or aromatic residues Phe and Tyr. Particularly preferred are variants in which several, 5-10, 1-5, 1-3, 1-2 or 1 amino acids are substituted, deleted, or added in any combination.
Polypeptides for use in the present invention can be prepared in any suitable manner. Such polypeptides include isolated naturally occurring polypeptides, recombinantly produced polypeptides, synthetically produced polypeptides, or polypeptides produced by a combination of these methods. Means for preparing such polypeptides are well understood in the art.
In a further aspect, the present invention relates to the use of CASB7439 polynucleotides in the treatment or diagnosis of breast cancer. Such polynucleotides include isolated polynucleotides comprising a nucleotide sequence encoding a polypeptide which has at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95% identity, to the amino acid sequence of SEQ ID ΝO:2 or SEQ ID NO:3 or SEQ ID NO:8, over the entire length of SEQ ID NO:2 or SEQ ID NO:3 or SEQ ID NO:8. In this regard, polypeptides which have at least 97% identity are highly preferred, whilst those with at least 98-99% identity are more highly preferred, and those with at least 99% identity are most highly preferred. Such polynucleotides include SEQ ID NO: 1 , or include a polynucleotide comprising the nucleotide sequence contained in SEQ ID NO:l encoding the polypeptide of SEQ ID NO:2 or comprising the nucleotide contained in SEQ ID NO:4 encoding the polypeptide of SEQ ID NO:3 or SEQ ID NO:8.
Further polynucleotides that find utility in the present invention include isolated polynucleotides comprising a nucleotide sequence that has at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95% identity, to a nucleotide sequence encoding a polypeptide of SEQ ID NO:2 or SEQ ID NO:3 or SEQ ID NO:8, over the entire coding region. In this regard, polynucleotides which have at least 97% identity are highly preferred, whilst those with at least 98-99% identity are more highly preferred, and those with at least 99% identity are most highly preferred.
Other polynucleotides include isolated polynucleotides comprising a nucleotide sequence which has at least 70% identity, preferably at least 80% identity, more preferably at least 90% identity, yet more preferably at least 95% identity, to SEQ ID NO: 1 or SEQ ID NO:4 or to the coding sequence of SEQ ID NO: 1 or SEQ ID NO:4 over the entire length of SEQ ID NO: 1 or SEQ ID NO:4 or over the entire length of the coding sequence of SEQ TD NO:l or SEQ ED NO:4 respectively. In this regard, polynucleotides which have at least 97% identity are highly preferred, whilst those with at least 98-99% identity are more highly preferred, and those with at least 99% identity are most highly preferred. Such polynucleotides include a polynucleotide comprising the polynucleotide of SEQ ID NO:l or SEQ ID NO:4 as well as the polynucleotide of SEQ ID NO:l or SEQ ID NO:4 or the coding region of SEQ LD NO: 1 or SEQ ID NO:4. Said polynucleotide can be inserted in a suitable plasmid or recombinant microrganism vector and used for immunisation ( see for example Wolff et. al, Science 247:1465-1468 (1990); Corr et. al., J. Exp. Med. 184:1555-1560 (1996); Doe et. al., Proc. Natl. Acad. Sci. 93:8578-8583 (1996)).
The present invention also provides a nucleic acid encoding the aforementioned xenogeneic proteins of the present invention and their use in medicine. In a preferred embodiment, the xenogeneic CASB7439 polynucleotide for use in pharmaceutical compositions has the sequence set forth in SEQ ID NO: 11 and SEQ ID NO: 12. The isolated xenogeneic CASB7439 polynucleotides according to the invention may be single-stranded (coding or antisense) or double-stranded, and maybe DNA (genomic, cDNA or synthetic) or RNA molecules. Additional coding or non-coding sequences may, but need not, be present within a polynucleotide of the present invention. In other related embodiments, the present invention provides polynucleotide variants having substantial identity to the sequences disclosed herein in SEQ LD NO:l 1 and SEQ ID NO: 12 for example those comprising at least 70% sequence identity, preferably at least 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% or higher, sequence identity compared to a polynucleotide sequence of this invention using the methods described herein, (e.g., BLAST analysis using standard parameters). In a related embodiment, the isolated xenogeneic polynucleotide of the invention will comprise a nucleotide sequence encoding a polypeptide that has at least 90%, preferably 95% and above, identity to the amino acid sequence of SEQ TD NO:l 1 and SEQ ED NO: 12, over the entire length of SEQ ED NO: 11 and SEQ LD NO: 12, respectively; or a nucleotide sequence complementary to said isolated polynucleotide.
The invention also contemplates the use of polynucleotides which are complementary to all the above described polynucleotides.
The invention also provides for the use of a fragment of a CASB7439 polynucleotide which when administered to a subject has the same immunogenic properties as the polynucleotide of SEQ LD NO:l or SEQ ID NO:4.
The invention also provides for the use of a polynucleotide encoding an immunological fragment of a CASB7439 polypeptide as hereinbefore defined. Also contemplated are the use of such fragments that have a level of immunogenic activity of at least about 50%, preferably at least about 70% and more preferably at least about 90% of the level of immunogenic activity of a polypeptide sequence encoded by a polynucleotide sequence set forth in SEQ LD NO: 1 or SEQ ED NO:4.
Polypeptide fragments for use according to the invention preferably comprise at least about 5, 10, 15, 20, 25, 50, or 100 contiguous amino acids, or more, including all intermediate lengths, of a polypeptide composition set forth herein, such as those set forth in SEQ ED NO:2 or SEQ ID NO:3 or SEQ ED NO:8, or those encoded by a polynucleotide sequence set forth in a sequence of SEQ ID NO:l or SEQ ED NO:4.
The nucleotide sequence of SEQ ID NO:l is a cDNA sequence which comprises a polypeptide encoding sequence (nucleotide 545 to 1126) encoding a polypeptide of 193 amino acids, the polypeptide of SEQ ED NO:2. The nucleotide sequence encoding the polypeptide of SEQ ED NO:2 may be identical to the polypeptide encoding sequence contained in SEQ ED NO: 1 or it may be a sequence other than the one contained in SEQ ED NO: 1, which, as a result of the redundancy (degeneracy) of the genetic code, also encodes the polypeptide of SEQ ED NO:2. The polypeptide of the SEQ ED NO:2 is structurally related to other proteins of the achaete scute family, and is also named "human Achaete Scute homologue 2" (HASH2) (accession number NP_005161 and AAB86993).
Human Achaete Scute homologue 2 (HASH2) gene, officially designated human ASCL2 (Achaete Scute complex like 2) is a homologue of the Drosophila Achaete and Scute genes. Human ASCL2 is expressed in the extravillus trophoblasts of the developing placenta only, and maps on chromosome 1 lpl5 close to IGF2 and H19.
The mouse achaete-scute homolog-2 gene (MASH2) encodes a transcription factor playing a role in the development of the trophoblast. The Mash2 gene is paternally imprinted in the mouse, and the lack of human ASCL2 expression in non-malignant hydatidiform (androgenetic) moles indicates that human Ascl2 is also imprinted in man.
Ascl2 genes are members of the basic helix-loop-helix (BHLH) family of transcription factors. They activate transcription by binding to the E box (5 - CANNTG-3'). Dimerization with other BHLH proteins is required for efficient DNA binding. They are involved in the determination of the neuronal precursors in the peripheral nervous system and the central nervous system in drosophila melanogaster, and probably in mammals as well.
The complementary strand of the nucleotide sequence of SEQ ED NO:l is the polynucleotide sequence of SEQ ED NO:4. This strand also comprises two other polypeptide encoding sequences. He first polypeptide encoding sequence (nucleotide 1184 to 399 of SEQ ED:1, nucleotide 608 to 1393 of SEQ LD NO:4) encodes a polypeptide of 262 amino acids, the polypeptide of SEQ ED NO:3. The second polypeptide encoding sequence (nucleotide 840 to 262 of SEQ LD NO:l, nucleotide 952 to 1530 of SEQ ED NO:4) encodes a polypeptide of 193 amino acids, the polypeptide of SEQ ED NO:8. The nucleotide sequence encoding the polypeptides of SEQ ID NO:3 and 8 may be identical to the polypeptides encoding sequence contained in SEQ ED NO: 1 or 4 or it may be a sequence other than the one contained in SEQ ID NO: 1 or 4, which, as a result of the redundancy (degeneracy) of the genetic code, also encodes the polypeptides of SEQ ID NO:3 and 8. The polypeptide of the SEQ ED NO:3 is structurally related to other proteins of the splicing coactivator protein family, having homology and/or structural similarity with homo sapiens splicing coactivator subunit srm300 (genbank accession AAF21439). The polypeptide of SEQ ID NO:8 is not related to any known protein.
Preferred polypeptides and polynucleotides for use in the present invention are expected to have, ter alia, similar biological functions/properties to their homologous polypeptides and polynucleotides. Furthermore, preferred polypeptides, immunological fragments and polynucleotides of the present invention have at least one activity of either SEQ ED NO: 1, SEQ ED NO:2, SEQ ED NO:3, SEQ ED NO:4 or SEQ ED NO:8 as appropriate.
Polynucleotides for use in the invention may be obtained, using standard cloning and screening techniques, from a cDNA library derived from mRNA in cells of human tumour tissue (breast for example), (for example Sambrook et al., Molecular Cloning: A Laboratory Manual, 2nd Ed., Cold Spring harbor Laboratory Press, Cold Spring harbor, N.Y. (1989)). Polynucleotides of the invention can also be obtained from natural sources such as genomic DNA libraries or can be synthesised using well known and commercially available techniques.
When polynucleotides of the present invention are used for the recombinant production of polypeptides, the polynucleotide may include the coding sequence for the mature polypeptide, by itself; or the coding sequence for the mature polypeptide in reading frame with other coding sequences, such as those encoding a leader or secretory sequence, a pre-, or pro- or prepro- protein sequence, or other fusion peptide portions. For example, a marker sequence which facilitates purification of the fused polypeptide can be encoded. In certain preferred embodiments of this aspect of the invention, the marker sequence is a hexa-histidine peptide, as provided in the pQE vector (Qiagen, hie.) and described in Gentz et al, Proc Natl Acad Sci USA (1989) 86:821-824, or is an HA tag. The polynucleotide may also contain non-coding 5' and 3' sequences, such as transcribed, non-translated sequences, splicing and polyadenylation signals, ribosome binding sites and sequences that stabilise mRNA.
Further embodiments of the present invention include the use of polynucleotides encoding polypeptide variants which comprise the amino acid sequence of SEQ ID NO:2, SEQ ED NO: 3 or SEQ LD NO:8 and in which several, for instance from 5 to 10, 1 to 5, 1 to 3, 1 to 2 or 1, amino acid residues are substituted, deleted or added, in any combination.
Polynucleotides which are identical or sufficiently identical to a nucleotide sequence contained in SEQ LD NO: 1 or SEQ ID NO:4, may be used as hybridisation probes for cDNA and genomic DNA or as primers for a nucleic acid amplification (PCR) reaction, to isolate full-length cDNAs and genomic clones encoding polypeptides according to the present invention and to isolate cDNA and genomic clones of other genes (including genes encoding paralogs from human sources and orthologs and paralogs from species other than human) that have a high sequence similarity to SEQ ED NO: 1 or SEQ LD NO:4. Typically these nucleotide sequences are 70% identical, preferably 80% identical, more preferably 90% identical, most preferably 95% identical to that of the referent. The probes or primers will generally comprise at least 15 nucleotides, preferably, at least 30 nucleotides and may have at least 50 nucleotides. Particularly preferred probes will have between 30 and 50 nucleotides. Particularly preferred primers wiU have between 20 and 25 nucleotides. In particular, polypeptides or polynucleotides derived from sequences from homologous animal origin could be used as immunogens to obtain a cross-reactive immune response to the human gene.
A polynucleotide encoding a polypeptide for use in of the present invention, including homologs from species other than human, may be obtained by a process which comprises the steps of screening an appropriate library under stringent hybridisation conditions with a labelled probe having the sequence of SEQ ED NO: 1 or SEQ ED NO:4 or a fragment thereof; and isolating full-length cDNA and genomic clones containing said polynucleotide sequence. Such hybridisation techniques are well known to the skilled artisan. Preferred stringent hybridisation conditions include overnight incubation at 42°C in a solution comprising: 50% formamide, 5xSSC (150mM NaCI, 15mM trisodium citrate), 50 mM sodium phosphate (pH7.6), 5x Denhardt's solution, 10 % dextran sulfate, and 20 microgram/ml denatured, sheared salmon sperm DNA; followed by washing the filters in O.lx SSC at about 65°C. Thus the present invention also includes the use of polynucleotides obtainable by screening an appropriate library under stringent hybridisation conditions with a labelled probe having the sequence of SEQ ED NO:l or SEQ ED NO:4 or a fragment thereof.
The skilled artisan will appreciate that, in many cases, an isolated cDNA sequence will be incomplete, in that the region coding for the polypeptide is short at the 5' end of the cDNA.
There are several methods available and well known to those skilled in the art to obtain full-length cDNAs, or extend short cDNAs, for example those based on the method of Rapid Amplification of cDNA ends (RACE) (see, for example, Frohman et al., PNAS USA 85, 8998-9002, 1988). Recent modifications of the technique, exemplified by the Marathon™ technology (Clontech Laboratories Inc.) for example, have significantly simplified the search for longer cDNAs. In the Marathon™ technology, cDNAs have been prepared from mRNA extracted from a chosen tissue and an 'adaptor' sequence ligated onto each end. Nucleic acid amplification (PCR) is then carried out to amplify the 'missing' 5' end of the cDNA using a combination of gene specific and adaptor specific oligonucleotide primers. The PCR reaction is then repeated using 'nested' primers, that is, primers designed to anneal within the amplified product (typically an adaptor specific primer that anneals further 3' in the adaptor sequence and a gene specific primer that anneals further 5' in the known gene sequence). The products of this reaction can then be analysed by DNA sequencing and a full-length cDNA constructed either by joining the product directly to the existing cDNA to give a complete sequence, or carrying out a separate full-length PCR using the new sequence information for the design of the 5' primer. Recombinant polypeptides for use in the present invention may be prepared by processes well known in the art from genetically engineered host cells comprising expression systems. Accordingly, in a further aspect, the present invention relates to an expression system which comprises a polynucleotide, to host cells which are genetically engineered with such expression systems and to the production of polypeptides of the invention by recombinant techniques. Cell-free translation systems can also be employed to produce such proteins using RNAs derived from the DNA constructs of the present invention.
For recombinant production, host cells can be genetically engineered to incorporate expression systems or portions thereof for polynucleotides of the present invention. Introduction of polynucleotides into host cells can be effected by methods described in many standard laboratory manuals, such as Davis et al., Basic Methods in Molecular Biology (1986) and Sambrook et al, Molecular Cloning: A Laboratory Manual, 2nd Ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY. (1989). Preferred such methods include, for instance, calcium phosphate transfection, DEAE-dextran mediated transfection, transvection, microinjection, cationic lipid-mediated transfection, electroporation, transduction, scrape loading, ballistic introduction or infection.
For recombinant expression, the nucleic acids transfected into the host cells can be genetically engineered to comprise the optimisation of codon usage involving the replacement of at least one non-preferred or less preferred codon in a given sequence encoding a CASB7439 protein or fusion or fragment or variant thereof, by a preferred codon encoding the same amino acid. The optimisation will be carried out to fit the preferred codons or more prefened codons for a given expression host, in particular for non-mammalian cell expression such as yeast or bacterial expression. Particularly contemplated is a CASB7439 sequence optimised for E.coli expression. At least one codon, and more prefereably all the codons of the CASB7439 protein or fragment or variant thereof can be changed to fit at best the host usage, especially the E.coli usage, that is, the one (or ones) that is the most prevalent. Accordingly, in another aspect of the invention there is provided the use of a codon-optimised CASB7439 polynucleotide or polypeptide sequence. Particularly preferred optimised CASB7439 sequences are given in SEQ ID NO: 6 coding for the polypeptide sequence set out in SEQ LD NO:7, and SEQ ED NO: 15 or SEQ ED NO:34 coding for the polypeptide sequence setu out in SEQ ED NO: 14.
Vectors comprising such DNA, hosts transformed thereby and the truncated or hybrid proteins themselves, expressed as described hereinbelow all form part of the invention.
Preferably the proteins are coexpressed with thioredoxin in trans (TIT). Coexpression of thioredoxin in trans versus in cis is preferred to keep antigen free of thioredoxin without the need for protease. Thioredoxin coexpression eases the solubilisation of the proteins of the invention. Thioredoxin coexpression has also a significant impact on protein purification yield, on purified-protein solubility and quality.
Representative examples of appropriate hosts include bacterial cells, such as Streptococci, Staphylococci, E. coli, Streptomyces and Bacillus subtilis cells; fungal cells, such as yeast cells and Aspergillus ceUs; insect cells such as Drosophila S2 and Spodoptera Sf9 cells; animal cells such as CHO, COS, HeLa, C127, 3T3, BHK, HEK 293 and Bowes melanoma cells; and plant cells.
A great variety of expression systems can be used, for instance, chromosomal, episomal and virus-derived systems, e.g., vectors derived from bacterial plasmids, from bacteriophage, from transposons, from yeast episomes, from insertion elements, from yeast chromosomal elements, from viruses such as baculoviruses, papova viruses, such as SV40, vaccinia viruses, adenoviruses, fowl pox viruses, pseudorabies viruses and retroviruses, and vectors derived from combinations thereof, such as those derived from plasmid and bacteriophage genetic elements, such as cosmids and phagemids. The expression systems may contain control regions that regulate as well as engender expression. Generally, any system or vector which is able to maintain, propagate or express a polynucleotide to produce a polypeptide in a host may be used. The appropriate nucleotide sequence may be inserted into an expression system by any of a variety of well-known and routine techniques, such as, for example, those set forth in Sambrook et al, Molecular Cloning, A Laboratory Manual (supra). Appropriate secretion signals may be incorporated into the desired polypeptide to allow secretion of the translated protein into the lumen of the endoplasmic reticulum, the periplasmic space or the extracellular environment. These signals may be endogenous to the polypeptide or they may be heterologous signals.
The expression system may also be a recombinant live microorganism, such as a virus or bacterium. The gene of interest can be inserted into the genome of a live recombinant virus or bacterium. Inoculation and in vivo infection with this live vector will lead to in vivo expression of the antigen and induction of immune responses.
Therefore, in certain embodiments, polynucleotides encoding immunogenic polypeptides for use according to the present invention are introduced into suitable mammalian host cells for expression using any of a number of known viral-based systems. In one illustrative embodiment, refroviruses provide a convenient and effective platform for gene delivery systems. A selected nucleotide sequence encoding a polypeptide for use in the present invention can be inserted into a vector and packaged in retroviral particles using techniques known in the art. The recombinant virus can then be isolated and delivered to a subject. A number of illustrative retroviral systems have been described (e.g., U.S. Pat. No. 5,219,740; Miller and Rosman (1989) BioTechniques 7:980-990; Miller, A. D. (1990) Human Gene Therapy 1:5-14; Scarpa et al. (1991) Nirology 180:849-852; Burns et al. (1993) Proc. atl. Acad. Sci. USA 90:8033-8037; and Boris-Lawrie and Temin (1993) Cur. Opin. Genet. Develop. 3:102-109.
In addition, a number of illustrative adenovirus-based systems have also been described. Unlike retroviruses which integrate into the host genome, adenoviruses persist extrachromosomally thus mimmising the risks associated with insertional mutagenesis (Haj-Ahmad and Graham (1986) J. Virol. 57:267-274; Bett et al. (1993) J. Virol. 67:5911-5921; Mittereder et al. (1994) Human Gene Therapy 5:717-729; Seth et al. (1994) J. Virol. 68:933-940; Barr et al. (1994) Gene Therapy 1 :51-58; Berkner, K. L. (1988) BioTechniques 6:616-629; and Rich et al.
(1993) Human Gene Therapy 4:461 -476).
Various adeno-associated virus (AAV) vector systems have also been developed for polynucleotide delivery. AAV vectors can be readily constructed using • techniques well known in the art. See, e.g., U.S. Pat. Nos. 5,173,414 and 5,139,941; International Publication Nos. WO 92/01070 and WO 93/03769; Lebkowski et al. (1988) Molec. Cell. Biol. 8:3988-3996; Vincent et al. (1990) Vaccines 90 (Cold Spring Harbor Laboratory Press); Carter, B. J. (1992) Current Opinion in Biotechnology 3:533-539; Muzyczka, N. (1992) Current Topics in Microbiol. and Immunol. 158:97-129; Kotin, R. M. (1994) Human Gene Therapy 5:793-801; Shelling and Smith (1994) Gene Therapy 1:165-169; and Zhou et al.
(1994) J. Exp. Med. 179:1867-1875.
Additional viral vectors useful for delivering the nucleic acid molecules encoding polypeptides for use in the present invention by gene transfer include those derived from the pox family of viruses, such as vaccinia virus and avian poxvirus. By way of example, vaccinia virus recombinants expressing the molecules of interest can be constructed as follows. The DNA encoding a polypeptide is first inserted into an appropriate vector so that it is adjacent to a vaccinia promoter and flanking vaccinia DNA sequences, such as the sequence encoding thymidine kinase (TK). This vector is then used to transfect cells which are simultaneously infected with vaccinia. Homologous recombination serves to insert the vaccinia promoter plus the gene encoding the polypeptide of interest into the viral genome. The resulting TK.sup.(-) recombinant can be selected by culturing the cells in the presence of 5-bromodeoxyuridine and picking viral plaques resistant thereto.
A vaccinia-based infection/transfection system can be conveniently used to provide for inducible, transient expression or coexpression of one or more polypeptides described herein in host cells of an organism. In this particular system, cells are first infected in vitro with a vaccinia virus recombinant that encodes the bacteriophage T7 RNA polymerase. This polymerase displays exquisite specificity in that it only transcribes templates bearing T7 promoters. Following infection, cells are transfected with the polynucleotide or polynucleotides of interest, driven by a T7 promoter. The polymerase expressed in the cytoplasm from the vaccinia virus recombinant transcribes the transfected DNA into RNA which is then translated into polypeptide by the host translational machinery. The method provides for high level, transient, cytoplasmic production of large quantities of RNA and its translation products. See, e.g., Elroy-Stein and Moss, Proc. Natl. Acad. Sci. USA (1990) 87:6743-6747; Fuerst et al. Proc. Natl. Acad. Sci. USA (1986) 83:8122-8126.
Alternatively, avipoxviruses, such as the fowlpox and canarypox viruses, can also be used to deliver the coding sequences of interest. Recombinant avipox viruses, expressing immunogens from mammalian pathogens, are known to confer protective immunity when administered to non-avian species. The use of an Avipox vector is particularly desirable in human and other mammalian species since members of the Avipox genus can only productively replicate in susceptible avian species and therefore are not infective in mammalian cells. Methods for producing recombinant Avipoxviruses are known in the art and employ genetic recombination, as described above with respect to the production of vaccinia viruses. See, e.g., WO 91/12882; WO 89/03429; and WO 92/03545.
Any of a number of alphavirus vectors can also be used for delivery of polynucleotide compositions for use in the present invention, such as those vectors described in U.S. Patent Nos. 5,843,723; 6,015,686; 6,008,035 and 6,015,694. Certain vectors based on Venezuelan Equine Encephalitis (NEE) can also be used, illustrative examples of which can be found in U.S. Patent Νos. 5,505,947 and 5,643,576.
Moreover, molecular conjugate vectors, such as the adenovirus chimeric vectors described in Michael et al. J. Biol. Chem. (1993) 268:6866-6869 and Wagner et al. Proc. Νatl. Acad. Sci. USA (1992) 89:6099-6103, can also be used for gene delivery under the invention.
Additional illustrative information on these and other known viral-based delivery systems can be found, for example, in Fisher-Hoch et al., Proc. Natl. Acad. Sci. USA 86:311-321, 1989; Flexner et al., Ann. N.Y. Acad. Sci. 55P:86-103, 1989; Flexner et al., Vaccine 8:11-21, 1990; U.S. Patent Nos. 4,603,112, 4,769,330, and 5,017,487; WO 89/01973; U.S. Patent No. 4,777,127; GB 2,200,651; EP 0,345,242; WO 91/02805; Berkner, Biotechniques 6:616-621, 1988; Rosenfeld " et al., Science 252:431-434, 1991; Kolls et al., Proc. Natl. Acad. Sci. USA
91:215-219, 1994; Kass-Eisler et al, Proc. Natl. Acad. Sci. USA O: 11498-11502, 1993; Guzman et al., Circulation 88:2838-2848, 1993; and Guzman et al., Cir. Res. 73:1202-1207, 1993.
The recombinant live microorganisms described above can be virulent, or attenuated in various ways in order to obtain live vaccines. Such live vaccines also form part of the invention.
In certain embodiments, a polynucleotide may be integrated into the genome of a target cell. This integration may be in the specific location and orientation via homologous recombination (gene replacement) or it may be integrated in a random, non-specific location (gene augmentation). In yet further embodiments, the polynucleotide may be stably maintained in the cell as a separate, episomal segment of DNA. Such polynucleotide segments or "episomes" encode sequences sufficient to permit maintenance and replication independent of or in synchronization with the host cell cycle. The manner in which the expression construct is delivered to a cell and where in the cell the polynucleotide remains is dependent on the type of expression construct employed.
In another embodiment of the invention, a polynucleotide is administered/delivered as "naked" DNA, for example as described in Ul er et al., Science 259:1145-1149, 1993 and reviewed by Cohen, Science 259:1691-1692, 1993. The uptake of naked DNA may be increased by coating the DNA onto biodegradable beads, which are efficiently transported into the cells.
In still another embodiment, a composition of the present invention can be delivered via a particle bombardment approach, many of which have been described. In one illustrative example, gas-driven particle acceleration can be achieved with devices such as those manufactured by Powderject Pharmaceuticals PLC (Oxford, UK) and Powderject Vaccines Inc. (Madison, WI), some examples of which are described in U.S. Patent Nos. 5,846,796; 6,010,478; 5,865,796; 5,584,807; and EP Patent No. 0500 799. This approach offers a needle-free delivery approach wherein a dry powder formulation of microscopic particles, such as polynucleotide or polypeptide particles, are accelerated to high speed within a helium gas jet generated by a hand held device, propelling the particles into a target tissue of interest.
In a related embodiment, other devices and methods that may be useful for gas- driven needle-less inj ection of compositions of the present invention include those provided by Bioject, Inc. (Portland, OR), some examples of which are described in U.S. Patent Nos. 4,790,824; 5,064,413; 5,312,335; 5,383,851; 5,399,163; 5,520,639 and 5,993,412.
Polypeptides for use in the present invention can be recovered and purified from recombinant cell cultures by well-known methods including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and lectin chromatography. Most preferably, ion metal affinity chromatography (EMAC) is employed for purification. Well known techniques for refolding proteins may be employed to regenerate active conformation when the polypeptide is denatured during intracellular synthesis, isolation and or purification.
Another important aspect of the invention relates to a method for inducing , reinforcing or modulating an immunological response in a mammal which comprises inoculating the mammal with a fragment or the entire polypeptide or polynucleotide of the invention, adequate to produce antibody and or T cell immune response for prophylaxis or for therapeutic treatment of breast cancer. Yet another aspect of the invention relates to a method of inducing, re-inforcing or modulating immunological response in a mammal which comprises, delivering a polypeptide as contemplated in the present invention via a vector or cell directing expression of the polynucleotide and coding for the polypeptide in vivo in order to induce such an immunological response to produce immune responses for prophylaxis or treatment of said mammal from breast cancer.
A further aspect of the invention relates to the use of an immunological/vaccine formulation (composition) which, when introduced into a mammalian host suffering from or susceptible to breast cancer, induces, re-inforces or modulates an immunological response in that mammal to a polypeptide according to the present invention wherein the composition comprises a polypeptide or polynucleotide of the invention or an immunological fragment thereof as herein before defined.The vaccine formulation for use in breast cancer indication may further comprise a suitable carrier. Since a polypeptide may be broken down in the stomach, it is preferably administered parenterally (for instance, subcutaneous, intramuscular, intravenous, or intradermal injection). Formulations suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents or thickening agents. The formulations may be presented in unit-dose or multi-dose containers, for example, sealed ampoules and vials and may be stored in a freeze-dried condition requiring only the addition of the sterile liquid carrier immediately prior to use.
A further aspect of the invention relates to the in vitro induction of immune responses to a fragment or the entire polypeptide or polynucleotide disclosed herein or a molecule comprising said polypeptide or polynucleotide, using cells from the immune system of a mammal, and reinfusing these activated immune cells of the mammal for the treatment of breast cancer. Activation of the cells from the immune system is achieved by in vitro incubation with the entire polypeptide or polynucleotide according to the present invention or a molecule comprising said polypeptide or polynucleotide in the presence or absence of various immunomodulator molecules.
A further aspect of the invention relates to the immunisation of a mammal suffering from or susceptible to breast cancer by administration of antigen presenting cells modified by in vitro loading with part or the entire polypeptide disclosed herein or a molecule comprising said polypeptide and administered in vivo in an immunogenic way. Alternatively, antigen presenting cells can be transfected in vitro with a vector containing a fragment or the entire polynucleotide according to the present invention or a molecule comprising said polynucleotide, such as to express the corresponding polypeptide, and administered in vivo in an immunogenic way.
According to another embodiment, the pharmaceutical compositions described herein will comprise one or more immunostimulants in addition to the immunogenic polynucleotide, polypeptide, antibody, T-cell and/or antigen presenting cell (APC) compositions of this invention. An immunostimulant refers to essentially any substance that enhances or potentiates an immune response (antibody and/or cell-mediated) to an exogenous antigen. One preferred type of immunostimulant comprises an adjuvant. Many adjuvants contain a substance designed to protect the antigen from rapid catabolism, such as aluminum hydroxide or mineral oil, and a stimulator of immune responses, such as lipid A, Bortadella pertussis or Mycobacterium tuberculosis derived proteins. Certain adjuvants are commercially available as, for example, Freund's Incomplete Adjuvant and Complete Adjuvant (Difco Laboratories, Detroit, MI); Merck Adjuvant 65 (Merck and Company, Inc., Rahway, NJ); AS-2 (SmithKline Beecham, Philadelphia, PA); aluminum salts such as aluminum hydroxide gel (alum) or aluminum phosphate; salts of calcium, iron or zinc; an insoluble suspension of acylated tyrosine; acylated sugars; cationically or anionically derivatized polysaccharides; polyphosphazenes; biodegradable microspheres; monophosphoryl lipid A and quil A. Cytokines, such as GM-CSF, interleukin-2, - 7, -12, and other like growth factors, may also be used as adjuvants.
Within certain embodiments of the invention, the adjuvant composition is preferably one that induces an immune response predominantly of the Thl type. High levels of Thl -type cytokines (e.g., IFN-γ, TNFα, IL-2 and IL-12) tend to favor the induction of cell mediated immune responses to an administered antigen. In contrast, high levels of Th2-type cytokines (e.g., IL-4, IL-5, IL-6 and IL-10) tend to favor the induction of humoral immune responses. Following application of a vaccine as provided herein, a patient will support an immune response that includes Thl- and Th2-type responses. Within a preferred embodiment, in which a response is predominantly Thl -type, the level of Thl -type cytokines will increase to a greater extent than the level of Th2-type cytokines. The levels of these cytokines may be readily assessed using standard assays. For a review of the families of cytokines, see Mosmann and Coffman, Ann. Rev. Immunol. 7:145- 173, 1989.
Certain preferred adjuvants for eliciting a predominantly Thl -type response include, for example, a combination of monophosphoryl lipid A, preferably 3-de- O-acylated monophosphoryl lipid A, together with an aluminum salt. MPL® adjuvants are available from Corixa Corporation (Seattle, WA; see, for example, US Patent Nos. 4,436,727; 4,877,611; 4,866,034 and 4,912,094). CpG-containing oligonucleotides (in which the CpG dinucleotide is unrnethylated) also induce a predominantly Thl response. Such oligonucleotides are well known and are described, for example, in WO 96/02555, WO 99/33488 and U.S. Patent Nos.
6,008,200 and 5,856,462. Immunostimulatory DNA sequences are also described, for example, by Sato et al., Science 273:352, 1996. Another preferred adjuvant comprises a saponin, such as Quil A, or derivatives thereof, including QS21 and QS7 (Aquila Biopharmaceuticals Inc., Framingham, MA); Escin; Digitonin; or Gypsophila or Chenopodium quinoa saponins . Other preferred formulations include more than one saponin in the adjuvant combinations of the present invention, for example combinations of at least two of the following group comprising QS21, QS7, Quil A, β-escin, or digitonin.
Alternatively the saponin formulations may be combined with vaccine vehicles composed of chitosan or other polycationic polymers, polylactide and polylactide- co-glycolide particles, poly-N-acetyl glucosamine-based polymer matrix, particles composed of polysaccharides or chemically modified polysaccharides, liposomes and lipid-based particles, particles composed of glycerol monoesters, etc. The saponins may also be formulated in the presence of cholesterol to form particulate structures such as liposomes or ISCOMs. Furthermore, the saponins may be formulated together with a polyoxyethylene ether or ester, in either a non- particulate solution or suspension, or in a particulate structure such as a paucilamelar liposome or ISCOM. The saponins may also be formulated with excipients such as CarbopolR to increase viscosity, or may be formulated in a dry powder form with a powder excipient such as lactose.
In one prefened embodiment, the adjuvant system includes the combination of a monophosphoryl lipid A and a saponin derivative, such as the combination of QS21 and 3D-MPL® adjuvant, as described in WO 94/00153, or a less reactogenic composition where the QS21 is quenched with cholesterol, as described in WO 96/33739. Other preferred formulations comprise an oil-in-water emulsion and tocopherol. Another particularly preferred adjuvant formulation employing QS21 , 3D-MPL® adjuvant and tocopherol in an oil-in-water emulsion is described in WO 95/17210.
Another enhanced adjuvant system involves the combination of a CpG-containing oligonucleotide and a saponin derivative particularly the combination of CpG and QS21 as disclosed in WO 00/09159. Preferably the formulation additionally comprises an oil in water emulsion and tocopherol.
Additional illustrative adjuvants for use in the pharmaceutical compositions of the invention include Montanide ISA 720 (Seppic, France), SAF (Chiron, California, United States), ISCOMS (CSL), MF-59 (Chiron), the SBAS series of adjuvants (e.g., SBAS-2 or SBAS-4, available from SmithKline Beecham, Rixensart, Belgium), Detox (Enhanzyn®) (Corixa, Hamilton, MT), RC-529 (Corixa, Hamilton, MT) and other aminoalkyl glucosaminide 4-phosphates (AGPs), such as those described in pending U.S. Patent Application Serial Nos. 08/853,826 and 09/074,720, the disclosures of which are incorporated herein by reference in their entireties, and polyoxyethylene ether adjuvants such as those described in WO 99/52549A1.
Other preferred adjuvants include adjuvant molecules of the general formula (I):
HO(CH2CH2O)n-A-R
Wherein, n is 1-50, A is a bond or -C(O)-, R is Ci-50 alkyl or Phenyl Cι-50 alkyl. One embodiment of the present invention consists of a vaccine formulation for use in the treatment or prophylaxis of breast cancer comprising a polyoxyethylene ether of general formula (I), wherein n is between 1 and 50, preferably 4-24, most preferably 9; the R component is C1-50, preferably C4-C20 alkyl and most preferably C12 alkyl, and A is a bond. The concentration of the polyoxyethylene ethers should be in the range 0.1-20%, preferably from 0.1-10%, and most preferably in the range 0.1-1%. Preferred polyoxyethylene ethers are selected from the following group: polyoxyethylene-9-lauryl ether, polyoxyethylene-9- steoryl ether, polyoxyethylene-8-steoryl ether, polyoxyethylene-4-lauryl ether, polyoxyethylene-35-lauryl ether, and polyoxyethylene-23-lauryl ether.
Polyoxyethylene ethers such as polyoxyethylene lauryl ether are described in the Merck index (12 edition: entry 7717). These adjuvant molecules are described in WO 99/52549.
The polyoxyethylene ether according to the general formula (I) above may, if desired, be combined with another adjuvant. For example, a prefened adjuvant combination is preferably with CpG as described in the pending UK patent application GB 9820956.2.
Preferably a carrier is also present in the vaccine composition for use according to the invention. The carrier may be an oil in water emulsion, or an aluminium salt, such as aluminium phosphate or aluminium hydroxide.
A prefened oil-in-water emulsion comprises a metabolisible oil, such as squalene, alpha tocopherol and Tween 80. In a particularly prefened aspect the antigens in the vaccine composition according to the invention are combined with QS21 and 3D-MPL in such an emulsion. Additionally the oil in water emulsion may contain span 85 and/or lecithin and/or tricaprylin.
Typically for human administration QS21 and 3D-MPL will be present in a vaccine in the range of lμg - 200μg, such as 10-100μg, preferably lOμg - 50μg per dose. Typically the oil in water will comprise from 2 to 10% squalene, from 2 to 10% alpha tocopherol and from 0.3 to 3% tween 80. Preferably the ratio of squalene: alpha tocopherol is equal to or less than 1 as this provides a more stable emulsion. Span 85 may also be present at a level of 1 %. In some cases it may be advantageous that the vaccines of the present invention will further contain a stabiliser.
Non-toxic oil in water emulsions preferably contain a non-toxic oil, e.g. squalane or squalene, an emulsifier, e.g. Tween 80, in an aqueous carrier. The aqueous carrier may be, for example, phosphate buffered saline.
A particularly potent adjuvant formulation involving QS21, 3D-MPL and tocopherol in an oil in water emulsion is described in WO 95/17210.
The present invention also provides the use of a polyvalent vaccine composition in the treatment or prophylaxis of breast cancer comprising a vaccine formulation of the invention in combination with other antigens, useful for treating breast cancers, . Such a polyvalent vaccine composition may include a TH-1 inducing adjuvant as hereinbefore described.
An immunogenic composition for use according to the invention is delivered to a host via antigen presenting cells (APCs), such as dendritic cells, macrophages, B cells, monocytes and other cells that may be engineered to be efficient APCs. Such cells may, but need not, be genetically modified to increase the capacity for presenting the antigen, to improve activation and or maintenance of the T cell response, to have anti-tumour effects per se and/or to be immunologically compatible with the receiver (i.e., matched HLA haplotype). APCs may generally be isolated from any of a variety of biological fluids and organs, including tumour and peritumoural tissues, and may be autologous, allogeneic, syngeneic or xenogeneic cells.
Certain preferred embodiments of the present invention use dendritic cells or progenitors thereof as antigen-presenting cells. Dendritic cells are highly potent APCs (Banchereau and Steinman, Nature 392:245-251, 1998) and have been shown to be effective as a physiological adjuvant for eliciting prophylactic or therapeutic antitumour immunity (see Timmerman and Levy, Ann. Rev. Med. 50:507-529, 1999). Ln general, dendritic cells may be identified based on their typical shape (stellate in situ, with marked cytoplasmic processes (dendrites) visible in vitro), their ability to take up, process and present antigens with high efficiency and their ability to activate naive T cell responses. Dendritic cells may, of course, be engineered to express specific cell-surface receptors or ligands that are not commonly found on dendritic cells in vivo or ex vivo, and such modified dendritic cells are contemplated by the present invention. As an alternative to dendritic cells, secreted vesicles antigen-loaded dendritic cells (called exosomes) may be used within a vaccine (see Zitvogel et al., Nature Med. 4:594-600, 1998).
Dendritic cells and progenitors may be obtained from peripheral blood, bone marrow, tumour-infiltrating cells, peritumoural tissues-infiltrating cells, lymph nodes, spleen, skin, umbilical cord blood or any other suitable tissue or fluid. For example, dendritic cells may be differentiated ex vivo by adding a combination of cytokines such as GM-CSF, IL-4, IL-13 and/or TNFα to cultures of monocytes harvested from peripheral blood. Alternatively, CD34 positive cells harvested from peripheral blood, umbilical cord blood or bone manow may be differentiated into dendritic cells by adding to the culture medium combinations of GM-CSF, EL-3, TNFα, CD40 ligand, LPS, flt3 ligand and/or other compound(s) that induce differentiation, maturation and proliferation of dendritic cells.
Dendritic cells are conveniently categorized as "immature" and "mature" cells, which allows a simple way to discriminate between two well characterized phenotypes. However, this nomenclature should not be construed to exclude all possible intermediate stages of differentiation. Immature dendritic cells are characterized as APC with a high capacity for antigen uptake and processing, which corcelates with the high expression of Fcγ receptor and mannose receptor. The mature phenotype is typically characterized by a lower expression of these markers, but a high expression of cell surface molecules responsible for T cell activation such as class I and class II MHC, adhesion molecules (e.g., CD54 and CD11) and costimulatory molecules (e.g., CD40, CD80, CD86 and 4-1BB). APCs may generally be transfected with a polynucleotide of the invention (or portion or other variant thereof) such that the encoded polypeptide, or an immunogenic portion thereof, is expressed on the cell surface. Such transfection may take place ex vivo, and a pharmaceutical composition comprising such transfected cells may then be used for therapeutic purposes, as described herein. Alternatively, a gene delivery vehicle that targets a dendritic or other antigen presenting cell may be administered to a patient, resulting in transfection that occurs in vivo. In vivo and ex vivo transfection of dendritic cells, for example, may generally be performed using any methods known in the art, such as those described in WO 97/24447, or the gene gun approach described by Mahvi et al., Immunology and cell Biology 75:456-460, 1997. Antigen loading of dendritic cells may be achieved by incubating dendritic cells or progenitor cells with the tumour polypeptide, DNA (naked or within a plasmid vector) or RNA; or with antigen-expressing recombinant bacterium or viruses (e.g., vaccinia, fowlpox, adenovirus or lentivirus vectors). Prior to loading, the polypeptide may be covalently conjugated to an immunological partner that provides T cell help (e.g., a carrier molecule). Alternatively, a dendritic cell may be pulsed with a non- conjugated immunological partner, separately or in the presence of the polypeptide.
While any suitable carrier known to those of ordinary skill in the art may be employed in the pharmaceutical compositions for use according to the present invention, the type of carrier will typically vary depending on the mode of administration. The compositions may be formulated for any appropriate manner of administration, including for example, topical, oral, nasal, mucosal, intravenous, intracranial, intraperitoneal, subcutaneous and intramuscular administration.
Carriers for use within such pharmaceutical compositions are biocompatible, and may also be biodegradable. In certain embodiments, the formulation preferably provides a relatively constant level of active component release. En other embodiments, however, a more rapid rate of release immediately upon administration may be desired. The formulation of such compositions is well within the level of ordinary skill in the art using known techniques. Illustrative carriers useful in this regard include microparticles of poly(lactide-co-glycolide), polyacrylate, latex, starch, cellulose, dextran and the like. Other illustrative delayed-release carriers include supramolecular biovectors, which comprise a non- liquid hydrophilic core (e.g., a cross-linked polysaccharide or oligosaccharide) and, optionally, an external layer comprising an amphiphihc compound, such as a phospholipid (see e.g., U.S. Patent No. 5,151,254 and PCT applications WO 94/20078, WO/94/23701 and WO 96/06638). The amount of active compound contained within a sustained release formulation depends upon the site of implantation, the rate and expected duration of release and the nature of the condition to be treated or prevented.
In another illustrative embodiment, biodegradable microspheres (e.g., polylactate polyglycolate) are employed as carriers for the compositions of this invention. Suitable biodegradable microspheres are disclosed, for example, in U.S. Patent Nos. 4,897,268; 5,075,109; 5,928,647; 5,811,128; 5,820,883; 5,853,763;
5,814,344, 5,407,609 and 5,942,252. Modified hepatitis B core protein carrier systems, such as described in WO/99 40934, and references cited therein, will also be useful for many applications. Another illustrative carrier/delivery system employs a carrier comprising particulate-protein complexes, such as those described in U.S. Patent No. 5,928,647, which are capable of inducing a class I- restricted cytotoxic T lymphocyte responses in a host.
The pharmaceutical compositions according to the invention will often further comprise one or more buffers (e.g., neutral buffered saline or phosphate buffered saline), carbohydrates (e.g., glucose, mannose, sucrose or dextrans), mannitol, proteins, polypeptides or amino acids such as glycine, antioxidants, bacteriostats, chelating agents such as EDTA or glutathione, adjuvants (e.g., aluminum hydroxide), solutes that render the formulation isotonic, hypotonic or weakly hypertonic with the blood of a recipient, suspending agents, thickening agents and/or preservatives. Alternatively, compositions of the present invention may be formulated as a lyophilizate.
The pharmaceutical compositions described herein may be presented in unit-dose or multi-dose containers, such as sealed ampoules or vials. Such containers are typically sealed in such a way to preserve the sterility and stability of the formulation until use. In general, formulations may be stored as suspensions, solutions or emulsions in oily or aqueous vehicles. Alternatively, a pharmaceutical composition may be stored in a freeze-dried condition requiring only the addition of a sterile liquid carrier immediately prior to use.
The development of suitable dosing and treatment regimens for using the particular compositions described herein in a variety of treatment regimens, including e.g., oral, parenteral, intravenous, intranasal, and intramuscular adπunistration and formulation, is well known in the art, some of which are briefly discussed below for general purposes of illustration.
En certain applications, the pharmaceutical compositions disclosed herein may be delivered via oral administration to an animal. As such, these compositions may be formulated with an inert diluent or with an assimilable edible carrier, or they may be enclosed in hard- or soft-shell gelatin capsule, or they may be compressed into tablets, or they may be incorporated directly with the food of the diet.
The active compounds may even be incorporated with excipients and used in the form of ingestible tablets, buccal tables, troches, capsules, elixirs, suspensions, syrups, wafers, and the like (see, for example, Mathiowitz et αl., Nature 1997 Mar 27;386(6623):410-4; Hwang et αl, CritRev Ther Drug Carrier Syst 1998;15(3):243-84; U. S. Patent 5,641,515; U. S. Patent 5,580,579 an U. S. Patent 5,792,451). Tablets, troches, pills, capsules and the like may also contain any of a variety of additional components, for example, a binder, such as gum tragacanth, acacia, cornstarch, or gelatin; excipients, such as dicalcium phosphate; a disintegrating agent, such as com starch, potato starch, alginic acid and the like; a lubricant, such as magnesium stearate; and a sweetening agent, such as sucrose, lactose or saccharin maybe added or a flavoring agent, such as peppermint, oil of wintergreen, or cheny flavoring. When the dosage unit form is a capsule, it may contain, in addition to materials of the above type, a liquid carrier. Various other materials may be present as coatings or to otherwise modify the physical form of the dosage unit. For instance, tablets, pills, or capsules may be coated with shellac, sugar, or both. Of course, any material used in preparing any dosage unit form should be pharmaceutically pure and substantially non-toxic in the amounts employed. In addition, the active compounds may be incorporated into sustained- release preparation and formulations.
Typically, these formulations will contain at least about 0.1% of the active compound or more, although the percentage of the active ingredient(s) may, of course, be varied and may conveniently be between about 1 or 2% and about 60% or 70% or more of the weight or volume of the total formulation. Naturally, the amount of active compound(s) in each therapeutically useful composition may be prepared is such a way that a suitable dosage will be obtained in any given unit dose of the compound. Factors such as solubility, bioavailability, biological half- life, route of administration, product shelf life, as well as other pharmacological considerations will be contemplated by one skilled in the art of preparing such pharmaceutical formulations, and as such, a variety of dosages and treatment regimens may be desirable.
For oral administration the compositions according to the present invention may alternatively be incorporated with one or more excipients in the form of a mouthwash, dentifrice, buccal tablet, oral spray, or sublingual orally-administered formulation. Alternatively, the active ingredient may be incorporated into an oral solution such as one containing sodium borate, glycerin and potassium bicarbonate, or dispersed in a dentifrice, or added in a therapeutically-effective amount to a composition that may include water, binders, abrasives, flavoring agents, foaming agents, and humectants. Alternatively the compositions may be fashioned into a tablet or solution form that may be placed under the tongue or otherwise dissolved in the mouth.
In certain circumstances it will be desirable to deliver the pharmaceutical compositions disclosed herein parenterally, intravenously, intramuscularly, or even intraperitoneally. Such approaches are well known to the skilled artisan, some of which are further described, for example, in U. S. Patent 5,543,158; U. S. Patent 5,641,515 and U. S. Patent 5,399,363. En certain embodiments, solutions of the active compounds as free base or pharmacologically acceptable salts may be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions may also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations generally will contain a preservative to prevent the growth of microorganisms.
Illustrative pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions (for example, see U. S. Patent 5,466,468). In all cases the form must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (e.g., glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and/or vegetable oils. Proper fluidity may be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and/or by the use of surfactants. The prevention of the action of microorganisms can be facilitated by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
In one embodiment, for parenteral administration in an aqueous solution, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose. These particular aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration. In this connection, a sterile aqueous medium that can be employed will be known to those of skill in the art in light of the present disclosure. For example, one dosage maybe dissolved in 1 ml of isotonic NaCI solution and either added to 1000 ml of hypodermoclysis fluid or injected at the proposed site of infusion, (see for example, "Remington's Pharmaceutical Sciences" 15th Edition, pages 1035-1038 and 1570-1580). Some variation in dosage will necessarily occur depending on the condition of the subject being treated. Moreover, for human administration, preparations will of course preferably meet sterility, pyrogenicity, and the general safety and purity standards as required by FDA Office of Biologies standards.
In another embodiment of the invention, the compositions disclosed herein may be formulated in a neutral or salt form. Illustrative pharmaceutically-acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like. Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective.
The carriers can further comprise any and all solvents, dispersion media, vehicles, coatings, diluents, antibacterial and antifungal agents, isotonic and absorption delaying agents, buffers, carrier solutions, suspensions, colloids, and the like. The use of such media and agents for pharmaceutical active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active ingredient, its use in the therapeutic compositions is contemplated. Supplementary active ingredients can also be incorporated into the compositions. The phrase "pharmaceutically-acceptable" refers to molecular entities and compositions that do not produce an allergic or similar untoward reaction when administered to a human.
In certain embodiments, the pharmaceutical compositions may be delivered by intranasal sprays, inhalation, and/or other aerosol delivery vehicles. Methods for delivering genes, nucleic acids, and peptide compositions directly to the lungs via nasal aerosol sprays has been described, e.g., inU. S. Patent 5,756,353 and U. S. Patent 5,804,212. Likewise, the delivery of drugs using intranasal microparticle resins (Takenaga et al, J Controlled Release 1998 Mar 2;52(l-2):81-7) and lysophosphatidyl-glycerol compounds (U. S. Patent 5,725,871) are also well- known in the pharmaceutical arts. Likewise, illustrative transmucosal drug delivery in the form of a polytetrafluoroetheylene support matrix is described in U. S. Patent 5,780,045.
In certain embodiments, liposomes, nanocapsules, microparticles, lipid particles, vesicles, and the like, are used for the introduction of the compositions of the present invention into suitable host cells/organisms. Ln particular, the compositions of the present invention may be formulated for delivery either encapsulated in a lipid particle, a liposome, a vesicle, a nanosphere, or a nanoparticle or the like. Alternatively, the compositions disclosed herein can be bound, either covalently or non-covalently, to the surface of such carrier vehicles.
The formation and use of liposome and liposome-like preparations as potential drag carriers is generally known to those of skill in the art (see for example, Lasic, Trends Biotechnol 1998 Jul; 16(7): 307-21; Takakura, Nippon Rinsho 1998 Mar;56(3):691-5; Chandran et al, Indian J Exp Biol. 1997 Aug;35(8):801-9; Margalit, Crit Rev Ther Drug Carrier Syst. 1995;12(2-3):233-61; U.S. Patent 5,567,434; U.S. Patent 5,552,157; U.S. Patent 5,565,213; U.S. Patent 5,738,868 and U.S. Patent 5,795,587, each specifically incorporated herein by reference in its entirety).
Liposomes have been used successfully with a number of cell types that are normally difficult to transfect by other procedures, including T cell suspensions, primary hepatocyte cultures and PC 12 cells (Renneisen et al, J Biol Chem. 1990 Sep 25;265(27): 16337-42; Muller et al, DNA Cell Biol. 1990 Apr;9(3):221-9). In addition, liposomes are free of the DNA length constraints that are typical of viral- based delivery systems. Liposomes have been used effectively to introduce genes, various drugs, radiotherapeutic agents, enzymes, viruses, transcription factors, allosteric effectors and the like, into a variety of cultured cell lines and animals. Furthermore, the use of liposomes does not appear to be associated with autoimmune responses or unacceptable toxicity after systemic delivery. In certain embodiments, liposomes are formed from phospholipids that are dispersed in an aqueous medium and spontaneously form multilamellar concentric bilayer vesicles (also termed multilamellar vesicles (MLVs).
Alternatively, in other embodiments, the invention provides the use of pharmaceutically-acceptable nanocapsule formulations of the compositions disclosed herein. Nanocapsules can generally entrap compounds in a stable and reproducible way (see, for example, Quintanar-Guenero et al., Drug Dev End Pharm. 1998 Dec;24(12): 1113-28). To avoid side effects due to intracellular polymeric overloading, such ultrafine particles (sized around 0.1 μm) may be designed using polymers able to be degraded in vivo. Such particles can be made as described, for example, by Couvreur et al, Crit Rev Ther Drug Carrier Syst. 1988;5(l):l-20; zur Muhlen et al, Eur J Pharm Biopharm. 1998 Mar;45(2):149- 55; Zambaux et al. J Controlled Release. 1998 Jan 2;50(l-3):31-40; and U. S. Patent 5,145,684.
This invention also relates to the use of polynucleotides, in the form of primers derived from the polynucleotides of the present invention, and of polypeptides, in the form of antibodies or reagents specific for the polypeptide of the present invention, as diagnostic reagents in the context of breast cancer.
The identification of genetic or biochemical markers in blood or tissues that will enable the detection of very early changes along the carcinogenesis pathway will help in determining the best treatment for the patient. Surrogate tumour markers, such as polynucleotide expression, can be used to diagnose different forms and states of breast cancer. The identification of expression levels of the polynucleotides of the invention will be useful in both the staging of the cancerous disorder and grading the nature of the cancerous tissue. The staging process monitors the advancement of the cancer of the breast and is determined on the presence or absence of malignant tissue in the areas biopsied. The polynucleotides of the invention can help to perfect the staging process by identifying markers for the aggressivity of said cancer, for example the presence in different areas of the body. The grading of the cancer describes how closely a tumour resembles normal tissue of its same type and is assessed by its cell morphology and other markers of differentiation. The polynucleotides of the invention can be useful in determining the tumour grade as they can help in the determination of the differentiation status of the cells of a tumour.
The diagnostic assays offer a process for diagnosing or determining a susceptibility to breast cancer, through diagnosis by methods comprising determining from a sample derived from a subject an abnormally decreased or increased level of polypeptide or mRNA. This method of diagnosis is known as differential expression. The expression of a particular gene is compared between a diseased tissue and a normal tissue. A difference between the polynucleotide-related gene, mRNA, or protein in the two tissues is compared, for example in molecular weight, amino acid or nucleotide sequence, or relative abundance, indicates a change in the gene, or a gene which regulates it, in the tissue of the human that was suspected of being diseased.
Decreased or increased expression can be measured at the RNA level. PolyA RNA is first isolated from the two tissues and the detection of mRNA encoded by a gene conesponding to a differentially expressed polynucleotide according to the invention can be performed by, for example, in situ hybridisation in tissue sections, reverse trascriptase-PCR, using Northern blots containing poly A+ mRNA, or any other direct or inderect RNA detection method. An increased or decreased expression of a given RNA in a diseased tissue compared to a normal tissue suggests that the transcript and/or the expressed protein has a role in the disease. Thus detection of a higher or lower level of mRNA conesponding to SEQ TD NO: 1 or SEQ ED NO:4 relative to normal level is indicative of the presence of cancer in the patient.
mRNA expression levels in a sample can be determined by generation of a library of expressed sequence tags (ESTs) from the sample. The relative representation of ESTs in the library can be used to assess the relative representation of the gene transcript in the starting sample. The EST analysis of the test can then be compared to the EST analysis of a reference sample to determine the relative expression levels of the polynucleotide of interest. Other mRNA analyses can be carried out using serial analysis of gene expression (SAGE) methodology (Velculescu et. Al. Science (1995) 270:484) , differential display methodology (For example, US 5,776,683) or hybridisation analysis which relies on the specificity of nucleotide interactions.
Alternatively, the comparison could be made at the protein level. The protein sizes in the two tissues may be compared using antibodies to detect polypeptides in Western blots of protein extracts from the two tissues. Expression levels and subcellular localization may also be detected immtmologically using antibodies to the conesponding protein. Further assay techniques that can be used to determine levels of a protein, such as a polypeptide of the present invention, in a sample derived from a host are well-known to those of skill in the art. A raised or decreased level of polypeptide expression in the diseased tissue compared with the same protein expression level in the normal tissue indicates that the expressed protein may be involved in the disease.
In the assays of the present invention, the diagnosis can be determined by detection of gene product expression levels encoded by at least one sequence set forth in SEQ ID NO: 1 or SEQ ED NO:4. A comparison of the mRNA or protein levels in a diseased versus normal tissue may also be used to follow the progression or remission of a disease.
A large number of polynucleotide sequences in a sample can be assayed using polynucleotide anays. These can be used to examine differential expression of genes and to determine gene function. For example, anays of the polynucleotide sequences SEQ ED NO: 1 or SEQ ED NO:4 can be used to determine if any of the polynucleotides are differentially expressed between a normal and cancer cell. In one embodiment of the invention, an anay of oligonucleotides probes comprising the SEQ TD NO: 1 or SEQ ED NO:4 nucleotide sequence or fragments thereof can be constructed to conduct efficient screening of e.g., genetic mutations. Anay technology methods are well known and have general applicability and can be used to address a variety of questions in molecular genetics including gene expression, genetic linkage, and genetic variability (see for example: M.Chee et al., Science, Vol 274, pp 610-613 (1996)).
"Diagnosis" as used herein includes determination of a subject's susceptibility to breast cancer disease, determination as to whether a subject presently has said disease, and also the prognosis of a subject affected by said disease.
The present invention, further relates to a diagnostic kit for performing a diagnostic assay in a patient suffering from or susceptible to breast cancer disease which comprises:
(a) a polynucleotide of the present invention, preferably the nucleotide sequence of SEQ ID NO: 1 or SEQ ED NO:4, or a fragment thereof ;
(b) a nucleotide sequence complementary to that of (a);
(c) a polypeptide of the present invention, preferably the polypeptide of SEQ TD NO: 2, SEQ ED NO:3 or SEQ ED NO:8, or a fragment thereof; or
(d) an antibody to a polypeptide of the present invention, preferably to the polypeptide of SEQ ID NO: 2, SEQ ED NO:3 or SEQ ED NO:8.
The nucleotide sequences for use in the present invention are also valuable for chromosomal localisation. The sequence is specifically targeted to, and can hybridize with, a particular location on an individual human chromosome. The mapping of relevant sequences to chromosomes according to the present invention is an important first step in conelating those sequences with gene associated disease. Once a sequence has been mapped to a precise chromosomal location, the physical position of the sequence on the chromosome can be conelated with genetic map data. Such data are found in, for example, V. McKusick, Mendelian Inheritance in Man (available on-line through Johns Hopkins University Welch Medical Library). The relationship between genes and diseases that have been mapped to the same chromosomal region are then identified through linkage analysis (coinheritance of physically adjacent genes).The differences in the cDNA or genomic sequence between affected and unaffected individuals can also be determined.
The polypeptides for use in the invention or their fragments or analogs thereof, or cells expressing them, can also be used as immunogens to produce antibodies immunospecific for polypeptides of the present invention. The term "immunospecific" means that the antibodies have substantially greater affinity for the polypeptides disclosed herein than their affinity for other related polypeptides in the prior art.
In a further aspect the invention provides for the use of an antibody immunospecific for a polypeptide according to the invention or an immunological fragment thereof as hereinbefore defined. Preferably the antibody is a monoclonal antibody
Antibodies generated against the polypeptides disclosed herein may be obtained by administering the polypeptides or epitope-bearing fragments, analogs or cells to an animal, preferably a non-human animal, using routine protocols. For preparation of monoclonal antibodies, any technique which provides antibodies produced by continuous cell line cultures can be used. Examples include the hybridoma technique (Kohler, G. and Milstein, C, Nature (1975) 256:495-497), the trioma technique, the human B-cell hybridoma technique (Kozbor et al, Immunology Today (1983) 4:72) and the EBV-hybridoma technique (Cole et al, Monoclonal Antibodies and Cancer Therapy, 77-96, Alan R. Liss, Inc., 1985).
Techniques for the production of single chain antibodies, such as those described in U.S. Patent No. 4,946,778, can also be adapted to produce single chain antibodies to polypeptides of this invention. Also, transgenic mice, or other organisms, including other mammals, may be used to express humanized antibodies.
The above-described antibodies may be employed to isolate or to identify clones expressing the polypeptide or to purify the polypeptides by affinity chromatography. The antibody of the invention may also be employed to prevent or treat breast cancer in a patient.
Another aspect of the invention relates to a method for inducing or modulating an immunological response in a mammal which comprises inoculating the mammal with a polypeptide of the present invention, adequate to produce antibody and/or T cell immune response to protect or ameliorate the symptoms or progression of the breast cancer. Yet another aspect of the invention relates to a method of inducing or modulating immunological response in a mammal which comprises, delivering a polypeptide of the present invention via a vector directing expression of the polynucleotide and coding for the polypeptide in vivo in order to induce such an immunological response to produce antibody to protect said animal from breast cancer.
It will be appreciated that the present invention therefore provides a method of treating breast cancer, related to either a presence of, an excess of, or an under- expression of, CASB7439 polypeptide activity.
The present invention further provides for a method of screening compounds to identify those which stimulate or which inhibit the function of the CASB7439 polypeptide. In general, agonists or antagonists may be employed for therapeutic and prophylactic purposes for breast cancer as hereinbefore mentioned. Compounds may be identified from a variety of sources, for example, cells, ceU-free preparations, chemical libraries, and natural product mixtures. Such agonists, antagonists or inhibitors so-identified may be natural or modified substrates, ligands, receptors, enzymes, etc., as the case may be, of the polypeptide; or may be structural or functional mimetics thereof (see Coligan et al, Current Protocols in Immunology l(2):Chapter 5 (1991)). Screening methods will be known to those skilled in the art. Further screening methods may be found in for example D. Bennett et al, J Mol Recognition, 8:52-58 (1995); and K. Johanson et al, J Biol Chem, 270(16):9459-9471 (1995) and references therein.
Thus the invention provides a method for screening to identify compounds which stimulate or which inhibit the function of the polypeptides according to the invention which comprises a method selected from the group consisting of:
(a) measuring the binding of a candidate compound to the polypeptide (or to the cells or membranes bearing the polypeptide) or a fusion protein thereof by means of a label directly or indirectly associated with the candidate compound;
(b) measuring the binding of a candidate compound to the polypeptide (or to the cells or membranes bearing the polypeptide) or a fusion protein thereof in the presense of a labeled competitior; (c) testing whether the candidate compound results in a signal generated by activation or inhibition of the polypeptide, using detection systems appropriate to the cells or cell membranes bearing the polypeptide;
(d) mixing a candidate compound with a solution containing a polypeptide of claim 1, to form a mixture, measuring activity of the polypeptide in the mixture, and comparing the activity of the mixture to a standard; or
(e) detecting the effect of a candidate compound on the production of mRNA encoding said polypeptide and said polypeptide in cells, using for instance, an ELISA assay.
The polypeptides according to the may be used to identify membrane bound or soluble receptors, if any, through standard receptor binding techniques known in the art. Well known screening methods may also be used to identify agonists and antagonists of the polypeptides disclosed herein which compete with the binding of the polypeptide of the invention to its receptors, if any.
Thus, in another aspect, the present invention relates to a screening kit for identifying agonists, antagonists, ligands, receptors, substrates, enzymes, etc. for polypeptides disclosed herein that are used for therapeutic and prophylactic purposes for breast cancer; or compounds which decrease or enhance the production of such polypeptides, which comprises:
(a) a polypeptide of the present invention;
(b) a recombinant cell expressing a polypeptide of the present invention;
(c) a cell membrane expressing a polypeptide of the present invention; or (d) antibody to a polypeptide of the present invention; which polypeptide is preferably that of SEQ ED NO: 2, SEQ ED NO:3 or SEQ ID NO:8.
It will be readily appreciated by the skilled artisan that a polypeptide according to the present invention may also be used in a method for the structure-based design of an agonist, antagonist or inhibitor of the polypeptide, by: (a) determining in the first instance the three-dimensional structure of the polypeptide; (b) deducing the three-dimensional structure for the likely reactive or binding site(s) of an agonist, antagonist or inhibitor;
(c) synthesing candidate compounds that are predicted to bind to or react with the deduced binding or reactive site; and (d) testing whether the candidate compounds are indeed agomsts, antagonists or inhibitors.
Gene therapy may also be employed to effect the endogenous production of CASB7439 polypeptide by the relevant cells in the subject. For an overview of gene therapy, see Chapter 20, Gene Therapy and other Molecular Genetic-based Therapeutic Approaches, (and references cited therein) in Human Molecular Genetics, T Strachan and A P Read, BIOS Scientific Publishers Ltd (1996).
Vaccine preparation is generally described in Pharmaceutical Biotechnology, Vol.61 Vaccine Design - the subunit and adjuvant approach, edited by Powell and
Newman, Plenum Press, 1995. New Trends and Developments in Vaccines, edited by Voller et al., University Park Press, Baltimore, Maryland, U.S.A. 1978.
Encapsulation within liposomes is described, for example, by Fullerton, U.S.
Patent 4,235,877. Conjugation of proteins to macromolecules is disclosed, for example, by Likhite, U.S. Patent 4,372,945 and by Armor et al, U.S. Patent
4,474,757.
The amount of protein in each vaccine dose is selected as an amount which induces an immunoprotective response without significant, adverse side effects in typical vaccinees. Such amount will vary depending upon which specific immunogen is employed. Generally, it is expected that each dose will comprise l-1000μg of protein, preferably 2- 1 OOμg, most preferably 4-40μg. An optimal amount for a particular vaccine can be ascertained by standard studies involving observation of antibody titres and other responses in subjects. Following an initial vaccination, subjects may receive a boost in about 4 weeks.
"Isolated" means altered "by the hand of man" from the natural state. If an "isolated" composition or substance occurs in nature, it has been changed or removed from its original environment, or both. For example, a polynucleotide or a polypeptide naturally present in a living animal is not "isolated," but the same polynucleotide or polypeptide separated from the coexisting materials of its natural state is "isolated", as the term is employed herein.
"Polynucleotide" generally refers to any polyribonucleotide or polydeoxribonucleotide, which may be unmodified RNA or DNA or modified RNA or DNA including single and double stranded regions.
"Variant" refers to a polynucleotide or polypeptide that differs from a reference polynucleotide or polypeptide, but retains essential properties. A typical variant of a polynucleotide differs in nucleotide sequence from another, reference polynucleotide. Changes in the nucleotide sequence of the variant may or may not alter the amino acid sequence of a polypeptide encoded by the reference polynucleotide. Nucleotide changes may result in amino acid substitutions, additions, deletions, fusions and truncations in the polypeptide encoded by the reference sequence, as discussed below. A typical variant of a polypeptide differs in amino acid sequence from another, reference polypeptide. Generally, differences are limited so that the sequences of the reference polypeptide and the variant are closely similar overall and, in many regions, identical. A variant and reference polypeptide may differ in amino acid sequence by one or more substitutions, additions, deletions in any combination. A substituted or inserted amino acid residue may or may not be one encoded by the genetic code. A variant of a polynucleotide or polypeptide may be a naturally occurring such as an allelic variant, or it may be a variant that is not known to occur naturally. Non-naturally occurring variants of polynucleotides and polypeptides may be made by mutagenesis techniques or by direct synthesis.
"Identity," as known in the art, is a relationship between two or more polypeptide sequences or two or more polynucleotide sequences, as determined by comparing the sequences. In the art, "identity" also means the degree of sequence relatedness between polypeptide or polynucleotide sequences, as the case may be, as determined by the match between strings of such sequences. "Identity" and "similarity" can be readily calculated by known methods, including but not limited to those described in (Computational Molecular Biology, Lesk, A.M., ed., Oxford University Press, New York, 1988; Biocomputing: liiformatics and Genome Projects, Smith, D.W., ed., Academic Press, New York, 1993; Computer Analysis of Sequence Data, Part I, Griffin, A.M., and Griffin, H.G., eds., Humana Press, New Jersey, 1994; Sequence Analysis in Molecular Biology, von Heinje, G., Academic Press, 1987; and Sequence Analysis Primer, Gribskov, M. and Devereux, J., eds., M Stockton Press, New York, 1991; and Carillo, H., and Lipman, D., SIAM J. Applied Math., 48: 1073 (1988). Prefened methods to determine identity are designed to give the largest match between the sequences tested. Methods to determine identity and similarity are codified in publicly available computer programs. Prefened computer program methods to determine identity and similarity between two sequences include, but are not limited to, the GCG program package (Devereux, J., et al., Nucleic Acids Research 12(1): 387 (1984)), BLASTP, BLASTN, and FASTA (Atschul, S.F. et al., J. Molec. Biol. 215: 403-410 (1990). The BLAST X program is publicly available from NCBI and other sources (BLAST Manual, Altschul, S., et al, NCBI NLM NEH Bethesda, MD 20894; Altschul, S., et al, J. Mol. Biol. 215: 403-410 (1990). The well known Smith Waterman algorithm may also be used to determine identity.
The prefened algorithm used is FASTA. The prefened parameters for polypeptide or polynuleotide sequence comparison using this algorithm include the following:
Gap Penalty: 12
Gap extension penalty: 4
Word size: 2, max 6
Prefened parameters for polypeptide sequence comparison with other methods include the following:
1) Algorithm: Needleman and Wunsch, J. Mol Biol. 48: 443-453 (1970)
Comparison matrix: BLOSSUM62 from Hentikoff and Hentikoff, Proc. Natl. Acad. Sci. USA. 89:10915-10919 (1992)
Gap Penalty: 12
Gap Length Penalty: 4 A program useful with these parameters is publicly available as the "gap" program from Genetics Computer Group, Madison WI. The aforementioned parameters are the default parameters for polypeptide comparisons (along with no penalty for end gaps).
Prefened parameters for polynucleotide comparison include the following: 1) Algorithm: Needleman and Wunsch, J. Mol Biol. 48: 443-453 (1970) Comparison matrix: matches = +10, mismatch = 0 Gap Penalty: 50 Gap Length Penalty: 3
A program useful with these parameters is publicly available as the "gap" program from Genetics Computer Group, Madison WI. The aforementioned parameters are the default parameters for polynucleotide comparisons.
By way of example, a polynucleotide sequence of the present invention may be identical to the reference sequence of SEQ TD NO: 1 or SEQ ED NO:4, that is be 100% identical, or it may include up to a certain integer number of nucleotide alterations as compared to the reference sequence. Such alterations are selected from the group consisting of at least one nucleotide deletion, substitution, including transition and transversion, or insertion, and wherein said alterations may occur at the 5' or 3' terminal positions of the reference nucleotide sequence or anywhere between those terminal positions, interspersed either individually among the nucleotides in the reference sequence or in one or more contiguous groups within the reference sequence. The number of nucleotide alterations is determined by multiplying the total number of nucleotides in SEQ TD NO:l or SEQ ED NO:4 by the numerical percent of the respective percent identity(divided by 100) and subtracting that product from said total number of nucleotides in SEQ TD NO:l or SEQ LD NO:4, or: ≤ xn - (xn » y), wherein nn is the number of nucleotide alterations, xn is the total number of nucleotides in SEQ TD NO:l or SEQ ED NO:4, and y is, for instance, 0.70 for 70%, 0.80 for 80%, 0.85 for 85%, 0.90 for 90%, 0.95 for 95%,etc, and wherein any non-integer product of xn and y is rounded down to the nearest integer prior to subtracting it from xn. Alterations of a polynucleotide sequence encoding the polypeptide of SEQ TD NO:2 or SEQ ED NO:3 or SEQ ED NO:8 may create nonsense, missense or frameshift mutations in this coding sequence and thereby alter the polypeptide encoded by the polynucleotide following such alterations.
Similarly, a polypeptide sequence of the present invention may be identical to the reference sequence of SEQ ID NO:2 or SEQ ED NO:3 or SEQ ED NO:8, that is be 100% identical, or it may include up to a certain integer number of amino acid alterations as compared to the reference sequence such that the % identity is less than 100%. Such alterations are selected from the group consisting of at least one amino acid deletion, substitution, including conservative and non-conservative substitution, or insertion, and wherein said alterations may occur at the amino- or carboxy-terminal positions of the reference polypeptide sequence or anywhere between those terminal positions, interspersed either individually among the amino acids in the reference sequence or in one or more contiguous groups within the reference sequence. The number of amino acid alterations for a given % identity is determined by multiplying the total number of amino acids in SEQ ED NO:2 or SEQ ED NO:3 or SEQ ED NO:8 by the numerical percent of the respective percent identity(divided by 100) and then subtracting that product from said total number of amino acids in SEQ ED NO:2 or SEQ ED NO:3 or SEQ ED NO:8, or: na<xa - (xa » y), wherein na is the number of amino acid alterations, xa is the total number of amino acids in SEQ TD NO:2 or SEQ ED NO:3 or SEQ ED NO:8, and y is, for instance 0.70 for 70%, 0.80 for 80%, 0.85 for 85% etc., and wherein any non- integer product of xa and y is rounded down to the nearest integer prior to subtracting it from xa.
"Homolog" is a generic term used in the art to indicate a polynucleotide or polypeptide sequence possessing a high degree of sequence relatedness to a subject sequence. Such relatedness may be quntified by determining the degree of identity and or similarity between the sequences being compared as hereinbefore described. Falling within this generic term are the terms "ortholog", meaning a polynucleotide or polypeptide that is the functional equivalent of a polynucleotide or polypeptide in another species and "paralog" meaning a functionally similar sequence when considered within the same species. The invention will be further illustrated with reference to the following, non-limiting, examples.
EXAMPLES
Example 1
Real-time RT-PCR analysis
Real-time RT-PCR (U. Gibson. 1996. Genome Research: 6,996) is used to compare mRNA transcript abundance of the candidate antigen in a panel of tumour and matched normal tissues of breast from multiple patient samples. Ln addition, mRNA levels of the candidate antigen in different normal and tumour cell lines of breast, in a panel of normal tissues, and finally in breast tumours grown in vivo in SCED mice are also evaluated by this approach. This analysis is important to establish the breast tumour specificity of CASB7439 expression, which is an important criterion a good antigen candidate must fulfil.
cDNA was derived from prostate, ovarian, colon and breast tumour cell lines as well as breast tumours, normal breast and other normal tissues. Human tumour explants from breast cancer patients (~lmm3) are implanted subcutaneously in SCID mice. Time-release hormone pellets (β-estradiol) are co-implanted subcutaneously. When tumours reach -5% of the total mouse body weight they are excised and a portion (~lmm3) is passaged into another set of SCED mice. This process can be repeated indefinitely. SCED-derived tumour specimens are designated by the passage number followed by 'p'. Real time PCR analysis was performed on matched pairs (early passage/late passage) of SCED-derived human tumours Total RNA is extracted from snap frozen biopsie or cell lines using TriPure reagent (Boehringer). Poly-A+ mRNA is purified from total RNA after DNAase treatment using oligo-dT magnetic beads (Dynal). Quantification of the mRNA is performed by spectrofluorimetry (VersaFluor, BioRad) using Sybrll dye (Molecular Probes).
Primers for real-time PCR amplification are designed with the Perkin-Elmer Primer Express software using default options for TaqMan amplification conditions. Real time PCR was performed with CASB7439 specific probes and the expression levels in the tissues determined : 2 distinct TaqMan primer pairs are designed (one pair in CASB7439 3' UTR region and one pair in CASB7439 ORF region) and used in subsequent real-time RT-PCR analysis.
Real-time reactions are assembled according to standard PCR protocols using 2 ng of reverse transcribed mRNA (Expand RT, Roche) for each reaction. Sybrl dye (Molecular Probes) is added at a final dilution of 1/75000 for real-time detection. Amplification (40 cycles) and real-time detection is performed in a Perkin-Elmer Biosystems PE7700 system using conventional instrument settings. Ct values are calculated using the PE7700 Sequence Detector software, and results are standardised with respect to Actin.
Figure 1 demonstrates while almost all colon cell lines expressed CASB7439 so did the breast tumour cell lines MCF-7, MDA-MB415, BT474 and T47D. These results on a panel of cell lines suggest that, in addition to colon cancer, CASB7439 transcript is also surprisingly over-expressed in breast tumour cell lines, as compared to other normal tissues. Expression is also seen in pooled breast tumour tissue, SCED-derived breast tumour 9987 (both passages) and 9077 (late passage only), as shown in figures 2 and 3. Primers derived from the CASB7439 ORF as well as the 3' end yielded similar profiles. Figure 4 confirms breast tumour specificity of CASB7439 expression on an extended panel of breast tumours and normal tissues. Taken together, these real-time RT-PCR results cleary indicates CASB7439 is over-expressed in an majority of breast tumours, as compared to normal tissues. Example 2
Differential Screening of cDNA arrays- Identification of tumour-associated genes in the subtracted cDNA library is accomplished by differential screening.
Total bacterial DNA is extracted from 100 μl over-night cultures. Bacteria are lysed with guanidium isothiocyanate and the bacterial DNA is affinity purified using magnetic glass (Boehringer). Plasmid inserts are recovered from the bacterial DNA by Advantage PCR amplification (Clontech). The PCR products are dotted onto two nylon membranes to produce high density cDNA anays using the Biomek 96 HDRT tool (Beekman). The spotted cDNA is covalently linked to the membrane by UV irradiation. The first membrane is hybridised with a mixed cDNA probe prepared from the tumour of a single patient. The second membrane is hybridised with an equivalent amount of mixed cDNA probe prepared from normal breast of the same patient. The probe cDNA is prepared by PCR amplification as described above and is labelled using the AlkPhos Direct System (Amersham). Hybridisation conditions and stringency washes are as described in the AlkPhos Direct kit. Hybridized probe is detected by chemiluminescence. Hybridisation intensities for each cDNA fragment on both blots are measured by film densitometry or direct measurement (BioRad Fluor-S Max). The ratio of the tumour to normal hybridisation intensities (T/N) is calculated for each gene to evaluate the degree of over-expression in the tumour. Genes which are significantly over-expressed in breast tumours are followed-up. Significance is arbitrarily defined as one standard deviation of the T/N frequency distribution. Differential screening experiments are repeated using RNA from multiple patient donors (>18) to estimate the frequency of over-expressing tumours in the patient population. In addition, the DNA anays are hybridised with mixed cDNA probes from normal tissues other than breast (see list above) to determine the level of expression of the candidate gene in these tissues.
Example 3
DNA microarrays
DNA micro-anays are used to examine mRNA expression profiles of large collections of genes in multiple samples. This information is used to complement the data obtained by real-time PCR and provides an independent measure of gene expression levels in tumours and normal tissues.
Examples of cunent technologies for production of DNA micro-arrays include 1) The Affymetrix "GeneChip" anays in which oligonucleotides are synthetized on the surface of the chip by solid phase chemical synthesis using a photolithographic process 2) DNA spotting technology in which small volumes of a DNA solution are robotically deposited and then immobilized onto the surface of a solid phase (e.g. glass). In both instances, the chips are hybridized with cDNA or cRNA which has been extracted from the tissue of interest (e.g. normal tissue, tumour etc...) and labeled with radioactivity or with a fluorescent reporter molecule. The labeled material is hybridized to the chip and the amount of probe bound to each sequence on the chip is determined using a specialized scanner. The experiment can be set-up with a single fluorescent reporter (or radioactivity) or, alternatively, can be performed using two fluorescent reporters. Ln this latter case, each of the two samples is labeled with one of the reporter molecules. The two labeled samples are then hybridized competitively to the sequences on the DNA chip. The ratio of the two fluorescent signals is determined for each sequence on the chip. This ratio is used to calculate the relative abundance of the transcript in the two samples. Detailed protocols are available from a number of sources including "DNA Microanays: A practical approach. Schena M. Oxford University Press 1999" and the World Wide Web
(http://cmgm.stanford.edu/pbrown/protocols/index.html), http://anayit.com/DNA- Microanay-Protocols/) and specialized distributors (e.g. Affymetrix).
Example 4
Northern-Southern blot analysis
Limited amounts of mixed tumour and matched normal breast cDNA are amplified by Advantage PCR (see above). Messenger RNA from multiple normal tissues is also amplified using the same procedure. The amplified cDNA (1 μg) is electrophoresed on a 1.2% agarose gel and transferred onto a nylon membrane. The membrane is hybridised (AlkPhos Direct System) with a probe prepared using a fragment of the candidate TAA cDNA. Northern-Southern analysis provides information on transcript size, presence of splice variants and transcript abundance in tumour and normal tissues.
Example 5 Northern blot analysis
Northern blots are produced according to standard protocols using 1 μg of poly A+ mRNA. Radioactive probes are prepared using the Ready-to-Go system (Pharmacia).
Example 6 :
6.1 Expression and purification of tumour-specific antigens
6.1.1 Expression of the present invention antigen in microbial hosts.
Expression in microbial hosts, or alternatively in vitro transcription/translation, is used to produce the antigen of the invention for vaccine purposes and to produce protein fragments or whole protein for rapid purification and generation of antibodies needed for characterization of the naturally expressed protein by western blot, immunohistochemistry or for follow-up of purification.
Recombinant proteins may be expressed in two microbial hosts, E. coli and in yeast (such as Saccharomyces cerevisiae or Pichia pastoris). This allows the selection of the expression system with the best features for this particular antigen production. In general, the recombinant antigen will be expressed in E. coli and the reagent protein expressed in yeast.
The expression strategy first involves the design of the primary structure of the recombinant antigen. In general an expression fusion partner (EFP) is placed at the N terminal extremity to improve levels of expression that could also include a region useful for modulating the immunogenic properties of the antigen, an immune fusion partner (EFP). In addition, an affinity fusion partner (AFP) useful for facilitating further purification is included at the C-terminal end. Moreover, polynucleotide sequence of the coding region of the antigen can also be modified to optimise its expression. Using a codon usage that is adapted to the chosen recombinant expression host can dramatically increase antigen expression yields.
A comparative evaluation of the different versions of the expressed antigen will allow the selection of the most promising candidate that is to be used for further purification and immunological evaluation.
Several constructs underwent comparative evaluation : For rapid expression and purification as well as generation of antibodies against CASB7439, it is proposed to generate in E. Coli a full length CASB7439 protein with Influenza protein NS 1 as EFP and a histidine tail as AFP. Five constructs have been designed:
Construct 1: Full length wild type CASB7439 cDNA in fusion with the N terminal fragment (the first 80 amino acids) of Influenza protein NSl coding cDNA as EFP and with a histidine tail coding cDNA as an AFP (SEQ ED NO:5). The encoded fusion protein sequence is SEQ ED NO:7. The CASB7439 protein design is shown in Figure 5.
Contruct 2 : Full length mutated CASB7439 cDNA in fusion with the N terminal fragment of Influenza protein NS 1 coding cDNA as EFP and with a histidine tail coding cDNA as an AFP (SEQ ED NO:6). This construct was designed in order to comprise the first 50 codons of native CASB7439 cDNA replaced by codons specific of the E. coli codon usage, to enhance expression potential of CASB7439 in its E. coli host. Only CASB7439 codons that are known to be problematic in E coli host by the man skilled in the art were replaced by an E. Coli codon. For each CASB7439 problematic codon, the E coli codon is chosen to encode for the same aminoacid and to have a low GC content. The encoded fusion protein sequence is SEQ ID NO:7.
Contruct 3 : Full length wild type CASB7439 cDNA in fusion with a ll amino acids T7*Tag® (Novagen, Madison, WI) coding cDNA as EFP and with a histidine tail coding cDNA as an AFP (SEQ ED NO: 13). The encoded fusion protein is SEQ ED NO: 14.
Construct 4 : Full length mutated CASB7439 cDNA in fusion with a 11 amino acids T7»Tag® (Novagen, Madison, WI) coding cDNA as EFP and with a histidine tail coding cDNA as an AFP (SEQ ID NO:34). The first 50 codons of native CASB7439 cDNA replaced by codons specific of the E. coli codon usage.The same substitutions as those performed in construct 2 have been carried out in contract 4. The encoded fusion protein sequence is SEQ ED NO: 14.
Construct 5 : Full length mutated CASB7439 cDNA is fusion with a l l amino acids T7»Tag® (Novagen, Madison, WTjcoding cDNA as EFP and a histine tail coding cDNA as AFP (SEQ ED NO: 15). This construct also has the first 50 codons of native CASB7439 cDNA replaced by codons specific of the E. coli codon usage. All CASB7439 codons are modified in this construct to have specific E Coli codons and to respect E. Coli codon frequency distribution. The encoded fusion protein sequence is SEQ TD NO: 14.
Tables of E Coli codon usage used for codon optimisation for the above- mentionned constructs is given in "Codon usage in regulatory genes in Ε Coli does nor reflect selcetion for rare codons", Sharp P. M., Wen-Hsiung L., Nucleic Acids Research, 14, 7737-7749 (1986).
Constructs 1 and 2 are cloned in pMGl plasmid. Plamid pMGl is a derivative of pMG27N. Plasmid pMG27N is a pBR322-derived expression vector which contains the PL promoter, an N utilization site (to relieve transcriptional polarity in the presence of N protein) and the ell ribisome binding site including the ell translation initiation codon incorporated in an Nde I restriction site (Gross et al. 1985, Mol.&Cell.Biol. 5:1015). Plasmid pMGl has been constructed by inserting the 81 first codons of the NSl coding region from influenza strain A/PR/8/34 cleaved from plasmid pASlΔΕH/801 (Young et al. 1983. Proc.Natl.Acad.Sci.USA 80:6105) by BamH I and Nco I into pMG27N digested by BamH I and Sac I. A synthetic DNA linker was introduced between the Nco I and the Sac I sites. A schematic represention of pMGl construction is shown in Figure 6. Construct 3 to 5 are cloned in a commercial pET24b(+) plamid from Novagen (Madison, WI).
Constructs 1 to 5 are transformed in the AR58 E. coli strain, which is a cryptic λ lysogen derived from N99 that is gal E::Tn 10,A-8(chlD-pgl),A-Hl(cro-chlA),Ni', and cI857 (Proc.Natl.Acad.Sci.USA vol82, pp.88-92, January 1985 Biochemistry).
When the recombinant AR58 strains are available, the recombinant product is characterized by the evaluation of the level of expression and the prediction of further solubility of the protein by analysis of the behavior in the crude extract. After growth on appropriate culture medium and induction of the recombinant protein expression, total extracts are analyzed by SDS-PAGE. The recombinant proteins are visualised in stained gels and identified by Western blot analysis using specific antibodies.
Expression of the recombinant protein from constructs 1 to 5 :
Bacteria was grown in LB medium + 50 μg/ml Kan at 30°C. When the culture reached OD = 0.5 (620nm), the culture was heated up to 39°C, after 5 hours of induction by ITPG, cells were harvested. The cell extract was prepared as follows: cells were resuspended in PBS buffer, disrupted (French press, 3 times), and centrifuged 30 min at 14000t. More than 90% of the protein was found in the supernatant of the cell extract.
Purification of the recombinant protein from constructs 1 to 5.
The purification scheme follows a classical approach based on the presence of an His affinity tail in the recombinant protein. In a typical experiment the disrupted cells are filtered and the acellular extracts loaded onto an Ion Metal Affinity Chromatography (EMAC; Ni'H TA from Qiagen) that will specifically retain the recombinant protein. The retained proteins are eluted by 0-500 mM imidazole gradient (possibly in presence of a detergent) in a phosphate buffer.
The supernatant from the harvested culture was denatured in 6M urea,100mM NaH2PO4, lOmM Tris, pH 8, and loaded on a chromatographic column EMAC Qiagen NTA Ni"1-1" under the following conditions :
Equilibration buffer :NaH2Po4 lOOmM (PH 8) Tris lOmM Urea6M.
Sample : supernatant in urea 6M, 100mM NaH2Po4, lOmMTris Wash buffers : 1) NaH2PO4 lOOmM PH 8Tris lOmM Urea 6 M Imidazole 25mM
2) NaH2Po4 lOOmM (PH8) Tris lOmM Urea 6mM Imidazole 50mM Elution buffer : NaH2PO4 lOOmM (PH 5,5) Tris lOmM Urea 6M Imidazole 500mM
The eluted protein in 500mM imidazole + 6M urea is dialysed under the following conditions :
1. PBS PH 7,2 + sarkosyl 0.5% + 4M Urea
2. idem at 2M Urea 2hrs 3. idem at OM Urea 2hrs
Recombinant expression in microbial hosts results :
Construct 1 (NS1-CASB7439-HIS fusion protein) :
The unpurified cell extracts is run on a 12.5% SDS PAGE, and subsequently stained with Coomassie blue. A Western blot is also performed using a monoclonal antibody directed against the 80 aminoacids N terminal fragment of the NSl protein in fusion with CASB7439 construct 1. The resulting gels (figures 7 and 8) show that the protein is expressed and visible in the cell extract supernatant.
The final purified material is freezed and stored. The protein content was quantified using a Lowry protein assay (0,9 mg/1.2 ml). The purity was assessed by a 12.5% PAGE SDS stained with Coomassie blue (figure 9), and the presence of the recombinant protein was checked by Western blot, using a monoclonal antibody directed against the 80 aminoacids N terminal fragment of the NSl protein in fusion with CASB7439 construct 1 (figure 10).
Constructs 3 to 5 (T7 Tag - CASB7439 - HIS fusion protein) :
Several dilutions of 15 μl of purified material for each construction is run on a 12.5% PAGE SDS gel. Presence and semi-quantification of recombinant protein is done by Western blot using SB600 anti-peptide antibody (see section 6.2.2 below for antibody description). Results, shown in figure 11, clearly indicate CASB7439 contracts that have optimized codons for their E. Coli host (contracts 3 and 4) yield higher quantities of recombinant protein. Using modified version of CASB7439 cDNAs therefore increases CASB7439 recombinant expression yields.
6.1.2 Expression of the present invention antigen in vaccination vectors
The immunogenicity of the antigen of the present invention has been verified by immunising rabbits and mice using various means of immunisation. Indeed, immunisation with CASB7439 forms, either peptide or recombinant protein could induce humoral immune response with the generation of specific antibodies against CASB7439 and/or could induce a CASB7439 specific cellular immune response. Additionally, in vivo delivery of CASB7439 protein using for instance, naked DNA in an appropriate vector encoding CASB7439 or fragments of CASB7439, CASB7439 gene delivered by a viral vector encoding CASB7439 or fragments of CASB7439, is equally appropriate to demonstrate CASB7439 immunogenicity.
Recombinant expression of CASB7439 in DNA vaccination vectors
Several vectors can be used to construct the vaccinating plasmid (for instance, pcDNA3.1 vector, pJB16 vector). The plasmid expression vector for CASB7439 (ρJB76) was constructed through the recombination of ρDOR207/CASB7439 with the destination vector pJB 16 (pVRl012-Dest) by the LR reaction of the Gateway Cloning Technology. HEK293T cells were transfected in 6-well plate with pJB76 DNA (0.25 or 1 μg), or empty vector (pJB16; lug). Cells were harvested at 48 hours post-infection and cell lysates were analyzed by Western blot for expression of CASB7439 using rabbit anti-NSl-CASB7439-His polyclonal antibody (dilution 1 :500, see section 6.2.1 for antibody description). As shown in figure 12, a band at the size expected for CASB7439 is observed at low level in fibroblasts transfected with 1 ug of CASB7439 DNA (ρJB76). No band is observed using with empty vector (pJB16).
Recombinant expression of CASB7439 in a adenoviral live vector
Recombinant adenoviruses are effective vectors for gene-based vaccination because they are capable of eliciting humoral and cellular immune responses against the encoded antigen. Recombining CASB7439 by such an approach is recommended for further immunological validations. Recombinant adenovirus for CASB7439 (AdC7439) was constructed using a Lac/GW-1 expression cassette under control of the CMV promoter.
A schematic representation of the recombinant expression vector is shown in Figure 13. In brief, 2 x 105 human fibroblasts from donor D93 were infected by the recombinant adenoviras for CASB7439 (AdC7439) in 6-well plate at various MOL Cells were harvested at 48 hours post-infection and cell lysates were analyzed by Western blot for expression of CASB7439 using rabbit anti-NSl- CASB7439-His polyclonal antibody (1:500). As shown in figure 14, A band at the size expected for CASB7439 is observed at low level in fibroblasts infected at an MOI of 200 and 300 and at high level at MOI of 400 or 500. No expression is seen in uninfected fibroblasts.
6.2 Antibody production and immunohistochemistry
Small amounts of relatively purified protein can be used to generate immunological tools in order to a) detect the expression by immunohistochemistry in normal or cancer tissue sections; b) detect the expression, and to follow the protein during the purification process (ELISA/ Western Blot); or c) characterise/ quantify the purified protein (ELISA).
6.2.1 Polyclonal antibodies: Immunisation Rabbits are immunised, intramuscularly (I.M.), 3 times at 3 weeks intervals with lOOμg of protein, formulated in the adjuvant 3D-MPL/QS21. Three weeks after each immunisation a blood sample is taken and the antibody titer estimated in the serum by ELISA using the protein as coating antigen following a standard protocol.
ELISA
96 well microplates (maxisorb Nunc) are coated with 5 μg of protein overnight at 4°C. After lhour saturation at 37°C with PBS NCS 1%, serial dilution of the rabbit sera is added for 1H 30 at 37°C (starting at 1/10). After 3 washings in PBS Tween, anti rabbit biotinylated anti serum (Amersham ) is added (1/5000). Plates are washed and peroxydase coupled streptavidin (1/5000) is added for 30 min at 37°C. After washing, 50μl TMB (BioRad) is added for 7 min and the reaction then stopped with H2SO4 0.2M. The OD can be measured at 450 nm and midpoint dilutions calculated by SoftmaxPro.
6.2.2 Anti-peptide antibodies. Immunisation
Rabbits are immunised with two synthetic peptides designed from CASB7439 aminoacid sequence SEQ ED NO:2. Chosen peptide are CASB7439 aminoacid positions 1 to 14 (SB595) and 157 to 172 (SB600). Synthetic peptides are then conjugated to a canier protein (KLH). Conjugates are formulated with Freund's adjuvant, and two rabbits are immunised with each of the conjugates. Four weeks after the second immunisation and four weeks after the third immunisation, a blood sample is taken and analysed.
ELISA
Anti-CASB7439 antibody titers are estimated in the serum by ELISA following standard protocols as described above. 6.2.3 Monoclonal antibodies: Immunisation
5 BALB/c mice are immunized 3 times at 3 week intervals with 5 μg of purified protein. Bleedings are performed 14 days post II and 1 week post 3. The sera are tested by Elisa on purified protein used as coated antigen. Based on these results (midpoint dilution >10000) one mouse is selected for fusion.
Fusion/ HATselection Spleen cells are fused with the SP2/0 myeloma according to a standard protocol using PEG 40% and DMSO 5%. Cells are then seeded in 96 well plates 2.5 xlO4 - 105 cells/well and resistant clones will be selected in HAT medium. The supernatant of these hybridomas will be tested for their content in specific antibodies and when positive, will be submitted to 2 cycles of limited dilution. After 2 rounds of screening, 3 hybridomas will be chosen for ascitis production.
6.2.3 frnmunohistochemistrv
When antibodies are available, immuno staining is performed on normal or cancer tissue sections, in order to determine : 0 the level of expression of the antigen of the invention in cancer relative to normal tissue or
0 the proportion of cancer of a certain type expressing the antigen
0 if other cancer types also express the antigen
0 the proportion of cells expressing the antigen in a cancer tissue
Tissue sample preparation
After dissection, the tissue sample is mounted on a cork disk in OCT compound and rapidly frozen in isopentane previously super cooled in liquid nitrogen (- 160°C). The block will then be conserved at -70°C until use. 7- 1 Oμm sections will be realised in a cryostat chamber (-20, -30°C).
Staining Tissue sections are dried for 5 min at room Temperature (RT), fixed in acetone for lOmin at RT, dried again, and saturated with PBS 0.5% BSA 5% serum. After 30 min at RT either a direct or indirect staining is performed using antigen specific antibodies. A direct staining leads to a better specificity but a less intense staining whilst an indirect staining leads to a more intense but less specific staining.
6.3 Analysis of human cellular immune responses to the antigen of the invention
The immunological relevance of the antigen of the invention can be assessed by in vitro priming of human T cells. All T cell lymphocyte lines and dendritic cells are derived from PBMCs (peripheral blood mononuclear cells) of healthy donors (prefened HLA-A2 subtype). An HLA-A2.1/Kb transgenic mouse model is also used for screening of HLA-A2.1 peptides.
CD8+ T-cell response
Newly discovered antigen-specific CD8+ T cell lines are raised and maintained by weekly in vitro stimulation. The lytic activity and the γ-EFN production of the CD8+ lines in response to the antigen or antigen derived-peptides is tested using standard procedures.
Two strategies to raise the CD8+ T cell lines are used: a peptide-based approach and a whole gene-based approach. Both approaches require the full-length cDNA of the newly discovered antigen in the conect reading frame to be either cloned in an appropriate delivery system or to be used to predict the sequence of HLA binding peptides.
Peptide-based approach
Briefly, transgenic mice are immunized with adjuvanted HLA-A2 peptides, those unable to induce a CD8+ response (as defined by an efficient lysis of peptide- pulsed autologous spleen cells) will be further analyzed in the human system. Human dendritic cells (cultured according to Romani et al.) will be pulsed with peptides and used to stimulate CD8+ -sorted T cells (by Facs). After several weekly stimulations, the CD8+ lines will be first tested on peptide-pulsed autologous BLCL (EBV-B transformed cell lines). To verify the proper in vivo processing of the peptide, the CD8+ lines will be tested on cDNA-transfected tumour cells (HLA-A2 transfected LnCaP, Skov3 or CAMA tumour cells).
Whole gene-based approach
CD8+ T cell lines will be primed and stimulated with either gene-gun transfected dendritic cells, retrovirally transduced B7.1 -transfected fibroblasts, recombinant pox virus or adenoviras infected dendritic cells. Virus infected cells are very efficient to present antigenic peptides since the antigen is expressed at high level but can only be used once to avoid the over-growth of viral T cells lines.
After alternated stimulations, the CD8+ lines are tested on cDNA-transfected tumour cells as indicated above. Peptide specificity and identity is determined to confirm the immunological validation.
CD4+ T-cell response
Similarly, the CD4+ T-cell immune response can also be assessed. Generation of specific CD4+ T-cells is made using dendritic cells loaded with recombinant purified protein or peptides to stimulate the T-cells.
Predicted epitopes (nonamers and decanters) binding HLA alleles :
The HLA Class I binding peptide sequences are predicted either by the Parker's algorithm (Parker, K. C, M. A. Bednarek, and J. E. Coligan. 1994. Scheme for ranking potential HLA-A2 binding peptides based on independent binding of individual peptide side-chains. J. Immunol. 152:163 and http:/^imas.dcrt.r-ih.gov/molbio/hla_bind/) or the Rammensee method (Rammensee, Friede, Stevanovic, MHC ligands and peptide motifs: 1st listing, Immunogenetics 41, 178-228, 1995 ; Rammensee, Bachmann, Stevanovic: MHC ligands and peptide motifs. Landes Bioscience 1997, and http://134.2.96.221/scripts/hlaserver.dll/home.htm). Peptides are then screened in the HLA-A2.1/Kb transgenic mice model (Nitiello et al.). The HLA Class II binding peptide sequences are predicted using the Tepitope algorithm, with a score cut-offset to 6 (Sturniolo, Hammer at al., Nature Biotechnology. 1999. 17;555-561).
The following tables gather the Class I and II predicted epitope sequences :
Figure imgf000066_0001
° : Estimate of Half Time of Disassociation of a Molecule Containing This Subsequence.
Figure imgf000066_0002
° : Estimate of Half Time of Disassociation of a Molecule Containing This Subsequence.
Figure imgf000066_0003
° : Estimate of Half Time of Disassociation of a Molecule Containing This Subsequence.
Figure imgf000066_0004
° : Estimate of Half Time of Disassociation of a Molecule Containing This Subsequence.
Figure imgf000067_0002
° : Estimate of Half Time of Disassociation of a Molecule Containing This Subsequence.
Figure imgf000067_0003
° : Estimate of Half Time of Disassociation of a Molecule Containing This Subsequence.
Figure imgf000067_0001
120 LRPQAVRPS 6.2 SEQ NO:26
Figure imgf000068_0002
HLA-DRB1*1106 : nonamers
Start Subsequence Residue Tepitope Score
Rank Position SEQ ID Listing
99 IRALQRLLA 5.8 SE ED NO:25
Figure imgf000068_0003
Figure imgf000068_0001
Figure imgf000068_0004
Figure imgf000068_0005
Figure imgf000068_0006
| 3 j 65 | LVNLGFQAL 4.5 |SEQEDNO:31 j
Figure imgf000069_0001
SEQUENCEINFORMATION
SEQIDNO:l
GTACCTTGCTTTGGGGGCGCACTAAGTACCTGCCGGGAGCAGGGGGCGCACCGGGAACTCGCAGAT TTCGCCAGTTGGGCGCACTGGGGATCTGTGGACTGCGTCCGGGGGATGGGCTAGGGGGACATGCGC ACGCTTTGGGCCTTACAGAATGTGATCGCGCGAGGGGGAGGGCGAAGCGTGGCGGGAGGGCGAGGC GAAGGAAGGAGGGCGTGAGAAAGGCGACGGCGGCGGCGCGGAGGAGGGTTATCTATACATTTAAAA ACCAGCCGCCTGCGCCGCGCCTGCGGAGACCTGGGAGAGTCCGGCCGCACGCGCGGGACACGAGCG TCCCACGCTCCCTGGCGCGTACGGCCTGCCACCACTAGGCCTCCTATCCCCGGGCTCCAGACGACC TAGGACGCGTGCCCTGGGGAGTTGCCTGGCGGCGCCGTGCCAGAAGCCCCCTTGGGGCGCCACAGT TTTCCCCGTCGCCTCCGGTTCCTCTGCCTGCACCTTCCTGCGGCGCGCCGGGACCTGGAGCGGGCG GGTGGATGCAGGCGCGatggacggcggcacactgcccaggtccgcgccccctgcgccccccgtccc tgtcggctgcgctgcccggcggagacccgcgtccccggaactgttgcgctgcagccggcggcggcg accggccaccgcagagaccggaggcggcgcagcggccgtagcgcggcgcaatgagcgcgagcgcaa ccgcgtgaagctggtgaacttgggcttccaggcgctgcggcagcacgtgccgcacggcggcgccag caagaagctgagcaaggtggagacgctgcgctcagccgtggagtacatccgcgcgctgcagcgcct gctggccgagcacgacgccgtgcgcaacgcgctggcgggagggctgaggccgcaggccgtgcggcc gtctgcgccccgcgggccgccagggaccaccccggtcgccgcctcgccctcccgcgcttcttcgtc cccgggccgcgggggcagctcggagcccggctccccgcgttccgcctactcgtcggacgacagcgg ctgcgaaggcgcgctgagtcctgcggagcgcgagctactcgacttctccagctggttagggggcta ctgaGCGCCCTCGACCTATGAGCCTCAGCCCCGGAAGCCGAGCGAGCGGCCGGCGCGCTCATCGCC GGGGAGCCCGCCAGGTGGACCGGCCCGCGCTCCGCCCCCAGCGAGCCGGGGACCCACCCACCACCC CCCGCACCGCCGACGCCGCCTCGTTCGTCCGGCCCAGCCTGACCAATGCCGCGGTGGAAACGGGCT TGGAGCTGGCCCCATAAGGGCTGGCGGCTTCCTCCGACGCCGCCCCTCCCCACAGCTTCTCGACTG CAGTGGGGCGGGGGGCACCAACACTTGGAGATTTTTCCGGAGGGGAGAGGATTTTCTAAGGGCACA GAGAATCCATTTTCTACACATTAACTTGAGCTGCTGGAGGGACACTGCTGGCAAACGGAGACCTAT TTTTGTACAAAGAACCCTTGACCTGGGGCGTAATAAAGATGACCTGGACCCCTGCCCCCACTATCT GGAGTTTTCCATGCTGGCCAAGATCTGGACACGAGCAGTCCCTGAGGGGCGGGGTCCCTGGCGTGA GGCCCCCGTGACAGCCCACCCTGGGGTGGGTTTGTGGGCACTGCTGCTCTGCTAGGGAGAAGCCTG TGTGGGGCACACCTCTTCAAGGGAGCGTGAACTTTATAAATAAATCAGTTCTGTTTAAAAAAAAAA AAAAAAAAA
SEQ ID NO:2 DGGTLPRSAPPAPPVPVGCAARRRPASPEL RCSRRRRPATAETGGGAAAVARRNE ERNRV V NLGFQ- iRQHVPHGGASKiπ-SKVET RSAVEYIRALQR LAEHDAVRNA AGGLRPQAVRPSAPRG PPGTTPVAASPSRASSSPGRGGSSEPGSPRSAYSSDDSGCEGA SPAERELLDFSS LGGY
SEQ ID NO:3 MSAPAARSASGAEAHRSRALSSPLTSWRSRVARAPQDSAR RSRCRPTSRRNAGSRAPSCPRGPGT KRGRARRRPG SLAARGAQTAARPAASA PPARCARRRARPAGAAARGCTPRLSAASPPCSASC RRRAARAAAAPGSPSSPASRGCARAHCAALRPLRRLRSLRWPVAAAGCSATVPGTRVSAGQRSRQG RGAQGART AVCRRPSRLHPPARSRSRRAAGRCRQRNRRRRGK WRPKGASGTAPPGNSPGHAS
SEQID NO:4
TTTTTTTTTTTTTTTTTTTAAACAGAACTGATTTATTTATAAAGTTCACGCTCCCTTGAAGAGGTG TGCCCCACACAGGCTTCTCCCTAGCAGAGCAGCAGTGCCCACAAACCCACCCCAGGGTGGGCTGTC ACGGGGGCCTCACGCCAGGGACCCCGCCCCTCAGGGACTGCTCGTGTCCAGATCTTGGCCAGCATG GAAAACTCCAGATAGTGGGGGCAGGGGTCCAGGTCATCTTTATTACGCCCCAGGTCAAGGGTTCTT TGTACAAAAATAGGTCTCCGTTTGCCAGCAGTGTCCCTCCAGCAGCTCAAGTTAATGTGTAGAAAA TGGATTCTCTGTGCCCTTAGAAAATCCTCTCCCCTCCGGAAAAATCTCCAAGTGTTGGTGCCCCCC GCCCCACTGCAGTCGAGAAGCTGTGGGGAGGGGCGGCGTCGGAGGAAGCCGCCAGCCCTTATGGGG CCAGCTCCAAGCCCGTTTCCACCGCGGCATTGGTCAGGCTGGGCCGGACGAACGAGGCGGCGTCGG CGGTGCGGGGGGTGGTGGGTGGGTCCCCGGCTCGCTGGGGGCGGAGCGCGGGCCGGTCCACCTGGC GGGCTCCCCGGCGATGAGCGCGCCGGCCGCTCGCTCGGCTTCCGGGGCTGAGGCTCATAGGTCGAG GGCGCTCAGTAGCCCCCTAACCAGCTGGAGAAGTCGAGTAGCTCGCGCTCCGCAGGACTCAGCGCG CCTTCGCAGCCGCTGTCGTCCGACGAGTAGGCGGAACGCGGGGAGCCGGGCTCCGAGCTGCCCCCG CGGCCCGGGGACGAAGAAGCGCGGGAGGGCGAGGCGGCGACCGGGGTGGTCCCTGGCGGCCCGCGG GGCGCAGACGGCCGCACGGCCTGCGGCCTCAGCCCTCCCGCCAGCGCGTTGCGCACGGCGTCGTGC TCGGCCAGCAGGCGCTGCAGCGCGCGGATGTACTCCACGGCTGAGCGCAGCGTCTCCACCTTGCTC AGCTTCTTGCTGGCGCCGCCGTGCGGCACGTGCTGCCGCAGCGCCTGGAAGCCCAAGTTCACCAGC TTCACGCGGTTGCGCTCGCGCTCATTGCGCCGCGCTACGGCCGCTGCGCCGCCTCCGGTCTCTGCG GTGGCCGGTCGCCGCCGCCGGCTGCAGCGCAACAGTTCCGGGGACGCGGGTCTCCGCCGGGCAGCG CAGCCGACAGGGACGGGGGGCGCAGGGGGCGCGGACCTGGGCAGTGTGCCGCCGTCCATCGCGCCT GCATCCACCCGCCCGCTCCAGGTCCCGGCGCGCCGCAGGAAGGTGCAGGCAGAGGAACCGGAGGCG ACGGGGAAAACTGTGGCGCCCCAAGGGGGCTTCTGGCACGGCGCCGCCAGGCAACTCCCCAGGGCA CGCGTCCTAGGTCGTCTGGAGCCCGGGGATAGGAGGCCTAGTGGTGGCAGGCCGTACGCGCCAGGG AGCGTGGGACGCTCGTGTCCCGCGCGTGCGGCCGGACTCTCCCAGGTCTCCGCAGGCGCGGCGCAG GCGGCTGGTTTTTAAATGTATAGATAACCCTCCTCCGCGCCGCCGCCGTCGCCTTTCTCACGCCCT CCTTCCTTCGCCTCGCCCTCCCGCCACGCTTCGCCCTCCCCCTCGCGCGATCACATTCTGTAAGGC CCAAAGCGTGCGCATGTCCCCCTAGCCCATCCCCCGGACGCAGTCCACAGATCCCCAGTGCGCCCA ACTGGCGAAATCTGCGAGTTCCCGGTGCGCCCCCTGCTCCCGGCAGGTACTTAGTGCGCCCCCAAA GCAAGGTAC
SEQIDNO: 5
ATGGATCCAAACACTGTGTCAAGCTTTCAGGTAGATTGCTTTCTTTGGCATGTCCGCAAACGAGTT GCAGACCAAGAACTAGGTGATGCCCCATTCCTTGATCGGCTTCGCCGAGATCAGAAATCCCTAAGA GGAAGGGGCAGCACcCTcGGTCTGGACATCGAGACAGCCACACGTGCTGGAAAGCAGATAGtGGAG CGGAttctGAAAGAAGAATCCGATGAGGCACTTAAAATGACCATGGACGGCGGCACACTGCCCAGG TCCGCGCCCCCTGCGCCCCCCGTCCCTGTCGGCTGCGCTGCCCGGCGGAGACCCGCGTCCCCGGAA CTGTTGCGCTGCAGCCGGCGGCGGCGACCGGCCACCGCAGAGACCGGAGGCGGCGCAGCGGCCGTA GCGCGGCGCAATGAGCGCGAGCGCAACCGCGTGAAGCTGGTGAACTTGGGCTTCCAGGCGCTGCGG CAGCACGTGCCGCACGGCGGCGCCAGCAAGAAGCTGAGCAAGGTGGAGACGCTGCGCTCAGCCGTG GAGTACATCCGCGCGCTGCAGCGCCTGCTGGCCGAGCACGACGCCGTGCGCAACGCGCTGGCGGGA GGGCTGAGGCCGCAGGCCGTGCGGCCGTCTGCGCCCCGCGGGCCGCCAGGGACCACCCCGGTCGCC GCCTCGCCCTCCCGCGCTTCTTCGTCCCCGGGCCGCGGGGGCAGCTCGGAGCCCGGCTCCCCGCGT TCCGCCTACTCGTCGGACGACAGCGGCTGCGAAGGCGCGCTGAGTCCTGCGGAGCGCGAGCTACTC GACTTCTCCAGCTGGTTAGGGGGCTACactagtggccaccatcaccatcaccattaa
SEQIDNO.6
ATGGATCCAAACACTGTGTCAAGCTTTCAGGTAGATTGCTTTCTTTGGCATGTCCGCAAACGAGTT GCAGACCAAGAACTAGGTGATGCCCCATTCCTTGATCGGCTTCGCCGAGATCAGAAATCCCTAAGA GGAAGGGGCAGCACCCTCGGTCTGGACATCGAGACAGCCACACGTGCTGGAAAGCAGATAGTGGAG CGGATTCTGAAAGAAGAATCCGATGAGGCACTTAAAATGACCATGGACGGCGGCACCCTGCCGCGT TCCGCGCCGCCGGCGCCGCCAGTTCCGGTTGGCTGCGCTGCCCGTCGCCGTCCCGCGTCCCCGGAA CTGCTGCGCTGCAGCCGTCGCCGTCGCCCGGCCACCGCAGAGACCGGAGGCGGCGCAGCGGCCGTA GCGCGGCGCAATGAGCGCGAGCGCAACCGCGTGAAGCTGGTGAACTTGGGCTTCCAGGCGCTGCGG CAGCACGTGCCGCACGGCGGCGCCAGCAAGAAGCTGAGCAAGGTGGAGACGCTGCGCTCAGCCGTG GAGTACATCCGCGCGCTGCAGCGCCTGCTGGCCGAGCACGACGCCGTGCGCAACGCGCTGGCGGGA GGGCTGAGGCCGCAGGCCGTGCGGCCGTCTGCGCCCCGCGGGCCGCCAGGGACCACCCCGGTCGCC GCCTCGCCCTCCCGCGCTTCTTCGTCCCCGGGCCGCGGGGGCAGCTCGGAGCCCGGCTCCCCGCGT TCCGCCTACTCGTCGGACGACAGCGGCTGCGAAGGCGCGCTGAGTCCTGCGGAGCGCGAGCTACTC GACTTCTCCAGCTGGTTAGGGGGCTACACTAGTGGCCACCATCACCATCACCATTAA
SEQIDNO:7 DPNTVSSFQVBCF WHVRKRVADQELGDAPF DRLRRDQKSLRGRGSTLGLDIETATRAGKQIVE RILKEESDEALKMTMDGGT PRSAPPAPPVPVGCAARRRPASPELLRCSRRRRPATAETGGGAAAV AR- ERERNRV-OiVN GFQ- iRQHVPHGGASKKLS-WETLRSAVEYIRALQR AEHDAV-^ALAG GLRPQAVRPSAPRGPPGTTPVAASPSRASSSPGRGGSSEPGSPRSAYSSDDSGCEGA SPAEREL DFSS GGYTSGHHHHHH
SEQID NO:8
MYSTAERSVST LSFLLAPPCGTCCRSA KPKFTSFTRLRSRSLRRATAAAPPPVSAVAGRRRRLQ RNSSGDAGLRRAAQPTGTGGAGGADLGSVPPSIAPASTRPLQVPARRRKVQAEEPEATGKTVAPQG GF HGAARQLPRARVLGRLEPGDRRPSGGRPYAPGSVGRSCPARAAGLSQVSAGAAQAAGF
SEQ ID NO:9 EAHLD YGVPGLQEASDACPRESCSSALPEAREGA VHFPPHPVPREHFSCAAPE VAGAQG NA SLMDGGA PR MPTS SGVAGACAARRRQAS PE-- RCSRRRRSGATE AS S S S AAVARRNERER RVK VN GFQALRQHVPHGG-^KIOjSK ^TLRSAVEYIRA QR LAEHDAVRAALAGG TPATPPSDE CAQPSASPASASLSCASTSPSPDR GCSEPTSPRSAYSSEESSCEGE SPMEQEL DFSS GGY
SEQ ID NO:10 ESHF WYGVPRLQKASDACPRESCSSALPEAREGANVHFPPHPVPREHFSCGAPKPVAGAPALNA SLMDGGALPRLVPTSSGVAGACTARRRPPSPELLRCSRRRRSGATEASSSSAAVARR ERERNRVK LV LGFQALRQHVPHGGANKKLSKVETLRSAVEYIRA QR AEHDAVRAA SGG LTPATRPSDV CTQPSASPASASLSCTSTSPDR GCSEPASPRSAYSSEDSSCEGETYPMGQMFDFSNWLGGY
SEQID NO.ll
GCCCGGAGCATGGAAGCACGTCAGCTAGGCCATGAACTGCACCCGGGAGGGGTGGGGGTGGAAGCG CACGGTGTCAGCTTTGCAGAATGTGTACACCAAGGGGAGGGCGAGGCGAAGGAAGGAGGGCGTAAG AAAGGAGGCGGTGGCGGGGCGGAGGAGATTATCTATACTTTTTAAAAAAAAGGAGCCTCTTAGCCG CGTAAAGGAGACTTGGGGAGCGCCTGACAGCACGCGCGGGACACGAGAGTACCACGCTTCCCTACT CTTTTCAGACCTTGACTGGTACGGGGTCCCAGGACTGCAGGAGGCCAGCGACGCGTGCCCTAGGGA GTCCTGCAGCAGTGCCCTGCCTGAGGCCCGTGAAGGTGCAAACGTCCACTTCCCACCGCACCCGGT TCCTCGCGAGCACTTTTCCTGTGCCGCACCAGAACTCGTAGCAGGGGCCCAGGGGCTGAATGCAAG CTTGATGGACGGCGGCGCGCTGCCCAGACTCATGCCCACCTCGTCTGGAGTCGCTGGAGCCTGCGC TGCTCGGCGGAGACAAGCGTCTCCGGAATTGCTGCGCTGCAGCCGGCGGCGGCGATCTGGAGCAAC CGAGGCCAGCAGCAGCTCGGCGTCCGTGGCACGCCGCAATGAGCGCGAGCGCAACCGCGTAAAGCT GGTAAACTTGGGCTTCCAGGCGCTGCGGCAGCACGTGCCGCACGGCGGCGCCAACAAGAAGCTGAG TAAGGTGGAGACGCTGCGCTCCGCGGTAGAGTACATTCGTGCGCTGCAGCGGCTGCTCGCAGAGCA CGACACGGTGCGGCCGGNGCTCGCTGGGGGGCTGTTAACACCCGCTACTCCGCCGTCCGATGAGTG CACGCAGCCCTCTGCCTCCCCTGCCAGCGGGTCTCTGTCCTGCGCCTCTACGTCTCCGTCCCGGAC CCTGGGCTGCTCTGAGCCTACCTCCCCGCGCTCCGCCTACTCGTCGGAGGAAAGCAGCTGCGAGGG AGAGCTAAGCCCGATGGAGCAGGAGCTGCTTGACTTTTCCAGTTGGTTAGGGGGCTACTGA
SEQ ID NO:12
TTCACCCGGCTGCAAGCGCTAGGTGTACGGAGACCTGGCAGCTCTTGGGGCTTAAGGACTGAGCRC CAGAGCCGGTGGAGGTTCCTGTGGAGTACATTCGGACCCTCTCACAGCCCCCGAGAGTGCGGGACG
TGCGGAGCGCAGTTCGGGATCTGCACTCGAGGACTTGTCGAGGACGCATTAAGCTAAGCATCTGCT
CGGAGCATGGAATCGCACTTTAACTGGTACGGGGTCCCAAGGCTCCAGAAGGCTAGCGACGCGTGC
CCTAGGGAATCCTGCAGCAGTGCCCTGCCTGAGGCCCGTGAAGGTGCGAACGTCCACTTCCCACCG
CACCCGGTTCCTCGCGAGCACTTTTCCTGTGGCGCACCGAAACCCGTAGCGGGGGCCCCGGCGCTG AATGCAAGCTTGATGGACGGCGGCGCGCTGCCCAGACTCGTGCCCACCTCGTCTGGAGTCGCTGGA
GCCTGCACTGCTCGGCGGAGACCCCCGTCCCCGGAACTGCTTCGCTGCAGCCGACGGCGGCGATCG
GGAGCAACCGAGGCCAGCAGCAGCTCGGCGGCCGTGGCACGCCGCAATGAGCGTGAGCGCAACCGC
GTAAAGCTGGTAAACTTGGGCTTCCAGGCGCTGCGGCAGCACGTGCCGCACGGCGGCGCCAACAAG
AAGCTGAGTAAGGTGGAGACGCTGCGCTCCGCGGTAGAGTACATCCGTGCGCTGCAGCGGCTGCTA GCAGAGCACGACGCGGTGCGTGCTGCGCTCTCTGGGGGTCTATTAACACCCGCTACTCGGCCGTCC
GATGTGTGCACGCAGCCCTCCGCCTCCCCTGCCAGCGCGTCTCTGTCCTGCACCTCTACATCCCCA GACCGCCTAGGCTGCTCCGAGCCTGCCTCTCCGCGCTCCGCCTACTCGTCGGAGGACAGCAGCTGC GAGGGAGAGACTTACCCGATGGGGCAGATGTTTGACTTTTCCAATTGGTTAGGGGGCTACTGAGCA CCCCACACCCCTAAGCTGCGTCCCTGGGTGTCCCCTGGTGGACCTACCTGCGTTTCTTGCCCAGGA AACCTGGGCCCATGCCTTACCCATGCTGTCTAGTGCAGCCTGACCAAATGCCAAGTACTGACCTCT GCTCGGCCTCCACGCCGCGGAATGACATCTTCCATCTCCCAGTCCTTGCCGAACCAGGACTTGGAA ATTTCTCAGGAGAAAGAATTTTACAATGACAATCTGCTTTTTATCAATTAACTTGAACTGCTGGAG GACTCTGCTGAAAATATGAAGAATTATTTTTATACAAAGGATCCTTAAGCTTGGAGCACAATAAAG ATGACCTCTGTCTCTCACCCCCACTGTCTAGAACTTTCCAACCTGGCCAAAGTGTGGACGGGTCGG GCCCTGAGGGCAAGATGCCTGGCTGCACCCTTCTTCCTCTTCCGAAGCCTATCCTGACGCTGATGT TTGGCCAGTGTGGGAACCCTGCTATTGCAAAGTGTACTATTCTATAAAAGTTGTTTTTCATTGGAA AGGAATTC
SEQIDNO:13
ATGGCTAGCATGACTGGTGGACAGCAAATGGGTCGGGATCCCATGGACGGCGGCACACTGCCCAGG TCCGCGCCCCCTGCGCCCCCCGTCCCTGTCGGCTGCGCTGCCCGGCGGAGACCCGCGTCCCCGGAA CTGTTGCGCTGCAGCCGGCGGCGGCGACCGGCCACCGCAGAGACCGGAGGCGGCGCAGCGGCCGTA GCGCGGCGCAATGAGCGCGAGCGCAACCGCGTGAAGCTGGTGAACTTGGGCTTCCAGGCGCTGCGG CAGCACGTGCCGCACGGCGGCGCCAGCAAGAAGCTGAGCAAGGTGGAGACGCTGCGCTCAGCCGTG GAGTACATCCGCGCGCTGCAGCGCCTGCTGGCCGAGCACGACGCCGTGCGCAACGCGCTGGCGGGA GGGCTGAGGCCGCAGGCCGTGCGGCCGTCTGCGCCCCGCGGGCCGCCAGGGACCACCCCGGTCGCC GCCTCGCCCTCCCGCGCTTCTTCGTCCCCGGGCCGCGGGGGCAGCTCGGAGCCCGGCTCCCCGCGT TCCGCCTACTCGTCGGACGACAGCGGCTGCGAAGGCGCGCTGAGTCCTGCGGAGCGCGAGCTACTC GACTTCTCCAGCTGGTTAGGGGGCTACACTAGTCTCGAGCACCACCACCACCACCACTGA
SEQ ID NO: 14
MASMTGGQQMGRDPMDGGTLPRSAPPAPPVPVGCAARRRPASPELLRCSRRRRPATAETGGGAAAV ARRNERERITOVIOJV GFQALRQHVPHGGASK-KIJSKΛ^TLRSAVEYIRALQRL AEHDAVR ALAG G RPQAVRPSAPRGPPGTTPVAASPSRASSSPGRGGSSEPGSPRSAYSSDDSGCEGALSPAERELL DFS S GGYTSLEHHHHHH
SEQ IDNO:15
ATGGCTAGCATGACTGGTGGACAGCAAATGGGTCGGGATCCCATGGACGGTGGTACCCTGCCGCGT TCCGCTCCGCCGGCTCCGCCGGTTCCGGTTGGTTGCGCTGCTCGTCGTCGTCCGGCTTCCCCGGAA CTGCTGCGTTGCTCCCGTCGTCGTCGTCCGGCTACCGCAGAGACCGGAGGCGGCGCAGCGGCCGTA GCGCGGCGCAATGAGCGCGAGCGCAACCGCGTGAAGCTGGTGAACTTGGGCTTCCAGGCGCTGCGG CAGCACGTGCCGCACGGCGGCGCCAGCAAGAAGCTGAGCAAGGTGGAGACGCTGCGCTCAGCCGTG GAGTACATCCGCGCGCTGCAGCGCCTGCTGGCCGAGCACGACGCCGTGCGCAACGCGCTGGCGGGA GGGCTGAGGCCGCAGGCCGTGCGGCCGTCTGCGCCCCGCGGGCCGCCAGGGACCACCCCGGTCGCC GCCTCGCCCTCCCGCGCTTCTTCGTCCCCGGGCCGCGGGGGCAGCTCGGAGCCCGGCTCCCCGCGT TCCGCCTACTCGTCGGACGACAGCGGCTGCGAAGGCGCGCTGAGTCCTGCGGAGCGCGAGCTACTC GACTTCTCCAGCTGGTTAGGGGGCTACACTAGTCTCGAGCACCACCACCACCACCACTGA SEQ ID NO:16
KLVNLGFQAL
SEQ ID NO:17
EL DFSSWL
SEQ ID NO:18
R LAEHDAV
SEQ D3 NO:19
KLVNLGFQA
SEQ ID NO:20 EYIRA QRL
SEQ D3 NO:21
EYIRALQRLL
SEQ ID NO:22
AVRNAAGG
SEQ ID NO:23
SEPGSPRSAY
SEQ π> NO:24
VET RSAVEY
SEQ ID NO:25 IRALQRLLA
SEQ ID NO:26
LRPQAVRPS
SEQ DD NO:27 LRQHVPHGG
SEQ ID NO:28
LGFQALRQH SEQ ID NO:29
VR ALAGGL
SEQ ID NO:30
YIRALQRLL
SEQ ID NO:31
LVNLGFQAL
SEQ ID NO:32
VEYIRALQR
SEQ ID NO:33 LLRCSRRRR
SEQ ID NO:34
ATGGCTAGCATGACTGGTGGACAGCAAATGGGTCGGGATCCCATGGATGGTGGTACTCTGCCACGT TCTGCTCCGCCAGCTCCACCGGTTCCGGTAGGTTGTGCTGCACGTCGCCGTCCAGCTTCTCCAGAA CTGCTTCGTTGTTCTCGTCGCAGACGTCCAGCTACCGCAGAGACCGGAGGCGGCGCAGCGGCCGTA GCGCGGCGCAATGAGCGCGAGCGCAACCGCGTGAAGCTGGTGAACTTGGGCTTCCAGGCGCTGCGG CAGCACGTGCCGCACGGCGGCGCCAGCAAGAAGCTGAGCAAGGTGGAGACGCTGCGCTCAGCCGTG GAGTACATCCGCGCGCTGCAGCGCCTGCTGGCCGAGCACGACGCCGTGCGCAACGCGCTGGCGGGA GGGCTGAGGCCGCAGGCCGTGCGGCCGTCTGCGCCCCGCGGGCCGCCAGGGACCACCCCGGTCGCC GCCTCGCCCTCCCGCGCTTCTTCGTCCCCGGGCCGCGGGGGCAGCTCGGAGCCCGGCTCCCCGCGT TCCGCCTACTCGTCGGACGACAGCGGCTGCGAAGGCGCGCTGAGTCCTGCGGAGCGCGAGCTACTC GACTTCTCCAGCTGGTTAGGGGGCTACACTAGTCTCGAGCACCACCACCACCACCACTGA

Claims

Claims
1. Use of a polynucleotide comprising a nucleotide sequence which has at least 70% identity to the nucleotide sequence as set forth in SEQ ED NO:l over the entire length of SEQ ED NO: 1 for the manufacture of a medicament for immunotherapeutically treating a patient suffering from or susceptible to breast carcinoma.
2. Use according to claim 1 wherein the identity is at least 95%.
3. Use according to claim 2 wherein the polynucleotide sequence is SEQ ED NO:l.
4. Use of a polynucleotide sequence for the manufacture of a medicament in the treatment or prevention of breast carcinoma, wherein the polynucleotide is selected from:
(a) a polynucleotide comprising a nucleotide sequence encoding the polypeptide of SEQ ED NO:2;
(b) the coding region of the polynucleotide of SEQ TD NO:l; and
(c) a polynucleotide obtainable by screening an appropriate library under stringent hybridisation conditions with a labeled probe having the sequence of SEQ ED NO:l or a fragment thereof, said polynucleotide encoding a protein which has similar properties to those oof the protein of
SEQ TD NO:2
5. Use of a polypeptide comprising an amino acid sequence which has at least 70% identity to the amino acid sequence as set forth in SEQ TD NO:2 for the manufacture of a medicament for immunotherapeutically treating a patient suffering from or susceptible to breast carcinoma.
6. Use according to claim 5 wherein the identity is at least 95%.
7. Use according to claim 6 wherein the polypeptide comprises the amino acid sequence of SEQ TD NO:2.
8. Use according to claim 7 wherein the polypeptide is that of SEQ ED NO:2.
9. Use according to any of claims 5 to 8 wherein the polypeptide comprises an immunogenic fragment of the polypeptide as defined in any of claims 5 to 8 in which the immunogenic activity of the immunogenic fragment is substantially the same as that of the polypeptide of SEQ TD NO:2.
10. Use according to claim 9 wherein the fragment comprises a sequence of one or more of SEQ ED NO: 16 to SEQ ED NO:33.
11. Use according to any of claims 5 to 9 wherein the polypeptide is encoded by a polynucleotide sequence whose sequence has been codon-optimised for the expression in E. coli.
12. Use according to any of claims 5 to 11 wherein the polypeptide is part of a larger fusion protein.
13. Use according to claim 11 wherein the fusion is the sequence set forth in SEQ TD NO:7 or SEQ ED NO: 14.
14. Use according to any of claims 5 to 10 wherein the polypeptide is chemically conjugated to a carrier.
15. A method for the treatment of breast cancer in a subject by immunoprophylaxis or therapy comprising in vitro induction of immune responses to a molecule as defined in any one of claims 5 to 14, using in vitro incubation of the polypeptide as defined in any of claims 5 to 13 or the polynucleotide as defined in any of claims 1 to 4 with cells from the immune system of a mammal, and reinfusing these activated immune cells to the mammal for the treatment of disease.
16. A process for diagnosing the presence of breast cancer or a susceptibility to breast cancer in a subject related to the expression or activity of a polypeptide as defined in any one of claims 5 to 13 in a subject comprising analysing for the presence or amount of said polypeptide in a sample derived from such subject.
7. A process for diagnosing the presence of breast cancer or a susceptibility to breast cancer in a subject related to the expression or activity of a polynucleotide as defined in any of claims 1 to 4 in a subject comprising analysing for the presence or amount of said polynucleotide in a sample derived from said subject.
PCT/EP2002/001649 2001-02-21 2002-02-15 Use of casb7439 (ash2) in the immunotherapy of breast cancer WO2002066506A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2002249221A AU2002249221A1 (en) 2001-02-21 2002-02-15 Use of casb7439 (ash2) in the immunotherapy of breast cancer

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
GBGB0104259.7A GB0104259D0 (en) 2001-02-21 2001-02-21 Novel compounds
GB0104259.7 2001-02-21

Publications (2)

Publication Number Publication Date
WO2002066506A2 true WO2002066506A2 (en) 2002-08-29
WO2002066506A3 WO2002066506A3 (en) 2002-12-19

Family

ID=9909202

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2002/001649 WO2002066506A2 (en) 2001-02-21 2002-02-15 Use of casb7439 (ash2) in the immunotherapy of breast cancer

Country Status (3)

Country Link
AU (1) AU2002249221A1 (en)
GB (1) GB0104259D0 (en)
WO (1) WO2002066506A2 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002092627A2 (en) * 2001-05-16 2002-11-21 Glaxosmithkline Biologicals S.A. Use of casb 7439 for treatment and diagnosis of lung cancer
WO2010136443A1 (en) * 2009-05-27 2010-12-02 Glaxosmithkline Biologicals S.A. Casb7439 constructs
US20140079774A1 (en) * 2011-04-28 2014-03-20 Stc.Unm Porous nanoparticle-supported lipid bilayers (protocells) for targeted delivery and methods of using same
US11344629B2 (en) 2017-03-01 2022-05-31 Charles Jeffrey Brinker Active targeting of cells by monosized protocells
US11672866B2 (en) 2016-01-08 2023-06-13 Paul N. DURFEE Osteotropic nanoparticles for prevention or treatment of bone metastases
EP3380505B1 (en) * 2015-11-23 2023-06-14 Immunocore Limited Peptides derived from achaete-scute homolog 2 (ascl2), complexes comprising such peptides bound to mhc molecules

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001002828A2 (en) * 1999-07-07 2001-01-11 Tularik Inc. Diagnosis of cancer by detecting ash2 polypeptides or polynucleotides

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2001002828A2 (en) * 1999-07-07 2001-01-11 Tularik Inc. Diagnosis of cancer by detecting ash2 polypeptides or polynucleotides

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
BERTHET F.-X. ET AL.: "Applied genome research in the field of human vaccines" JOURNAL OF BIOTECHNOLOGY, vol. 85, no. 2, 13 February 2001 (2001-02-13), pages 213-226, XP002214628 *
KOBAYASHI H. ET AL.: "Defining promiscuous MHC class II Helper T-cell epitopes for the Her2/neu Tumor Antigen" CANCER RESEARCH, vol. 60, 15 September 2000 (2000-09-15), pages 5228-5236, XP002214629 *

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002092627A2 (en) * 2001-05-16 2002-11-21 Glaxosmithkline Biologicals S.A. Use of casb 7439 for treatment and diagnosis of lung cancer
WO2002092627A3 (en) * 2001-05-16 2003-02-06 Glaxosmithkline Biolog Sa Use of casb 7439 for treatment and diagnosis of lung cancer
WO2010136443A1 (en) * 2009-05-27 2010-12-02 Glaxosmithkline Biologicals S.A. Casb7439 constructs
CN102459324A (en) * 2009-05-27 2012-05-16 葛兰素史密丝克莱恩生物有限公司 Casb7439 constructs
CN102459324B (en) * 2009-05-27 2014-07-09 葛兰素史密丝克莱恩生物有限公司 Casb7439 constructs
US8916514B2 (en) 2009-05-27 2014-12-23 Glaxosmithkline Biologicals, S.A. CASB7439 constructs
US20140079774A1 (en) * 2011-04-28 2014-03-20 Stc.Unm Porous nanoparticle-supported lipid bilayers (protocells) for targeted delivery and methods of using same
US9579283B2 (en) * 2011-04-28 2017-02-28 Stc.Unm Porous nanoparticle-supported lipid bilayers (protocells) for targeted delivery and methods of using same
US10022327B2 (en) 2011-04-28 2018-07-17 Stc.Unm Porous nanoparticle-supported lipid bilayers (protocells) for targeted delivery and methods of using same
EP3380505B1 (en) * 2015-11-23 2023-06-14 Immunocore Limited Peptides derived from achaete-scute homolog 2 (ascl2), complexes comprising such peptides bound to mhc molecules
US11672866B2 (en) 2016-01-08 2023-06-13 Paul N. DURFEE Osteotropic nanoparticles for prevention or treatment of bone metastases
US11344629B2 (en) 2017-03-01 2022-05-31 Charles Jeffrey Brinker Active targeting of cells by monosized protocells

Also Published As

Publication number Publication date
WO2002066506A3 (en) 2002-12-19
AU2002249221A1 (en) 2002-09-04
GB0104259D0 (en) 2001-04-11

Similar Documents

Publication Publication Date Title
US8535690B2 (en) Tumor specific animal proteins
AU2001256156A1 (en) Novel compounds
WO2002050103A2 (en) Tumour-related antigens
WO2002066506A2 (en) Use of casb7439 (ash2) in the immunotherapy of breast cancer
WO2002006338A1 (en) Vaccine comprising a lung tumour associated antigen
US7811574B2 (en) Tumour-specific animal proteins
WO2001034794A1 (en) Casb7434, an antigen over-expressed in colon cancer
WO2001029214A1 (en) Colon-tumour-associated antigens
WO2002092627A2 (en) Use of casb 7439 for treatment and diagnosis of lung cancer
EP1222198A2 (en) Human tumor-associated lak-4p related polynucleotides and polypeptides and their uses
WO2001080879A2 (en) Colorectal cancer vaccines and diagnosis
WO2002098913A2 (en) Polypeptides and corresponding polynucleotides for prophylaxis and treatment of colon cancer
WO2001057077A1 (en) Proteins that are specifically expressed or highly overexpressed in tumors and nucleic acids encoding them
ZA200206746B (en) Tumour-specific animal proteins.
WO2001034795A2 (en) Casb7435 polypeptide, nucleic acid encoding it, and their uses in treatment and diagnosis
WO2003016344A2 (en) Use of lung carcinoma antigen

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
AK Designated states

Kind code of ref document: A3

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NO NZ OM PH PL PT RO RU SD SE SG SI SK SL TJ TM TR TT TZ UA UG US UZ VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A3

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE CH CY DE DK ES FI FR GB GR IE IT LU MC NL PT SE TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP