US20040138112A1 - Vaccines - Google Patents

Vaccines Download PDF

Info

Publication number
US20040138112A1
US20040138112A1 US10/407,481 US40748103A US2004138112A1 US 20040138112 A1 US20040138112 A1 US 20040138112A1 US 40748103 A US40748103 A US 40748103A US 2004138112 A1 US2004138112 A1 US 2004138112A1
Authority
US
United States
Prior art keywords
seq
cells
polypeptide
cripto
nonamer
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US10/407,481
Inventor
Jean-Pol Cassart
Thierry Coche
Remi Palmantier
Carlota Bassols
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
GlaxoSmithKline Biologicals SA
Original Assignee
GlaxoSmithKline Biologicals SA
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by GlaxoSmithKline Biologicals SA filed Critical GlaxoSmithKline Biologicals SA
Priority to US10/407,481 priority Critical patent/US20040138112A1/en
Assigned to GLAXOSMITHKLINE BIOLOGICALS S.A. reassignment GLAXOSMITHKLINE BIOLOGICALS S.A. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: COCHE, THIERRY, CASSART, JEAN-POL, PALMANTIER, REMI M., VINALS Y DE BASSOLS, CARLOTA
Publication of US20040138112A1 publication Critical patent/US20040138112A1/en
Abandoned legal-status Critical Current

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
    • C07K14/475Growth factors; Growth regulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5158Antigen-pulsed cells, e.g. T-cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/53DNA (RNA) vaccination
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/80Vaccine for a specifically defined cancer
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/80Vaccine for a specifically defined cancer
    • A61K2039/812Breast
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/80Vaccine for a specifically defined cancer
    • A61K2039/82Colon
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/80Vaccine for a specifically defined cancer
    • A61K2039/86Lung
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide

Definitions

  • the present invention relates generally to therapy of cancer, such as colon, colorectal, breast, bladder, lung and endometrial cancer.
  • the invention is more specifically related to polypeptides, comprising at least a portion of a cripto protein tumor protein, and to polynucleotides encoding such polypeptides, in particular pharmaceutical compositions, e.g., vaccines, and other compositions for the treatment of cancer that are cripto-expressing carcinomas such as certain non-small long cell carcinoma, breast, colon, colorectal cancer.
  • CRIPTO is a 188 amino acid protein shares homologies with the epidermal growth factor (EGF) family (EMBO Journal (1989) Vol 8 (7) pp1987-1991).
  • huCRIPTO mRNA is detected only in undifferentiated cells and disappear after cell differentiation mCRIPTO is expressed during pregnancy and lactation (induces branching morphogenesis in mammary epithelial cells) and is suspected to be an autocrine growth factor for normal breast cells. CRIPTO is required for correct orientation of the anterior-posterior axis in the mouse embryo.
  • Cancer is a significant health problem throughout the world. Although advances have been made in detection and therapy of cancer, no vaccine or other universally successful method for prevention and/or treatment is currently available. Current therapies, which are generally based on a combination of chemotherapy or surgery and radiation, continue to prove inadequate in many patients.
  • Colon cancer is the second most frequently diagnosed malignancy in the United Sates as well as the second most common cause of cancer death.
  • An estimated 95,600 new cases of colon cancer will have been diagnosed in 1998, with an estimated 47,700 deaths.
  • the five-year survival rate for patients with colorectal cancer detected in an early-localised stage is 92%, unfortunately, only 37% of colorectal cancer is diagnosed at this stage, the survival stage.
  • the survival rate drops to 64% if the cancer is allowed to spread to adjacent organs or lymph nodes, and to 7% in patients with distant metastases.
  • breast cancer is a significant health problem for women in the United States and throughout the world. Although advances have been made in detection and treatment of the disease, breast cancer remains the second leading cause of cancer-related deaths in women, affecting more than 180,000 women in the United States each year. For women in North America, the lifetime odds of getting breast cancer are now one in eight.
  • Lung cancer is the primary cause of cancer death among both men and women in the US, with an estimated 172,000 new cases being reported in 1994.
  • the five-year survival rate among all lung cancer patients, regardless of the state of disease at diagnosis is only 13%. This contrasts with a five-year survival rate of 46% among cases detected while the disease is still localised. However, only 16% of lung cancers are discovered before the disease has spread.
  • the present invention provides a method for inhibiting the development of a cancer in a patient, comprising the steps of:
  • the invention provides a method for producing an immunogenic response to a tumor-associated antigen in an animal comprising administering a first component comprising a polynucleotide encoding a fragment of the tumor-associated antigen to the animal.
  • the invention provides a method of inducing an immunoresponse to Cripto in an animal comprising repeatedly administering a composition comprising a first component comprising SEQ ID NO:11 or SEQ ID NO:12.
  • the present invention in another aspect, provides polypeptide compositions comprising an amino acid sequence that is encoded by a polynucleotide sequence described above.
  • polypeptides and/or polynucleotides of the present invention are immunogenic, i.e., they are capable of eliciting an immune response, particularly a humoral and/or cellular immune response, when if necessary, they are conjugated to a suitable carrier and/or adjuvanted.
  • the present invention further provides fragments, variants and/or derivatives of the disclosed polypeptide and/or polynucleotide sequences, wherein the fragments, variants and/or derivatives preferably 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 set forth in SEQ ID No: 2 or 4 or a polypeptide sequence encoded by a polynucleotide sequence set forth in SEQ ID NO: 1 or 3.
  • the present invention further provides polynucleotides that encode a polypeptide described above, expression vectors comprising such polynucleotides and host cells transformed or transfected with such expression vectors.
  • compositions comprising a polypeptide or polynucleotide as described above and a physiologically acceptable carrier.
  • compositions e.g., vaccine compositions
  • Such compositions generally comprise an immunogenic polypeptide or polynucleotide of the invention and an immunostimulant, such as an adjuvant.
  • compositions comprising: (a) an antigen presenting cell that expresses a polypeptide as described above and (b) a pharmaceutically acceptable carrier or excipient.
  • antigen presenting cells include dendritic cells, macrophages, monocytes, fibroblasts and B cells.
  • compositions comprise: (a) an antigen presenting cell that expresses a polypeptide as described above and (b) an immunostimulant.
  • the present invention further provides, in other aspects, fusion proteins that comprise at least one polypeptide as described above, as well as polynucleotides encoding such fusion proteins, typically in the form of pharmaceutical compositions, e.g., vaccine compositions, comprising a physiologically acceptable carrier and/or an immunostimulant.
  • the fusion proteins may comprise multiple immunogenic polypeptides or portions/variants thereof, as described herein, and may further comprise one or more polypeptide segments for facilitating the expression, purification and/or immunogenicity of the polypeptide(s).
  • the present invention provides methods for stimulating an immune response in a patient, preferably a T cell response in a human patient, comprising administering a pharmaceutical composition described herein.
  • the patient may be afflicted with lung or colon cancer or colorectal cancer or breast cancer, in which case the methods provide treatment for the disease, or patient considered at risk for such a disease may be treated prophylactically.
  • the patient will be afflicted with a tumour expressing cripto antigens.
  • the present invention provides methods for inhibiting the development of a cancer in a patient, comprising administering to a patient a pharmaceutical composition as recited above.
  • the patient may be afflicted with lung, colon, colorectal or breast cancer, in which case the methods provide treatment for the disease, or patient considered at risk for such a disease may be treated prophylactically.
  • the present invention further provides, within other aspects, methods for removing tumor cells from a biological sample, comprising contacting a biological sample with T cells that specifically react with a polypeptide of the present invention, wherein the step of contacting is performed under conditions and for a time sufficient to permit the removal of cells expressing the protein from the sample.
  • methods for inhibiting the development of a cancer in a patient, comprising administering to a patient a biological sample treated as described above.
  • Methods are further provided, within other aspects, for stimulating and/or expanding T cells specific for a polypeptide of the present invention, comprising contacting T cells with one or more of: (i) a polypeptide as described above; (ii) a polynucleotide encoding such a polypeptide; and/or (iii) an antigen presenting cell that expresses such a polypeptide; under conditions and for a time sufficient to permit the stimulation and/or expansion of T cells.
  • Isolated T cell populations comprising T cells prepared as described above are also provided.
  • the present invention provides methods for inhibiting the development of a cancer in a patient, comprising administering to a patient an effective amount of a T cell population as described above.
  • the present invention further provides methods for inhibiting the development of a cancer in a patient, comprising the steps of: (a) incubating CD4+ and/or CD8+ T cells isolated from a patient with one or more of: (i) a polypeptide comprising at least an immunogenic portion of a cripto polypeptide disclosed herein; (ii) a polynucleotide encoding such a polypeptide; and (iii) an antigen-presenting cell that expressed such a polypeptide; and (b) administering to the patient an effective amount of the proliferated T cells, and thereby inhibiting the development of a cancer in the patient.
  • Proliferated cells may, but need not, be cloned prior to administration to the patient.
  • SEQ ID NO: 5 Cripto Polynucleotide as described in U.S. Pat. No. 5,654,140
  • compositions of the present invention are directed generally to compositions and their use in the therapy and diagnosis of cancer, particularly cripto expressing cancer and metastases including Cripto expressing lung, colon, colorectal and breast cancers.
  • illustrative compositions of the present invention include, but are not restricted to, polypeptides, particularly immunogenic polypeptides, polynucleotides encoding such polypeptides, antibodies and other binding agents, antigen presenting cells (APCs) and immune system cells (e.g., T cells).
  • APCs antigen presenting cells
  • T cells immune system cells
  • the present invention provides a method for inhibiting the development of a cancer in a patient, comprising the steps of:
  • the T cells are proliferated prior to being administered ot the patient.
  • a method for producing an immunogenic response to a tumor-associated antigen in an animal comprising administering a first component comprising a polynucleotide encoding a fragment of the tumor-associated antigen to the animal.
  • the tumor-associated antigen is Cripto.
  • the polynucleotide encodes SEQ ID NO:11 and does not encode SEQ ID NO:3 or SEQ ID NO:4.
  • the polynucleotide encodes SEQ ID NO:12 and does not encode SEQ ID NO:3 or SEQ ID NO:4.
  • the polynucleotide is recombinant DNA.
  • the polynucleotide is admixed with a second component comprising an immunostimulant, adjuvant and physiologically acceptable carrier.
  • a method for inducing an immunoresponse to Cripto in an animal comprising repeatedly administering a composition comprising a first component comprising SEQ ID NO:11 or SEQ ID NO:12.
  • the first component is admixed with a second component comprising a physiologically acceptable carrier, adjuvant and immunostimulant prior to administering the first component to the animal.
  • Polypeptide(s) refers to any peptide or protein comprising two or more amino acids joined to each other by peptide bonds or modified peptide bonds. “Polypeptide(s)” refers to both short chains, commonly referred to as peptides, oligopeptides and oligomers and to longer chains generally referred to as proteins. Polypeptides may comprise amino acids other than the 20 gene encoded amino acids. “Polypeptide(s)” include those modified either by natural processes, such as processing and other post-translational modifications, but also by chemical modification techniques. Such modifications are well described in basic texts and in more detailed monographs, as well as in a voluminous research literature, and they are well known to those of skill in the art.
  • a given polypeptide may comprise many types of modifications. Modifications can occur anywhere in a polypeptide, including the peptide backbone, the amino acid side-chains, and the amino or carboxyl termini.
  • Modifications include, for example, acetylation, acylation, ADP-ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphotidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cysteine, formation of pyroglutamate, formylation, gamma-carboxylation, GPI anchor formation, hydroxylation, iodination, methylation, myristoylation, oxidation, proteolytic processing, phosphorylation, prenylation, racemization, glycosylation, lipid attachment, sulfation, gamma-carboxylation of glutamic acid residues, hydroxylation and ADP-ribosylation, selenoylation
  • Polypeptides may be branched or cyclic, with or without branching. Cyclic, branched and branched circular polypeptides may result from post-translational natural processes and may be made by entirely synthetic methods, as well.
  • polypeptides of the present invention comprise a sequence of at least 10 contiguous amino acids, preferably 20, more preferably 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180 amino acids of the cirpto protein of ID NO: 2 or 4.
  • the polypeptides of the invention are immunogenic, i.e., they react detectably within an immunoassay (such as an ELISA or T-cell stimulation assay) with antisera and/or T-cells from a patient with cripto expressing cancer.
  • an immunoassay such as an ELISA or T-cell stimulation assay
  • Screening for immunogenic activity can be performed using techniques well known to the skilled artisan. For example, such screens can be performed using methods such as those described in Harlow and Lane, Antibodies: A Laboratory Manual , Cold Spring Harbor Laboratory, 1988.
  • a polypeptide may be immobilized on a solid support and contacted with patient sera to allow binding of antibodies within the sera to the immobilized polypeptide. Unbound sera may then be removed and bound antibodies detected using, for example, 125 I-labeled Protein A.
  • immunogenic portions of the polypeptides disclosed herein are also encompassed by the present invention.
  • An “immunogenic portion,” as used herein, is a fragment of an immunogenic polypeptide of the invention that itself is immunologically reactive (i.e., specifically binds) with the B-cells and/or T-cell surface antigen receptors that recognize the polypeptide. Immunogenic portions may generally be identified using well known techniques, such as those summarized in Paul, Fundamental Immunology, 3rd ed., 243247 (Raven Press, 1993) and references cited therein. Such techniques include screening polypeptides for the ability to react with antigen-specific antibodies, antisera and/or T-cell lines or clones.
  • antisera and antibodies are “antigen-specific” if they specifically bind to an antigen (i.e., they react with the protein in an ELISA or other immunoassay, and do not react detectably with unrelated proteins).
  • antisera and antibodies may be prepared as described herein, and using well-known techniques.
  • an immunogenic portion of a polypeptide of the present invention is a portion that reacts with antisera and/or T-cells at a level that is not substantially less than the reactivity of the full-length polypeptide (e.g., in an ELISA and/or T-cell reactivity assay).
  • the level of immunogenic activity of the immunogenic portion is at least about 50%, preferably at least about 70% and most preferably greater than about 90% of the immunogenicity for the full-length polypeptide.
  • preferred immunogenic portions will be identified that have a level of immunogenic activity greater than that of the corresponding full-length polypeptide, e.g., having greater than about 100% or 150% or more immunogenic activity.
  • illustrative immunogenic portions may include peptides in which an N-terminal leader sequence and/or transmembrane domain have been deleted.
  • Other illustrative immunogenic portions will contain a small N- and/or C-terminal deletion (e.g., 1-30 amino acids, preferably 5-15 amino acids), relative to the mature protein.
  • a polypeptide composition of the invention may also comprise one or more polypeptides that are immunologically reactive with T cells and/or antibodies generated against a polypeptide of the invention, particularly a polypeptide having an amino acid sequence disclosed herein, or to an immunogenic fragment or variant thereof.
  • polypeptides comprise one or more polypeptides that are capable of eliciting T cells and/or antibodies that are immunologically reactive with one or more polypeptides described herein, or one or more polypeptides encoded by contiguous nucleic acid sequences contained in the polynucleotide sequences disclosed herein, or immunogenic fragments or variants thereof, or to one or more nucleic acid sequences which hybridize to one or more of these sequences under conditions of moderate to high stringency.
  • the present invention in another aspect, provides polypeptide fragments comprising at least about 5, 10, 15, 20, 25, 50, 100, or 150 contiguous amino acids, or more, including all intermediate lengths, of a polypeptide compositions set forth herein, such as those set forth in SEQ ID NO: 2 or 4 or those encoded by a polynucleotide sequence set forth in a sequence of SEQ ID NO:1 or 3. It is preferred that the polypeptides comprise at least one preferably a pluarality of epitopes as set forth in sequence ID no 13 to 94. Optionally the frgaments are fused or otherwise conjugated to a heterologous carrier.
  • the present invention provides variants of the polypeptide compositions described herein.
  • Polypeptide variants generally encompassed by the present invention will typically exhibit at least about 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% or more identity (determined as described below), along its length, to a polypeptide sequences set forth herein.
  • polypeptide fragments and variants provided by the present invention are immunologically reactive with an antibody and/or T-cell that reacts with a full-length polypeptide specifically set for the herein.
  • polypeptide fragments and variants provided by the present invention exhibit a level of immunogenic activity of at least about 50%, preferably at least about 70%, and most preferably at least about 90% or more of that exhibited by a full-length polypeptide sequence specifically set forth herein.
  • a polypeptide “variant,” as the term is used herein, is a polypeptide that typically differs from a polypeptide specifically disclosed herein in one or more substitutions, deletions, additions and/or insertions. Such variants may be naturally occurring or may be synthetically generated, for example, by modifying one or more of the above polypeptide sequences of the invention and evaluating their immunogenic activity as described herein and/or using any of a number of techniques well known in the art.
  • certain illustrative variants of the polypeptides of the invention include those in which one or more portions, such as an N-terminal leader sequence or transmembrane domain, have been removed.
  • Other illustrative variants include variants in which a small portion (e.g., 1-30 amino acids, preferably 5-15 amino acids) has been removed from the N- and/or C-terminal of the mature protein.
  • a variant will contain conservative substitutions.
  • a “conservative substitution” is one in which an amino acid is substituted for another amino acid that has similar properties, such that one skilled in the art of peptide chemistry would expect the secondary structure and hydropathic nature of the polypeptide to be substantially unchanged.
  • modifications may be made in the structure of the polynucleotides and polypeptides of the present invention and still obtain a functional molecule that encodes a variant or derivative polypeptide with desirable characteristics, e.g., with immunogenic characteristics.
  • amino acids may be substituted for other amino acids in a protein structure without appreciable loss of interactive binding capacity with structures such as, for example, antigen-binding regions of antibodies or binding sites on substrate molecules. Since it is the interactive capacity and nature of a protein that defines that protein's biological functional activity, certain amino acid sequence substitutions can be made in a protein sequence, and, of course, its underlying DNA coding sequence, and nevertheless obtain a protein with like properties. It is thus contemplated that various changes may be made in the peptide sequences of the disclosed compositions, or corresponding DNA sequences which encode said peptides without appreciable loss of their biological utility or activity.
  • the hydropathic index of amino acids may be considered.
  • the importance of the hydropathic amino acid index in conferring interactive biologic function on a protein is generally understood in the art (Kyte and Doolittle, 1982, incorporated herein by reference). It is accepted that the relative hydropathic character of the amino acid contributes to the secondary structure of the resultant protein, which in turn defines the interaction of the protein with other molecules, for example, enzymes, substrates, receptors, DNA, antibodies, antigens, and the like.
  • Each amino acid has been assigned a hydropathic index on the basis of its hydrophobicity and charge characteristics (Kyte and Doolittle, 1982).
  • hydrophilicity values have been assigned to amino acid residues: arginine (+3.0); lysine (+3.0); aspartate (+3.0 ⁇ 1); glutamate (+3.0 ⁇ 1); serine (+0.3); asparagine (+0.2); glutamine (+0.2); glycine (0); threonine ( ⁇ 0.4); proline ( ⁇ 0.5 ⁇ 1); alanine ( ⁇ 0.5); histidine ( ⁇ 0.5); cysteine ( ⁇ 1.0); methionine ( ⁇ 1.3); valine ( ⁇ 1.5); leucine ( ⁇ 1.8); isoleucine ( ⁇ 1.8); tyrosine ( ⁇ 2.3); phenylalanine ( ⁇ 2.5); tryptophan ( ⁇ 3.4).
  • an amino acid can be substituted for another having a similar hydrophilicity value and still obtain a biologically equivalent, and in particular, an immunologically equivalent protein.
  • substitution of amino acids whose hydrophilicity values are within ⁇ 2 is preferred, those within ⁇ 1 are particularly preferred, and those within ⁇ 0.5 are even more particularly preferred.
  • amino acid substitutions are generally therefore based on the relative similarity of the amino acid side-chain substituents, for example, their hydrophobicity, hydrophilicity, charge, size, and the like.
  • Exemplary substitutions that take various of the foregoing characteristics into consideration are well known to those of skill in the art and include: arginine and lysine; glutamate and aspartate; serine and threonine; glutamine and asparagine; and valine, leucine and isoleucine.
  • any polynucleotide may be further modified to increase stability in vivo. Possible modifications include, but are not limited to, the addition of flanking sequences at the 5′ and/or 3′ ends; the use of phosphorothioate or 2′ O-methyl rather than phosphodiesterase linkages in the backbone; and/or the inclusion of nontraditional bases such as inosine, queosine and wybutosine, as well as acetyl-methyl-, thio- and other modified forms of adenine, cytidine, guanine, thymine and uridine.
  • Amino acid substitutions may further be made on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity and/or the amphipathic nature of the residues.
  • negatively charged amino acids include aspartic acid and glutamic acid
  • positively charged amino acids include lysine and arginine
  • amino acids with uncharged polar head groups having similar hydrophilicity values include leucine, isoleucine and valine; glycine and alanine; asparagine and glutamine; and serine, threonine, phenylalanine and tyrosine.
  • variant polypeptides differ from a native sequence by substitution, deletion or addition of five amino acids or fewer.
  • Variants may also (or alternatively) be modified by, for example, the deletion or addition of amino acids that have minimal influence on the immunogenicity, secondary structure and hydropathic nature of the polypeptide.
  • polypeptides may comprise a signal (or leader) sequence at the N-terminal end of the protein, which co-translationally or post-translationally directs transfer of the protein.
  • the polypeptide may also be conjugated to a linker or other sequence for ease of synthesis, purification or identification of the polypeptide (e.g., poly-His), or to enhance binding of the polypeptide to a solid support.
  • a polypeptide may be conjugated to an immunoglobulin Fc region.
  • two sequences are said to be “identical” if the sequence of amino acids in the two sequences is the same when aligned for maximum correspondence, as described below. Comparisons between two sequences are typically performed by comparing the sequences over a comparison window to identify and compare local regions of sequence similarity.
  • a “comparison window” as used herein refers to a segment of at least about 20 contiguous positions, usually 30 to about 75, 40 to about 50, in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned.
  • Optimal alignment of sequences for comparison may be conducted using the Megalign program in the Lasergene suite of bioinformatics software (DNASTAR, Inc., Madison, Wis.), using default parameters.
  • This program embodies several alignment schemes described in the following references: Dayhoff, M. O. (1978) A model of evolutionary change in proteins—Matrices for detecting distant relationships. In Dayhoff, M. O. (ed.) Atlas of Protein Sequence and Structure, National Biomedical Research Foundation, Washington D.C. Vol. 5, Suppl. 3, pp. 345-358; Hein J. (1990) Unified Approach to Alignment and Phylogenes pp. 626-645 Methods in Enzymology vol.
  • optimal alignment of sequences for comparison may be conducted by the local identity algorithm of Smith and Waterman (1981) Add. APL. Math 2:482, by the identity alignment algorithm of Needleman and Wunsch (1970) J. Mol. Biol. 48:443, by the search for similarity methods of Pearson and Lipman (1988) Proc. Natl. Acad. Sci. USA 85: 2444, by computerized implementations of these algorithms (GAP, BESTFIT, BLAST, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group (GCG), 575 Science Dr., Madison, Wis.), or by inspection.
  • BLAST and BLAST 2.0 are described in Altschul et al. (1977) Nucl. Acids Res. 25:3389-3402 and Altschul et al. (1990) J. Mol. Biol. 215:403-410, respectively.
  • BLAST and BLAST 2.0 can be used, for example with the parameters described herein, to determine percent sequence identity for the polynucleotides and polypeptides of the invention.
  • Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information. For amino acid sequences, a scoring matrix can be used to calculate the cumulative score.
  • Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached.
  • the BLAST algorithm parameters W, T and X determine the sensitivity and speed of the alignment.
  • the “percentage of sequence identity” is determined by comparing two optimally aligned sequences over a window of comparison of at least 20 positions, wherein the portion of the polypeptide sequence in the comparison window may comprise additions or deletions (i.e., gaps) of 20 percent or less, usually 5 to 15 percent, or 10 to 12 percent, as compared to the reference sequences (which does not comprise additions or deletions) for optimal alignment of the two sequences.
  • the percentage is calculated by determining the number of positions at which the identical amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the reference sequence (i.e., the window size) and multiplying the results by 100 to yield the percentage of sequence identity.
  • a polypeptide may be a fusion polypeptide that comprises multiple polypeptides as described herein, or that comprises at least one polypeptide as described herein and an unrelated sequence, such as a known tumor protein.
  • a fusion partner may, for example, assist in providing T helper epitopes (an immunological fusion partner), preferably T helper epitopes recognized by humans, or may assist in expressing the protein (an expression enhancer) at higher yields than the native recombinant protein.
  • Certain preferred fusion partners are both immunological and expression enhancing fusion partners.
  • Other fusion partners may be selected so as to increase the solubility of the polypeptide or to enable the polypeptide to be targeted to desired intracellular compartments.
  • Still further fusion partners include affinity tags, which facilitate purification of the polypeptide.
  • Fusion polypeptides may generally be prepared using standard techniques, including chemical conjugation.
  • a fusion polypeptide is expressed as a recombinant polypeptide, allowing the production of increased levels, relative to a non-fused polypeptide, in an expression system.
  • DNA sequences encoding the polypeptide components may be assembled separately, and ligated into an appropriate expression vector.
  • the 3′ end of the DNA sequence encoding one polypeptide component is ligated, with or without a peptide linker, to the 5′ end of a DNA sequence encoding the second polypeptide component so that the reading frames of the sequences are in phase. This permits translation into a single fusion polypeptide that retains the biological activity of both component polypeptides.
  • a peptide linker sequence may be employed to separate the first and second polypeptide components by a distance sufficient to ensure that each polypeptide folds into its secondary and tertiary structures.
  • Such a peptide linker sequence is incorporated into the fusion polypeptide using standard techniques well known in the art.
  • Suitable peptide linker sequences may be chosen based on the following factors: (1) their ability to adopt a flexible extended conformation; (2) their inability to adopt a secondary structure that could interact with functional epitopes on the first and second polypeptides; and (3) the lack of hydrophobic or charged residues that might react with the polypeptide functional epitopes.
  • Preferred peptide linker sequences contain Gly, Asn and Ser residues.
  • linker sequences which may be usefully employed as linkers include those disclosed in Maratea et al., Gene 40:39-46, 1985; Murphy et al., Proc. Natl. Acad. Sci. USA 83:8258-8262, 1986; U.S. Pat. No. 4,935,233 and U.S. Pat. No. 4,751,180.
  • the linker sequence may generally be from 1 to about 50 amino acids in length. Linker sequences are not required when the first and second polypeptides have non-essential N-terminal amino acid regions that can be used to separate the functional domains and prevent steric interference.
  • the ligated DNA sequences are operably linked to suitable transcriptional or translational regulatory elements.
  • the regulatory elements responsible for expression of DNA are located only 5′ to the DNA sequence encoding the first polypeptides.
  • stop codons required to end translation and transcription termination signals are only present 3′ to the DNA sequence encoding the second polypeptide.
  • the fusion polypeptide can comprise a polypeptide as described herein together with an unrelated immunogenic protein, such as an immunogenic protein capable of eliciting a recall response.
  • an immunogenic protein capable of eliciting a recall response.
  • immunogenic proteins include tetanus, tuberculosis and hepatitis proteins (see, for example, Stoute et al. New Engl. J. Med., 336:86-91, 1997).
  • the immunological fusion partner is derived from a Mycobacterium sp., such as a Mycobacterium tuberculosis -derived Ra12 fragment.
  • a Mycobacterium sp. such as a Mycobacterium tuberculosis -derived Ra12 fragment.
  • Ra12 compositions and methods for their use in enhancing the expression and/or immunogenicity of heterologous polynucleotide/polypeptide sequences is described in U.S. Patent Application 60/158,585, the disclosure of which is incorporated herein by reference in its entirety.
  • Ra12 refers to a polynucleotide region that is a subsequence of a Mycobacterium tuberculosis MTB32A nucleic acid.
  • MTB32A is a serine protease of 32 KD molecular weight encoded by a gene in virulent and avirulent strains of M. tuberculosis .
  • the nucleotide sequence and amino acid sequence of MTB32A have been described (for example, U.S. Patent Application 60/158,585; see also, Skeiky et al., Infection and Immun. (1999) 67:3998-4007, incorporated herein by reference).
  • C-terminal fragments of the MTB32A coding sequence express at high levels and remain as a soluble polypeptides throughout the purification process.
  • Ra12 may enhance the immunogenicity of heterologous immunogenic polypeptides with which it is fused.
  • Ra12 fusion polypeptide comprises a 14 KD C-terminal fragment corresponding to amino acid residues 192 to 323 of MTB32A.
  • Other preferred Ra12 polynucleotides generally comprise at least about 15 consecutive nucleotides, at least about 30 nucleotides, at least about 60 nucleotides, at least about 100 nucleotides, at least about 200 nucleotides, or at least about 300 nucleotides that encode a portion of a Ra12 polypeptide.
  • Ra12 polynucleotides may comprise a native sequence (i.e., an endogenous sequence that encodes a Ra12 polypeptide or a portion thereof) or may comprise a variant of such a sequence.
  • Ra12 polynucleotide variants may contain one or more substitutions, additions, deletions and/or insertions such that the biological activity of the encoded fusion polypeptide is not substantially diminished, relative to a fusion polypeptide comprising a native Ra12 polypeptide.
  • Variants preferably exhibit at least about 70% identity, more preferably at least about 80% identity and most preferably at least about 90% identity to a polynucleotide sequence that encodes a native Ra12 polypeptide or a portion thereof.
  • an immunological fusion partner is derived from protein D, a surface protein of the gram-negative bacterium Haemophilus influenza B (WO 91/18926).
  • a protein D derivative comprises approximately the first third of the protein (e.g., the first N-terminal 100-110 amino acids), and a protein D derivative may be lipidated.
  • the first 109 residues of a Lipoprotein D fusion partner is included on the N-terminus to provide the polypeptide with additional exogenous T-cell epitopes and to increase the expression level in E. coli (thus functioning as an expression enhancer).
  • the lipid tail ensures optimal presentation of the antigen to antigen presenting cells.
  • Other fusion partners include the non-structural protein from influenzae virus, NS1 (hemaglutinin). Typically, the N-terminal 81 amino acids are used, although different fragments that include T-helper epitopes may be used.
  • the immunological fusion partner is the protein known as LYTA, or a portion thereof (preferably a C-terminal portion).
  • LYTA is derived from Streptococcus pneumoniae , which synthesizes an N-acetyl-L-alanine amidase known as amidase LYTA (encoded by the LytA gene; Gene 43:265-292, 1986).
  • LYTA is 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 the amino terminus has been described (see Biotechnology 10:795-798, 1992).
  • a repeat portion of LYTA may be incorporated into a fusion polypeptide. A repeat portion is found in the C-terminal region starting at residue 178. A particularly preferred repeat portion incorporates residues 188-305.
  • Yet another illustrative embodiment involves fusion polypeptides, and the polynucleotides encoding them, wherein the fusion partner comprises a targeting signal capable of directing a polypeptide to the endosomal/lysosomal compartment, as described in U.S. Pat. No. 5,633,234.
  • a targeting signal capable of directing a polypeptide to the endosomal/lysosomal compartment, as described in U.S. Pat. No. 5,633,234.
  • An immunogenic polypeptide of the invention when fused with this targeting signal, will associate more efficiently with MHC class II molecules and thereby provide enhanced in vivo stimulation of CD4+ T-cells specific for the polypeptide.
  • the cripto part of the fusion molecule may prefreably be the whole length 188 aa protein of Cripto 1 or Cripto 3 or a fragment thereof as described herein.
  • Polypeptides of the invention are prepared using any of a variety of well known synthetic and/or recombinant techniques, the latter of which are further described below. Polypeptides, portions and other variants generally less than about 150 amino acids can be generated by synthetic means, using techniques well known to those of ordinary skill in the art. In one illustrative example, such polypeptides are synthesized using any of the commercially available solid-phase techniques, such as the Merrifield solid-phase synthesis method, where amino acids are sequentially added to a growing amino acid chain. See Merrifield, J. Am. Chem. Soc. 85:2149-2146, 1963. Equipment for automated synthesis of polypeptides is commercially available from suppliers such as Perkin Elmer/Applied BioSystems Division (Foster City, Calif.), and may be operated according to the manufacturer's instructions.
  • polypeptide compositions including fusion polypeptides of the invention are isolated.
  • An “isolated” polypeptide is one that is removed from its original environment.
  • a naturally-occurring protein or polypeptide is isolated if it is separated from some or all of the coexisting materials in the natural system.
  • polypeptides are also purified, e.g., are at least about 90% pure, more preferably at least about 95% pure and most preferably at least about 99% pure.
  • Polynucleotide(s) generally refers to any polyribonucleotide or polydeoxyribonucleotide, that may be unmodified RNA or DNA or modified RNA or DNA.
  • Polynucleotide(s)” include, without limitation, single- and double-stranded DNA, DNA that is a mixture of single- and double-stranded regions or single-, double- and triple-stranded regions, single- and double-stranded RNA, and RNA that is mixture of single- and double-stranded regions, hybrid molecules comprising DNA and RNA that may be single-stranded or, more typically, double-stranded, or triple-stranded regions, or a mixture of single- and double-stranded regions.
  • polynucleotide refers to triple-stranded regions comprising RNA or DNA or both RNA and DNA.
  • the strands in such regions may be from the same molecule or from different molecules.
  • the regions may include all of one or more of the molecules, but more typically involve only a region of some of the molecules.
  • One of the molecules of a triple-helical region often is an oligonucleotide.
  • the term “polynucleotide(s)” also includes DNAs or RNAs as described above that comprise one or more modified bases. Thus, DNAs or RNAs with backbones modified for stability or for other reasons are “polynucleotide(s)” as that term is intended herein.
  • DNAs or RNAs comprising unusual bases, such as inosine, or modified bases, such as tritylated bases, to name just two examples, are polynucleotides as the term is used herein. It will be appreciated that a great variety of modifications have been made to DNA and RNA that serve many useful purposes known to those of skill in the art.
  • the term “polynucleotide(s)” as it is employed herein embraces such chemically, enzymatically or metabolically modified forms of polynucleotides, as well as the chemical forms of DNA and RNA characteristic of viruses and cells, including, for example, simple and complex cells. “Polynucleotide(s)” also embraces short polynucleotides often referred to as oligonucleotide(s).
  • the present invention provides polynucleotide compositions that encode for the polypeptides of the invention.
  • DNA and “polynucleotide” are used essentially interchangeably herein to refer to a DNA molecule that has been isolated free of total genomic DNA of a particular species. “Isolated,” as used herein, means that a polynucleotide is substantially away from other coding sequences, and that the DNA molecule does not contain large portions of unrelated coding DNA, such as large chromosomal fragments or other functional genes or polypeptide coding regions. Of course, this refers to the DNA molecule as originally isolated, and does not exclude genes or coding regions later added to the segment by the hand of man.
  • polynucleotide compositions of this invention can include genomic sequences, extra-genomic and plasmid-encoded sequences and smaller engineered gene segments that express, or may be adapted to express, proteins, polypeptides, peptides and the like. Such segments may be naturally isolated, or modified synthetically by the hand of man.
  • polynucleotides of the invention may be single-stranded (coding or antisense) or double-stranded, and may be DNA (genomic, cDNA or synthetic) or RNA molecules.
  • RNA molecules may include HnRNA molecules, which contain introns and correspond to a DNA molecule in a one-to-one manner, and mRNA molecules, which do not contain introns. Additional coding or non-coding sequences may, but need not, be present within a polynucleotide of the present invention, and a polynucleotide may, but need not, be linked to other molecules and/or support materials.
  • Polynucleotides may comprise a native sequence (i.e., an endogenous sequence that encodes a polypeptide/protein of the invention or a portion thereof) or may comprise a sequence that encodes a variant or derivative, preferably and immunogenic variant or derivative, of such a sequence.
  • polynucleotide variants will contain one or more substitutions, additions, deletions and/or insertions, preferably such that the immunogenicity of the polypeptide encoded by the variant polynucleotide is not substantially diminished relative to a polypeptide encoded by a polynucleotide sequence specifically set forth herein).
  • variants should also be understood to encompasses homologous genes of xenogenic origin.
  • the present invention provides polynucleotide fragments comprising various lengths of contiguous stretches of sequence identical to or complementary to one or more of the SEQ ID NO:1 or 3.
  • polynucleotides are provided by this invention that comprise at least about 10, 15, 20, 30, 40, 50, 75, 100, 150, 200, 300, 400, the SEQ ID NO: 1 as well as all intermediate lengths there between.
  • intermediate lengths means any length between the quoted values, such as 16, 17, 18, 19, etc.; 21, 22, 23, etc.; 30, 31, 32, etc.; 50, 51, 52, 53, etc.; 100, 101, 102, 103, etc.; 150, 151, 152, 153, etc.; including all integers through 200-500; 500-1,000, and the like.
  • Particularly preferred polynucleotides are those which encode the epitopes as set forth in SEQ ID NOS: 13-94.
  • polynucleotide compositions are provided that are capable of hybridizing under moderate to high stringency conditions to a polynucleotide sequence provided herein, or a fragment thereof, or a complementary sequence thereof.
  • Hybridization techniques are well known in the art of molecular biology.
  • suitable moderately stringent conditions for testing the hybridization of a polynucleotide of this invention with other polynucleotides include prewashing in a solution of 5 ⁇ SSC, 0.5% SDS, 1.0 mM EDTA (pH 8.0); hybridizing at 50° C.-60° C., 5 ⁇ SSC, overnight; followed by washing twice at 65° C.
  • hybridization can be readily manipulated, such as by altering the salt content of the hybridization solution and/or the temperature at which the hybridization is performed.
  • suitable highly stringent hybridization conditions include those described above, with the exception that the temperature of hybridization is increased, e.g., to 60-65° C. or 65-70° C.
  • the polynucleotides described above e.g., polynucleotide variants, fragments and hybridizing sequences, encode polypeptides that are immunologically cross-reactive with a polypeptide sequence specifically set forth in SEQ ID NO:1 or SEQ ID NO:3.
  • such polynucleotides encode polypeptides 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 that for a polypeptide sequence specifically set forth herein.
  • polynucleotides of the present invention may be combined with other DNA sequences, such as promoters, polyadenylation signals, additional restriction enzyme sites, multiple cloning sites, other coding segments, and the like, such that their overall length may vary considerably. It is therefore contemplated that a nucleic acid fragment of almost any length may be employed, with the total length preferably being limited by the ease of preparation and use in the intended recombinant DNA protocol.
  • illustrative polynucleotide segments with total lengths of about 10,000, about 5000, about 3000, about 2,000, about 1,000, about 500, about 200, about 100, about 50 base pairs in length, and the like, (including all intermediate lengths) are contemplated to be useful in many implementations of this invention.
  • two sequences are said to be “identical” if the sequence of nucleotides in the two sequences is the same when aligned for maximum correspondence, as described below. Comparisons between two sequences are typically performed by comparing the sequences over a comparison window to identify and compare local regions of sequence similarity.
  • a “comparison window” as used herein refers to a segment of at least about 20 contiguous positions, usually 30 to about 75, 40 to about 50, in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned.
  • Optimal alignment of sequences for comparison may be conducted using the Megalign program in the Lasergene suite of bioinformatics software (DNASTAR, Inc., Madison, Wis.), using default parameters.
  • This program embodies several alignment schemes described in the following references: Dayhoff, M. O. (1978) A model of evolutionary change in proteins—Matrices for detecting distant relationships. In Dayhoff, M. O. (ed.) Atlas of Protein Sequence and Structure, National Biomedical Research Foundation, Washington D.C. Vol. 5, Suppl. 3, pp. 345-358; Hein J. (1990) Unified Approach to Alignment and Phylogenes pp. 626-645 Methods in Enzymology vol.
  • optimal alignment of sequences for comparison may be conducted by the local identity algorithm of Smith and Waterman (1981) Add. APL. Math 2:482, by the identity alignment algorithm of Needleman and Wunsch (1970) J. Mol. Biol. 48:443, by the search for similarity methods of Pearson and Lipman (1988) Proc. Natl. Acad. Sci. USA 85: 2444, by computerized implementations of these algorithms (GAP, BESTFIT, BLAST, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group (GCG), 575 Science Dr., Madison, Wis.), or by inspection.
  • BLAST and BLAST 2.0 are described in Altschul et al. (1977) Nucl. Acids Res. 25:3389-3402 and Altschul et al. (1990) J. Mol. Biol. 215:403-410, respectively.
  • BLAST and BLAST 2.0 can be used, for example with the parameters described herein, to determine percent sequence identity for the polynucleotides of the invention.
  • Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information.
  • cumulative scores can be calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always >0) and N (penalty score for mismatching residues; always ⁇ 0). Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached.
  • the BLAST algorithm parameters W, T and X determine the sensitivity and speed of the alignment.
  • the “percentage of sequence identity” is determined by comparing two optimally aligned sequences over a window of comparison of at least 20 positions, wherein the portion of the polynucleotide sequence in the comparison window may comprise additions or deletions (i.e., gaps) of 20 percent or less, usually 5 to 15 percent, or 10 to 12 percent, as compared to the reference sequences (which does not comprise additions or deletions) for optimal alignment of the two sequences.
  • additions or deletions i.e., gaps
  • the percentage is calculated by determining the number of positions at which the identical nucleic acid bases occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the reference sequence (i.e., the window size) and multiplying the results by 100 to yield the percentage of sequence identity.
  • a mutagenesis approach such as site-specific mutagenesis, is employed for the preparation of immunogenic variants and/or derivatives of the polypeptides described herein.
  • site-specific mutagenesis By this approach, specific modifications in a polypeptide sequence can be made through mutagenesis of the underlying polynucleotides that encode them.
  • Site-specific mutagenesis allows the production of mutants through the use of specific oligonucleotide sequences which encode the DNA sequence of the desired mutation, as well as a sufficient number of adjacent nucleotides, to provide a primer sequence of sufficient size and sequence complexity to form a stable duplex on both sides of the deletion junction being traversed. Mutations may be employed in a selected polynucleotide sequence to improve, alter, decrease, modify, or otherwise change the properties of the polynucleotide itself, and/or alter the properties, activity, composition, stability, or primary sequence of the encoded polypeptide.
  • the inventors contemplate the mutagenesis of the disclosed polynucleotide sequences to alter one or more properties of the encoded polypeptide, such as the immunogenicity of a polypeptide vaccine.
  • the techniques of site-specific mutagenesis are well-known in the art, and are widely used to create variants of both polypeptides and polynucleotides.
  • site-specific mutagenesis is often used to alter a specific portion of a DNA molecule.
  • a primer comprising typically about 14 to about 25 nucleotides or so in length is employed, with about 5 to about 10 residues on both sides of the junction of the sequence being altered.
  • site-specific mutagenesis techniques have often employed a phage vector that exists in both a single stranded and double stranded form.
  • Typical vectors useful in site-directed mutagenesis include vectors such as the M13 phage. These phage are readily commercially-available and their use is generally well-known to those skilled in the art.
  • Double-stranded plasmids are also routinely employed in site directed mutagenesis that eliminates the step of transferring the gene of interest from a plasmid to a phage.
  • site-directed mutagenesis in accordance herewith is performed by first obtaining a single-stranded vector or melting apart of two strands of a double-stranded vector that includes within its sequence a DNA sequence that encodes the desired peptide.
  • An oligonucleotide primer bearing the desired mutated sequence is prepared, generally synthetically. This primer is then annealed with the single-stranded vector, and subjected to DNA polymerizing enzymes such as E. coli polymerase I Klenow fragment, in order to complete the synthesis of the mutation-bearing strand.
  • DNA polymerizing enzymes such as E. coli polymerase I Klenow fragment
  • sequence variants of the selected peptide-encoding DNA segments using site-directed mutagenesis provides a means of producing potentially useful species and is not meant to be limiting as there are other ways in which sequence variants of peptides and the DNA sequences encoding them may be obtained.
  • recombinant vectors encoding the desired peptide sequence may be treated with mutagenic agents, such as hydroxylamine, to obtain sequence variants.
  • mutagenic agents such as hydroxylamine
  • oligonucleotide directed mutagenesis procedure refers to template-dependent processes and vector-mediated propagation which result in an increase in the concentration of a specific nucleic acid molecule relative to its initial concentration, or in an increase in the concentration of a detectable signal, such as amplification.
  • oligonucleotide directed mutagenesis procedure is intended to refer to a process that involves the template-dependent extension of a primer molecule.
  • template dependent process refers to nucleic acid synthesis of an RNA or a DNA molecule wherein the sequence of the newly synthesized strand of nucleic acid is dictated by the well-known rules of complementary base pairing (see, for example, Watson, 1987).
  • vector mediated methodologies involve the introduction of the nucleic acid fragment into a DNA or RNA vector, the clonal amplification of the vector, and the recovery of the amplified nucleic acid fragment. Examples of such methodologies are provided by U.S. Pat. No. 4,237,224, specifically incorporated herein by reference in its entirety.
  • recursive sequence recombination as described in U.S. Pat. No. 5,837,458, may be employed.
  • iterative cycles of recombination and screening or selection are performed to “evolve” individual polynucleotide variants of the invention having, for example, enhanced immunogenic activity.
  • polynucleotide compositions comprising antisense oligonucleotides are provided.
  • Antisense oligonucleotides have been demonstrated to be effective and targeted inhibitors of protein synthesis, and, consequently, provide a therapeutic approach by which a disease can be treated by inhibiting the synthesis of proteins that contribute to the disease.
  • the efficacy of antisense oligonucleotides for inhibiting protein synthesis is well established. For example, the synthesis of polygalactauronase and the muscarine type 2 acetylcholine receptor are inhibited by antisense oligonucleotides directed to their respective mRNA sequences (U.S. Pat. No.
  • Antisense constructs have also been described that inhibit and can be used to treat a variety of abnormal cellular proliferations, e.g. cancer (U.S. Pat. No. 5,747,470; U.S. Pat. No. 5,591,317 and U.S. Pat. No. 5,783,683).
  • the present invention provides oligonucleotide sequences that comprise all, or a portion of, any sequence that is capable of specifically binding to polynucleotide sequence described herein, or a complement thereof.
  • the antisense oligonucleotides comprise DNA or derivatives thereof.
  • the oligonucleotides comprise RNA or derivatives thereof.
  • the oligonucleotides are modified DNAs comprising a phosphorothioated modified backbone.
  • the oligonucleotide sequences comprise peptide nucleic acids or derivatives thereof.
  • preferred compositions comprise a sequence region that is complementary, and more preferably substantially-complementary, and even more preferably, completely complementary to one or more portions of polynucleotides disclosed herein.
  • Selection of antisense compositions specific for a given gene sequence is based upon analysis of the chosen target sequence (i.e. in these illustrative examples the rat and human sequences) and determination of secondary structure, Tm, binding energy, relative stability, and antisense compositions were selected based upon their relative inability to form dimers, hairpins, or other secondary structures that would reduce or prohibit specific binding to the target mRNA in a host cell.
  • Highly preferred target regions of the mRNA are those which are at or near the AUG translation initiation codon, and those sequences which are substantially complementary to 5′ regions of the mRNA.
  • These secondary structure analyses and target site selection considerations can be performed, for example, using v.4 of the OLIGO primer analysis software and/or the BLASTN 2.0.5 algorithm software (Altschul et al., Nucleic Acids Res. 1997 Sep. 1;25(17):3389-402).
  • MPG short peptide vector
  • the MPG peptide contains a hydrophobic domain derived from the fusion sequence of HIV gp41 and a hydrophilic domain from the nuclear localization sequence of SV40 T-antigen (Morris et al., Nucleic Acids Res. 1997 Jul. 15;25(14):2730-6). It has been demonstrated that several molecules of the MPG peptide coat the antisense oligonucleotides and can be delivered into cultured mammalian cells in less than 1 hour with relatively high efficiency (90%). Further, the interaction with MPG strongly increases both the stability of the oligonucleotide to nuclease and the ability to cross the plasma membrane.
  • the polynucleotide compositions described herein are used in the design and preparation of ribozyme molecules for inhibiting expression of the tumor polypeptides and proteins of the present invention in tumor cells.
  • Ribozymes are RNA-protein complexes that cleave nucleic acids in a site-specific fashion. Ribozymes have specific catalytic domains that possess endonuclease activity (Kim and Cech, Proc Natl Acad Sci USA. 1987 December;84(24):8788-92; Forster and Symons, Cell. 1987 Apr. 24;49(2):211-20).
  • ribozymes accelerate phosphoester transfer reactions with a high degree of specificity, often cleaving only one of several phosphoesters in an oligonucleotide substrate (Cech et al., Cell. 1981 December;27(3 Pt 2):487-96; Michel and Westhof, J Mol Biol. 1990 Dec. 5;216(3):585-610; Reinhold-Hurek and Shub, Nature. 1992 May 14;357(6374):173-6).
  • This specificity has been attributed to the requirement that the substrate bind via specific base-pairing interactions to the internal guide sequence (“IGS”) of the ribozyme prior to chemical reaction.
  • IGS internal guide sequence
  • enzymatic nucleic acids act by first binding to a target RNA. Such binding occurs through the target binding portion of a enzymatic nucleic acid which is held in close proximity to an enzymatic portion of the molecule that acts to cleave the target RNA. Thus, the enzymatic nucleic acid first recognizes and then binds a target RNA through complementary base-pairing, and once bound to the correct site, acts enzymatically to cut the target RNA.
  • RNA Strategic cleavage of such a target RNA will destroy its ability to direct synthesis of an encoded protein. After an enzymatic nucleic acid has bound and cleaved its RNA target, it is released from that RNA to search for another target and can repeatedly bind and cleave new targets.
  • ribozyme The enzymatic nature of a ribozyme is advantageous over many technologies, such as antisense technology (where a nucleic acid molecule simply binds to a nucleic acid target to block its translation) since the concentration of ribozyme necessary to affect a therapeutic treatment is lower than that of an antisense oligonucleotide.
  • This advantage reflects the ability of the ribozyme to act enzymatically.
  • a single ribozyme molecule is able to cleave many molecules of target RNA.
  • the ribozyme is a highly specific inhibitor, with the specificity of inhibition depending not only on the base pairing mechanism of binding to the target RNA, but also on the mechanism of target RNA cleavage.
  • the enzymatic nucleic acid molecule may be formed in a hammerhead, hairpin, a hepatitis ⁇ virus, group I intron or RNaseP RNA (in association with an RNA guide sequence) or Neurospora VS RNA motif.
  • hammerhead motifs are described by Rossi et al. Nucleic Acids Res. 1992 Sep. 11;20(17):4559-65.
  • hairpin motifs are described by Hampel et al. (Eur. Pat. Appl. Publ. No. EP 0360257), Hampel and Tritz, Biochemistry 1989 Jun. 13;28(12):4929-33; Hampel et al., Nucleic Acids Res. 1990 Jan.
  • Ribozymes may be designed as described in Int. Pat. Appl. Publ. No. WO 93/23569 and Int. Pat. Appl. Publ. No. WO 94/02595, each specifically incorporated herein by reference) and synthesized to be tested in vitro and in vivo, as described. Such ribozymes can also be optimized for delivery. While specific examples are provided, those in the art will recognize that equivalent RNA targets in other species can be utilized when necessary.
  • Ribozyme activity can be optimized by altering the length of the ribozyme binding arms, or chemically synthesizing ribozymes with modifications that prevent their degradation by serum ribonucleases (see e.g., Int. Pat. Appl. Publ. No. WO 92/07065; Int. Pat. Appl. Publ. No. WO 93/15187; Int. Pat. Appl. Publ. No. WO 91/03162; Eur. Pat. Appl. Publ. No. 92110298.4; U.S. Pat. No. 5,334,711; and Int. Pat. Appl. Publ. No. WO 94/13688, which describe various chemical modifications that can be made to the sugar moieties of enzymatic RNA molecules), modifications which enhance their efficacy in cells, and removal of stem II bases to shorten RNA synthesis times and reduce chemical requirements.
  • Ribozymes may be administered to cells by a variety of methods known to those familiar to the art, including, but not restricted to, encapsulation in liposomes, by iontophoresis, or by incorporation into other vehicles, such as hydrogels, cyclodextrins, biodegradable nanocapsules, and bioadhesive microspheres.
  • ribozymes may be directly delivered ex vivo to cells or tissues with or without the aforementioned vehicles.
  • the RNA/vehicle combination may be locally delivered by direct inhalation, by direct injection or by use of a catheter, infusion pump or stent.
  • routes of delivery include, but are not limited to, intravascular, intramuscular, subcutaneous or joint injection, aerosol inhalation, oral (tablet or pill form), topical, systemic, ocular, intraperitoneal and/or intrathecal delivery. More detailed descriptions of ribozyme delivery and administration are provided in Int. Pat. Appl. Publ. No. WO 94/02595 and Int. Pat. Appl. Publ. No. WO 93/23569, each specifically incorporated herein by reference.
  • RNA polymerase I RNA polymerase I
  • RNA polymerase II RNA polymerase II
  • RNA polymerase III RNA polymerase III
  • Transcripts from pol II or pol III promoters will be expressed at high levels in all cells; the levels of a given pol II promoter in a given cell type will depend on the nature of the gene regulatory sequences (enhancers, silencers, etc.) present nearby.
  • Prokaryotic RNA polymerase promoters may also be used, providing that the prokaryotic RNA polymerase enzyme is expressed in the appropriate cells Ribozymes expressed from such promoters have been shown to function in mammalian cells.
  • Such transcription units can be incorporated into a variety of vectors for introduction into mammalian cells, including but not restricted to, plasmid DNA vectors, viral DNA vectors (such as adenovirus or adeno-associated vectors), or viral RNA vectors (such as retroviral, semliki forest virus, Sindbis virus vectors).
  • PNAs peptide nucleic acids compositions.
  • PNA is a DNA mimic in which the nucleobases are attached to a pseudopeptide backbone (Good and Nielsen, Antisense Nucleic Acid Drug Dev. 1997 7(4) 431-37).
  • PNA is able to be utilized in a number methods that traditionally have used RNA or DNA. Often PNA sequences perform better in techniques than the corresponding RNA or DNA sequences and have utilities that are not inherent to RNA or DNA.
  • a review of PNA including methods of making, characteristics of, and methods of using, is provided by Corey ( Trends Biotechnol 1997 June;15(6):224-9).
  • PNAs have 2-aminoethyl-glycine linkages replacing the normal phosphodiester backbone of DNA (Nielsen et al., Science 1991 Dec. 6;254(5037):1497-500; Hanvey et al., Science. 1992 Nov. 27;258(5087):1481-5; Hyrup and Nielsen, Bioorg Med Chem. 1996 January;4(1):5-23).
  • PNAs are neutral molecules; secondly, PNAs are achiral, which avoids the need to develop a stereoselective synthesis; and thirdly, PNA synthesis uses standard Boc or Fmoc protocols for solid-phase peptide synthesis, although other methods, including a modified Merrifield method, have been used.
  • PNA monomers or ready-made oligomers are commercially available from PerSeptive Biosystems (Framingham, Mass.). PNA syntheses by either Boc or Fmoc protocols are straightforward using manual or automated protocols (Norton et al., Bioorg Med Chem. 1995 April;3(4):437-45). The manual protocol lends itself to the production of chemically modified PNAs or the simultaneous synthesis of families of closely related PNAs.
  • PNAs can incorporate any combination of nucleotide bases
  • the presence of adjacent purines can lead to deletions of one or more residues in the product.
  • Modifications of PNAs for a given application may be accomplished by coupling amino acids during solid-phase synthesis or by attaching compounds that contain a carboxylic acid group to the exposed N-terminal amine.
  • PNAs can be modified after synthesis by coupling to an introduced lysine or cysteine. The ease with which PNAs can be modified facilitates optimization for better solubility or for specific functional requirements.
  • the identity of PNAs and their derivatives can be confirmed by mass spectrometry.
  • Several studies have made and utilized modifications of PNAs (for example, Norton et al., Bioorg Med Chem. 1995 April;3(4):437-45; Petersen et al., J Pept Sci.
  • U.S. Pat. No. 5,700,922 discusses PNA-DNA-PNA chimeric molecules and their uses in diagnostics, modulating protein in organisms, and treatment of conditions susceptible to therapeutics.
  • PNAs include use in DNA strand invasion, antisense inhibition, mutational analysis, enhancers of transcription, nucleic acid purification, isolation of transcriptionally active genes, blocking of transcription factor binding, genome cleavage, biosensors, in situ hybridization, and the like.
  • compositions of the present invention may be prepared and/or manipulated using any of a variety of well established techniques (see generally, Sambrook et al., Molecular Cloning: A Laboratory Manual , Cold Spring Harbor Laboratories, Cold Spring Harbor, N.Y., 1989, and other like references).
  • polynucleotide sequences or fragments thereof which encode polypeptides of the invention, or fusion proteins or functional equivalents thereof may be used in recombinant DNA molecules to direct expression of a polypeptide in appropriate host cells. Due to the inherent degeneracy of the genetic code, other DNA sequences that encode substantially the same or a functionally equivalent amino acid sequence may be produced and these sequences may be used to clone and express a given polypeptide.
  • codons preferred by a particular prokaryotic or eukaryotic host can be selected to increase the rate of protein expression or to produce a recombinant RNA transcript having desirable properties, such as a half-life which is longer than that of a transcript generated from the naturally occurring sequence.
  • polynucleotide sequences of the present invention can be engineered using methods generally known in the art in order to alter polypeptide encoding sequences for a variety of reasons, including but not limited to, alterations which modify the cloning, processing, and/or expression of the gene product.
  • DNA shuffling by random fragmentation and PCR reassembly of gene fragments and synthetic oligonucleotides may be used to engineer the nucleotide sequences.
  • site-directed mutagenesis may be used to insert new restriction sites, alter glycosylation patterns, change codon preference, produce splice variants, or introduce mutations, and so forth.
  • natural, modified, or recombinant nucleic acid sequences may be ligated to a heterologous sequence to encode a fusion protein.
  • a heterologous sequence For example, to screen peptide libraries for inhibitors of polypeptide activity, it may be useful to encode a chimeric protein that can be recognized by a commercially available antibody.
  • a fusion protein may also be engineered to contain a cleavage site located between the polypeptide-encoding sequence and the heterologous protein sequence, so that the polypeptide may be cleaved and purified away from the heterologous moiety.
  • Sequences encoding a desired polypeptide may be synthesized, in whole or in part, using chemical methods well known in the art (see Caruthers, M. H. et al. (1980) Nucl. Acids Res. Symp. Ser. 215-223, Horn, T. et al. (1980) Nucl. Acids Res. Symp. Ser. 225-232).
  • the protein itself may be produced using chemical methods to synthesize the amino acid sequence of a polypeptide, or a portion thereof.
  • peptide synthesis can be performed using various solid-phase techniques (Roberge, J. Y. et al. (1995) Science 269:202-204) and automated synthesis may be achieved, for example, using the ABI 431A Peptide Synthesizer (Perkin Elmer, Palo Alto, Calif.).
  • a newly synthesized peptide may be substantially purified by preparative high performance liquid chromatography (e.g., Creighton, T. (1983) Proteins, Structures and Molecular Principles, WH Freeman and Co., New York, N.Y.) or other comparable techniques available in the art.
  • the composition of the synthetic peptides may be confirmed by amino acid analysis or sequencing (e.g., the Edman degradation procedure). Additionally, the amino acid sequence of a polypeptide, or any part thereof, may be altered during direct synthesis and/or combined using chemical methods with sequences from other proteins, or any part thereof, to produce a variant polypeptide.
  • the nucleotide sequences encoding the polypeptide, or functional equivalents may be inserted into appropriate expression vector, i.e., a vector which contains the necessary elements for the transcription and translation of the inserted coding sequence.
  • appropriate expression vector i.e., a vector which contains the necessary elements for the transcription and translation of the inserted coding sequence.
  • Methods which are well known to those skilled in the art may be used to construct expression vectors containing sequences encoding a polypeptide of interest and appropriate transcriptional and translational control elements. These methods include in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination. Such techniques are described, for example, in Sambrook, J. et al.
  • a variety of expression vector/host systems may be utilized to contain and express polynucleotide sequences. These include, but are not limited to, microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid, or cosmid DNA expression vectors; yeast transformed with yeast expression vectors; insect cell systems infected with virus expression vectors (e.g., baculovirus); plant cell systems transformed with virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or with bacterial expression vectors (e.g., Ti or pBR322 plasmids); or animal cell systems.
  • microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid, or cosmid DNA expression vectors
  • yeast transformed with yeast expression vectors e.g., insect cell systems infected with virus expression vectors (e.g., baculovirus)
  • plant cell systems transformed with virus expression vectors e.g., cauliflower mosaic virus
  • control elements or “regulatory sequences” present in an expression vector are those non-translated regions of the vector—enhancers, promoters, 5′ and 3′ untranslated regions—which interact with host cellular proteins to carry out transcription and translation. Such elements may vary in their strength and specificity. Depending on the vector system and host utilized, any number of suitable transcription and translation elements, including constitutive and inducible promoters, may be used.
  • inducible promoters such as the hybrid lacZ promoter of the PBLUESCRIPT phagemid (Stratagene, La Jolla, Calif.) or PSPORT1 plasmid (Gibco BRL, Gaithersburg, Md.) and the like may be used.
  • promoters from mammalian genes or from mammalian viruses are generally preferred. If it is necessary to generate a cell line that contains multiple copies of the sequence encoding a polypeptide, vectors based on SV40 or EBV may be advantageously used with an appropriate selectable marker.
  • any of a number of expression vectors may be selected depending upon the use intended for the expressed polypeptide.
  • vectors which direct high level expression of fusion proteins that are readily purified may be used.
  • Such vectors include, but are not limited to, the multifunctional E. coli cloning and expression vectors such as BLUESCRIPT (Stratagene), in which the sequence encoding the polypeptide of interest may be ligated into the vector in frame with sequences for the amino-terminal Met and the subsequent 7 residues of .beta.galactosidase so that a hybrid protein is produced; pIN vectors (Van Heeke, G. and S. M.
  • pGEX Vectors may also be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST).
  • GST glutathione S-transferase
  • fusion proteins are soluble and can easily be purified from lysed cells by adsorption to glutathione-agarose beads followed by elution in the presence of free glutathione.
  • Proteins made in such systems may be designed to include heparin, thrombin, or factor XA protease cleavage sites so that the cloned polypeptide of interest can be released from the GST moiety at will.
  • yeast Saccharomyces cerevisiae
  • a number of vectors containing constitutive or inducible promoters such as alpha factor, alcohol oxidase, and PGH may be used.
  • constitutive or inducible promoters such as alpha factor, alcohol oxidase, and PGH
  • sequences encoding polypeptides may be driven by any of a number of promoters.
  • viral promoters such as the 35S and 19S promoters of CaMV may be used alone or in combination with the omega leader sequence from TMV (Takamatsu, N. (1987) EMBO J. 3:173-311.
  • plant promoters such as the small subunit of RUBISCO or heat shock promoters may be used (Coruzzi, G. et al. (1984) EMBO J. 3:1671-1680; Broglie, R. et al. (1984) Science 224:838-843; and Winter, J. et al. (1991) Results Probl.
  • An insect system may also be used to express a polypeptide of interest.
  • Autographa californica nuclear polyhedrosis virus (AcNPV) is used as a vector to express foreign genes in Spodoptera frugiperda cells or in Trichoplusia larvae.
  • the sequences encoding the polypeptide may be cloned into a non-essential region of the virus, such as the polyhedrin gene, and placed under control of the polyhedrin promoter. Successful insertion of the polypeptide-encoding sequence will render the polyhedrin gene inactive and produce recombinant virus lacking coat protein.
  • the recombinant viruses may then be used to infect, for example, S. frugiperda cells or Trichoplusia larvae in which the polypeptide of interest may be expressed (Engelhard, E. K. et al. (1994) Proc. Natl. Acad. Sci. 91:3224-3227).
  • a number of viral-based expression systems are generally available.
  • sequences encoding a polypeptide of interest may be ligated into an adenovirus transcription/translation complex consisting of the late promoter and tripartite leader sequence. Insertion in a non-essential E1 or E3 region of the viral genome may be used to obtain a viable virus which is capable of expressing the polypeptide in infected host cells (Logan, J. and Shenk, T. (1984) Proc. Natl. Acad. Sci. 81:3655-3659).
  • transcription enhancers such as the Rous sarcoma virus (RSV) enhancer, may be used to increase expression in mammalian host cells.
  • RSV Rous sarcoma virus
  • Specific initiation signals may also be used to achieve more efficient translation of sequences encoding a polypeptide of interest. Such signals include the ATG initiation codon and adjacent sequences. In cases where sequences encoding the polypeptide, its initiation codon, and upstream sequences are inserted into the appropriate expression vector, no additional transcriptional or translational control signals may be needed. However, in cases where only coding sequence, or a portion thereof, is inserted, exogenous translational control signals including the ATG initiation codon should be provided. Furthermore, the initiation codon should be in the correct reading frame to ensure translation of the entire insert. Exogenous translational elements and initiation codons may be of various origins, both natural and synthetic. The efficiency of expression may be enhanced by the inclusion of enhancers which are appropriate for the particular cell system which is used, such as those described in the literature (Scharf, D. et al. (1994) Results Probl. Cell Differ. 20:125-162).
  • a host cell strain may be chosen for its ability to modulate the expression of the inserted sequences or to process the expressed protein in the desired fashion.
  • modifications of the polypeptide include, but are not limited to, acetylation, carboxylation, glycosylation, phosphorylation, lipidation, and acylation.
  • Post-translational processing which cleaves a “prepro” form of the protein may also be used to facilitate correct insertion, folding and/or function.
  • Different host cells such as CHO, COS, HeLa, MDCK, HEK293, and WI38, which have specific cellular machinery and characteristic mechanisms for such post-translational activities, may be chosen to ensure the correct modification and processing of the foreign protein.
  • cell lines which stably express a polynucleotide of interest may be transformed using expression vectors which may contain viral origins of replication and/or endogenous expression elements and a selectable marker gene on the same or on a separate vector. Following the introduction of the vector, cells may be allowed to grow for 1-2 days in an enriched media before they are switched to selective media.
  • the purpose of the selectable marker is to confer resistance to selection, and its presence allows growth and recovery of cells which successfully express the introduced sequences.
  • Resistant clones of stably transformed cells may be proliferated using tissue culture techniques appropriate to the cell type.
  • any number of selection systems may be used to recover transformed cell lines. These include, but are not limited to, the herpes simplex virus thymidine kinase (Wigler, M. et al. (1977) Cell 11:223-32) and adenine phosphoribosyltransferase (Lowy, I. et al. (1990) Cell 22:817-23) genes which can be employed in tk.sup.- or aprt.sup.-cells, respectively. Also, antimetabolite, antibiotic or herbicide resistance can be used as the basis for selection; for example, dhfr which confers resistance to methotrexate (Wigler, M. et al. (1980) Proc.
  • npt which confers resistance to the aminoglycosides, neomycin and G-418 (Colbere-Garapin, F. et al (1981) J. Mol. Biol. 150:1-14); and als or pat, which confer resistance to chlorsulfuron and phosphinotricin acetyltransferase, respectively (Murry, supra). Additional selectable genes have been described, for example, trpB, which allows cells to utilize indole in place of tryptophan, or hisD, which allows cells to utilize histinol in place of histidine (Hartman, S. C. and R. C. Mulligan (1988) Proc.
  • marker gene expression suggests that the gene of interest is also present, its presence and expression may need to be confirmed.
  • sequence encoding a polypeptide is inserted within a marker gene sequence, recombinant cells containing sequences can be identified by the absence of marker gene function.
  • a marker gene can be placed in tandem with a polypeptide-encoding sequence under the control of a single promoter. Expression of the marker gene in response to induction or selection usually indicates expression of the tandem gene as well.
  • host cells that contain and express a desired polynucleotide sequence may be identified by a variety of procedures known to those of skill in the art. These procedures include, but are not limited to, DNA-DNA or DNA-RNA hybridizations and protein bioassay or immunoassay techniques which include, for example, membrane, solution, or chip based technologies for the detection and/or quantification of nucleic acid or protein.
  • Host cells transformed with a polynucleotide sequence of interest may be cultured under conditions suitable for the expression and recovery of the protein from cell culture.
  • the protein produced by a recombinant cell may be secreted or contained intracellularly depending on the sequence and/or the vector used.
  • expression vectors containing polynucleotides of the invention may be designed to contain signal sequences which direct secretion of the encoded polypeptide through a prokaryotic or eukaryotic cell membrane.
  • Other recombinant constructions may be used to join sequences encoding a polypeptide of interest to nucleotide sequence encoding a polypeptide domain which will facilitate purification of soluble proteins.
  • Such purification facilitating domains include, but are not limited to, metal chelating peptides such as histidine-tryptophan modules that allow purification on immobilized metals, protein A domains that allow purification on immobilized immunoglobulin, and the domain utilized in the FLAGS extension/affinity purification system (Immunex Corp., Seattle, Wash.).
  • metal chelating peptides such as histidine-tryptophan modules that allow purification on immobilized metals
  • protein A domains that allow purification on immobilized immunoglobulin
  • the domain utilized in the FLAGS extension/affinity purification system Immunex Corp., Seattle, Wash.
  • cleavable linker sequences such as those specific for Factor XA or enterokinase (Invitrogen. San Diego, Calif.) between the purification domain and the encoded polypeptide may be used to facilitate purification.
  • One such expression vector provides for expression of a fusion protein containing a polypeptide of interest and a nucleic acid encoding 6 histidine residues preceding a thioredoxin or an enterokinase cleavage site.
  • the histidine residues facilitate purification on IMIAC (immobilized metal ion affinity chromatography) as described in Porath, J. et al. (1992, Prot. Exp. Purif. 3:263-281) while the enterokinase cleavage site provides a means for purifying the desired polypeptide from the fusion protein.
  • IMIAC immobilized metal ion affinity chromatography
  • polypeptides of the invention may be produced by direct peptide synthesis using solid-phase techniques (Merrifield J. (1963) J. Am. Chem. Soc. 85:2149-2154). Protein synthesis may be performed using manual techniques or by automation. Automated synthesis may be achieved, for example, using Applied Biosystems 431A Peptide Synthesizer (Perkin Elmer). Alternatively, various fragments may be chemically synthesized separately and combined using chemical methods to produce the full length molecule.
  • the present invention in another aspect, provides T cells specific for a tumor polypeptide disclosed herein, or for a variant or derivative thereof.
  • T cells may generally be prepared in vitro or ex vivo, using standard procedures.
  • T cells may be isolated from bone marrow, peripheral blood, or a fraction of bone marrow or peripheral blood of a patient, using a commercially available cell separation system, such as the IsolexTM System, available from Nexell Therapeutics, Inc. (Irvine, Calif.; see also U.S. Pat. No. 5,240,856; U.S. Pat. No. 5,215,926; WO 89/06280; WO 91/16116 and WO 92/07243).
  • T cells may be derived from related or unrelated humans, non-human mammals, cell lines or cultures.
  • T cells may be stimulated with a polypeptide, polynucleotide encoding a polypeptide and/or an antigen presenting cell (APC) that expresses such a polypeptide.
  • APC antigen presenting cell
  • Such stimulation is performed under conditions and for a time sufficient to permit the generation of T cells that are specific for the polypeptide of interest.
  • a tumor polypeptide or polynucleotide of the invention is present within a delivery vehicle, such as a microsphere, to facilitate the generation of specific T cells.
  • T cells are considered to be specific for a polypeptide of the present invention if the T cells specifically proliferate, secrete cytokines or kill target cells coated with the polypeptide or expressing a gene encoding the polypeptide.
  • T cell specificity may be evaluated using any of a variety of standard techniques. For example, within a chromium release assay or proliferation assay, a stimulation index of more than two fold increase in lysis and/or proliferation, compared to negative controls, indicates T cell specificity. Such assays may be performed, for example, as described in Chen et al., Cancer Res. 54:1065-1070, 1994. Alternatively, detection of the proliferation of T cells may be accomplished by a variety of known techniques.
  • T cell proliferation can be detected by measuring an increased rate of DNA synthesis (e.g., by pulse-labeling cultures of T cells with tritiated thymidine and measuring the amount of tritiated thymidine incorporated into DNA).
  • a tumor polypeptide 100 ng/ml-100 ⁇ g/ml, preferably 200 ng/ml-25 ⁇ g/ml
  • 3-7 days will typically result in at least a two fold increase in proliferation of the T cells.
  • T cells that have been activated in response to a tumor polypeptide, polynucleotide or polypeptide-expressing APC may be CD4 + and/or CD8 + .
  • Tumor polypeptide-specific T cells may be expanded using standard techniques.
  • the T cells are derived from a patient, a related donor or an unrelated donor, and are administered to the patient following stimulation and expansion.
  • CD4 + or CD8 + T cells that proliferate in response to a tumor polypeptide, polynucleotide or APC can be expanded in number either in vitro or in vivo. Proliferation of such T cells in vitro may be accomplished in a variety of ways. For example, the T cells can be re-exposed to a tumor polypeptide, or a short peptide corresponding to an immunogenic portion of such a polypeptide, with or without the addition of T cell growth factors, such as interleukin-2, and/or stimulator cells that synthesize a tumor polypeptide. Alternatively, one or more T cells that proliferate in the presence of the tumor polypeptide can be expanded in number by cloning. Methods for cloning cells are well known in the art, and include limiting dilution.
  • the present invention concerns formulation of one or more of the polynucleotide, polypeptide or T-cell disclosed herein in pharmaceutically-acceptable solutions for administration to a cell or an animal, either alone, or in combination with one or more other modalities of therapy.
  • the present invention concerns, the use of cripto polynucleotides, polypeptides, fragments, fusions and variants in a pharmaceutical composition for the treatment of tumours.
  • the Cripto is Cripto 1.
  • compositions as disclosed herein may be administered in combination with other agents as well, such as, e.g., other proteins or polypeptides or various pharmaceutically-active agents.
  • agents such as, e.g., other proteins or polypeptides or various pharmaceutically-active agents.
  • additional agents do not cause a significant adverse effect upon contact with the target cells or host tissues.
  • the compositions may thus be delivered along with various other agents as required in the particular instance.
  • Such compositions may be purified from host cells or other biological sources, or alternatively may be chemically synthesized as described herein.
  • such compositions may further comprise substituted or derivatized RNA or DNA compositions.
  • compositions comprising one or more of the polynucleotide, polypeptide and/or T-cell compositions described herein in combination with a physiologically acceptable carrier.
  • the pharmaceutical compositions of the invention comprise immunogenic polynucleotide and/or polypeptide compositions of the invention for use in prophylactic and theraputic vaccine applications.
  • Vaccine preparation is generally described in, for example, M. F. Powell and M. J. Newman, eds., “Vaccine Design (the subunit and adjuvant approach),” Plenum Press (NY, 1995).
  • such compositions will comprise one or more polynucleotide and/or polypeptide compositions of the present invention in combination with one or more immunostimulants.
  • any of the pharmaceutical compositions described herein can contain pharmaceutically acceptable salts of the polynucleotides and polypeptides of the invention.
  • Such salts can be prepared, for example, from pharmaceutically acceptable non-toxic bases, including organic bases (eg., salts of primary, secondary and tertiary amines and basic amino acids) and inorganic bases (eg., sodium, potassium, lithium, ammonium, calcium and magnesium salts).
  • illustrative immunogenic compositions e.g., vaccine compositions, of the present invention comprise DNA encoding one or more of the polypeptides as described above, such that the polypeptide is generated in situ.
  • the polynucleotide may be administered within any of a variety of delivery systems known to those of ordinary skill in the art. Indeed, numerous gene delivery techniques are well known in the art, such as those described by Rolland, Crit. Rev. Therap. Drug Carrier Systems 15:143-198, 1998, and references cited therein. Appropriate polynucleotide expression systems will, of course, contain the necessary regulatory DNA regulatory sequences for expression in a patient (such as a suitable promoter and terminating signal).
  • bacterial delivery systems may involve the administration of a bacterium (such as Bacillus-Calmette-Guerrin) that expresses an immunogenic portion of the polypeptide on its cell surface or secretes such an epitope.
  • polynucleotides encoding immunogenic polypeptides described herein are introduced into suitable mammalian host cells for expression using any of a number of known viral-based systems.
  • retroviruses provide a convenient and effective platform for gene delivery systems.
  • a selected nucleotide sequence encoding a polypeptide of 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 minimizing 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).
  • 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.
  • Additional viral vectors useful for delivering the nucleic acid molecules encoding polypeptides of 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 novel molecules 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.
  • any of a number of alphavirus vectors can also be used for delivery of polynucleotide compositions of the present invention, such as those vectors described in U.S. Pat. Nos. 5,843,723; 6,015,686; 6,008,035 and 6,015,694.
  • Certain vectors based on Venezuelan Equine Encephalitis (VEE) can also be used, illustrative examples of which can be found in U.S. Pat. Nos. 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. Natl. Acad. Sci. USA (1992) 89:6099-6103, can also be used for gene delivery under the invention.
  • a polynucleotide is administered/delivered as “naked” DNA, for example as described in Ulmer et al., Science 259:1745-1749, 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, Wis.), some examples of which are described in U.S. Pat. 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.
  • the particles when delivering nucleic acid are preferably gold beads of a 0.4-4.0 um, more preferably 0.6-2.0 um diameter and the DNA conjugate coated onto these and then encased in a cartridge for placing into the “gene gun”.
  • the particles are typically and preferably delivered to the skin.
  • Other means of delivery to the skin comprise utilising needle delivery via a needle of a liquid formulation.
  • DNA vaccines usually consist of a bacterial plasmid vector into which is inserted a strong, normally viral, promoter, the gene of interest which encodes for an antigenic peptide and a polyadenylation/transcriptional termination sequences.
  • gene of interest may encode a full cripto protein as described or simply an antigenic peptide sequence such as described in seq ID no 13-94.
  • the plasmid can be grown in bacteria, such as for example E. coli and then isolated and prepared in an appropriate medium, depending upon the intended route of administration, before being administered to the host. Following administration the plasmid is taken up by cells of the host where the encoded peptide is produced.
  • the plasmid vector will preferably be made without an origin of replication which is functional in eukaryotic cells, in order to prevent plasmid replication in the mammalian host and integration within chromosomal DNA of the animal concerned.
  • DNA vaccination there are a number of advantages of DNA vaccination relative to traditional vaccination techniques. First, it is predicted that because of the proteins which are encoded by the DNA sequence are synthesised in the host, the structure or conformation of the protein will be similar to the native protein associated with the disease state. It is also likely that DNA vaccination will offer protection against different strains of a virus, by generating cytotoxic T lymphocyte response that recognise epitopes from conserved proteins. Furthermore, because the plasmids are taken up by the host cells where antigenic protein can be produced, a long-lasting immune response will be elicited. The technology also offers the possibility of combing diverse immunogens/epitopes into a single preparation.
  • compositions of the present invention include those provided by Bioject, Inc. (Portland, Oreg.), some examples of which are described in U.S. Pat. Nos. 4,790,824; 5,064,413; 5,312,335; 5,383,851; 5,399,163; 5,520,639 and 5,993,412.
  • the pharmaceutical compositions described herein will comprise one or more immunostimulants in addition to the immunogenic polynucleotide, polypeptide, T-cell and/or 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, Bordatella pertussis or Mycobacterium tuberculosis derived proteins.
  • adjuvants are commercially available as, for example, Freund's Incomplete Adjuvant and Complete Adjuvant (Difco Laboratories, Detroit, Mich.); Merck Adjuvant 65 (Merck and Company, Inc., Rahway, N.J.); 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.
  • GM-CSF interleukin-2, -7, -12, and other like growth factors
  • the adjuvant composition is preferably one that induces an immune response predominantly of the Th1 type.
  • High levels of Th1-type cytokines eg., IFN- ⁇ , TNF ⁇ , IL-2 and IL-12
  • high levels of Th2-type cytokines eg., IL-4, IL-5, IL-6 and IL-10
  • a patient will support an immune response that includes Th1- and Th2-type responses.
  • Th1-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 Th1-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, Wash.; see, for example, U.S. Pat. Nos. 4,436,727; 4,877,611; 4,866,034 and 4,912,094).
  • CpG-containing oligonucleotides in which the CpG dinucleotide is unmethylated also induce a predominantly Th1 response.
  • oligonucleotides are well known and are described, for example, in WO 96/02555, WO 99/33488 and U.S. Pat. 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, Mass.); 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® 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
  • 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, Calif., United States), ISCOMS (CSL), MF-59 (Chiron), the SBAS series of adjuvants (eg., SBAS-2 or SBAS-4, available from SmithKline Beecham, Rixensart, Belgium), Detox (Enhanzyn®) (Corixa, Hamilton, Mont.), RC-529 (Corixa, Hamilton, Mont.) and other aminoalkyl glucosaminide 4-phosphates (AGPs), such as those described in pending U.S. patent application Ser. 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 is 1-50
  • A is a bond or —C(O)—
  • R is C 1-50 alkyl or Phenyl C 1-50 alkyl.
  • One embodiment of the present invention consists of a vaccine formulation 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 1-2 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 th 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 preferred adjuvant combination is preferably with CpG as described in the pending UK patent application GB 9820956.2.
  • an immunogenic composition described herein 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-tumor 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 tumor and peritumoral 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 antitumor immunity (see Timmerman and Levy, Ann. Rev. Med. 50:507-529, 1999).
  • 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 na ⁇ ve 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, tumor-infiltrating cells, peritumoral 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 marrow may be differentiated into dendritic cells by adding to the culture medium combinations of GM-CSF, IL-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 correlates with the high expression of Fc ⁇ 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 (eg., CD54 and CD11) and costimulatory molecules (eg., CD40, CD80, CD86 and 4-1BB).
  • cell surface molecules responsible for T cell activation such as class I and class II MHC, adhesion molecules (eg., CD54 and CD11) and costimulatory molecules (eg., 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 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 tumor polypeptide, DNA (naked or within a plasmid vector) or RNA; or with antigen-expressing recombinant bacterium or viruses (eg., 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 (eg., a carrier molecule).
  • an immunological partner that provides T cell help eg., a carrier molecule.
  • a dendritic cell may be pulsed with a non-conjugated immunological partner, separately or in the presence of the polypeptide.
  • compositions of this invention 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. In 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 amphiphilic compound, such as a phospholipid (see eg., U.S. Pat. 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. Pat. 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 for many applications.
  • Another illustrative carrier/delivery system employs a carrier comprising particulate-protein complexes, such as those described in U.S. Pat. No. 5,928,647, which are capable of inducing a class I-restricted cytotoxic T lymphocyte responses in a host.
  • compositions of the invention will often further comprise one or more buffers (eg., 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 (eg., 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., 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., mannitol
  • 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 al., Nature 1997 Mar. 27;386(6623):410-4; Hwang et al., Crit Rev Ther Drug Carrier Syst 1998;15(3):243-84; U.S. Pat. No. 5,641,515; U.S. Pat. No. 5,580,579 and U.S. Pat. No. 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 corn starch, potato starch, alginic acid and the like; a lubricant, such as magnesium stearate; and a sweetening agent, such as sucrose, lactose or saccharin may be added or a flavoring agent, such as peppermint, oil of wintergreen, or cherry flavoring.
  • a binder such as gum tragacanth, acacia, cornstarch, or gelatin
  • excipients such as dicalcium phosphate
  • a disintegrating agent such as corn starch, potato starch, alginic acid and the like
  • a lubricant such as magnesium stearate
  • a sweetening agent such as sucrose, lactose
  • 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 of 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. Pat. No. 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.
  • the solution for parenteral administration in an aqueous solution, should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose.
  • aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration.
  • 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 may be dissolved in 1 ml of isotonic NaCl 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 Biologics 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.
  • 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., in U.S. Pat. No. 5,756,353 and U.S. Pat. No. 5,804,212.
  • the delivery of drugs using intranasal microparticle resins Takenaga et al., J Controlled Release 1998 Mar. 2;52(1-2):81-7) and lysophosphatidyl-glycerol compounds (U.S. Pat. No. 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. Pat. No. 5,780,045.
  • compositions of the present invention 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.
  • compositions of the present invention can be bound, either covalently or non-covalently, to the surface of such carrier vehicles.
  • 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 April;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. Furthermore, he use of 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 for pharmaceutically-acceptable nanocapsule formulations of the compositions of the present invention.
  • Nanocapsules can generally entrap compounds in a stable and reproducible way (see, for example, Quintanar-Guerrero et al., Drug Dev Ind Pharm. 1998 December;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.
  • the pharmaceutical compositions described herein may be used for the treatment of cancer, particularly for the immunotherapy of colon or colorectal cancer.
  • the compositions can be used to treat breast, or non-small cell lung carcinoma.
  • the composition will be useful for treating patients whose cancers express cripto antigen and/or whose metastases express cripto antigen.
  • the pharmaceutical compositions described herein are administered to a patient, typically a warm-blooded animal, preferably a human.
  • a patient may or may not be afflicted with cancer.
  • the above pharmaceutical compositions may be used to prevent the development of a cancer or to treat a patient afflicted with a cancer.
  • compositions and vaccines may be administered either prior to or following surgical removal of primary tumors and/or treatment such as administration of radiotherapy or conventional chemotherapeutic drugs.
  • administration of the pharmaceutical compositions may be by any suitable method, including administration by intravenous, intraperitoneal, intramuscular, subcutaneous, intranasal, intradermal, anal, vaginal, topical and oral routes.
  • immunotherapy may be active immunotherapy, in which treatment relies on the in vivo stimulation of the endogenous host immune system to react against tumors with the administration of immune response-modifying agents (such as polypeptides and polynucleotides as provided herein).
  • immune response-modifying agents such as polypeptides and polynucleotides as provided herein.
  • immunotherapy may be passive immunotherapy, in which treatment involves the delivery of agents with established tumor-immune reactivity (such as effector cells) that can directly or indirectly mediate antitumor effects and does not necessarily depend on an intact host immune system.
  • effector cells include T cells as discussed above, T lymphocytes (such as CD8+ cytotoxic T lymphocytes and CD4+ T-helper tumor-infiltrating lymphocytes), killer cells (such as Natural Killer cells and lymphokine-activated killer cells), B cells and antigen-presenting cells (such as dendritic cells and macrophages) expressing a polypeptide provided herein.
  • T cell receptors and antibody receptors specific for the polypeptides recited herein may be cloned, expressed and transferred into other vectors or effector cells for adoptive immunotherapy.
  • Effector cells may generally be obtained in sufficient quantities for adoptive immunotherapy by growth in vitro, as described herein.
  • Culture conditions for expanding single antigen-specific effector cells to several billion in number with retention of antigen recognition in vivo are well known in the art.
  • Such in vitro culture conditions typically use intermittent stimulation with antigen, often in the presence of cytokines (such as IL-2) and non-dividing feeder cells.
  • cytokines such as IL-2
  • immunoreactive polypeptides as provided herein may be used to rapidly expand antigen-specific T cell cultures in order to generate a sufficient number of cells for immunotherapy.
  • antigen-presenting cells such as dendritic, macrophage, monocyte, fibroblast and/or B cells
  • antigen-presenting cells may be pulsed with immunoreactive polypeptides or transfected with one or more polynucleotides using standard techniques well known in the art.
  • antigen-presenting cells can be transfected with a polynucleotide having a promoter appropriate for increasing expression in a recombinant virus or other expression system.
  • Cultured effector cells for use in therapy must be able to grow and distribute widely, and to survive long term in vivo.
  • a vector expressing a polypeptide recited herein may be introduced into antigen presenting cells taken from a patient and clonally propagated ex vivo for transplant back into the same patient.
  • Transfected cells may be reintroduced into the patient using any means known in the art, preferably in sterile form by intravenous, intracavitary, intraperitoneal or intratumor administration.
  • compositions and vaccines may be administered by injection (e.g., intracutaneous, intramuscular, intravenous, intradermally or subcutaneous), intranasally (e.g., by aspiration) or orally.
  • injection e.g., intracutaneous, intramuscular, intravenous, intradermally or subcutaneous
  • intranasally e.g., by aspiration
  • between 1 and 10 doses may be administered over a 52 week period.
  • 6 doses are administered, at intervals of 1 month, and booster vaccinations may be given periodically thereafter.
  • Alternate protocols may be appropriate for individual patients.
  • a suitable dose is an amount of a compound that, when administered as described above, is capable of promoting an anti-tumor immune response, and is at least 10-50% above the basal (i.e., untreated) level.
  • Such response can be monitored by measuring the anti-tumor antibodies in a patient or by vaccine-dependent generation of cytolytic effector cells capable of killing the patient's tumor cells in vitro.
  • Such vaccines should also be capable of causing an immune response that leads to an improved clinical outcome (e.g., more frequent remissions, complete or partial or longer disease-free survival) in vaccinated patients as compared to non-vaccinated patients.
  • the amount of each polypeptide present in a dose ranges from about 25 ⁇ g to 5 mg per kg of host. Suitable dose sizes will vary with the size of the patient, but will typically range from about 0.1 ml to about 5 ml.
  • an appropriate dosage and treatment regimen provides the active compound(s) in an amount sufficient to provide therapeutic and/or prophylactic benefit.
  • a response can be monitored by establishing an improved clinical outcome (e.g., more frequent remissions, complete or partial, or longer disease-free survival) in treated patients as compared to non-treated patients.
  • Increases in preexisting immune responses to a tumor protein generally correlate with an improved clinical outcome.
  • Such immune responses may generally be evaluated using standard proliferation, cytotoxicity or cytokine assays, which may be performed using samples obtained from a patient before and after treatment.
  • transcripto refers to polypeptides having about 188 amino acids and being expressed in carcinoma cells.
  • “cripto” refers to polypeptides having at least 70%, but preferably 80%, and more preferably 90%, and more preferably 95%, and yet most preferably 98% sequence identity to SEQ ID NOs: 3 or 4 or variants thereof, including Cripto 1 and Cripto 3.
  • Real-time RT-PCR (U. Gibson. 1996. Genome Research: 6,996) is used to compare mRNA transcript abundance of the target protein in a panel of normal and tumor tissues and/or cell lines. This analysis is critical to establish the tumor specificity of Cripto expression, which is an important criterion a good vaccine candidate must fulfil.
  • RNA is extracted from snap frozen biopsies or cell lines using TriPure reagent (Roche). Total RNA from normal tissues is also purchased from InVitrogen. 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 RiboGreen dye (Molecular Probes).
  • TaqMan primers forward primer sequence: TGGGTAGGAAAGAGGAAGCAAAT, SEQ ID NO:7; reverse primer sequence: TGCTTCTCTACCACCACCTAATCA, SEQ ID NO:8
  • probe for real-time RT-PCR amplification are designed with the Perkin-Elmer Primer Express software using default options for TaqMan amplification conditions.
  • Real-time reactions are assembled according to standard PCR protocols using 2 ng of reverse transcribed mRNA (Expand RT, Roche) for each reaction. Either SybrI or TaqMan detection is undergone, depending on the evaluated sample. In case of SybrI detection, SybrI dye (Molecular Probes) is added at a final dilution of ⁇ fraction (1/75000) ⁇ for real-time detection, and TaqMan probe is omitted. 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. Ct values are obtained from each tissue sample for the target mRNA (CtX) and for the actin mRNA (CtA).
  • CtX target mRNA
  • CtA actin mRNA
  • RT-PCR analysis using SybrI detection, was performed on a set of colon tumor and matched normal colon from 6 different patients and 48 normal tissue samples.
  • a TaqMan detection was run on a set of colon tumor and matched normal colon from 6 other patients (reactions were run in triplicates) and 48 normal tissue samples. Tested normal tissues (and the abbreviations used in graphics) are shown below:
  • Adrenal gland Adrenal gland
  • Brain Brain (Bra, Bra1, Bra2, Bra3, Bra4, Bra5)
  • Thyroid (Thyr, Thy, Thy1, Thy2)
  • CRL-5803 Carcinoma, Non-Small Cell Lung Cancer, large cell, neuroendocrine, metastatic site: lymph node
  • CRL-5810 Adocarcinoma, Non-Small Cell Lung Cancer
  • CRL-5865 (Adenocarcinoma, metastatic site: pleural effusion)
  • CRL-9609 Normal lung, bronchus, epithelial, virus transformed
  • HTB-177 Carcinoma, large cell lung cancer, pleural effusion
  • 2 colon cell lines
  • RT-PCR results on colorectal biopsies and normal tissues are shown in FIGS. 1, 2, 3 and 4, and in Table 1.
  • RT-PCR results on cell lines are shown in FIG. 5.
  • FIGS. 1, 2, 3 & 4 clearly show that Cripto, while being marginally expressed in normal adult tissues, is highly over-expressed in a majority of colorectal tumors, with an over-expression rate of more than ten fold.
  • FIG. 5 indicates Cripto is dramatically over-expressed in a lung tumor cell line (CRL-5815). Cripto tumor associated antigen is therefore a suitable vaccine candidate to treat both colorectal and lung cancer patients.
  • mRNA 5′ ends were decapped with TAP (Tobacco Acid Pyrophosphatase) and were ligated to a specific RNA oligonucleotide.
  • the ligated mRNA was reverse transcribed into cDNA using an oligod(T)-tailed primer.
  • Amplification of cDNA was performed using both GeneRacer flanking primers (Advantage, Clontech). Cripto amplification was performed on 10 ng of GeneRacer cDNA using gene specific PCR primers (forward primer sequence: CGTCCAAGGCCGAAAGCCCTCCAGTT, SEQ ID NO:9; and reverse primer sequence: TTGGGAGAGGGCAGGGCAAAGAAGTAAGAA, SEQ ID NO:10).
  • PCR reaction was done with Advantage II Taq DNA polymerase (Clontech) under standard conditions.
  • PCR product was cloned in pCR4-TOPO plasmid (Invitrogen). Amplified sequence (SEQ ID NO:95) was shown to display a variation at codon 22 (SEQ ID NO:96): Ala (GCC) instead of Val (GTC) in the native version (SeqID6). Native version was restored by PCR mutagenesis.
  • the immunogenicity of the antigen of the present invention can be verified by immunizing rabbits and mice using various means of immunization. Indeed, immunization with Cripto forms, either peptide or recombinant protein could induce humoral immune response with the generation of specific antibodies against Cripto and/or could induce a Cripto specific cellular immune response. In vivo delivery of Cripto protein using for instance, naked DNA in an appropriate vector encoding Cripto or fragments of Cripto, Cripto genes delivered by a viral vector encoding Cripto or fragments of Critpo, could also be useful to demonstrate Cripto immunogenicity.
  • the synthetic peptides from human Cripto-1 amino-acid sequence selected to immunize rabbits are GHQEFARPSRGYL (13 amino acids, SEQ ID NO:11), and QEEPAIRPRSSQRVPPMG (18 amino acids, SEQ ID NO:12). Synthetic peptides are then conjugated to a carrier protein (KLH). Conjugates are formulated with Freund's adjuvant, and two rabbits are immunized with each of the conjugates. Four weeks after the second immunization and four weeks after the third immunization, a blood sample is taken. Anti-Cripto antibody titers are estimated in the serum by ELISA and/or Western Blot following standard protocols (see section 3.5).
  • pcDNA3.1 vector (Invitrogen) is used to construct the vaccinating plasmid.
  • nucleic acid sequence encoding Cripto-1 with its own signal peptide is inserted into the vector polycloning site.
  • the recombinant expression plasmid is used to transform a host E. coli strain such as BL21.
  • the above recombinant strain is grown in conventional cell culture medium. Bacteria are harvested before reaching the stationary phase. Plasmid preparation using Quiagen system is undergone for injection in mice.
  • Recombinant adenoviruses are effective vectors for gene-based vaccination because they are capable of eliciting humoral and cellular immune responses against the encoded antigen.
  • the nucleotide sequence coding for Cripto-1 protein with its own signal sequence could be inserted in an appropriated Adeno derivative viral vector.
  • the adenoviral recombinant vector could be administrated to mice by different routes (intramuscular, intranasal, intradermal, subcutaneous or intraeperitoneal) After two week, blood samples could be taken and the titer of antibodies elicited examined. Additional experiments to measure the cellular immune response could also be performed.
  • Expression in microbial hosts is used to produce the whole protein or fragments of the invention antigen for immunization purposes.
  • 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.
  • the expression strategy first involves the design of the primary structure of the recombinant antigen.
  • an expression fusion partner EFP to improve levels of expression and/or an immune fusion partner (IFP) to modulate the immunogenic properties of the antigen, are placed at the N-terminal extremity.
  • an affinity fusion partner AFP useful for facilitating further purification is included at the C-terminal end.
  • the recombinant product is characterized by the evaluation of the level of expression and the prediction of further solubility of the engineered protein by analysis of its behavior in the crude extract.
  • a comparative evaluation of the different versions of the expressed antigen and expression hosts will allow the selection of the most promising candidate and host that is to be used for further purification and further immunological evaluation.
  • the purification schemes follow a classical approach based on the presence of a Histidine affinity tail in the recombinant protein.
  • the disrupted cells are filtered and the acellular extracts loaded onto an Ion Metal Affinity Chromatography (IMAC; Ni++NTA from Qiagen) that will specifically retain the recombinant protein.
  • IMAC 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.
  • Rabbits are immunized, intramuscularly several times at several week intervals with recombinant purified protein, formulated in the adjuvant 3D-MPL/QS21. Three weeks after each immunization, blood samples are taken. Anti-Cripto antibody titer is estimated in the serum by ELISA. The specificity of the anti-Cripto antibodies generated is tested by Western Blot (see section 3.5) using the purified protein and including appropriated controls.
  • ELISA reactions one of the above mentioned antigen is coated on microtiter plates (purified recombinant protein is preferred). ELISA is then performed using standard protocol.
  • the cellular response can also be assessed by stimulating in vitro vaccinated mouse spleen cells with the peptides used to immunize the mice, or with antigen-derived overlapping peptides covering the whole antigen sequence.
  • T cell lymphocytes, T cell lines and dendritic cells are derived from PBMCs (peripheral blood mononuclear cells) of healthy donors or cancer patients (preferred donors are from HLA-A2 subtype).
  • HLA Class I binding peptide sequences are predicted either by the Parker's algorithm (Parker, et al. J. Immunol. 152:163 (1994)) (http://bimas.dcrt.nih.gov/molbio/hla_bind/), and the Rammensee method (Rammensee et al. (1997). Austin Bioscience (1997); Rammensee et al., Immunogenetics 41,178-228 (1995)). (http://syfpeithi.bmi-heidelberg.com/Scripts/MHCServer.dll/EpPredict.htm).
  • HLA Class II binding peptide sequences are predicted using the Tepitope algorithm (Sturniolo et al. Nat Biotechnol. 17: 555-61 (1999).
  • HLA-A2.1/Kb transgenic mouse model is used for screening of the HLA-A2.1 peptides. Briefly, transgenic mice are immunized with adjuvanted HLA-A2 peptides, those able to induce a CD8 response (as defined by an efficient lysis or g-IFN production on peptide-pulsed target cells) are further analyzed in the human system.
  • Human dendritic cells (cultured according to Romani et al. J. Exp. Med. 180:83-93(1994)) will be pulsed with the selected peptides and used to stimulate CD8+-sorted T cells (by Facs). After several weekly stimulation, the CD8+ lines are 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 are then tested on cDNA-transfected tumor cells (HLA-A2 transfected LnCaP, Skov3 or CAMA tumor cells).
  • cDNA-transfected tumor cells HLA-A2 transfected LnCaP, Skov3 or CAMA tumor cells.
  • CD8+ T cell lines are primed and stimulated with either gene-gun transfected dendritic cells, retrovirally transduced B7.1-transfected fibroblasts, recombinant pox virus (Kim, et al., J. Immunother. 20:276-286 (1997)) or adenovirus (Butterfield, et al. J. Immunol. 161:5607-13 (1998)). 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 tumor cells as indicated above. Peptide specificity and identity is determined to confirm the immunological validation of antigen of the present invention.
  • 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.
  • A-2 Cripto-1 specific class I epitopes start HLA allele Epitope size position Sequence Score SEQ ID NO: A0201 Nonamer 5 KMARFSYSV 668.086 SEQ ID NO:31 A0201 Decamer 16 IMAISKVFEL 349.885 SEQ ID NO:32 A0201 Decamer 13 VIWIMAISKV 310.361 SEQ ID NO:33 A0201 Decamer 175 FMLVGICLSI 128.242 SEQ ID NO:34 B2702 Nonamer 7 ARFSYSVIW 500.000 SEQ ID NO:35 B2702 Nonamer 3 CRKMARFSY 200.000 SEQ ID NO:36 B2702 Decamer 7 ARFSYSVIWI 300.000 SEQ ID NO:37 B2702 Decamer 37 ARPSRGYLAF 200.000 SEQ ID NO:38 B2705 Nonamer 7 ARFSYSVIW 1000.000 SEQ ID NO:35 B2705 Nonamer 3 CRKMARFSY 1000.000 SEQ ID NO:36 B2705
  • a series of experiments aimed at assessing the inhibitory effect of the Cripto-specific humoral response on the pathophysiological activities of Cripto in cancer could be carried out by using various standards in vitro assays.
  • the in vitro assays could be, but not restricted to, growth inhibition assay, cell motility inhibition assay, chemotaxis inhibition assay, inhibition of the invasion through extracellular matrix protein (ECM), and growth signal transduction pathway inhibition.
  • ECM extracellular matrix protein
  • adenoviral vector, encoding the Cripto protein could be assessed in these in vitro assays that gauge Cripto-mediated biological effects on Cripto-expressing cell lines. In parallel, the effect of the pre-immune sera from the same animals will be tested as negative control.
  • the cell lines used in these assays could be, but not limited to, human tumor cells expressing Cripto such as GEO cells, NTERA2 cells, the CRL-5815 cell line, or murine tumor cell lines that naturally over-express Cripto.
  • mice or rabbits would be immunized on day 0, 14, and 21 by intra-footpad injections of either peptides or protein in adjuvant, intra-dermal injections of a plasmid DNA encoding Cripto using gene-gun devices, or intra-dermal injections of a viral vector delivery system, e.g. adenoviral vector, encoding Cripto.
  • a viral vector delivery system e.g. adenoviral vector, encoding Cripto.
  • the sera from immunized animals will be collected and added at different dilutions to the culture medium of cells platted in 96-well plates. Similarly, pre-immune sera of these animals will also be added to cells as negative control. The cells will be treated for 3 to 7 days. The cell growth will be measured by standard methods such as 3 H-thymidine incorporation assay, MTT assay, crystal violet staining, or colony counting for proliferation in soft agar-medium.
  • the sera from immunized animals will be collected pre- and post-immunization and added at different dilutions to cell suspensions used for invasion, motility, and chemotaxis standards assays.
  • the inhibition of Cripto-mediated invasion, motility, and chemotaxis could be assessed with the use of, for instance, but limited to, commercially available Falcon chambers with matrigel inserts (Collaborative Research), agarose droplet motility assay (Yamamoto et al. Ophthalmology 97: 1204-1210 (1994)), and commercially available Boyden apparatus (Neuro Probe), respectively.
  • the sera from immunized animals will be collected pre- and post-immunization and added at different dilutions to Cripto-expressing tumor cells in culture.
  • Exogenous Cripto protein or human sera or milk containing the highest concentration of Cripto detected by ELISA (Bianco, et al. Breast Cancer Res Treat 66:1-7 (2001)) will be added to the culture medium of the cells. After various incubation times, the cells will be harvested and processed by standard methods in order to perform immunoblot analysis. The phosphorylation status of various key signal transduction pathways involved in cell proliferation will be assessed in these Cripto-simulated cells in the presence of pre- or post-immune sera.
  • MAP mitogen-activated protein
  • the prophylactic or therapeutic potential of vaccines containing the human Cripto protein, Cripto peptides, or Cripto gene can be evaluated in mice challenged with syngeneic murine tumor cell line that express Cripto.
  • the tumor cell lines could be a murine tumor cell line transfected with the human Cripto gene.
  • the transfected cell lines could be the TC1 cell line transfected with human Cripto gene.
  • the high level of homology between human Cripto and its murine homologue suggests that cross-reactive immune responses induced by the human vaccine can protect against mouse Cripto-expressing tumors.
  • Cripto gene encodes a 171-amino acid protein which has 93% identity with its human counterpart (Liguori, et al Mamm Genome 7:344-348 (1996)). Therefore, mice could be protected by immunization with a human Critpo vaccine from murine tumor challenge or spontaneously arising tumor known to endogenously over-express the Cripto protein. With this respect, spontaneous tumors in transgenic mice designed to over-express several different oncogenes such as MMTV-Polyoma virus middle T antigen, MMTV-c-ErbB2, and MT-hTGF alpha, have been shown also to over-express Cripto. (Kenney, et al. Mol Carcinog 15: 44-56 (1996); Niemeye, et al. Int. J Cancer 81:588-591 (1999)).
  • the tumor-bearing mice could be vaccinated with a plasmid DNA or a viral delivery system, e.g. adenoviral vector, encoding the murine Cripto protein to protect from murine tumor growth.
  • a viral delivery system e.g. adenoviral vector
  • mice would be vaccinated on day 0 and 14, prior tumor challenge, by either intra footpad injections of 5 ⁇ g of Cripto protein in adjuvant, intra-dermal injections of DNA plasmid encoding Cripto using gene-gun technology, or intra-dermal injections of a viral vector, e.g. adenoviral vector, encoding Cripto. Then, 10 6 TC1-CR-1 cells, human Cripto-expressing tumor cells, could be injected subcutaneously in the flank of C57BL/6 immunocompetent mice 1 or 2 weeks post vaccination. The tumor growth should be monitored in vivo by measuring individual tumors twice a week for several weeks post-tumor challenge.
  • a viral vector e.g. adenoviral vector
  • the efficacy of prophylactic vaccination could be assessed by inhibiting the development of spontaneous Cripto-expressing tumors in transgenic mice (Niemeye, et al. Int J Cancer 81:588-591 (1999)) immunized multiple times before onset of the palpable tumor with either form of Cripto vaccines.
  • mice 10 6 TC1-CR-1 cells, human-Critpo expressing tumor cells, would be injected subcutaneously in the flank of C57BL/6 immunocompetent mice. Mice could be vaccinated on days 7 and 14, post-tumor challenge, by either intra-footpad injections of 5 ⁇ g Cripto protein in adjuvant, intra-dermal injections of DNA plasmid encoding Cripto using gene-gun technology, or intra-dermal injections of a viral vector, e.g. adenoviral vector, encoding Cripto. The tumor growth should be monitored in vivo by measuring individual tumors twice a week.
  • a viral vector e.g. adenoviral vector
  • mice per group will be sacrificed to harvest spleen cells, draining lymph nodes, and sera for analysis of the immune responses to establish a correlation between the induction of a Cripto-specific immune response and the antitumor effect.
  • the analysis of the Cripto-specific immune response induced by immunization could be assessed by measuring the antibody titers, antibody isotypic profile, the CD4 + T-cell proliferation response, and CTL CD8 + T-cell responses including cytokines production and lysis activity against Cripto-expressing target cells. All assays would be performed according to standards protocols.
  • This experiment demonstrates that two selected polypeptide fragments of Cripto (SEQ ID NO:11 and SEQ ID NO:12) mixed with a vaccine adjuvant induces Cripto peptide specific antibody response in immunized animals. Furthermore, the assessment of the recognition of Cripto protein shows that antibodies recognize the tumour-associated antigen itself. Finally, an immunostaining study on cancer tissues using anti-Cripto antibodies shows Cripto-specific immune response to target cripto naturally expressed in cancer cells.
  • Two peptides were selected and prepared from the amino acid sequence of human Cripto-1: a 13 mer synthetic peptide (GHQEFARPSRGYL—SEQ ID NO:11) that corresponds to amino acids 32 to 44 and an 18 mer synthetic peptide (QEEPAIRPRSSQRVPPMG—SEQ ID NO:12) that corresponds to amino acids 54 to 71 of human Cripto-1 sequence. These peptides were coupled to KLH using gluteraldehyde.
  • the KLH-coupled synthetic peptides were used to immunize two rabbits for each synthetic peptide. Antibodies were produced in rabbits during 3 months according to the protocol described in section 3.1 of the specification. Antisera were tested by ELISA against the peptide and the carrier protein.
  • E. coli recombinant protein Cripto-1 (18 Kd) was detected with the sera of rabbits injected with the synthetic peptides aa32->44 (SEQ ID NO:11) or aa54->71 (SEQ ID NO:12).
  • Coommassie Blue stained polyacrylamide gel indicated the presence of a polypeptide ( ⁇ 18 kD). No polypeptide having a molecular weight of 18 kD was detected by antibodies, antisera, or Coommassie Blue stain in the negative control or cellular extract supernatant.
  • SCID mice subcutaneous xenograft tumors (mRNA cripto-expressing CRL5815 cell line—human small cell lung carcinoma—expression determined by real time PCR methods) were incubated in buffered formalin fixative for 24 h at room temperature and paraffin embedded. Five micron thick sections were attached to superfrost glass slides and deparaffinized. Prior to antibody binding, the sections were heated in glyca buffer (Biogenex) (microwave for 10 min), then saturated with PBS buffer containing 0.5% blocking reagent (Bo ⁇ rhinger) for 30 min. Two rabbit anti-human CR-1 peptides (32-44; 54-71) were separately added (dil.
  • Cripto-1 peptide was detected using rabbit anti-human CR-1 peptide 54-71 (dil. ⁇ fraction (1/50) ⁇ ) (SEQ ID NO:12) by immunostaining. The immunostaining specificity was confirmed by preabsorption of the anti-CR-1 peptide (54-71) antibody with the corresponding purified Cripto-1 peptide (50 ⁇ g/ml).
  • Cripto peptides formulated in vaccine adjuvant are immunogenic in animals. Antibodies raised in immunized rabbits may recognize not only the peptide used as immunogen, but also a recombinant Cripto protein expressed in E. coli and Cripto naturally expressed in tumour tissue. Cripto is a suitable target for active immunotherapy.
  • Serum from vaccinated mice was taken at day 0 and 42, for Western blot analyses for the presence of anti-cripto antibodies. Proteins contained in the solubilized pellet of an acellular extract, were separated by SDS-PAGE and used to test the sera of mice by Western blot. The pre-immune and post-immune serum (42 days post-immunisation) of eight mice, and a pool of the negative control group sera were tested at the dilution ⁇ fraction (1/500) ⁇ .

Abstract

Compositions and methods for the therapy and diagnosis of cancer, particularly lung, colon, colorectal and breast cancer, are disclosed. Illustrative compositions comprise one or more Cripto tumor polypeptides, immunogenic portions thereof, polynucleotides that encode such polypeptides, antigen presenting cell that expresses such polypeptides, and T cells that are specific for cells expressing such polypeptides. The disclosed compositions are useful, for example, in the diagnosis, prevention and/or treatment of diseases, particularly lung, colon, colorectal and breast cancer.

Description

    CROSS REFERENCES
  • This application is a continuation-in-part of international application PCT/EP01/09646, filed 20 Aug. 2000, and filed in the United States on 20 Aug. 2000, application Ser. No. 10/362,597, the entirety of which is incorporated herein by reference.[0001]
  • TECHNICAL FIELD OF THE INVENTION
  • The present invention relates generally to therapy of cancer, such as colon, colorectal, breast, bladder, lung and endometrial cancer. The invention is more specifically related to polypeptides, comprising at least a portion of a cripto protein tumor protein, and to polynucleotides encoding such polypeptides, in particular pharmaceutical compositions, e.g., vaccines, and other compositions for the treatment of cancer that are cripto-expressing carcinomas such as certain non-small long cell carcinoma, breast, colon, colorectal cancer. [0002]
  • BACKGROUND OF THE INVENTION
  • CRIPTO is a 188 amino acid protein shares homologies with the epidermal growth factor (EGF) family (EMBO Journal (1989) Vol 8 (7) pp1987-1991). [0003]
  • huCRIPTO mRNA is detected only in undifferentiated cells and disappear after cell differentiation mCRIPTO is expressed during pregnancy and lactation (induces branching morphogenesis in mammary epithelial cells) and is suspected to be an autocrine growth factor for normal breast cells. CRIPTO is required for correct orientation of the anterior-posterior axis in the mouse embryo. [0004]
  • Human cripto gene has been expressed U.S. Pat. No. 5,654,140 herein incorporated by reference in its entirety. [0005]
  • Cancer is a significant health problem throughout the world. Although advances have been made in detection and therapy of cancer, no vaccine or other universally successful method for prevention and/or treatment is currently available. Current therapies, which are generally based on a combination of chemotherapy or surgery and radiation, continue to prove inadequate in many patients. [0006]
  • Colon cancer is the second most frequently diagnosed malignancy in the United Sates as well as the second most common cause of cancer death. An estimated 95,600 new cases of colon cancer will have been diagnosed in 1998, with an estimated 47,700 deaths. The five-year survival rate for patients with colorectal cancer detected in an early-localised stage is 92%, unfortunately, only 37% of colorectal cancer is diagnosed at this stage, the survival stage. The survival rate drops to 64% if the cancer is allowed to spread to adjacent organs or lymph nodes, and to 7% in patients with distant metastases. [0007]
  • The prognosis of colon cancer is directly related to the degree of penetration of the tumour through the bowel wall, to the level of metastasis, to the presence or absence of nodal involvement, consequently, early detection and treatment are especially important. Currently, diagnosis is aided by the use of screening assays for fecal occult blood, sigmoidoscopy, colonoscopy and double contrast barium enemas. Treatment regimens are determined by the type and stage of the cancer, and include surgery, radiation therapy and/or chemotherapy. Recurrence following surgery (the most common form of therapy) is a major problem and is often the ultimate cause of death. In spite of considerable research into therapies for the disease, colon cancer remains difficult to diagnose and treat. Accordingly, there is a need in the art for improved methods for treating such cancers. The present invention fulfils these needs and further provides other related advantages. [0008]
  • Breast cancer is a significant health problem for women in the United States and throughout the world. Although advances have been made in detection and treatment of the disease, breast cancer remains the second leading cause of cancer-related deaths in women, affecting more than 180,000 women in the United States each year. For women in North America, the lifetime odds of getting breast cancer are now one in eight. [0009]
  • No vaccine or other universally successful method for the prevention or treatment of breast cancer is currently available. Management of the disease currently relies on a combination of early diagnosis (through routine breast screening procedures) and aggressive treatment which may including one or more of a variety of treatments such as surgery, radiotherapy, chemotherapy and hormone therapy. The course of treatment for a particular breast cancer is often selected based on a variety of prognostic parameters, including an analysis of specific tumour markers. See, eg Porter-Jordan and Lippman, Breast Cancer 8:73-100 (1994). However, the use of established markers often leads to a result that this is difficult to interpret, and the high mortality observed in breast cancer patients indicates that improvements are needed in the treatment and prevention of the disease. [0010]
  • Lung cancer is the primary cause of cancer death among both men and women in the US, with an estimated 172,000 new cases being reported in 1994. The five-year survival rate among all lung cancer patients, regardless of the state of disease at diagnosis is only 13%. This contrasts with a five-year survival rate of 46% among cases detected while the disease is still localised. However, only 16% of lung cancers are discovered before the disease has spread. [0011]
  • In spite of considerable research into therapies for these and other cancers, breast, colon and colorectal remains difficult to diagnose and treat effectively. Accordingly, there is a need in the art for improved methods for treating and preventing such cancers. The present invention fulfills these needs and further provides other related advantages. [0012]
  • SUMMARY OF THE INVENTION
  • In one aspect, the present invention provides a method for inhibiting the development of a cancer in a patient, comprising the steps of: [0013]
  • (a) incubating CD4+ and/or CD8+ T cells isolated from a patient with at least one component selected from the group consisting of SEQ ID NO:11 or SEQ ID NO:12; and [0014]
  • (b) administering to the patient an effective amount of the T cells, and thereby inhibiting the development of a cancer in the patient. [0015]
  • In another aspect, the invention provides a method for producing an immunogenic response to a tumor-associated antigen in an animal comprising administering a first component comprising a polynucleotide encoding a fragment of the tumor-associated antigen to the animal. [0016]
  • In another aspect, the invention provides a method of inducing an immunoresponse to Cripto in an animal comprising repeatedly administering a composition comprising a first component comprising SEQ ID NO:11 or SEQ ID NO:12. [0017]
  • The present invention, in another aspect, provides polypeptide compositions comprising an amino acid sequence that is encoded by a polynucleotide sequence described above. [0018]
  • The polypeptides and/or polynucleotides of the present invention are immunogenic, i.e., they are capable of eliciting an immune response, particularly a humoral and/or cellular immune response, when if necessary, they are conjugated to a suitable carrier and/or adjuvanted. [0019]
  • The present invention further provides fragments, variants and/or derivatives of the disclosed polypeptide and/or polynucleotide sequences, wherein the fragments, variants and/or derivatives preferably 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 set forth in SEQ ID No: 2 or 4 or a polypeptide sequence encoded by a polynucleotide sequence set forth in SEQ ID NO: 1 or 3. [0020]
  • The present invention further provides polynucleotides that encode a polypeptide described above, expression vectors comprising such polynucleotides and host cells transformed or transfected with such expression vectors. [0021]
  • Within other aspects, the present invention provides pharmaceutical compositions comprising a polypeptide or polynucleotide as described above and a physiologically acceptable carrier. [0022]
  • Within a related aspect of the present invention, the pharmaceutical compositions, e.g., vaccine compositions, are provided for prophylactic or therapeutic applications. Such compositions generally comprise an immunogenic polypeptide or polynucleotide of the invention and an immunostimulant, such as an adjuvant. [0023]
  • Within further aspects, the present invention provides pharmaceutical compositions comprising: (a) an antigen presenting cell that expresses a polypeptide as described above and (b) a pharmaceutically acceptable carrier or excipient. Illustrative antigen presenting cells include dendritic cells, macrophages, monocytes, fibroblasts and B cells. [0024]
  • Within related aspects, pharmaceutical compositions are provided that comprise: (a) an antigen presenting cell that expresses a polypeptide as described above and (b) an immunostimulant. [0025]
  • The present invention further provides, in other aspects, fusion proteins that comprise at least one polypeptide as described above, as well as polynucleotides encoding such fusion proteins, typically in the form of pharmaceutical compositions, e.g., vaccine compositions, comprising a physiologically acceptable carrier and/or an immunostimulant. The fusion proteins may comprise multiple immunogenic polypeptides or portions/variants thereof, as described herein, and may further comprise one or more polypeptide segments for facilitating the expression, purification and/or immunogenicity of the polypeptide(s). [0026]
  • Within further aspects, the present invention provides methods for stimulating an immune response in a patient, preferably a T cell response in a human patient, comprising administering a pharmaceutical composition described herein. The patient may be afflicted with lung or colon cancer or colorectal cancer or breast cancer, in which case the methods provide treatment for the disease, or patient considered at risk for such a disease may be treated prophylactically. In particular the patient will be afflicted with a tumour expressing cripto antigens. [0027]
  • Within further aspects, the present invention provides methods for inhibiting the development of a cancer in a patient, comprising administering to a patient a pharmaceutical composition as recited above. The patient may be afflicted with lung, colon, colorectal or breast cancer, in which case the methods provide treatment for the disease, or patient considered at risk for such a disease may be treated prophylactically. [0028]
  • The present invention further provides, within other aspects, methods for removing tumor cells from a biological sample, comprising contacting a biological sample with T cells that specifically react with a polypeptide of the present invention, wherein the step of contacting is performed under conditions and for a time sufficient to permit the removal of cells expressing the protein from the sample. [0029]
  • Within related aspects, methods are provided for inhibiting the development of a cancer in a patient, comprising administering to a patient a biological sample treated as described above. [0030]
  • Methods are further provided, within other aspects, for stimulating and/or expanding T cells specific for a polypeptide of the present invention, comprising contacting T cells with one or more of: (i) a polypeptide as described above; (ii) a polynucleotide encoding such a polypeptide; and/or (iii) an antigen presenting cell that expresses such a polypeptide; under conditions and for a time sufficient to permit the stimulation and/or expansion of T cells. Isolated T cell populations comprising T cells prepared as described above are also provided. [0031]
  • Within further aspects, the present invention provides methods for inhibiting the development of a cancer in a patient, comprising administering to a patient an effective amount of a T cell population as described above. [0032]
  • The present invention further provides methods for inhibiting the development of a cancer in a patient, comprising the steps of: (a) incubating CD4+ and/or CD8+ T cells isolated from a patient with one or more of: (i) a polypeptide comprising at least an immunogenic portion of a cripto polypeptide disclosed herein; (ii) a polynucleotide encoding such a polypeptide; and (iii) an antigen-presenting cell that expressed such a polypeptide; and (b) administering to the patient an effective amount of the proliferated T cells, and thereby inhibiting the development of a cancer in the patient. Proliferated cells may, but need not, be cloned prior to administration to the patient. [0033]
  • These and other aspects of the present invention will become apparent upon reference to the following detailed description. All references disclosed herein are hereby incorporated by reference in their entirety as if each was incorporated individually. [0034]
  • BRIEF DESCRIPTION OF THE DRAWINGS AND SEQUENCE IDENTIFIERS
  • SEQ ID NO: 1 Cripto 1 Polynucleotide [0035]
  • SEQ ID NO: 2 Cripto 3 Polynucleotide [0036]
  • SEQ ID NO: 2 Cripto 3 Polypeptide1 [0037]
  • SEQ ID NO: 3 Cripto 1 Polypeptide [0038]
  • SEQ ID NO: 4 Cripto 3 Polypeptide [0039]
  • SEQ ID NO: 5 Cripto Polynucleotide as described in U.S. Pat. No. 5,654,140 [0040]
  • SEQ ID NO: 6 Cripto Polynucleotide as described in U.S. Pat. No. 5,654,140 [0041]
  • SEQ ID NOS: 7-10 PCR primers [0042]
  • SEQ ID NOS: 11 & 12 synthetic Cripto 1 peptides [0043]
  • SEQ ID NOS: 13-94 Epitopes from Cripto 1 and 3 [0044]
  • SEQ ID NO: 95 Cripto 1 variant Polynucleotide [0045]
  • SEQ ID NO: 96 Cripto 1 variant Polypeptide[0046]
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention is directed generally to compositions and their use in the therapy and diagnosis of cancer, particularly cripto expressing cancer and metastases including Cripto expressing lung, colon, colorectal and breast cancers. As described further below, illustrative compositions of the present invention include, but are not restricted to, polypeptides, particularly immunogenic polypeptides, polynucleotides encoding such polypeptides, antibodies and other binding agents, antigen presenting cells (APCs) and immune system cells (e.g., T cells). [0047]
  • The present invention provides a method for inhibiting the development of a cancer in a patient, comprising the steps of: [0048]
  • (c) incubating CD4+ and/or CD8+ T cells isolated from a patient with at least one component selected from the group consisting of SEQ ID NO:11 or SEQ ID NO:12; and [0049]
  • (d) administering to the patient an effective amount of the T cells, and thereby inhibiting the development of a cancer in the patient. [0050]
  • In one aspect of the invention, the T cells are proliferated prior to being administered ot the patient. [0051]
  • In another embodiment, a method is provided for producing an immunogenic response to a tumor-associated antigen in an animal comprising administering a first component comprising a polynucleotide encoding a fragment of the tumor-associated antigen to the animal. In one aspect of the invention, the tumor-associated antigen is Cripto. In another aspect, the polynucleotide encodes SEQ ID NO:11 and does not encode SEQ ID NO:3 or SEQ ID NO:4. In another aspect, the polynucleotide encodes SEQ ID NO:12 and does not encode SEQ ID NO:3 or SEQ ID NO:4. In yet another aspect of the invention, the polynucleotide is recombinant DNA. In yet another aspect, the polynucleotide is admixed with a second component comprising an immunostimulant, adjuvant and physiologically acceptable carrier. [0052]
  • In another embodiment, a method is provided for inducing an immunoresponse to Cripto in an animal comprising repeatedly administering a composition comprising a first component comprising SEQ ID NO:11 or SEQ ID NO:12. In yet another aspect, the first component is admixed with a second component comprising a physiologically acceptable carrier, adjuvant and immunostimulant prior to administering the first component to the animal. [0053]
  • The practice of the present invention will employ, unless indicated specifically to the contrary, conventional methods of virology, immunology, microbiology, molecular biology and recombinant DNA techniques within the skill of the art, many of which are described below for the purpose of illustration. Such techniques are explained fully in the literature. See, e.g., Sambrook, et al. Molecular Cloning: A Laboratory Manual (2nd Edition, 1989); Maniatis et al. Molecular Cloning: A Laboratory Manual (1982); DNA Cloning: A Practical Approach, vol. I & II (D. Glover, ed.); Oligonucleotide Synthesis (N. Gait, ed., 1984); Nucleic Acid Hybridization (B. Hames & S. Higgins, eds., 1985); Transcription and Translation (B. Hames & S. Higgins, eds., 1984); Animal Cell Culture (R. Freshney, ed., 1986); Perbal, A Practical Guide to Molecular Cloning (1984). [0054]
  • All publications, patents and patent applications cited herein, whether supra or infra, are hereby incorporated by reference in their entirety. [0055]
  • As used in this specification and the appended claims, the singular forms “a,” “an” and “the” include plural references unless the content clearly dictates otherwise. [0056]
  • Polypeptide Compositions [0057]
  • “Polypeptide(s)” refers to any peptide or protein comprising two or more amino acids joined to each other by peptide bonds or modified peptide bonds. “Polypeptide(s)” refers to both short chains, commonly referred to as peptides, oligopeptides and oligomers and to longer chains generally referred to as proteins. Polypeptides may comprise amino acids other than the 20 gene encoded amino acids. “Polypeptide(s)” include those modified either by natural processes, such as processing and other post-translational modifications, but also by chemical modification techniques. Such modifications are well described in basic texts and in more detailed monographs, as well as in a voluminous research literature, and they are well known to those of skill in the art. It will be appreciated that the same type of modification may be present in the same or varying degree at several sites in a given polypeptide. Also, a given polypeptide may comprise many types of modifications. Modifications can occur anywhere in a polypeptide, including the peptide backbone, the amino acid side-chains, and the amino or carboxyl termini. Modifications include, for example, acetylation, acylation, ADP-ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphotidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cysteine, formation of pyroglutamate, formylation, gamma-carboxylation, GPI anchor formation, hydroxylation, iodination, methylation, myristoylation, oxidation, proteolytic processing, phosphorylation, prenylation, racemization, glycosylation, lipid attachment, sulfation, gamma-carboxylation of glutamic acid residues, hydroxylation and ADP-ribosylation, selenoylation, sulfation, transfer-RNA mediated addition of amino acids to proteins, such as arginylation, and ubiquitination. See, for instance, PROTEINS—STRUCTURE AND MOLECULAR PROPERTIES, 2nd Ed., T. E. Creighton, W. H. Freeman and Company, New York (1993) and Wold, F., Posttranslational Protein Modifications: Perspectives and Prospects, pgs. 1-12 in POSTTRANSLATIONAL COVALENT MODIFICATION OF PROTEINS, B. C. Johnson, Ed., Academic Press, New York (1983); Seifter et al., Meth. Enzymol. 182:626-646 (1990) and Rattan et al., Protein Synthesis: Posttranslational Modifications and Aging, Ann. N.Y. Acad. Sci. 663: 48-62 (1992). Polypeptides may be branched or cyclic, with or without branching. Cyclic, branched and branched circular polypeptides may result from post-translational natural processes and may be made by entirely synthetic methods, as well. [0058]
  • Particularly illustrative polypeptides of the present invention comprise a sequence of at least 10 contiguous amino acids, preferably 20, more preferably 30, 40, 50, 60, 70, 80, 90, 100, 110, 120, 130, 140, 150, 160, 170, 180 amino acids of the cirpto protein of ID NO: 2 or 4. [0059]
  • In certain preferred embodiments, the polypeptides of the invention are immunogenic, i.e., they react detectably within an immunoassay (such as an ELISA or T-cell stimulation assay) with antisera and/or T-cells from a patient with cripto expressing cancer. Screening for immunogenic activity can be performed using techniques well known to the skilled artisan. For example, such screens can be performed using methods such as those described in Harlow and Lane, [0060] Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory, 1988. In one illustrative example, a polypeptide may be immobilized on a solid support and contacted with patient sera to allow binding of antibodies within the sera to the immobilized polypeptide. Unbound sera may then be removed and bound antibodies detected using, for example, 125I-labeled Protein A.
  • As would be recognized by the skilled artisan, immunogenic portions of the polypeptides disclosed herein are also encompassed by the present invention. An “immunogenic portion,” as used herein, is a fragment of an immunogenic polypeptide of the invention that itself is immunologically reactive (i.e., specifically binds) with the B-cells and/or T-cell surface antigen receptors that recognize the polypeptide. Immunogenic portions may generally be identified using well known techniques, such as those summarized in Paul, [0061] Fundamental Immunology, 3rd ed., 243247 (Raven Press, 1993) and references cited therein. Such techniques include screening polypeptides for the ability to react with antigen-specific antibodies, antisera and/or T-cell lines or clones. As used herein, antisera and antibodies are “antigen-specific” if they specifically bind to an antigen (i.e., they react with the protein in an ELISA or other immunoassay, and do not react detectably with unrelated proteins). Such antisera and antibodies may be prepared as described herein, and using well-known techniques.
  • In one preferred embodiment, an immunogenic portion of a polypeptide of the present invention is a portion that reacts with antisera and/or T-cells at a level that is not substantially less than the reactivity of the full-length polypeptide (e.g., in an ELISA and/or T-cell reactivity assay). Preferably, the level of immunogenic activity of the immunogenic portion is at least about 50%, preferably at least about 70% and most preferably greater than about 90% of the immunogenicity for the full-length polypeptide. In some instances, preferred immunogenic portions will be identified that have a level of immunogenic activity greater than that of the corresponding full-length polypeptide, e.g., having greater than about 100% or 150% or more immunogenic activity. [0062]
  • In certain other embodiments, illustrative immunogenic portions may include peptides in which an N-terminal leader sequence and/or transmembrane domain have been deleted. Other illustrative immunogenic portions will contain a small N- and/or C-terminal deletion (e.g., 1-30 amino acids, preferably 5-15 amino acids), relative to the mature protein. [0063]
  • In another embodiment, a polypeptide composition of the invention may also comprise one or more polypeptides that are immunologically reactive with T cells and/or antibodies generated against a polypeptide of the invention, particularly a polypeptide having an amino acid sequence disclosed herein, or to an immunogenic fragment or variant thereof. [0064]
  • In another embodiment of the invention, polypeptides are provided that comprise one or more polypeptides that are capable of eliciting T cells and/or antibodies that are immunologically reactive with one or more polypeptides described herein, or one or more polypeptides encoded by contiguous nucleic acid sequences contained in the polynucleotide sequences disclosed herein, or immunogenic fragments or variants thereof, or to one or more nucleic acid sequences which hybridize to one or more of these sequences under conditions of moderate to high stringency. [0065]
  • The present invention, in another aspect, provides polypeptide fragments comprising at least about 5, 10, 15, 20, 25, 50, 100, or 150 contiguous amino acids, or more, including all intermediate lengths, of a polypeptide compositions set forth herein, such as those set forth in SEQ ID NO: 2 or 4 or those encoded by a polynucleotide sequence set forth in a sequence of SEQ ID NO:1 or 3. It is preferred that the polypeptides comprise at least one preferably a pluarality of epitopes as set forth in sequence ID no 13 to 94. Optionally the frgaments are fused or otherwise conjugated to a heterologous carrier. [0066]
  • In another aspect, the present invention provides variants of the polypeptide compositions described herein. Polypeptide variants generally encompassed by the present invention will typically exhibit at least about 70%, 75%, 80%, 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% or more identity (determined as described below), along its length, to a polypeptide sequences set forth herein. [0067]
  • In one preferred embodiment, the polypeptide fragments and variants provide by the present invention are immunologically reactive with an antibody and/or T-cell that reacts with a full-length polypeptide specifically set for the herein. [0068]
  • In another preferred embodiment, the polypeptide fragments and variants provided by the present invention exhibit a level of immunogenic activity of at least about 50%, preferably at least about 70%, and most preferably at least about 90% or more of that exhibited by a full-length polypeptide sequence specifically set forth herein. [0069]
  • A polypeptide “variant,” as the term is used herein, is a polypeptide that typically differs from a polypeptide specifically disclosed herein in one or more substitutions, deletions, additions and/or insertions. Such variants may be naturally occurring or may be synthetically generated, for example, by modifying one or more of the above polypeptide sequences of the invention and evaluating their immunogenic activity as described herein and/or using any of a number of techniques well known in the art. [0070]
  • For example, certain illustrative variants of the polypeptides of the invention include those in which one or more portions, such as an N-terminal leader sequence or transmembrane domain, have been removed. Other illustrative variants include variants in which a small portion (e.g., 1-30 amino acids, preferably 5-15 amino acids) has been removed from the N- and/or C-terminal of the mature protein. [0071]
  • In many instances, a variant will contain conservative substitutions. A “conservative substitution” is one in which an amino acid is substituted for another amino acid that has similar properties, such that one skilled in the art of peptide chemistry would expect the secondary structure and hydropathic nature of the polypeptide to be substantially unchanged. As described above, modifications may be made in the structure of the polynucleotides and polypeptides of the present invention and still obtain a functional molecule that encodes a variant or derivative polypeptide with desirable characteristics, e.g., with immunogenic characteristics. When it is desired to alter the amino acid sequence of a polypeptide to create an equivalent, or even an improved, immunogenic variant or portion of a polypeptide of the invention, one skilled in the art will typically change one or more of the codons of the encoding DNA sequence according to Table 1. [0072]
  • For example, certain amino acids may be substituted for other amino acids in a protein structure without appreciable loss of interactive binding capacity with structures such as, for example, antigen-binding regions of antibodies or binding sites on substrate molecules. Since it is the interactive capacity and nature of a protein that defines that protein's biological functional activity, certain amino acid sequence substitutions can be made in a protein sequence, and, of course, its underlying DNA coding sequence, and nevertheless obtain a protein with like properties. It is thus contemplated that various changes may be made in the peptide sequences of the disclosed compositions, or corresponding DNA sequences which encode said peptides without appreciable loss of their biological utility or activity. [0073]
    TABLE 1
    Amino Acid Codons
    Alanine Ala A GCA GCC GCG GCU
    Cysteine Cys C UGC UGU
    Aspartic acid Asp D GAC GAU
    Glutamic acid Glu E GAA GAG
    Phenylalanine Phe F UUC UUU
    Glycine Gly G GGA GGC GGG GGU
    Histidine His H CAC CAU
    Isoleucine Ile I AUA AUC AUU
    Lysine Lys K AAA AAG
    Leucine Leu L UUA UUG CUA CUC CUG CUU
    Methionine Met M AUG
    Asparagine Asn N AAC AAU
    Proline Pro P CCA CCC CCG CCU
    Glutamine Gln Q CAA CAG
    Arginine Arg R AGA AGG CGA CGC CGG CGU
    Serine Ser S AGC AGU UCA UCC UCG UCU
    Threonine Thr T ACA ACC ACG ACU
    Valine Val V GUA GUC GUG GUU
    Tryptophan Trp W UGG
    Tyrosine Tyr Y UAC UAU
  • In making such changes, the hydropathic index of amino acids may be considered. The importance of the hydropathic amino acid index in conferring interactive biologic function on a protein is generally understood in the art (Kyte and Doolittle, 1982, incorporated herein by reference). It is accepted that the relative hydropathic character of the amino acid contributes to the secondary structure of the resultant protein, which in turn defines the interaction of the protein with other molecules, for example, enzymes, substrates, receptors, DNA, antibodies, antigens, and the like. Each amino acid has been assigned a hydropathic index on the basis of its hydrophobicity and charge characteristics (Kyte and Doolittle, 1982). These values are: isoleucine (+4.5); valine (+4.2); leucine (+3.8); phenylalanine (+2.8); cysteine/cystine (+2.5); methionine (+1.9); alanine (+1.8); glycine (−0.4); threonine (−0.7); serine (−0.8); tryptophan (−0.9); tyrosine (−1.3); proline (−1.6); histidine (−3.2); glutamate (−3.5); glutamine (−3.5); aspartate (−3.5); asparagine (−3.5); lysine (−3.9); and arginine (−4.5). [0074]
  • It is known in the art that certain amino acids may be substituted by other amino acids having a similar hydropathic index or score and still result in a protein with similar biological activity, i.e. still obtain a biological functionally equivalent protein. In making such changes, the substitution of amino acids whose hydropathic indices are within ±2 is preferred, those within +1 are particularly preferred, and those within ±0.5 are even more particularly preferred. It is also understood in the art that the substitution of like amino acids can be made effectively on the basis of hydrophilicity. U.S. Pat. No. 4,554,101 (specifically incorporated herein by reference in its entirety), states that the greatest local average hydrophilicity of a protein, as governed by the hydrophilicity of its adjacent amino acids, correlates with a biological property of the protein. [0075]
  • As detailed in U.S. Pat. No. 4,554,101, the following hydrophilicity values have been assigned to amino acid residues: arginine (+3.0); lysine (+3.0); aspartate (+3.0±1); glutamate (+3.0±1); serine (+0.3); asparagine (+0.2); glutamine (+0.2); glycine (0); threonine (−0.4); proline (−0.5±1); alanine (−0.5); histidine (−0.5); cysteine (−1.0); methionine (−1.3); valine (−1.5); leucine (−1.8); isoleucine (−1.8); tyrosine (−2.3); phenylalanine (−2.5); tryptophan (−3.4). It is understood that an amino acid can be substituted for another having a similar hydrophilicity value and still obtain a biologically equivalent, and in particular, an immunologically equivalent protein. In such changes, the substitution of amino acids whose hydrophilicity values are within ±2 is preferred, those within ±1 are particularly preferred, and those within ±0.5 are even more particularly preferred. [0076]
  • As outlined above, amino acid substitutions are generally therefore based on the relative similarity of the amino acid side-chain substituents, for example, their hydrophobicity, hydrophilicity, charge, size, and the like. Exemplary substitutions that take various of the foregoing characteristics into consideration are well known to those of skill in the art and include: arginine and lysine; glutamate and aspartate; serine and threonine; glutamine and asparagine; and valine, leucine and isoleucine. [0077]
  • In addition, any polynucleotide may be further modified to increase stability in vivo. Possible modifications include, but are not limited to, the addition of flanking sequences at the 5′ and/or 3′ ends; the use of phosphorothioate or 2′ O-methyl rather than phosphodiesterase linkages in the backbone; and/or the inclusion of nontraditional bases such as inosine, queosine and wybutosine, as well as acetyl-methyl-, thio- and other modified forms of adenine, cytidine, guanine, thymine and uridine. [0078]
  • Amino acid substitutions may further be made on the basis of similarity in polarity, charge, solubility, hydrophobicity, hydrophilicity and/or the amphipathic nature of the residues. For example, negatively charged amino acids include aspartic acid and glutamic acid; positively charged amino acids include lysine and arginine; and amino acids with uncharged polar head groups having similar hydrophilicity values include leucine, isoleucine and valine; glycine and alanine; asparagine and glutamine; and serine, threonine, phenylalanine and tyrosine. Other groups of amino acids that may represent conservative changes include: (1) ala, pro, gly, glu, asp, gln, asn, ser, thr; (2) cys, ser, tyr, thr; (3) val, ile, leu, met, ala, phe; (4) lys, arg, his; and (5) phe, tyr, trp, his. A variant may also, or alternatively, contain nonconservative changes. In a preferred embodiment, variant polypeptides differ from a native sequence by substitution, deletion or addition of five amino acids or fewer. Variants may also (or alternatively) be modified by, for example, the deletion or addition of amino acids that have minimal influence on the immunogenicity, secondary structure and hydropathic nature of the polypeptide. [0079]
  • As noted above, polypeptides may comprise a signal (or leader) sequence at the N-terminal end of the protein, which co-translationally or post-translationally directs transfer of the protein. The polypeptide may also be conjugated to a linker or other sequence for ease of synthesis, purification or identification of the polypeptide (e.g., poly-His), or to enhance binding of the polypeptide to a solid support. For example, a polypeptide may be conjugated to an immunoglobulin Fc region. [0080]
  • When comparing polypeptide sequences, two sequences are said to be “identical” if the sequence of amino acids in the two sequences is the same when aligned for maximum correspondence, as described below. Comparisons between two sequences are typically performed by comparing the sequences over a comparison window to identify and compare local regions of sequence similarity. A “comparison window” as used herein, refers to a segment of at least about 20 contiguous positions, usually 30 to about 75, 40 to about 50, in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned. [0081]
  • Optimal alignment of sequences for comparison may be conducted using the Megalign program in the Lasergene suite of bioinformatics software (DNASTAR, Inc., Madison, Wis.), using default parameters. This program embodies several alignment schemes described in the following references: Dayhoff, M. O. (1978) A model of evolutionary change in proteins—Matrices for detecting distant relationships. In Dayhoff, M. O. (ed.) Atlas of Protein Sequence and Structure, National Biomedical Research Foundation, Washington D.C. Vol. 5, Suppl. 3, pp. 345-358; Hein J. (1990) Unified Approach to Alignment and Phylogenes pp. 626-645 Methods in Enzymology vol. 183, Academic Press, Inc., San Diego, Calif.; Higgins, D. G. and Sharp, P. M. (1989) CABIOS 5:151-153; Myers, E. W. and Muller W. (1988) CABIOS 4:11-17; Robinson, E. D. (1971) Comb. Theor 11:105; Santou, N. Nes, M. (1987) Mol. Biol. Evol. 4:406-425; Sneath, P. H. A. and Sokal, R. R. (1973) Numerical Taxonomy—the Principles and Practice of Numerical Taxonomy, Freeman Press, San Francisco, Calif.; Wilbur, W. J. and Lipman, D. J. (1983) Proc. Natl. Acad., Sci. USA 80:726-730. [0082]
  • Alternatively, optimal alignment of sequences for comparison may be conducted by the local identity algorithm of Smith and Waterman (1981) Add. APL. Math 2:482, by the identity alignment algorithm of Needleman and Wunsch (1970) J. Mol. Biol. 48:443, by the search for similarity methods of Pearson and Lipman (1988) Proc. Natl. Acad. Sci. USA 85: 2444, by computerized implementations of these algorithms (GAP, BESTFIT, BLAST, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group (GCG), 575 Science Dr., Madison, Wis.), or by inspection. [0083]
  • One preferred example of algorithms that are suitable for determining percent sequence identity and sequence similarity are the BLAST and BLAST 2.0 algorithms, which are described in Altschul et al. (1977) Nucl. Acids Res. 25:3389-3402 and Altschul et al. (1990) J. Mol. Biol. 215:403-410, respectively. BLAST and BLAST 2.0 can be used, for example with the parameters described herein, to determine percent sequence identity for the polynucleotides and polypeptides of the invention. Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information. For amino acid sequences, a scoring matrix can be used to calculate the cumulative score. Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached. The BLAST algorithm parameters W, T and X determine the sensitivity and speed of the alignment. [0084]
  • In one preferred approach, the “percentage of sequence identity” is determined by comparing two optimally aligned sequences over a window of comparison of at least 20 positions, wherein the portion of the polypeptide sequence in the comparison window may comprise additions or deletions (i.e., gaps) of 20 percent or less, usually 5 to 15 percent, or 10 to 12 percent, as compared to the reference sequences (which does not comprise additions or deletions) for optimal alignment of the two sequences. The percentage is calculated by determining the number of positions at which the identical amino acid residue occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the reference sequence (i.e., the window size) and multiplying the results by 100 to yield the percentage of sequence identity. [0085]
  • Within other illustrative embodiments, a polypeptide may be a fusion polypeptide that comprises multiple polypeptides as described herein, or that comprises at least one polypeptide as described herein and an unrelated sequence, such as a known tumor protein. A fusion partner may, for example, assist in providing T helper epitopes (an immunological fusion partner), preferably T helper epitopes recognized by humans, or may assist in expressing the protein (an expression enhancer) at higher yields than the native recombinant protein. Certain preferred fusion partners are both immunological and expression enhancing fusion partners. Other fusion partners may be selected so as to increase the solubility of the polypeptide or to enable the polypeptide to be targeted to desired intracellular compartments. Still further fusion partners include affinity tags, which facilitate purification of the polypeptide. [0086]
  • Fusion polypeptides may generally be prepared using standard techniques, including chemical conjugation. Preferably, a fusion polypeptide is expressed as a recombinant polypeptide, allowing the production of increased levels, relative to a non-fused polypeptide, in an expression system. Briefly, DNA sequences encoding the polypeptide components may be assembled separately, and ligated into an appropriate expression vector. The 3′ end of the DNA sequence encoding one polypeptide component is ligated, with or without a peptide linker, to the 5′ end of a DNA sequence encoding the second polypeptide component so that the reading frames of the sequences are in phase. This permits translation into a single fusion polypeptide that retains the biological activity of both component polypeptides. [0087]
  • A peptide linker sequence may be employed to separate the first and second polypeptide components by a distance sufficient to ensure that each polypeptide folds into its secondary and tertiary structures. Such a peptide linker sequence is incorporated into the fusion polypeptide using standard techniques well known in the art. Suitable peptide linker sequences may be chosen based on the following factors: (1) their ability to adopt a flexible extended conformation; (2) their inability to adopt a secondary structure that could interact with functional epitopes on the first and second polypeptides; and (3) the lack of hydrophobic or charged residues that might react with the polypeptide functional epitopes. Preferred peptide linker sequences contain Gly, Asn and Ser residues. Other near neutral amino acids, such as Thr and Ala may also be used in the linker sequence. Amino acid sequences which may be usefully employed as linkers include those disclosed in Maratea et al., Gene 40:39-46, 1985; Murphy et al., Proc. Natl. Acad. Sci. USA 83:8258-8262, 1986; U.S. Pat. No. 4,935,233 and U.S. Pat. No. 4,751,180. The linker sequence may generally be from 1 to about 50 amino acids in length. Linker sequences are not required when the first and second polypeptides have non-essential N-terminal amino acid regions that can be used to separate the functional domains and prevent steric interference. [0088]
  • The ligated DNA sequences are operably linked to suitable transcriptional or translational regulatory elements. The regulatory elements responsible for expression of DNA are located only 5′ to the DNA sequence encoding the first polypeptides. Similarly, stop codons required to end translation and transcription termination signals are only present 3′ to the DNA sequence encoding the second polypeptide. [0089]
  • The fusion polypeptide can comprise a polypeptide as described herein together with an unrelated immunogenic protein, such as an immunogenic protein capable of eliciting a recall response. Examples of such proteins include tetanus, tuberculosis and hepatitis proteins (see, for example, Stoute et al. New Engl. J. Med., 336:86-91, 1997). [0090]
  • In one preferred embodiment, the immunological fusion partner is derived from a Mycobacterium sp., such as a [0091] Mycobacterium tuberculosis-derived Ra12 fragment. Ra12 compositions and methods for their use in enhancing the expression and/or immunogenicity of heterologous polynucleotide/polypeptide sequences is described in U.S. Patent Application 60/158,585, the disclosure of which is incorporated herein by reference in its entirety. Briefly, Ra12 refers to a polynucleotide region that is a subsequence of a Mycobacterium tuberculosis MTB32A nucleic acid. MTB32A is a serine protease of 32 KD molecular weight encoded by a gene in virulent and avirulent strains of M. tuberculosis. The nucleotide sequence and amino acid sequence of MTB32A have been described (for example, U.S. Patent Application 60/158,585; see also, Skeiky et al., Infection and Immun. (1999) 67:3998-4007, incorporated herein by reference). C-terminal fragments of the MTB32A coding sequence express at high levels and remain as a soluble polypeptides throughout the purification process. Moreover, Ra12 may enhance the immunogenicity of heterologous immunogenic polypeptides with which it is fused. One preferred Ra12 fusion polypeptide comprises a 14 KD C-terminal fragment corresponding to amino acid residues 192 to 323 of MTB32A. Other preferred Ra12 polynucleotides generally comprise at least about 15 consecutive nucleotides, at least about 30 nucleotides, at least about 60 nucleotides, at least about 100 nucleotides, at least about 200 nucleotides, or at least about 300 nucleotides that encode a portion of a Ra12 polypeptide. Ra12 polynucleotides may comprise a native sequence (i.e., an endogenous sequence that encodes a Ra12 polypeptide or a portion thereof) or may comprise a variant of such a sequence. Ra12 polynucleotide variants may contain one or more substitutions, additions, deletions and/or insertions such that the biological activity of the encoded fusion polypeptide is not substantially diminished, relative to a fusion polypeptide comprising a native Ra12 polypeptide. Variants preferably exhibit at least about 70% identity, more preferably at least about 80% identity and most preferably at least about 90% identity to a polynucleotide sequence that encodes a native Ra12 polypeptide or a portion thereof.
  • Within other preferred embodiments, an immunological fusion partner is derived from protein D, a surface protein of the gram-negative bacterium [0092] Haemophilus influenza B (WO 91/18926). Preferably, a protein D derivative comprises approximately the first third of the protein (e.g., the first N-terminal 100-110 amino acids), and a protein D derivative may be lipidated. Within certain preferred embodiments, the first 109 residues of a Lipoprotein D fusion partner is included on the N-terminus to provide the polypeptide with additional exogenous T-cell epitopes and to increase the expression level in E. coli (thus functioning as an expression enhancer). The lipid tail ensures optimal presentation of the antigen to antigen presenting cells. Other fusion partners include the non-structural protein from influenzae virus, NS1 (hemaglutinin). Typically, the N-terminal 81 amino acids are used, although different fragments that include T-helper epitopes may be used.
  • In another embodiment, the immunological fusion partner is the protein known as LYTA, or a portion thereof (preferably a C-terminal portion). LYTA is derived from [0093] Streptococcus pneumoniae, which synthesizes an N-acetyl-L-alanine amidase known as amidase LYTA (encoded by the LytA gene; Gene 43:265-292, 1986). LYTA is 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 the amino terminus has been described (see Biotechnology 10:795-798, 1992). Within a preferred embodiment, a repeat portion of LYTA may be incorporated into a fusion polypeptide. A repeat portion is found in the C-terminal region starting at residue 178. A particularly preferred repeat portion incorporates residues 188-305.
  • Yet another illustrative embodiment involves fusion polypeptides, and the polynucleotides encoding them, wherein the fusion partner comprises a targeting signal capable of directing a polypeptide to the endosomal/lysosomal compartment, as described in U.S. Pat. No. 5,633,234. An immunogenic polypeptide of the invention, when fused with this targeting signal, will associate more efficiently with MHC class II molecules and thereby provide enhanced in vivo stimulation of CD4+ T-cells specific for the polypeptide. [0094]
  • The cripto part of the fusion molecule may prefreably be the whole length 188 aa protein of Cripto 1 or Cripto 3 or a fragment thereof as described herein. [0095]
  • Polypeptides of the invention are prepared using any of a variety of well known synthetic and/or recombinant techniques, the latter of which are further described below. Polypeptides, portions and other variants generally less than about 150 amino acids can be generated by synthetic means, using techniques well known to those of ordinary skill in the art. In one illustrative example, such polypeptides are synthesized using any of the commercially available solid-phase techniques, such as the Merrifield solid-phase synthesis method, where amino acids are sequentially added to a growing amino acid chain. See Merrifield, J. Am. Chem. Soc. 85:2149-2146, 1963. Equipment for automated synthesis of polypeptides is commercially available from suppliers such as Perkin Elmer/Applied BioSystems Division (Foster City, Calif.), and may be operated according to the manufacturer's instructions. [0096]
  • In general, polypeptide compositions (including fusion polypeptides) of the invention are isolated. An “isolated” polypeptide is one that is removed from its original environment. For example, a naturally-occurring protein or polypeptide is isolated if it is separated from some or all of the coexisting materials in the natural system. Preferably, such polypeptides are also purified, e.g., are at least about 90% pure, more preferably at least about 95% pure and most preferably at least about 99% pure. [0097]
  • Polynucleotide Compositions [0098]
  • “Polynucleotide(s)” generally refers to any polyribonucleotide or polydeoxyribonucleotide, that may be unmodified RNA or DNA or modified RNA or DNA. “Polynucleotide(s)” include, without limitation, single- and double-stranded DNA, DNA that is a mixture of single- and double-stranded regions or single-, double- and triple-stranded regions, single- and double-stranded RNA, and RNA that is mixture of single- and double-stranded regions, hybrid molecules comprising DNA and RNA that may be single-stranded or, more typically, double-stranded, or triple-stranded regions, or a mixture of single- and double-stranded regions. In addition, “polynucleotide” as used herein refers to triple-stranded regions comprising RNA or DNA or both RNA and DNA. The strands in such regions may be from the same molecule or from different molecules. The regions may include all of one or more of the molecules, but more typically involve only a region of some of the molecules. One of the molecules of a triple-helical region often is an oligonucleotide. As used herein, the term “polynucleotide(s)” also includes DNAs or RNAs as described above that comprise one or more modified bases. Thus, DNAs or RNAs with backbones modified for stability or for other reasons are “polynucleotide(s)” as that term is intended herein. Moreover, DNAs or RNAs comprising unusual bases, such as inosine, or modified bases, such as tritylated bases, to name just two examples, are polynucleotides as the term is used herein. It will be appreciated that a great variety of modifications have been made to DNA and RNA that serve many useful purposes known to those of skill in the art. The term “polynucleotide(s)” as it is employed herein embraces such chemically, enzymatically or metabolically modified forms of polynucleotides, as well as the chemical forms of DNA and RNA characteristic of viruses and cells, including, for example, simple and complex cells. “Polynucleotide(s)” also embraces short polynucleotides often referred to as oligonucleotide(s). [0099]
  • The present invention, in other aspects, provides polynucleotide compositions that encode for the polypeptides of the invention. The terms “DNA” and “polynucleotide” are used essentially interchangeably herein to refer to a DNA molecule that has been isolated free of total genomic DNA of a particular species. “Isolated,” as used herein, means that a polynucleotide is substantially away from other coding sequences, and that the DNA molecule does not contain large portions of unrelated coding DNA, such as large chromosomal fragments or other functional genes or polypeptide coding regions. Of course, this refers to the DNA molecule as originally isolated, and does not exclude genes or coding regions later added to the segment by the hand of man. [0100]
  • As will be understood by those skilled in the art, the polynucleotide compositions of this invention can include genomic sequences, extra-genomic and plasmid-encoded sequences and smaller engineered gene segments that express, or may be adapted to express, proteins, polypeptides, peptides and the like. Such segments may be naturally isolated, or modified synthetically by the hand of man. [0101]
  • As will be also recognized by the skilled artisan, polynucleotides of the invention may be single-stranded (coding or antisense) or double-stranded, and may be DNA (genomic, cDNA or synthetic) or RNA molecules. RNA molecules may include HnRNA molecules, which contain introns and correspond to a DNA molecule in a one-to-one manner, and mRNA molecules, which do not contain introns. Additional coding or non-coding sequences may, but need not, be present within a polynucleotide of the present invention, and a polynucleotide may, but need not, be linked to other molecules and/or support materials. [0102]
  • Polynucleotides may comprise a native sequence (i.e., an endogenous sequence that encodes a polypeptide/protein of the invention or a portion thereof) or may comprise a sequence that encodes a variant or derivative, preferably and immunogenic variant or derivative, of such a sequence. [0103]
  • Typically, polynucleotide variants will contain one or more substitutions, additions, deletions and/or insertions, preferably such that the immunogenicity of the polypeptide encoded by the variant polynucleotide is not substantially diminished relative to a polypeptide encoded by a polynucleotide sequence specifically set forth herein). The term “variants” should also be understood to encompasses homologous genes of xenogenic origin. [0104]
  • In additional embodiments, the present invention provides polynucleotide fragments comprising various lengths of contiguous stretches of sequence identical to or complementary to one or more of the SEQ ID NO:1 or 3. For example, polynucleotides are provided by this invention that comprise at least about 10, 15, 20, 30, 40, 50, 75, 100, 150, 200, 300, 400, the SEQ ID NO: 1 as well as all intermediate lengths there between. It will be readily understood that “intermediate lengths”, in this context, means any length between the quoted values, such as 16, 17, 18, 19, etc.; 21, 22, 23, etc.; 30, 31, 32, etc.; 50, 51, 52, 53, etc.; 100, 101, 102, 103, etc.; 150, 151, 152, 153, etc.; including all integers through 200-500; 500-1,000, and the like. Particularly preferred polynucleotides are those which encode the epitopes as set forth in SEQ ID NOS: 13-94. [0105]
  • In another embodiment of the invention, polynucleotide compositions are provided that are capable of hybridizing under moderate to high stringency conditions to a polynucleotide sequence provided herein, or a fragment thereof, or a complementary sequence thereof. Hybridization techniques are well known in the art of molecular biology. For purposes of illustration, suitable moderately stringent conditions for testing the hybridization of a polynucleotide of this invention with other polynucleotides include prewashing in a solution of 5×SSC, 0.5% SDS, 1.0 mM EDTA (pH 8.0); hybridizing at 50° C.-60° C., 5×SSC, overnight; followed by washing twice at 65° C. for 20 minutes with each of 2×, 0.5× and 0.2×SSC containing 0.1% SDS. One skilled in the art will understand that the stringency of hybridization can be readily manipulated, such as by altering the salt content of the hybridization solution and/or the temperature at which the hybridization is performed. For example, in another embodiment, suitable highly stringent hybridization conditions include those described above, with the exception that the temperature of hybridization is increased, e.g., to 60-65° C. or 65-70° C. [0106]
  • In certain preferred embodiments, the polynucleotides described above, e.g., polynucleotide variants, fragments and hybridizing sequences, encode polypeptides that are immunologically cross-reactive with a polypeptide sequence specifically set forth in SEQ ID NO:1 or SEQ ID NO:3. In other preferred embodiments, such polynucleotides encode polypeptides 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 that for a polypeptide sequence specifically set forth herein. [0107]
  • The polynucleotides of the present invention, or fragments thereof, regardless of the length of the coding sequence itself, may be combined with other DNA sequences, such as promoters, polyadenylation signals, additional restriction enzyme sites, multiple cloning sites, other coding segments, and the like, such that their overall length may vary considerably. It is therefore contemplated that a nucleic acid fragment of almost any length may be employed, with the total length preferably being limited by the ease of preparation and use in the intended recombinant DNA protocol. For example, illustrative polynucleotide segments with total lengths of about 10,000, about 5000, about 3000, about 2,000, about 1,000, about 500, about 200, about 100, about 50 base pairs in length, and the like, (including all intermediate lengths) are contemplated to be useful in many implementations of this invention. [0108]
  • When comparing polynucleotide sequences, two sequences are said to be “identical” if the sequence of nucleotides in the two sequences is the same when aligned for maximum correspondence, as described below. Comparisons between two sequences are typically performed by comparing the sequences over a comparison window to identify and compare local regions of sequence similarity. A “comparison window” as used herein, refers to a segment of at least about 20 contiguous positions, usually 30 to about 75, 40 to about 50, in which a sequence may be compared to a reference sequence of the same number of contiguous positions after the two sequences are optimally aligned. [0109]
  • Optimal alignment of sequences for comparison may be conducted using the Megalign program in the Lasergene suite of bioinformatics software (DNASTAR, Inc., Madison, Wis.), using default parameters. This program embodies several alignment schemes described in the following references: Dayhoff, M. O. (1978) A model of evolutionary change in proteins—Matrices for detecting distant relationships. In Dayhoff, M. O. (ed.) Atlas of Protein Sequence and Structure, National Biomedical Research Foundation, Washington D.C. Vol. 5, Suppl. 3, pp. 345-358; Hein J. (1990) Unified Approach to Alignment and Phylogenes pp. 626-645 Methods in Enzymology vol. 183, Academic Press, Inc., San Diego, Calif.; Higgins, D. G. and Sharp, P. M. (1989) CABIOS 5:151-153; Myers, E. W. and Muller W. (1988) CABIOS 4:11-17; Robinson, E. D. (1971) Comb. Theor 11:105; Santou, N. Nes, M. (1987) Mol. Biol. Evol. 4:406-425; Sneath, P. H. A. and Sokal, R. R. (1973) Numerical Taxonomy—the Principles and Practice of Numerical Taxonomy, Freeman Press, San Francisco, Calif.; Wilbur, W. J. and Lipman, D. J. (1983) Proc. Natl. Acad., Sci. USA 80:726-730. [0110]
  • Alternatively, optimal alignment of sequences for comparison may be conducted by the local identity algorithm of Smith and Waterman (1981) Add. APL. Math 2:482, by the identity alignment algorithm of Needleman and Wunsch (1970) J. Mol. Biol. 48:443, by the search for similarity methods of Pearson and Lipman (1988) Proc. Natl. Acad. Sci. USA 85: 2444, by computerized implementations of these algorithms (GAP, BESTFIT, BLAST, FASTA, and TFASTA in the Wisconsin Genetics Software Package, Genetics Computer Group (GCG), 575 Science Dr., Madison, Wis.), or by inspection. [0111]
  • One preferred example of algorithms that are suitable for determining percent sequence identity and sequence similarity are the BLAST and BLAST 2.0 algorithms, which are described in Altschul et al. (1977) Nucl. Acids Res. 25:3389-3402 and Altschul et al. (1990) J. Mol. Biol. 215:403-410, respectively. BLAST and BLAST 2.0 can be used, for example with the parameters described herein, to determine percent sequence identity for the polynucleotides of the invention. Software for performing BLAST analyses is publicly available through the National Center for Biotechnology Information. In one illustrative example, cumulative scores can be calculated using, for nucleotide sequences, the parameters M (reward score for a pair of matching residues; always >0) and N (penalty score for mismatching residues; always <0). Extension of the word hits in each direction are halted when: the cumulative alignment score falls off by the quantity X from its maximum achieved value; the cumulative score goes to zero or below, due to the accumulation of one or more negative-scoring residue alignments; or the end of either sequence is reached. The BLAST algorithm parameters W, T and X determine the sensitivity and speed of the alignment. The BLASTN program (for nucleotide sequences) uses as defaults a wordlength (W) of 11, and expectation (E) of 10, and the BLOSUM62 scoring matrix (see Henikoff and Henikoff (1989) Proc. Natl. Acad. Sci. USA 89:10915) alignments, (B) of 50, expectation (E) of 10, M=5, N=−4 and a comparison of both strands. [0112]
  • Preferably, the “percentage of sequence identity” is determined by comparing two optimally aligned sequences over a window of comparison of at least 20 positions, wherein the portion of the polynucleotide sequence in the comparison window may comprise additions or deletions (i.e., gaps) of 20 percent or less, usually 5 to 15 percent, or 10 to 12 percent, as compared to the reference sequences (which does not comprise additions or deletions) for optimal alignment of the two sequences. The percentage is calculated by determining the number of positions at which the identical nucleic acid bases occurs in both sequences to yield the number of matched positions, dividing the number of matched positions by the total number of positions in the reference sequence (i.e., the window size) and multiplying the results by 100 to yield the percentage of sequence identity. [0113]
  • It will be appreciated by those of ordinary skill in the art that, as a result of the degeneracy of the genetic code, there are many nucleotide sequences that encode a polypeptide as described herein. Some of these polynucleotides bear minimal homology to the nucleotide sequence of any native gene. Nonetheless, polynucleotides that vary due to differences in codon usage are specifically contemplated by the present invention. Further, alleles of the genes comprising the polynucleotide sequences provided herein are within the scope of the present invention. Alleles are endogenous genes that are altered as a result of one or more mutations, such as deletions, additions and/or substitutions of nucleotides. The resulting mRNA and protein may, but need not, have an altered structure or function. Alleles may be identified using standard techniques (such as hybridization, amplification and/or database sequence comparison). [0114]
  • Therefore, in another embodiment of the invention, a mutagenesis approach, such as site-specific mutagenesis, is employed for the preparation of immunogenic variants and/or derivatives of the polypeptides described herein. By this approach, specific modifications in a polypeptide sequence can be made through mutagenesis of the underlying polynucleotides that encode them. These techniques provides a straightforward approach to prepare and test sequence variants, for example, incorporating one or more of the foregoing considerations, by introducing one or more nucleotide sequence changes into the polynucleotide. [0115]
  • Site-specific mutagenesis allows the production of mutants through the use of specific oligonucleotide sequences which encode the DNA sequence of the desired mutation, as well as a sufficient number of adjacent nucleotides, to provide a primer sequence of sufficient size and sequence complexity to form a stable duplex on both sides of the deletion junction being traversed. Mutations may be employed in a selected polynucleotide sequence to improve, alter, decrease, modify, or otherwise change the properties of the polynucleotide itself, and/or alter the properties, activity, composition, stability, or primary sequence of the encoded polypeptide. [0116]
  • In certain embodiments of the present invention, the inventors contemplate the mutagenesis of the disclosed polynucleotide sequences to alter one or more properties of the encoded polypeptide, such as the immunogenicity of a polypeptide vaccine. The techniques of site-specific mutagenesis are well-known in the art, and are widely used to create variants of both polypeptides and polynucleotides. For example, site-specific mutagenesis is often used to alter a specific portion of a DNA molecule. In such embodiments, a primer comprising typically about 14 to about 25 nucleotides or so in length is employed, with about 5 to about 10 residues on both sides of the junction of the sequence being altered. [0117]
  • As will be appreciated by those of skill in the art, site-specific mutagenesis techniques have often employed a phage vector that exists in both a single stranded and double stranded form. Typical vectors useful in site-directed mutagenesis include vectors such as the M13 phage. These phage are readily commercially-available and their use is generally well-known to those skilled in the art. Double-stranded plasmids are also routinely employed in site directed mutagenesis that eliminates the step of transferring the gene of interest from a plasmid to a phage. [0118]
  • In general, site-directed mutagenesis in accordance herewith is performed by first obtaining a single-stranded vector or melting apart of two strands of a double-stranded vector that includes within its sequence a DNA sequence that encodes the desired peptide. An oligonucleotide primer bearing the desired mutated sequence is prepared, generally synthetically. This primer is then annealed with the single-stranded vector, and subjected to DNA polymerizing enzymes such as [0119] E. coli polymerase I Klenow fragment, in order to complete the synthesis of the mutation-bearing strand. Thus, a heteroduplex is formed wherein one strand encodes the original non-mutated sequence and the second strand bears the desired mutation. This heteroduplex vector is then used to transform appropriate cells, such as E. coli cells, and clones are selected which include recombinant vectors bearing the mutated sequence arrangement.
  • The preparation of sequence variants of the selected peptide-encoding DNA segments using site-directed mutagenesis provides a means of producing potentially useful species and is not meant to be limiting as there are other ways in which sequence variants of peptides and the DNA sequences encoding them may be obtained. For example, recombinant vectors encoding the desired peptide sequence may be treated with mutagenic agents, such as hydroxylamine, to obtain sequence variants. Specific details regarding these methods and protocols are found in the teachings of Maloy et al., 1994; Segal, 1976; Prokop and Bajpai, 1991; Kuby, 1994; and Maniatis et al., 1982, each incorporated herein by reference, for that purpose. [0120]
  • As used herein, the term “oligonucleotide directed mutagenesis procedure” refers to template-dependent processes and vector-mediated propagation which result in an increase in the concentration of a specific nucleic acid molecule relative to its initial concentration, or in an increase in the concentration of a detectable signal, such as amplification. As used herein, the term “oligonucleotide directed mutagenesis procedure” is intended to refer to a process that involves the template-dependent extension of a primer molecule. The term template dependent process refers to nucleic acid synthesis of an RNA or a DNA molecule wherein the sequence of the newly synthesized strand of nucleic acid is dictated by the well-known rules of complementary base pairing (see, for example, Watson, 1987). Typically, vector mediated methodologies involve the introduction of the nucleic acid fragment into a DNA or RNA vector, the clonal amplification of the vector, and the recovery of the amplified nucleic acid fragment. Examples of such methodologies are provided by U.S. Pat. No. 4,237,224, specifically incorporated herein by reference in its entirety. [0121]
  • In another approach for the production of polypeptide variants of the present invention, recursive sequence recombination, as described in U.S. Pat. No. 5,837,458, may be employed. In this approach, iterative cycles of recombination and screening or selection are performed to “evolve” individual polynucleotide variants of the invention having, for example, enhanced immunogenic activity. [0122]
  • According to another embodiment of the present invention, polynucleotide compositions comprising antisense oligonucleotides are provided. Antisense oligonucleotides have been demonstrated to be effective and targeted inhibitors of protein synthesis, and, consequently, provide a therapeutic approach by which a disease can be treated by inhibiting the synthesis of proteins that contribute to the disease. The efficacy of antisense oligonucleotides for inhibiting protein synthesis is well established. For example, the synthesis of polygalactauronase and the muscarine type 2 acetylcholine receptor are inhibited by antisense oligonucleotides directed to their respective mRNA sequences (U.S. Pat. No. 5,739,119 and U.S. Pat. No. 5,759,829). Further, examples of antisense inhibition have been demonstrated with the nuclear protein cyclin, the multiple drug resistance gene (MDG1), ICAM-1, E-selectin, STK-1, striatal GABAA receptor and human EGF (Jaskulski et al., Science. 1988 Jun. 10;240(4858):1544-6; Vasanthakumar and Ahmed, Cancer Commun. 1989;1(4):225-32; Peris et al., Brain Res Mol Brain Res. 1998 Jun. 15;57(2):310-20; U.S. Pat. No. 5,801,154; U.S. Pat. No. 5,789,573; U.S. Pat. No. 5,718,709 and U.S. Pat. No. 5,610,288). Antisense constructs have also been described that inhibit and can be used to treat a variety of abnormal cellular proliferations, e.g. cancer (U.S. Pat. No. 5,747,470; U.S. Pat. No. 5,591,317 and U.S. Pat. No. 5,783,683). [0123]
  • Therefore, in certain embodiments, the present invention provides oligonucleotide sequences that comprise all, or a portion of, any sequence that is capable of specifically binding to polynucleotide sequence described herein, or a complement thereof. In one embodiment, the antisense oligonucleotides comprise DNA or derivatives thereof. In another embodiment, the oligonucleotides comprise RNA or derivatives thereof. In a third embodiment, the oligonucleotides are modified DNAs comprising a phosphorothioated modified backbone. In a fourth embodiment, the oligonucleotide sequences comprise peptide nucleic acids or derivatives thereof. In each case, preferred compositions comprise a sequence region that is complementary, and more preferably substantially-complementary, and even more preferably, completely complementary to one or more portions of polynucleotides disclosed herein. [0124]
  • Selection of antisense compositions specific for a given gene sequence is based upon analysis of the chosen target sequence (i.e. in these illustrative examples the rat and human sequences) and determination of secondary structure, Tm, binding energy, relative stability, and antisense compositions were selected based upon their relative inability to form dimers, hairpins, or other secondary structures that would reduce or prohibit specific binding to the target mRNA in a host cell. [0125]
  • Highly preferred target regions of the mRNA, are those which are at or near the AUG translation initiation codon, and those sequences which are substantially complementary to 5′ regions of the mRNA. These secondary structure analyses and target site selection considerations can be performed, for example, using v.4 of the OLIGO primer analysis software and/or the BLASTN 2.0.5 algorithm software (Altschul et al., Nucleic Acids Res. 1997 Sep. 1;25(17):3389-402). [0126]
  • The use of an antisense delivery method employing a short peptide vector, termed MPG (27 residues), is also contemplated. The MPG peptide contains a hydrophobic domain derived from the fusion sequence of HIV gp41 and a hydrophilic domain from the nuclear localization sequence of SV40 T-antigen (Morris et al., Nucleic Acids Res. 1997 Jul. 15;25(14):2730-6). It has been demonstrated that several molecules of the MPG peptide coat the antisense oligonucleotides and can be delivered into cultured mammalian cells in less than 1 hour with relatively high efficiency (90%). Further, the interaction with MPG strongly increases both the stability of the oligonucleotide to nuclease and the ability to cross the plasma membrane. [0127]
  • According to another embodiment of the invention, the polynucleotide compositions described herein are used in the design and preparation of ribozyme molecules for inhibiting expression of the tumor polypeptides and proteins of the present invention in tumor cells. Ribozymes are RNA-protein complexes that cleave nucleic acids in a site-specific fashion. Ribozymes have specific catalytic domains that possess endonuclease activity (Kim and Cech, Proc Natl Acad Sci USA. 1987 December;84(24):8788-92; Forster and Symons, Cell. 1987 Apr. 24;49(2):211-20). For example, a large number of ribozymes accelerate phosphoester transfer reactions with a high degree of specificity, often cleaving only one of several phosphoesters in an oligonucleotide substrate (Cech et al., Cell. 1981 December;27(3 Pt 2):487-96; Michel and Westhof, J Mol Biol. 1990 Dec. 5;216(3):585-610; Reinhold-Hurek and Shub, Nature. 1992 May 14;357(6374):173-6). This specificity has been attributed to the requirement that the substrate bind via specific base-pairing interactions to the internal guide sequence (“IGS”) of the ribozyme prior to chemical reaction. [0128]
  • Six basic varieties of naturally-occurring enzymatic RNAs are known presently. Each can catalyze the hydrolysis of RNA phosphodiester bonds in trans (and thus can cleave other RNA molecules) under physiological conditions. In general, enzymatic nucleic acids act by first binding to a target RNA. Such binding occurs through the target binding portion of a enzymatic nucleic acid which is held in close proximity to an enzymatic portion of the molecule that acts to cleave the target RNA. Thus, the enzymatic nucleic acid first recognizes and then binds a target RNA through complementary base-pairing, and once bound to the correct site, acts enzymatically to cut the target RNA. Strategic cleavage of such a target RNA will destroy its ability to direct synthesis of an encoded protein. After an enzymatic nucleic acid has bound and cleaved its RNA target, it is released from that RNA to search for another target and can repeatedly bind and cleave new targets. [0129]
  • The enzymatic nature of a ribozyme is advantageous over many technologies, such as antisense technology (where a nucleic acid molecule simply binds to a nucleic acid target to block its translation) since the concentration of ribozyme necessary to affect a therapeutic treatment is lower than that of an antisense oligonucleotide. This advantage reflects the ability of the ribozyme to act enzymatically. Thus, a single ribozyme molecule is able to cleave many molecules of target RNA. In addition, the ribozyme is a highly specific inhibitor, with the specificity of inhibition depending not only on the base pairing mechanism of binding to the target RNA, but also on the mechanism of target RNA cleavage. Single mismatches, or base-substitutions, near the site of cleavage can completely eliminate catalytic activity of a ribozyme. Similar mismatches in antisense molecules do not prevent their action (Woolf et al., Proc Natl Acad Sci USA. 1992 Aug. 15;89(16):7305-9). Thus, the specificity of action of a ribozyme is greater than that of an antisense oligonucleotide binding the same RNA site. [0130]
  • The enzymatic nucleic acid molecule may be formed in a hammerhead, hairpin, a hepatitis δ virus, group I intron or RNaseP RNA (in association with an RNA guide sequence) or Neurospora VS RNA motif. Examples of hammerhead motifs are described by Rossi et al. Nucleic Acids Res. 1992 Sep. 11;20(17):4559-65. Examples of hairpin motifs are described by Hampel et al. (Eur. Pat. Appl. Publ. No. EP 0360257), Hampel and Tritz, Biochemistry 1989 Jun. 13;28(12):4929-33; Hampel et al., Nucleic Acids Res. 1990 Jan. 25;18(2):299-304 and U.S. Pat. No. 5,631,359. An example of the hepatitis 6 virus motif is described by Perrotta and Been, Biochemistry. 1992 Dec. 1;31(47):11843-52; an example of the RNaseP motif is described by Guerrier-Takada et al., Cell. 1983 December;35(3 Pt 2):849-57; Neurospora VS RNA ribozyme motif is described by Collins (Saville and Collins, Cell. 1990 May 18;61(4):685-96; Saville and Collins, Proc Natl Acad Sci USA. 1991 Oct. 1;88(19):8826-30; Collins and Olive, Biochemistry. 1993 Mar. 23;32(11):2795-9); and an example of the Group I intron is described in (U.S. Pat. No. 4,987,071). All that is important in an enzymatic nucleic acid molecule of this invention is that it has a specific substrate binding site which is complementary to one or more of the target gene RNA regions, and that it have nucleotide sequences within or surrounding that substrate binding site which impart an RNA cleaving activity to the molecule. Thus the ribozyme constructs need not be limited to specific motifs mentioned herein. [0131]
  • Ribozymes may be designed as described in Int. Pat. Appl. Publ. No. WO 93/23569 and Int. Pat. Appl. Publ. No. WO 94/02595, each specifically incorporated herein by reference) and synthesized to be tested in vitro and in vivo, as described. Such ribozymes can also be optimized for delivery. While specific examples are provided, those in the art will recognize that equivalent RNA targets in other species can be utilized when necessary. [0132]
  • Ribozyme activity can be optimized by altering the length of the ribozyme binding arms, or chemically synthesizing ribozymes with modifications that prevent their degradation by serum ribonucleases (see e.g., Int. Pat. Appl. Publ. No. WO 92/07065; Int. Pat. Appl. Publ. No. WO 93/15187; Int. Pat. Appl. Publ. No. WO 91/03162; Eur. Pat. Appl. Publ. No. 92110298.4; U.S. Pat. No. 5,334,711; and Int. Pat. Appl. Publ. No. WO 94/13688, which describe various chemical modifications that can be made to the sugar moieties of enzymatic RNA molecules), modifications which enhance their efficacy in cells, and removal of stem II bases to shorten RNA synthesis times and reduce chemical requirements. [0133]
  • Sullivan et al. (Int. Pat. Appl. Publ. No. WO 94/02595) describes the general methods for delivery of enzymatic RNA molecules. Ribozymes may be administered to cells by a variety of methods known to those familiar to the art, including, but not restricted to, encapsulation in liposomes, by iontophoresis, or by incorporation into other vehicles, such as hydrogels, cyclodextrins, biodegradable nanocapsules, and bioadhesive microspheres. For some indications, ribozymes may be directly delivered ex vivo to cells or tissues with or without the aforementioned vehicles. Alternatively, the RNA/vehicle combination may be locally delivered by direct inhalation, by direct injection or by use of a catheter, infusion pump or stent. Other routes of delivery include, but are not limited to, intravascular, intramuscular, subcutaneous or joint injection, aerosol inhalation, oral (tablet or pill form), topical, systemic, ocular, intraperitoneal and/or intrathecal delivery. More detailed descriptions of ribozyme delivery and administration are provided in Int. Pat. Appl. Publ. No. WO 94/02595 and Int. Pat. Appl. Publ. No. WO 93/23569, each specifically incorporated herein by reference. [0134]
  • Another means of accumulating high concentrations of a ribozyme(s) within cells is to incorporate the ribozyme-encoding sequences into a DNA expression vector. Transcription of the ribozyme sequences are driven from a promoter for eukaryotic RNA polymerase I (pol I), RNA polymerase II (pol II), or RNA polymerase III (pol III). Transcripts from pol II or pol III promoters will be expressed at high levels in all cells; the levels of a given pol II promoter in a given cell type will depend on the nature of the gene regulatory sequences (enhancers, silencers, etc.) present nearby. Prokaryotic RNA polymerase promoters may also be used, providing that the prokaryotic RNA polymerase enzyme is expressed in the appropriate cells Ribozymes expressed from such promoters have been shown to function in mammalian cells. Such transcription units can be incorporated into a variety of vectors for introduction into mammalian cells, including but not restricted to, plasmid DNA vectors, viral DNA vectors (such as adenovirus or adeno-associated vectors), or viral RNA vectors (such as retroviral, semliki forest virus, sindbis virus vectors). [0135]
  • In another embodiment of the invention, peptide nucleic acids (PNAs) compositions are provided. PNA is a DNA mimic in which the nucleobases are attached to a pseudopeptide backbone (Good and Nielsen, Antisense Nucleic Acid Drug Dev. 1997 7(4) 431-37). PNA is able to be utilized in a number methods that traditionally have used RNA or DNA. Often PNA sequences perform better in techniques than the corresponding RNA or DNA sequences and have utilities that are not inherent to RNA or DNA. A review of PNA including methods of making, characteristics of, and methods of using, is provided by Corey ([0136] Trends Biotechnol 1997 June;15(6):224-9). As such, in certain embodiments, one may prepare PNA sequences that are complementary to one or more portions of the ACE mRNA sequence, and such PNA compositions may be used to regulate, alter, decrease, or reduce the translation of ACE-specific mRNA, and thereby alter the level of ACE activity in a host cell to which such PNA compositions have been administered.
  • PNAs have 2-aminoethyl-glycine linkages replacing the normal phosphodiester backbone of DNA (Nielsen et al., [0137] Science 1991 Dec. 6;254(5037):1497-500; Hanvey et al., Science. 1992 Nov. 27;258(5087):1481-5; Hyrup and Nielsen, Bioorg Med Chem. 1996 January;4(1):5-23). This chemistry has three important consequences: firstly, in contrast to DNA or phosphorothioate oligonucleotides, PNAs are neutral molecules; secondly, PNAs are achiral, which avoids the need to develop a stereoselective synthesis; and thirdly, PNA synthesis uses standard Boc or Fmoc protocols for solid-phase peptide synthesis, although other methods, including a modified Merrifield method, have been used.
  • PNA monomers or ready-made oligomers are commercially available from PerSeptive Biosystems (Framingham, Mass.). PNA syntheses by either Boc or Fmoc protocols are straightforward using manual or automated protocols (Norton et al., Bioorg Med Chem. 1995 April;3(4):437-45). The manual protocol lends itself to the production of chemically modified PNAs or the simultaneous synthesis of families of closely related PNAs. [0138]
  • As with peptide synthesis, the success of a particular PNA synthesis will depend on the properties of the chosen sequence. For example, while in theory PNAs can incorporate any combination of nucleotide bases, the presence of adjacent purines can lead to deletions of one or more residues in the product. In expectation of this difficulty, it is suggested that, in producing PNAs with adjacent purines, one should repeat the coupling of residues likely to be added inefficiently. This should be followed by the purification of PNAs by reverse-phase high-pressure liquid chromatography, providing yields and purity of product similar to those observed during the synthesis of peptides. [0139]
  • Modifications of PNAs for a given application may be accomplished by coupling amino acids during solid-phase synthesis or by attaching compounds that contain a carboxylic acid group to the exposed N-terminal amine. Alternatively, PNAs can be modified after synthesis by coupling to an introduced lysine or cysteine. The ease with which PNAs can be modified facilitates optimization for better solubility or for specific functional requirements. Once synthesized, the identity of PNAs and their derivatives can be confirmed by mass spectrometry. Several studies have made and utilized modifications of PNAs (for example, Norton et al., Bioorg Med Chem. 1995 April;3(4):437-45; Petersen et al., J Pept Sci. 1995 May-Jun;1(3):175-83; Orum et al., Biotechniques. 1995 September;19(3):472-80; Footer et al., Biochemistry. 1996 Aug. 20;35(33):10673-9; Griffith et al., Nucleic Acids Res. 1995 Aug. 11;23(15):3003-8; Pardridge et al., Proc Natl Acad Sci USA. 1995 Jun. 6;92(12):5592-6; Boffa et al., Proc Natl Acad Sci USA. 1995 Mar. 14;92(6):1901-5; Gambacorti-Passerini et al., Blood. 1996 Aug. 15;88(4):1411-7; Armitage et al., Proc Natl Acad Sci USA. 1997 Nov. 11;94(23):12320-5; Seeger et al., Biotechniques. 1997 September;23(3):512-7). U.S. Pat. No. 5,700,922 discusses PNA-DNA-PNA chimeric molecules and their uses in diagnostics, modulating protein in organisms, and treatment of conditions susceptible to therapeutics. [0140]
  • Methods of characterizing the antisense binding properties of PNAs are discussed in Rose (Anal Chem. 1993 Dec. 15;65(24):3545-9) and Jensen et al. (Biochemistry. 1997 Apr. 22;36(16):5072-7). Rose uses capillary gel electrophoresis to determine binding of PNAs to their complementary oligonucleotide, measuring the relative binding kinetics and stoichiometry. Similar types of measurements were made by Jensen et al. using BIAcore™ technology. [0141]
  • Other applications of PNAs that have been described and will be apparent to the skilled artisan include use in DNA strand invasion, antisense inhibition, mutational analysis, enhancers of transcription, nucleic acid purification, isolation of transcriptionally active genes, blocking of transcription factor binding, genome cleavage, biosensors, in situ hybridization, and the like. [0142]
  • Polynucleotide Characterization and Expression [0143]
  • Polynucleotides compositions of the present invention may be prepared and/or manipulated using any of a variety of well established techniques (see generally, Sambrook et al., [0144] Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratories, Cold Spring Harbor, N.Y., 1989, and other like references).
  • In other embodiments of the invention, polynucleotide sequences or fragments thereof which encode polypeptides of the invention, or fusion proteins or functional equivalents thereof, may be used in recombinant DNA molecules to direct expression of a polypeptide in appropriate host cells. Due to the inherent degeneracy of the genetic code, other DNA sequences that encode substantially the same or a functionally equivalent amino acid sequence may be produced and these sequences may be used to clone and express a given polypeptide. [0145]
  • As will be understood by those of skill in the art, it may be advantageous in some instances to produce polypeptide-encoding nucleotide sequences possessing non-naturally occurring codons. For example, codons preferred by a particular prokaryotic or eukaryotic host can be selected to increase the rate of protein expression or to produce a recombinant RNA transcript having desirable properties, such as a half-life which is longer than that of a transcript generated from the naturally occurring sequence. [0146]
  • Moreover, the polynucleotide sequences of the present invention can be engineered using methods generally known in the art in order to alter polypeptide encoding sequences for a variety of reasons, including but not limited to, alterations which modify the cloning, processing, and/or expression of the gene product. For example, DNA shuffling by random fragmentation and PCR reassembly of gene fragments and synthetic oligonucleotides may be used to engineer the nucleotide sequences. In addition, site-directed mutagenesis may be used to insert new restriction sites, alter glycosylation patterns, change codon preference, produce splice variants, or introduce mutations, and so forth. [0147]
  • In another embodiment of the invention, natural, modified, or recombinant nucleic acid sequences may be ligated to a heterologous sequence to encode a fusion protein. For example, to screen peptide libraries for inhibitors of polypeptide activity, it may be useful to encode a chimeric protein that can be recognized by a commercially available antibody. A fusion protein may also be engineered to contain a cleavage site located between the polypeptide-encoding sequence and the heterologous protein sequence, so that the polypeptide may be cleaved and purified away from the heterologous moiety. [0148]
  • Sequences encoding a desired polypeptide may be synthesized, in whole or in part, using chemical methods well known in the art (see Caruthers, M. H. et al. (1980) [0149] Nucl. Acids Res. Symp. Ser. 215-223, Horn, T. et al. (1980) Nucl. Acids Res. Symp. Ser. 225-232). Alternatively, the protein itself may be produced using chemical methods to synthesize the amino acid sequence of a polypeptide, or a portion thereof. For example, peptide synthesis can be performed using various solid-phase techniques (Roberge, J. Y. et al. (1995) Science 269:202-204) and automated synthesis may be achieved, for example, using the ABI 431A Peptide Synthesizer (Perkin Elmer, Palo Alto, Calif.).
  • A newly synthesized peptide may be substantially purified by preparative high performance liquid chromatography (e.g., Creighton, T. (1983) Proteins, Structures and Molecular Principles, WH Freeman and Co., New York, N.Y.) or other comparable techniques available in the art. The composition of the synthetic peptides may be confirmed by amino acid analysis or sequencing (e.g., the Edman degradation procedure). Additionally, the amino acid sequence of a polypeptide, or any part thereof, may be altered during direct synthesis and/or combined using chemical methods with sequences from other proteins, or any part thereof, to produce a variant polypeptide. [0150]
  • In order to express a desired polypeptide, the nucleotide sequences encoding the polypeptide, or functional equivalents, may be inserted into appropriate expression vector, i.e., a vector which contains the necessary elements for the transcription and translation of the inserted coding sequence. Methods which are well known to those skilled in the art may be used to construct expression vectors containing sequences encoding a polypeptide of interest and appropriate transcriptional and translational control elements. These methods include in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination. Such techniques are described, for example, in Sambrook, J. et al. (1989) Molecular Cloning, A Laboratory Manual, Cold Spring Harbor Press, Plainview, N.Y., and Ausubel, F. M. et al. (1989) Current Protocols in Molecular Biology, John Wiley & Sons, New York. N.Y. [0151]
  • A variety of expression vector/host systems may be utilized to contain and express polynucleotide sequences. These include, but are not limited to, microorganisms such as bacteria transformed with recombinant bacteriophage, plasmid, or cosmid DNA expression vectors; yeast transformed with yeast expression vectors; insect cell systems infected with virus expression vectors (e.g., baculovirus); plant cell systems transformed with virus expression vectors (e.g., cauliflower mosaic virus, CaMV; tobacco mosaic virus, TMV) or with bacterial expression vectors (e.g., Ti or pBR322 plasmids); or animal cell systems. [0152]
  • The “control elements” or “regulatory sequences” present in an expression vector are those non-translated regions of the vector—enhancers, promoters, 5′ and 3′ untranslated regions—which interact with host cellular proteins to carry out transcription and translation. Such elements may vary in their strength and specificity. Depending on the vector system and host utilized, any number of suitable transcription and translation elements, including constitutive and inducible promoters, may be used. For example, when cloning in bacterial systems, inducible promoters such as the hybrid lacZ promoter of the PBLUESCRIPT phagemid (Stratagene, La Jolla, Calif.) or PSPORT1 plasmid (Gibco BRL, Gaithersburg, Md.) and the like may be used. In mammalian cell systems, promoters from mammalian genes or from mammalian viruses are generally preferred. If it is necessary to generate a cell line that contains multiple copies of the sequence encoding a polypeptide, vectors based on SV40 or EBV may be advantageously used with an appropriate selectable marker. [0153]
  • In bacterial systems, any of a number of expression vectors may be selected depending upon the use intended for the expressed polypeptide. For example, when large quantities are needed, for example for the induction of antibodies, vectors which direct high level expression of fusion proteins that are readily purified may be used. Such vectors include, but are not limited to, the multifunctional [0154] E. coli cloning and expression vectors such as BLUESCRIPT (Stratagene), in which the sequence encoding the polypeptide of interest may be ligated into the vector in frame with sequences for the amino-terminal Met and the subsequent 7 residues of .beta.galactosidase so that a hybrid protein is produced; pIN vectors (Van Heeke, G. and S. M. Schuster (1989) J. Biol. Chem. 264:5503-5509); and the like. pGEX Vectors (Promega, Madison, Wis.) may also be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST). In general, such fusion proteins are soluble and can easily be purified from lysed cells by adsorption to glutathione-agarose beads followed by elution in the presence of free glutathione. Proteins made in such systems may be designed to include heparin, thrombin, or factor XA protease cleavage sites so that the cloned polypeptide of interest can be released from the GST moiety at will.
  • In the yeast, [0155] Saccharomyces cerevisiae, a number of vectors containing constitutive or inducible promoters such as alpha factor, alcohol oxidase, and PGH may be used. For reviews, see Ausubel et al. (supra) and Grant et al. (1987) Methods Enzymol 153:516-544.
  • In cases where plant expression vectors are used, the expression of sequences encoding polypeptides may be driven by any of a number of promoters. For example, viral promoters such as the 35S and 19S promoters of CaMV may be used alone or in combination with the omega leader sequence from TMV (Takamatsu, N. (1987) [0156] EMBO J. 6:307-311. Alternatively, plant promoters such as the small subunit of RUBISCO or heat shock promoters may be used (Coruzzi, G. et al. (1984) EMBO J. 3:1671-1680; Broglie, R. et al. (1984) Science 224:838-843; and Winter, J. et al. (1991) Results Probl. Cell Differ. 17:85-105). These constructs can be introduced into plant cells by direct DNA transformation or pathogen-mediated transfection. Such techniques are described in a number of generally available reviews (see, for example, Hobbs, S. or Murry, L. E. in McGraw Hill Yearbook of Science and Technology (1992) McGraw Hill, New York, N.Y.; pp. 191-196).
  • An insect system may also be used to express a polypeptide of interest. For example, in one such system, [0157] Autographa californica nuclear polyhedrosis virus (AcNPV) is used as a vector to express foreign genes in Spodoptera frugiperda cells or in Trichoplusia larvae. The sequences encoding the polypeptide may be cloned into a non-essential region of the virus, such as the polyhedrin gene, and placed under control of the polyhedrin promoter. Successful insertion of the polypeptide-encoding sequence will render the polyhedrin gene inactive and produce recombinant virus lacking coat protein. The recombinant viruses may then be used to infect, for example, S. frugiperda cells or Trichoplusia larvae in which the polypeptide of interest may be expressed (Engelhard, E. K. et al. (1994) Proc. Natl. Acad. Sci. 91:3224-3227).
  • In mammalian host cells, a number of viral-based expression systems are generally available. For example, in cases where an adenovirus is used as an expression vector, sequences encoding a polypeptide of interest may be ligated into an adenovirus transcription/translation complex consisting of the late promoter and tripartite leader sequence. Insertion in a non-essential E1 or E3 region of the viral genome may be used to obtain a viable virus which is capable of expressing the polypeptide in infected host cells (Logan, J. and Shenk, T. (1984) [0158] Proc. Natl. Acad. Sci. 81:3655-3659). In addition, transcription enhancers, such as the Rous sarcoma virus (RSV) enhancer, may be used to increase expression in mammalian host cells.
  • Specific initiation signals may also be used to achieve more efficient translation of sequences encoding a polypeptide of interest. Such signals include the ATG initiation codon and adjacent sequences. In cases where sequences encoding the polypeptide, its initiation codon, and upstream sequences are inserted into the appropriate expression vector, no additional transcriptional or translational control signals may be needed. However, in cases where only coding sequence, or a portion thereof, is inserted, exogenous translational control signals including the ATG initiation codon should be provided. Furthermore, the initiation codon should be in the correct reading frame to ensure translation of the entire insert. Exogenous translational elements and initiation codons may be of various origins, both natural and synthetic. The efficiency of expression may be enhanced by the inclusion of enhancers which are appropriate for the particular cell system which is used, such as those described in the literature (Scharf, D. et al. (1994) [0159] Results Probl. Cell Differ. 20:125-162).
  • In addition, a host cell strain may be chosen for its ability to modulate the expression of the inserted sequences or to process the expressed protein in the desired fashion. Such modifications of the polypeptide include, but are not limited to, acetylation, carboxylation, glycosylation, phosphorylation, lipidation, and acylation. Post-translational processing which cleaves a “prepro” form of the protein may also be used to facilitate correct insertion, folding and/or function. Different host cells such as CHO, COS, HeLa, MDCK, HEK293, and WI38, which have specific cellular machinery and characteristic mechanisms for such post-translational activities, may be chosen to ensure the correct modification and processing of the foreign protein. [0160]
  • For long-term, high-yield production of recombinant proteins, stable expression is generally preferred. For example, cell lines which stably express a polynucleotide of interest may be transformed using expression vectors which may contain viral origins of replication and/or endogenous expression elements and a selectable marker gene on the same or on a separate vector. Following the introduction of the vector, cells may be allowed to grow for 1-2 days in an enriched media before they are switched to selective media. The purpose of the selectable marker is to confer resistance to selection, and its presence allows growth and recovery of cells which successfully express the introduced sequences. Resistant clones of stably transformed cells may be proliferated using tissue culture techniques appropriate to the cell type. [0161]
  • Any number of selection systems may be used to recover transformed cell lines. These include, but are not limited to, the herpes simplex virus thymidine kinase (Wigler, M. et al. (1977) [0162] Cell 11:223-32) and adenine phosphoribosyltransferase (Lowy, I. et al. (1990) Cell 22:817-23) genes which can be employed in tk.sup.- or aprt.sup.-cells, respectively. Also, antimetabolite, antibiotic or herbicide resistance can be used as the basis for selection; for example, dhfr which confers resistance to methotrexate (Wigler, M. et al. (1980) Proc. Natl. Acad. Sci. 77:3567-70); npt, which confers resistance to the aminoglycosides, neomycin and G-418 (Colbere-Garapin, F. et al (1981) J. Mol. Biol. 150:1-14); and als or pat, which confer resistance to chlorsulfuron and phosphinotricin acetyltransferase, respectively (Murry, supra). Additional selectable genes have been described, for example, trpB, which allows cells to utilize indole in place of tryptophan, or hisD, which allows cells to utilize histinol in place of histidine (Hartman, S. C. and R. C. Mulligan (1988) Proc. Natl. Acad. Sci. 85:8047-51). The use of visible markers has gained popularity with such markers as anthocyanins, beta-glucuronidase and its substrate GUS, and luciferase and its substrate luciferin, being widely used not only to identify transformants, but also to quantify the amount of transient or stable protein expression attributable to a specific vector system (Rhodes, C. A. et al. (1995) Method Mol. Biol. 55:121-131).
  • Although the presence/absence of marker gene expression suggests that the gene of interest is also present, its presence and expression may need to be confirmed. For example, if the sequence encoding a polypeptide is inserted within a marker gene sequence, recombinant cells containing sequences can be identified by the absence of marker gene function. Alternatively, a marker gene can be placed in tandem with a polypeptide-encoding sequence under the control of a single promoter. Expression of the marker gene in response to induction or selection usually indicates expression of the tandem gene as well. [0163]
  • Alternatively, host cells that contain and express a desired polynucleotide sequence may be identified by a variety of procedures known to those of skill in the art. These procedures include, but are not limited to, DNA-DNA or DNA-RNA hybridizations and protein bioassay or immunoassay techniques which include, for example, membrane, solution, or chip based technologies for the detection and/or quantification of nucleic acid or protein. [0164]
  • Host cells transformed with a polynucleotide sequence of interest may be cultured under conditions suitable for the expression and recovery of the protein from cell culture. The protein produced by a recombinant cell may be secreted or contained intracellularly depending on the sequence and/or the vector used. As will be understood by those of skill in the art, expression vectors containing polynucleotides of the invention may be designed to contain signal sequences which direct secretion of the encoded polypeptide through a prokaryotic or eukaryotic cell membrane. Other recombinant constructions may be used to join sequences encoding a polypeptide of interest to nucleotide sequence encoding a polypeptide domain which will facilitate purification of soluble proteins. Such purification facilitating domains include, but are not limited to, metal chelating peptides such as histidine-tryptophan modules that allow purification on immobilized metals, protein A domains that allow purification on immobilized immunoglobulin, and the domain utilized in the FLAGS extension/affinity purification system (Immunex Corp., Seattle, Wash.). The inclusion of cleavable linker sequences such as those specific for Factor XA or enterokinase (Invitrogen. San Diego, Calif.) between the purification domain and the encoded polypeptide may be used to facilitate purification. One such expression vector provides for expression of a fusion protein containing a polypeptide of interest and a nucleic acid encoding 6 histidine residues preceding a thioredoxin or an enterokinase cleavage site. The histidine residues facilitate purification on IMIAC (immobilized metal ion affinity chromatography) as described in Porath, J. et al. (1992, [0165] Prot. Exp. Purif. 3:263-281) while the enterokinase cleavage site provides a means for purifying the desired polypeptide from the fusion protein. A discussion of vectors which contain fusion proteins is provided in Kroll, D. J. et al. (1993; DNA Cell Biol. 12:441-453).
  • In addition to recombinant production methods, polypeptides of the invention, and fragments thereof, may be produced by direct peptide synthesis using solid-phase techniques (Merrifield J. (1963) [0166] J. Am. Chem. Soc. 85:2149-2154). Protein synthesis may be performed using manual techniques or by automation. Automated synthesis may be achieved, for example, using Applied Biosystems 431A Peptide Synthesizer (Perkin Elmer). Alternatively, various fragments may be chemically synthesized separately and combined using chemical methods to produce the full length molecule.
  • T Cells Compositions [0167]
  • The present invention, in another aspect, provides T cells specific for a tumor polypeptide disclosed herein, or for a variant or derivative thereof. Such cells may generally be prepared in vitro or ex vivo, using standard procedures. For example, T cells may be isolated from bone marrow, peripheral blood, or a fraction of bone marrow or peripheral blood of a patient, using a commercially available cell separation system, such as the Isolex™ System, available from Nexell Therapeutics, Inc. (Irvine, Calif.; see also U.S. Pat. No. 5,240,856; U.S. Pat. No. 5,215,926; WO 89/06280; WO 91/16116 and WO 92/07243). Alternatively, T cells may be derived from related or unrelated humans, non-human mammals, cell lines or cultures. [0168]
  • T cells may be stimulated with a polypeptide, polynucleotide encoding a polypeptide and/or an antigen presenting cell (APC) that expresses such a polypeptide. Such stimulation is performed under conditions and for a time sufficient to permit the generation of T cells that are specific for the polypeptide of interest. Preferably, a tumor polypeptide or polynucleotide of the invention is present within a delivery vehicle, such as a microsphere, to facilitate the generation of specific T cells. [0169]
  • T cells are considered to be specific for a polypeptide of the present invention if the T cells specifically proliferate, secrete cytokines or kill target cells coated with the polypeptide or expressing a gene encoding the polypeptide. T cell specificity may be evaluated using any of a variety of standard techniques. For example, within a chromium release assay or proliferation assay, a stimulation index of more than two fold increase in lysis and/or proliferation, compared to negative controls, indicates T cell specificity. Such assays may be performed, for example, as described in Chen et al., [0170] Cancer Res. 54:1065-1070, 1994. Alternatively, detection of the proliferation of T cells may be accomplished by a variety of known techniques. For example, T cell proliferation can be detected by measuring an increased rate of DNA synthesis (e.g., by pulse-labeling cultures of T cells with tritiated thymidine and measuring the amount of tritiated thymidine incorporated into DNA). Contact with a tumor polypeptide (100 ng/ml-100 μg/ml, preferably 200 ng/ml-25 μg/ml) for 3-7 days will typically result in at least a two fold increase in proliferation of the T cells. Contact as described above for 2-3 hours should result in activation of the T cells, as measured using standard cytokine assays in which a two fold increase in the level of cytokine release (e.g., TNF or IFN-γ) is indicative of T cell activation (see Coligan et al., Current Protocols in Immunology, vol. 1, Wiley Interscience (Greene 1998)). T cells that have been activated in response to a tumor polypeptide, polynucleotide or polypeptide-expressing APC may be CD4+ and/or CD8+. Tumor polypeptide-specific T cells may be expanded using standard techniques. Within preferred embodiments, the T cells are derived from a patient, a related donor or an unrelated donor, and are administered to the patient following stimulation and expansion.
  • For therapeutic purposes, CD4[0171] + or CD8+ T cells that proliferate in response to a tumor polypeptide, polynucleotide or APC can be expanded in number either in vitro or in vivo. Proliferation of such T cells in vitro may be accomplished in a variety of ways. For example, the T cells can be re-exposed to a tumor polypeptide, or a short peptide corresponding to an immunogenic portion of such a polypeptide, with or without the addition of T cell growth factors, such as interleukin-2, and/or stimulator cells that synthesize a tumor polypeptide. Alternatively, one or more T cells that proliferate in the presence of the tumor polypeptide can be expanded in number by cloning. Methods for cloning cells are well known in the art, and include limiting dilution.
  • Pharmaceutical Compositions [0172]
  • In additional embodiments, the present invention concerns formulation of one or more of the polynucleotide, polypeptide or T-cell disclosed herein in pharmaceutically-acceptable solutions for administration to a cell or an animal, either alone, or in combination with one or more other modalities of therapy. In particular, the present invention concerns, the use of cripto polynucleotides, polypeptides, fragments, fusions and variants in a pharmaceutical composition for the treatment of tumours. In particular it is preferred that the Cripto is Cripto 1. [0173]
  • It will be understood that, if desired, a composition as disclosed herein may be administered in combination with other agents as well, such as, e.g., other proteins or polypeptides or various pharmaceutically-active agents. In fact, there is virtually no limit to other components that may also be included, given that the additional agents do not cause a significant adverse effect upon contact with the target cells or host tissues. The compositions may thus be delivered along with various other agents as required in the particular instance. Such compositions may be purified from host cells or other biological sources, or alternatively may be chemically synthesized as described herein. Likewise, such compositions may further comprise substituted or derivatized RNA or DNA compositions. [0174]
  • Therefore, in another aspect of the present invention, pharmaceutical compositions are provided comprising one or more of the polynucleotide, polypeptide and/or T-cell compositions described herein in combination with a physiologically acceptable carrier. In certain preferred embodiments, the pharmaceutical compositions of the invention comprise immunogenic polynucleotide and/or polypeptide compositions of the invention for use in prophylactic and theraputic vaccine applications. Vaccine preparation is generally described in, for example, M. F. Powell and M. J. Newman, eds., “Vaccine Design (the subunit and adjuvant approach),” Plenum Press (NY, 1995). Generally, such compositions will comprise one or more polynucleotide and/or polypeptide compositions of the present invention in combination with one or more immunostimulants. [0175]
  • It will be apparent that any of the pharmaceutical compositions described herein can contain pharmaceutically acceptable salts of the polynucleotides and polypeptides of the invention. Such salts can be prepared, for example, from pharmaceutically acceptable non-toxic bases, including organic bases (eg., salts of primary, secondary and tertiary amines and basic amino acids) and inorganic bases (eg., sodium, potassium, lithium, ammonium, calcium and magnesium salts). [0176]
  • In another embodiment, illustrative immunogenic compositions, e.g., vaccine compositions, of the present invention comprise DNA encoding one or more of the polypeptides as described above, such that the polypeptide is generated in situ. As noted above, the polynucleotide may be administered within any of a variety of delivery systems known to those of ordinary skill in the art. Indeed, numerous gene delivery techniques are well known in the art, such as those described by Rolland, [0177] Crit. Rev. Therap. Drug Carrier Systems 15:143-198, 1998, and references cited therein. Appropriate polynucleotide expression systems will, of course, contain the necessary regulatory DNA regulatory sequences for expression in a patient (such as a suitable promoter and terminating signal). Alternatively, bacterial delivery systems may involve the administration of a bacterium (such as Bacillus-Calmette-Guerrin) that expresses an immunogenic portion of the polypeptide on its cell surface or secretes such an epitope.
  • Therefore, in certain embodiments, polynucleotides encoding immunogenic polypeptides described herein are introduced into suitable mammalian host cells for expression using any of a number of known viral-based systems. In one illustrative embodiment, retroviruses provide a convenient and effective platform for gene delivery systems. A selected nucleotide sequence encoding a polypeptide of 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) Virology 180:849-852; Burns et al. (1993) Proc. Natl. Acad. Sci. USA 90:8033-8037; and Boris-Lawrie and Temin (1993) Cur. Opin. Genet. Develop. 3:102-109. [0178]
  • 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 minimizing 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). [0179]
  • 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. [0180]
  • Additional viral vectors useful for delivering the nucleic acid molecules encoding polypeptides of 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 novel molecules 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. [0181]
  • 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. [0182]
  • 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. [0183]
  • Any of a number of alphavirus vectors can also be used for delivery of polynucleotide compositions of the present invention, such as those vectors described in U.S. Pat. Nos. 5,843,723; 6,015,686; 6,008,035 and 6,015,694. Certain vectors based on Venezuelan Equine Encephalitis (VEE) can also be used, illustrative examples of which can be found in U.S. Pat. Nos. 5,505,947 and 5,643,576. [0184]
  • 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. Natl. Acad. Sci. USA (1992) 89:6099-6103, can also be used for gene delivery under the invention. [0185]
  • Additional illustrative information on these and other known viral-based delivery systems can be found, for example, in Fisher-Hoch et al., [0186] Proc. Natl. Acad. Sci. USA 86:317-321, 1989; Flexner et al., Ann. N.Y. Acad. Sci. 569:86-103, 1989; Flexner et al., Vaccine 8:17-21, 1990; U.S. Pat. Nos. 4,603,112, 4,769,330, and 5,017,487; WO 89/01973; U.S. Pat. No. 4,777,127; GB 2,200,651; EP 0,345,242; WO 91/02805; Berkner, Biotechniques 6:616-627, 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 90:11498-11502, 1993; Guzman et al., Circulation 88:2838-2848, 1993; and Guzman et al., Cir. Res. 73:1202-1207, 1993.
  • In another embodiment of the invention, a polynucleotide is administered/delivered as “naked” DNA, for example as described in Ulmer et al., [0187] Science 259:1745-1749, 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, Wis.), some examples of which are described in U.S. Pat. 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. The particles, when delivering nucleic acid are preferably gold beads of a 0.4-4.0 um, more preferably 0.6-2.0 um diameter and the DNA conjugate coated onto these and then encased in a cartridge for placing into the “gene gun”. The particles are typically and preferably delivered to the skin. Other means of delivery to the skin, comprise utilising needle delivery via a needle of a liquid formulation. [0188]
  • DNA vaccines usually consist of a bacterial plasmid vector into which is inserted a strong, normally viral, promoter, the gene of interest which encodes for an antigenic peptide and a polyadenylation/transcriptional termination sequences. Thus gene of interest may encode a full cripto protein as described or simply an antigenic peptide sequence such as described in seq ID no 13-94. The plasmid can be grown in bacteria, such as for example [0189] E. coli and then isolated and prepared in an appropriate medium, depending upon the intended route of administration, before being administered to the host. Following administration the plasmid is taken up by cells of the host where the encoded peptide is produced. The plasmid vector will preferably be made without an origin of replication which is functional in eukaryotic cells, in order to prevent plasmid replication in the mammalian host and integration within chromosomal DNA of the animal concerned.
  • There are a number of advantages of DNA vaccination relative to traditional vaccination techniques. First, it is predicted that because of the proteins which are encoded by the DNA sequence are synthesised in the host, the structure or conformation of the protein will be similar to the native protein associated with the disease state. It is also likely that DNA vaccination will offer protection against different strains of a virus, by generating cytotoxic T lymphocyte response that recognise epitopes from conserved proteins. Furthermore, because the plasmids are taken up by the host cells where antigenic protein can be produced, a long-lasting immune response will be elicited. The technology also offers the possibility of combing diverse immunogens/epitopes into a single preparation. [0190]
  • Helpful background information in relation to DNA vaccination is provided in Donnelly et al “DNA vaccines” Ann. Rev Immunol. 1997 15: 617-648, the disclosure of which is included herein in its entirety by way of reference. [0191]
  • In a related embodiment, other devices and methods that may be useful for gas-driven needle-less injection of compositions of the present invention include those provided by Bioject, Inc. (Portland, Oreg.), some examples of which are described in U.S. Pat. Nos. 4,790,824; 5,064,413; 5,312,335; 5,383,851; 5,399,163; 5,520,639 and 5,993,412. [0192]
  • According to another embodiment, the pharmaceutical compositions described herein will comprise one or more immunostimulants in addition to the immunogenic polynucleotide, polypeptide, T-cell and/or 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, [0193] Bordatella pertussis or Mycobacterium tuberculosis derived proteins. Certain adjuvants are commercially available as, for example, Freund's Incomplete Adjuvant and Complete Adjuvant (Difco Laboratories, Detroit, Mich.); Merck Adjuvant 65 (Merck and Company, Inc., Rahway, N.J.); 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 Th1 type. High levels of Th1-type cytokines (eg., 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 (eg., 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 Th1- and Th2-type responses. Within a preferred embodiment, in which a response is predominantly Th1-type, the level of Th1-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, [0194] Ann. Rev. Immunol. 7:145-173, 1989.
  • Certain preferred adjuvants for eliciting a predominantly Th1-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, Wash.; see, for example, U.S. Pat. Nos. 4,436,727; 4,877,611; 4,866,034 and 4,912,094). CpG-containing oligonucleotides (in which the CpG dinucleotide is unmethylated) also induce a predominantly Th1 response. Such oligonucleotides are well known and are described, for example, in WO 96/02555, WO 99/33488 and U.S. Pat. Nos. 6,008,200 and 5,856,462. Immunostimulatory DNA sequences are also described, for example, by Sato et al., [0195] 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, Mass.); 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 Carbopol® to increase viscosity, or may be formulated in a dry powder form with a powder excipient such as lactose. [0196]
  • In one preferred 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. [0197]
  • 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. [0198]
  • Additional illustrative adjuvants for use in the pharmaceutical compositions of the invention include Montanide ISA 720 (Seppic, France), SAF (Chiron, Calif., United States), ISCOMS (CSL), MF-59 (Chiron), the SBAS series of adjuvants (eg., SBAS-2 or SBAS-4, available from SmithKline Beecham, Rixensart, Belgium), Detox (Enhanzyn®) (Corixa, Hamilton, Mont.), RC-529 (Corixa, Hamilton, Mont.) and other aminoalkyl glucosaminide 4-phosphates (AGPs), such as those described in pending U.S. patent application Ser. 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. [0199]
  • Other preferred adjuvants include adjuvant molecules of the general formula (I): [0200]
  • HO(CH2CH2O)n-A-R
  • Wherein, n is 1-50, A is a bond or —C(O)—, R is C[0201] 1-50 alkyl or Phenyl C1-50 alkyl.
  • One embodiment of the present invention consists of a vaccine formulation 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[0202] 1-50, preferably C4-C20 alkyl and most preferably C1-2 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 (12th 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 preferred adjuvant combination is preferably with CpG as described in the pending UK patent application GB 9820956.2. [0203]
  • According to another embodiment of this invention, an immunogenic composition described herein 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-tumor 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 tumor and peritumoral tissues, and may be autologous, allogeneic, syngeneic or xenogeneic cells. [0204]
  • 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, [0205] Nature 392:245-251, 1998) and have been shown to be effective as a physiological adjuvant for eliciting prophylactic or therapeutic antitumor immunity (see Timmerman and Levy, Ann. Rev. Med. 50:507-529, 1999). In 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 naïve 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. 4:594600, 1998).
  • Dendritic cells and progenitors may be obtained from peripheral blood, bone marrow, tumor-infiltrating cells, peritumoral 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 marrow may be differentiated into dendritic cells by adding to the culture medium combinations of GM-CSF, IL-3, TNFα, CD40 ligand, LPS, flt3 ligand and/or other compound(s) that induce differentiation, maturation and proliferation of dendritic cells. [0206]
  • 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 correlates 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 (eg., CD54 and CD11) and costimulatory molecules (eg., CD40, CD80, CD86 and 4-1BB). [0207]
  • 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., [0208] 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 tumor polypeptide, DNA (naked or within a plasmid vector) or RNA; or with antigen-expressing recombinant bacterium or viruses (eg., vaccinia, fowlpox, adenovirus or lentivirus vectors). Prior to loading, the polypeptide may be covalently conjugated to an immunological partner that provides T cell help (eg., 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 of this invention, the type of carrier will typically vary depending on the mode of administration. Compositions of the present invention may be formulated for any appropriate manner of administration, including for example, topical, oral, nasal, mucosal, intravenous, intracranial, intraperitoneal, subcutaneous and intramuscular administration. [0209]
  • 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. In 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 amphiphilic compound, such as a phospholipid (see eg., U.S. Pat. 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. [0210]
  • 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. Pat. 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 for many applications. Another illustrative carrier/delivery system employs a carrier comprising particulate-protein complexes, such as those described in U.S. Pat. No. 5,928,647, which are capable of inducing a class I-restricted cytotoxic T lymphocyte responses in a host. [0211]
  • The pharmaceutical compositions of the invention will often further comprise one or more buffers (eg., 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 (eg., 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. [0212]
  • 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. [0213]
  • 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 administration and formulation, is well known in the art, some of which are briefly discussed below for general purposes of illustration. [0214]
  • In 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. [0215]
  • 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 al., Nature 1997 Mar. 27;386(6623):410-4; Hwang et al., Crit Rev Ther Drug Carrier Syst 1998;15(3):243-84; U.S. Pat. No. 5,641,515; U.S. Pat. No. 5,580,579 and U.S. Pat. No. 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 corn starch, potato starch, alginic acid and the like; a lubricant, such as magnesium stearate; and a sweetening agent, such as sucrose, lactose or saccharin may be added or a flavoring agent, such as peppermint, oil of wintergreen, or cherry 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. [0216]
  • 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. [0217]
  • For oral administration the compositions of 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. [0218]
  • In certain circumstances it will be desirable to deliver the pharmaceutical compositions disclosed herein parenterally, intravenously, intramuscularly, or even intraperitoneally or intradermally. Such approaches are well known to the skilled artisan, some of which are further described, for example, in U.S. Pat. No. 5,543,158; U.S. Pat. No. 5,641,515 and U.S. Pat. No. 5,399,363. In 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. [0219]
  • 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. Pat. No. 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. [0220]
  • 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 may be dissolved in 1 ml of isotonic NaCl 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 Biologics standards. [0221]
  • 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. [0222]
  • 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. [0223]
  • 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., in U.S. Pat. No. 5,756,353 and U.S. Pat. No. 5,804,212. Likewise, the delivery of drugs using intranasal microparticle resins (Takenaga et al., J Controlled Release 1998 Mar. 2;52(1-2):81-7) and lysophosphatidyl-glycerol compounds (U.S. Pat. No. 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. Pat. No. 5,780,045. [0224]
  • 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. In 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, compositions of the present invention can be bound, either covalently or non-covalently, to the surface of such carrier vehicles. [0225]
  • The formation and use of liposome and liposome-like preparations as potential drug carriers is generally known to those of skill in the art (see for example, Lasic, Trends Biotechnol 1998 July;16(7):307-21; Takakura, Nippon Rinsho 1998 March;56(3):691-5; Chandran et al., Indian J Exp Biol. 1997 August;35(8):801-9; Margalit, Crit Rev Ther Drug Carrier Syst. 1995;12(2-3):233-61; U.S. Pat. No. 5,567,434; U.S. Pat. No. 5,552,157; U.S. Pat. No. 5,565,213; U.S. Pat. No. 5,738,868 and U.S. Pat. No. 5,795,587, each specifically incorporated herein by reference in its entirety). [0226]
  • 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 April;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, he use of liposomes does not appear to be associated with autoimmune responses or unacceptable toxicity after systemic delivery. [0227]
  • 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). [0228]
  • Alternatively, in other embodiments, the invention provides for pharmaceutically-acceptable nanocapsule formulations of the compositions of the present invention. Nanocapsules can generally entrap compounds in a stable and reproducible way (see, for example, Quintanar-Guerrero et al., Drug Dev Ind Pharm. 1998 December;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 viva. Such particles can be made as described, for example, by Couvreur et al., Crit Rev Ther Drug Carrier Syst. 1988;5(1):1-20; zur Muhlen et al., Eur J Pharm Biopharm. 1998 March;45(2):149-55; Zambaux et al. J Controlled Release. 1998 Jan. 2;50(1-3):31-40; and U.S. Pat. No. 5,145,684. [0229]
  • Cancer Therapeutic Methods [0230]
  • In further aspects of the present invention, the pharmaceutical compositions described herein may be used for the treatment of cancer, particularly for the immunotherapy of colon or colorectal cancer. In other embodiments the compositions can be used to treat breast, or non-small cell lung carcinoma. Typically the composition will be useful for treating patients whose cancers express cripto antigen and/or whose metastases express cripto antigen. Within such methods, the pharmaceutical compositions described herein are administered to a patient, typically a warm-blooded animal, preferably a human. A patient may or may not be afflicted with cancer. Accordingly, the above pharmaceutical compositions may be used to prevent the development of a cancer or to treat a patient afflicted with a cancer. Pharmaceutical compositions and vaccines may be administered either prior to or following surgical removal of primary tumors and/or treatment such as administration of radiotherapy or conventional chemotherapeutic drugs. As discussed above, administration of the pharmaceutical compositions may be by any suitable method, including administration by intravenous, intraperitoneal, intramuscular, subcutaneous, intranasal, intradermal, anal, vaginal, topical and oral routes. [0231]
  • Within certain embodiments, immunotherapy may be active immunotherapy, in which treatment relies on the in vivo stimulation of the endogenous host immune system to react against tumors with the administration of immune response-modifying agents (such as polypeptides and polynucleotides as provided herein). [0232]
  • Within other embodiments, immunotherapy may be passive immunotherapy, in which treatment involves the delivery of agents with established tumor-immune reactivity (such as effector cells) that can directly or indirectly mediate antitumor effects and does not necessarily depend on an intact host immune system. Examples of effector cells include T cells as discussed above, T lymphocytes (such as CD8+ cytotoxic T lymphocytes and CD4+ T-helper tumor-infiltrating lymphocytes), killer cells (such as Natural Killer cells and lymphokine-activated killer cells), B cells and antigen-presenting cells (such as dendritic cells and macrophages) expressing a polypeptide provided herein. T cell receptors and antibody receptors specific for the polypeptides recited herein may be cloned, expressed and transferred into other vectors or effector cells for adoptive immunotherapy. [0233]
  • Effector cells may generally be obtained in sufficient quantities for adoptive immunotherapy by growth in vitro, as described herein. Culture conditions for expanding single antigen-specific effector cells to several billion in number with retention of antigen recognition in vivo are well known in the art. Such in vitro culture conditions typically use intermittent stimulation with antigen, often in the presence of cytokines (such as IL-2) and non-dividing feeder cells. As noted above, immunoreactive polypeptides as provided herein may be used to rapidly expand antigen-specific T cell cultures in order to generate a sufficient number of cells for immunotherapy. In particular, antigen-presenting cells, such as dendritic, macrophage, monocyte, fibroblast and/or B cells, may be pulsed with immunoreactive polypeptides or transfected with one or more polynucleotides using standard techniques well known in the art. For example, antigen-presenting cells can be transfected with a polynucleotide having a promoter appropriate for increasing expression in a recombinant virus or other expression system. Cultured effector cells for use in therapy must be able to grow and distribute widely, and to survive long term in vivo. Studies have shown that cultured effector cells can be induced to grow in vivo and to survive long term in substantial numbers by repeated stimulation with antigen supplemented with IL-2 (see, for example, Cheever et al., Immunological Reviews 157:177, 1997). [0234]
  • Alternatively, a vector expressing a polypeptide recited herein may be introduced into antigen presenting cells taken from a patient and clonally propagated ex vivo for transplant back into the same patient. Transfected cells may be reintroduced into the patient using any means known in the art, preferably in sterile form by intravenous, intracavitary, intraperitoneal or intratumor administration. [0235]
  • Routes and frequency of administration of the therapeutic compositions described herein, as well as dosage, will vary from individual to individual, and may be readily established using standard techniques. In general, the pharmaceutical compositions and vaccines may be administered by injection (e.g., intracutaneous, intramuscular, intravenous, intradermally or subcutaneous), intranasally (e.g., by aspiration) or orally. Preferably, between 1 and 10 doses may be administered over a 52 week period. Preferably, 6 doses are administered, at intervals of 1 month, and booster vaccinations may be given periodically thereafter. Alternate protocols may be appropriate for individual patients. A suitable dose is an amount of a compound that, when administered as described above, is capable of promoting an anti-tumor immune response, and is at least 10-50% above the basal (i.e., untreated) level. Such response can be monitored by measuring the anti-tumor antibodies in a patient or by vaccine-dependent generation of cytolytic effector cells capable of killing the patient's tumor cells in vitro. Such vaccines should also be capable of causing an immune response that leads to an improved clinical outcome (e.g., more frequent remissions, complete or partial or longer disease-free survival) in vaccinated patients as compared to non-vaccinated patients. In general, for pharmaceutical compositions and vaccines comprising one or more polypeptides, the amount of each polypeptide present in a dose ranges from about 25 μg to 5 mg per kg of host. Suitable dose sizes will vary with the size of the patient, but will typically range from about 0.1 ml to about 5 ml. [0236]
  • In general, an appropriate dosage and treatment regimen provides the active compound(s) in an amount sufficient to provide therapeutic and/or prophylactic benefit. Such a response can be monitored by establishing an improved clinical outcome (e.g., more frequent remissions, complete or partial, or longer disease-free survival) in treated patients as compared to non-treated patients. Increases in preexisting immune responses to a tumor protein generally correlate with an improved clinical outcome. Such immune responses may generally be evaluated using standard proliferation, cytotoxicity or cytokine assays, which may be performed using samples obtained from a patient before and after treatment. [0237]
  • As used herein “cripto” refers to polypeptides having about 188 amino acids and being expressed in carcinoma cells. In addition, “cripto” refers to polypeptides having at least 70%, but preferably 80%, and more preferably 90%, and more preferably 95%, and yet most preferably 98% sequence identity to SEQ ID NOs: 3 or 4 or variants thereof, including Cripto 1 and Cripto 3. [0238]
  • EXAMPLES
  • The following examples illustrate various aspects of this invention. These examples do not limit the scope of this invention which is defined by the appended claims. [0239]
  • Example 1a
  • Expression of Immunoreactive Cripto-1 in Human Lesions [0240]
    Non-involved Premalignant
    Tissue Epithelium Lesion Carcinoma
    TA TVA
    Colon 25/193 (13) 26/65 (40) 10/13 (77) 122/168 (73)**
    IM  17/37 (46)**
    Stomach  1/37 (3) 16/30 (53)
    Pancreas  10/58 (17)  58/98 (59)**
    Hyperplasias Adenomas
    Gall Bladder N.D.  6/9 (67)  4/7 (58)  89/132 (68)**
    DCIS
    Breast  5/33 (15) 26/55 (47) 497/631 (79)**
    Non Small 178/195 (91)**
    Cell Lung
    Non-involved
    Tissue Epithelium Cystadenomas Serous Mucinous
    Serous Mucinous
    Ovary  6/7 (86) post- 6/14 (43)  0/7  4/10 (40)  4/5 (80)
    menopausal
    23/40 (58)
    25/48 (52)
     3/9 (33)  3/8 (38)  4/8 (50)  9/10 (90)
    Borderline: 10/10
    Endometrium 10/28 (36) 53/91 (58)*
    post-menopausal
    Hyperplasias
    18/30 (60) 68/96 (71)*
    Cervix  4/25 (17) 40/74 (54)*
    Testis  0/3 29/51 (57)**
    Embryonal
    Carcinomas Seminomas
    19/19 (100)** 10/32 (31)
    Adrenal Cortex  1/3 (33)
    Bladder  0/6 23/39 (60)**
    Renal  0/18
    Prostate  0/9
  • Example 1.6 Over-Expression of Cripto in Cancerous Tissues
  • Real-time RT-PCR (U. Gibson. 1996. Genome Research: 6,996) is used to compare mRNA transcript abundance of the target protein in a panel of normal and tumor tissues and/or cell lines. This analysis is critical to establish the tumor specificity of Cripto expression, which is an important criterion a good vaccine candidate must fulfil. [0241]
  • Total RNA is extracted from snap frozen biopsies or cell lines using TriPure reagent (Roche). Total RNA from normal tissues is also purchased from InVitrogen. 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 RiboGreen dye (Molecular Probes). [0242]
  • TaqMan primers (forward primer sequence: TGGGTAGGAAAGAGGAAGCAAAT, SEQ ID NO:7; reverse primer sequence: TGCTTCTCTACCACCACCTAATCA, SEQ ID NO:8) and probe for real-time RT-PCR amplification are designed with the Perkin-Elmer Primer Express software using default options for TaqMan amplification conditions. [0243]
  • Real-time reactions are assembled according to standard PCR protocols using 2 ng of reverse transcribed mRNA (Expand RT, Roche) for each reaction. Either SybrI or TaqMan detection is undergone, depending on the evaluated sample. In case of SybrI detection, SybrI dye (Molecular Probes) is added at a final dilution of {fraction (1/75000)} for real-time detection, and TaqMan probe is omitted. 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. Ct values are obtained from each tissue sample for the target mRNA (CtX) and for the actin mRNA (CtA). [0244]
  • As the efficiency of PCR amplification under the prevailing experimental conditions is close to the theoretical amplification efficiency, 2[0245] (CtA−CtX) value is an estimate of the relative target transcript level of the sample, standardized with respect to Actin transcript level. A value of 1 thus suggests the candidate antigen and Actin have the same expression level.
  • RT-PCR analysis, using SybrI detection, was performed on a set of colon tumor and matched normal colon from 6 different patients and 48 normal tissue samples. A TaqMan detection was run on a set of colon tumor and matched normal colon from 6 other patients (reactions were run in triplicates) and 48 normal tissue samples. Tested normal tissues (and the abbreviations used in graphics) are shown below: [0246]
  • Adrenal gland (Ad_Gl) [0247]
  • Aorta (Ao) [0248]
  • Bladder (Bl) [0249]
  • Bone marrow Bo_Ma [0250]
  • Brain (Bra, Bra1, Bra2, Bra3, Bra4, Bra5) [0251]
  • Cervix (Ce) [0252]
  • Colon (Co) [0253]
  • Fallopian tube (Fa_Tu) [0254]
  • Heart (He) [0255]
  • Ileum (Il) [0256]
  • Kidney (Ki) [0257]
  • Liver (Li, Li 1, Li2) [0258]
  • Lung (Lu) [0259]
  • Lymph node (Ly_No) [0260]
  • Esophagus (Oe) [0261]
  • Ovary (Ov) [0262]
  • Pancreas (Pa, Panc1, Panc2) [0263]
  • Parathyrois (Pa_Thy) [0264]
  • Placenta (Pl) [0265]
  • Prostate (Pr) [0266]
  • Rectum (Re) [0267]
  • Skin (Sk) [0268]
  • Skeletal muscle (Sk_Mu) [0269]
  • Small intestine (Sm_In) [0270]
  • Spleen (Sp) [0271]
  • Stomach (St) [0272]
  • Testis (Te) [0273]
  • Thyroid (Thyr, Thy, Thy1, Thy2) [0274]
  • Thymus (Thym1,)Thym2 [0275]
  • Trachea (Tr, Tra) [0276]
  • Real-time RT-PCR reactions, using SybrI detection, were also performed on a set of 7 lung cell lines: [0277]
  • CRL-5803 (Carcinoma, Non-Small Cell Lung Cancer, large cell, neuroendocrine, metastatic site: lymph node) [0278]
  • CRL-5807 (Bronchioalveolar carcinoma, Non-Small Cell Lung Cancer) [0279]
  • CRL-5810 (Adenocarcinoma, Non-Small Cell Lung Cancer) [0280]
  • CRL-5815 (Carcinoid, lung bronchus) [0281]
  • CRL-5865 (Adenocarcinoma, metastatic site: pleural effusion) [0282]
  • CRL-9609 (Normal lung, bronchus, epithelial, virus transformed) [0283]
  • HTB-177 (Carcinoma, large cell lung cancer, pleural effusion) and 2 colon cell lines: [0284]
  • CRL-2159 (Carcinoma, Cecum, Dukes' B) [0285]
  • CCL-250 [0286]
  • Fresh biopsy normal lung tissues (Lu(ucl), Lu(IVG)) and a lung tumor tissue (LuTum) were also performed as control. [0287]
  • RT-PCR results on colorectal biopsies and normal tissues are shown in FIGS. 1, 2, 3 and 4, and in Table 1. RT-PCR results on cell lines are shown in FIG. 5. [0288]
    TABLE 1
    Cripto expression in colorectal tumors and normal tissues.
    Sybr detection1 TaqMan detection1
    Colorectal tumor versus
    adjacent normal colon2
    Number of over-expressing 5/6 5/6
    patients
    Average over-expression fold 200 90
    in over-expressing patients
    Median over-expression fold  64 (22-724) 20 (4-397)
    (minimum-maximum)
    Colorectal tumor versus
    average normal tissues2
    Number of over-expressing 5/6 4/6
    patients
    Average over-expression fold  10 12
    in over-expressing patients
    Median over-expression fold  11 (3-22)  7 (3-32)
    (minimum-maximum)
    Normal tissues with a high Spleen (0.5) Spleen (0.75), ovary (2)4
    transcript level3 (normal-to-
    tumor ratio)
    # TaqMan detection involved 6 patients and measures were done in triplicates, whereas Sybr detection was undergone on 6 different patients.
  • Table 1, FIGS. 1, 2, 3 & 4 clearly show that Cripto, while being marginally expressed in normal adult tissues, is highly over-expressed in a majority of colorectal tumors, with an over-expression rate of more than ten fold. Moreover, FIG. 5 indicates Cripto is dramatically over-expressed in a lung tumor cell line (CRL-5815). Cripto tumor associated antigen is therefore a suitable vaccine candidate to treat both colorectal and lung cancer patients. [0289]
  • Example 2 Cloning of Cripto-1 c-DNA from Lung Tumor Cell Lines
  • Total RNA was extracted using TriPure reagent from 10[0290] 7 cultured cells of 7 different lung cell lines (see section 1 for the complete list of cell lines). Total RNAs were pooled, and mRNA was purified from pooled total RNA on oligo-d(T) magnetic beads (Dynal). 250 ng of mRNA were used for cDNA synthesis. Quantification of the mRNA is performed by spectrofluorimetry (VersaFluor, BioRad) using RiboGreen dye (Molecular Probes). cDNA was synthesized using the GeneRacer technology (Invitrogen) which ensures the amplification of only full-length transcripts. mRNA was treated with CIP. mRNA 5′ ends were decapped with TAP (Tobacco Acid Pyrophosphatase) and were ligated to a specific RNA oligonucleotide. The ligated mRNA was reverse transcribed into cDNA using an oligod(T)-tailed primer. Amplification of cDNA was performed using both GeneRacer flanking primers (Advantage, Clontech). Cripto amplification was performed on 10 ng of GeneRacer cDNA using gene specific PCR primers (forward primer sequence: CGTCCAAGGCCGAAAGCCCTCCAGTT, SEQ ID NO:9; and reverse primer sequence: TTGGGAGAGGGCAGGGCAAAGAAGTAAGAA, SEQ ID NO:10). PCR reaction was done with Advantage II Taq DNA polymerase (Clontech) under standard conditions. PCR product was cloned in pCR4-TOPO plasmid (Invitrogen). Amplified sequence (SEQ ID NO:95) was shown to display a variation at codon 22 (SEQ ID NO:96): Ala (GCC) instead of Val (GTC) in the native version (SeqID6). Native version was restored by PCR mutagenesis.
  • Example 3 Immunogenicity of Cripto Tumor-Associated Antigen in Animal Models
  • The immunogenicity of the antigen of the present invention can be verified by immunizing rabbits and mice using various means of immunization. Indeed, immunization with Cripto forms, either peptide or recombinant protein could induce humoral immune response with the generation of specific antibodies against Cripto and/or could induce a Cripto specific cellular immune response. In vivo delivery of Cripto protein using for instance, naked DNA in an appropriate vector encoding Cripto or fragments of Cripto, Cripto genes delivered by a viral vector encoding Cripto or fragments of Critpo, could also be useful to demonstrate Cripto immunogenicity. [0291]
  • 3.1: Synthetic Peptide Immunization. [0292]
  • The synthetic peptides from human Cripto-1 amino-acid sequence selected to immunize rabbits are GHQEFARPSRGYL (13 amino acids, SEQ ID NO:11), and QEEPAIRPRSSQRVPPMG (18 amino acids, SEQ ID NO:12). Synthetic peptides are then conjugated to a carrier protein (KLH). Conjugates are formulated with Freund's adjuvant, and two rabbits are immunized with each of the conjugates. Four weeks after the second immunization and four weeks after the third immunization, a blood sample is taken. Anti-Cripto antibody titers are estimated in the serum by ELISA and/or Western Blot following standard protocols (see section 3.5). [0293]
  • 3.2: Nucleic Acids Immunization. [0294]
  • pcDNA3.1 vector (Invitrogen) is used to construct the vaccinating plasmid. To promote secretion of the in viva translated protein and to therefore induce the humoral response against the present invention antigen, nucleic acid sequence encoding Cripto-1 with its own signal peptide is inserted into the vector polycloning site. The recombinant expression plasmid is used to transform a host [0295] E. coli strain such as BL21.
  • The above recombinant strain is grown in conventional cell culture medium. Bacteria are harvested before reaching the stationary phase. Plasmid preparation using Quiagen system is undergone for injection in mice. [0296]
  • Six to eight weeks-old Balb/c mice receive intramuscular injections of recombinant expression plasmid. Two weeks after the last injection, a blood sample is taken. Titers of specific antibodies elicited against the present invention antigen are determined by ELISA and/or Western Blot (see section 3.5). [0297]
  • 3.3 Viral Vector Immunization Using Adenoviruses. [0298]
  • Recombinant adenoviruses are effective vectors for gene-based vaccination because they are capable of eliciting humoral and cellular immune responses against the encoded antigen. The nucleotide sequence coding for Cripto-1 protein with its own signal sequence could be inserted in an appropriated Adeno derivative viral vector. The adenoviral recombinant vector could be administrated to mice by different routes (intramuscular, intranasal, intradermal, subcutaneous or intraeperitoneal) After two week, blood samples could be taken and the titer of antibodies elicited examined. Additional experiments to measure the cellular immune response could also be performed. [0299]
  • 3.4: Recombinant Protein Immunization. [0300]
  • 3.4.1: Expression and Purification of Cripto-1 Recombinant Protein [0301]
  • Expression in microbial hosts, is used to produce the whole protein or fragments of the invention antigen for immunization purposes. Recombinant proteins may be expressed in two microbial hosts, [0302] 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.
  • The expression strategy first involves the design of the primary structure of the recombinant antigen. In general, an expression fusion partner (EFP) to improve levels of expression and/or an immune fusion partner (IFP) to modulate the immunogenic properties of the antigen, are placed at the N-terminal extremity. In addition, an affinity fusion partner (AFP) useful for facilitating further purification is included at the C-terminal end. [0303]
  • When the recombinant strains are available, the recombinant product is characterized by the evaluation of the level of expression and the prediction of further solubility of the engineered protein by analysis of its behavior in the crude extract. [0304]
  • After growth in appropriate culture medium and induction of the recombinant protein expression, total extracts are analyzed by SDS-PAGE. The recombinant proteins are visualized in stained gels and identified by Western blot analysis using the specific anti-peptide antibodies generated by peptide immunization in rabbit (see section 3.1). [0305]
  • A comparative evaluation of the different versions of the expressed antigen and expression hosts will allow the selection of the most promising candidate and host that is to be used for further purification and further immunological evaluation. [0306]
  • The purification schemes follow a classical approach based on the presence of a Histidine 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 (IMAC; Ni++NTA 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. [0307]
  • 3.4.2: Protein Immunization [0308]
  • Rabbits are immunized, intramuscularly several times at several week intervals with recombinant purified protein, formulated in the adjuvant 3D-MPL/QS21. Three weeks after each immunization, blood samples are taken. Anti-Cripto antibody titer is estimated in the serum by ELISA. The specificity of the anti-Cripto antibodies generated is tested by Western Blot (see section 3.5) using the purified protein and including appropriated controls. [0309]
  • 3.5: Immunological Response Assays of Cripto-Immunized Animals. [0310]
  • Humoral response to Cripto immunization is assessed by measuring Cripto specific antibody titers in animal sera using ELISA and Western Blot. The following material harboring Cripto-1 derived peptides or full protein could be used for such test: [0311]
  • Cripto synthetic peptides (see section 3.1 for possible peptides), or [0312]
  • protein extracts from cultures of Cripto-expressing cell lines (see section 1 for possible cell lines), or [0313]
  • lysates of COS cells that have been engineered to transiently express a Cripto recombinant plasmid (see below), or [0314]
  • protein extracts of recombinant [0315] E. coli or yeast strains (see section 3.3.1 for recombinant strain generation), or
  • purified recombinant antigen (see section 3.3.1 for antigen purification). [0316]
  • Transient expression of Cripto in COS cells is obtained by transiently transfecting COS cells with recombinant pcDNA3.1 plasmid prepared for nucleic acid vaccination (see section 3.2). [0317]
  • For ELISA reactions, one of the above mentioned antigen is coated on microtiter plates (purified recombinant protein is preferred). ELISA is then performed using standard protocol. [0318]
  • Western Blots are realized under standard conditions with one of the above mentioned antigen (purified recombinant protein is preferred, synthetic peptides are not used) [0319]
  • The cellular response can also be assessed by stimulating in vitro vaccinated mouse spleen cells with the peptides used to immunize the mice, or with antigen-derived overlapping peptides covering the whole antigen sequence. [0320]
  • Example 4 Demonstration of Existing Cripto Specific Human T-Cell by In Vitro Priming
  • Immunological relevance of Cripto-1 can be further confirmed by in vitro priming of human T cells. All T cell lymphocytes, T cell lines and dendritic cells are derived from PBMCs (peripheral blood mononuclear cells) of healthy donors or cancer patients (preferred donors are from HLA-A2 subtype). [0321]
  • Epitopes Binding of HLA Alleles Prediction: [0322]
  • The HLA Class I binding peptide sequences (nonamers, decamers) are predicted either by the Parker's algorithm (Parker, et al. [0323] J. Immunol. 152:163 (1994)) (http://bimas.dcrt.nih.gov/molbio/hla_bind/), and the Rammensee method (Rammensee et al. (1997). Landes Bioscience (1997); Rammensee et al., Immunogenetics 41,178-228 (1995)). (http://syfpeithi.bmi-heidelberg.com/Scripts/MHCServer.dll/EpPredict.htm).
  • The HLA Class II binding peptide sequences (nonamers) are predicted using the Tepitope algorithm (Sturniolo et al. [0324] Nat Biotechnol. 17: 555-61 (1999).
  • CD8+ T-Cell Response: [0325]
  • 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 interest in the correct reading frame to be cloned in an appropriate delivery system and to be used to predict the sequence of the HLA binding peptides. [0326]
  • Peptide-Based Approach: [0327]
  • For this approach, an HLA-A2.1/Kb transgenic mouse model is used for screening of the HLA-A2.1 peptides. Briefly, transgenic mice are immunized with adjuvanted HLA-A2 peptides, those able to induce a CD8 response (as defined by an efficient lysis or g-IFN production on peptide-pulsed target cells) are further analyzed in the human system. [0328]
  • Human dendritic cells (cultured according to Romani et al. [0329] J. Exp. Med. 180:83-93(1994)) will be pulsed with the selected peptides and used to stimulate CD8+-sorted T cells (by Facs). After several weekly stimulation, the CD8+ lines are 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 are then tested on cDNA-transfected tumor cells (HLA-A2 transfected LnCaP, Skov3 or CAMA tumor cells).
  • Whole Gene-Based Approach: [0330]
  • CD8+ T cell lines are primed and stimulated with either gene-gun transfected dendritic cells, retrovirally transduced B7.1-transfected fibroblasts, recombinant pox virus (Kim, et al., [0331] J. Immunother. 20:276-286 (1997)) or adenovirus (Butterfield, et al. J. Immunol. 161:5607-13 (1998)). 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 stimulation, the CD8+ lines are tested on cDNA-transfected tumor cells as indicated above. Peptide specificity and identity is determined to confirm the immunological validation of antigen of the present invention. [0332]
  • CD4+ T-Cell Response: [0333]
  • 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. [0334]
  • Results [0335]
    A - Prediction of Class I epitopes using the Parker method.
    A-1 Cripto-1 and -3 Class I epitopes.
    Start
    HLA allele Epitope size position Sequence Score SEQ ID NO:
    A68 Decamer 12 SVIWIMAISK 240.000 SEQ ID NO: 13
    A68 Decamer 117 SVPHDTWLPK 120.000 SEQ ID NO: 14
    B2705 Nonamer 33 HQEFARPSR 100.000 SEQ ID NO: 15
    B2705 Nonamer 59 IRPRSSQRV 600.000 SEQ ID NO: 16
    B2705 Nonamer 72 IQHSKELNR 100.000 SEQ ID NO: 17
    B2705 Nonamer 103 GRNCEHDVR 1000.000 SEQ ID NO: 18
    B2705 Nonamer 110 VRKENCGSV 600.000 SEQ ID NO: 19
    B2705 Nonamer 138 LRCFPQAFL 2000.000 SEQ ID NO: 20
    B2705 Nonamer 161 SRTPELPPS 200.000 SEQ ID NO: 21
    B2705 Decamer 65 QRVPPMGIQH 200.000 SEQ ID NO: 22
    B2705 Decamer 79 NRTCCLNGGT 200.000 SEQ ID NO: 23
    B2705 Decamer 103 GRNCEHDVRK 2000.000 SEQ ID NO: 93
    B2705 Decamer 136 GQLRCFPQAF 100.000 SEQ ID NO: 24
    B2705 Decamer 161 SRTPELPPSA 200.000 SEQ ID NO: 94
    B5101 Decamer 143 QAFLPGCDGL 110.000 SEQ IDNO: 25
    B5102 Decamer 143 QAFLPGCDGL 302.500 SEQ ID NO: 25
    B5102 Decamer 150 DGLVMDEHLV 120.000 SEQ ID NO: 26
    B60 Nonamer 23 FELGLVAGL 325.000 SEQ ID NO: 27
    B60 Nonamer 76 KELNRTCCL 320.000 SEQ ID NO: 28
    B62 Nonamer 137 QLRCFPQAF 240.000 SEQ ID NO: 29
    B62 Decamer 136 GQLRCFPQAF 160.000 SEQ ID NO: 24
    B7 Nonamer 169 SARTTTFML 120.000 SEQ ID NO: 30
  • [0336]
    A-2 Cripto-1 specific class I epitopes.
    Start
    HLA allele Epitope size position Sequence Score SEQ ID NO:
    A0201 Nonamer 5 KMARFSYSV 668.086 SEQ ID NO:31
    A0201 Decamer 16 IMAISKVFEL 349.885 SEQ ID NO:32
    A0201 Decamer 13 VIWIMAISKV 310.361 SEQ ID NO:33
    A0201 Decamer 175 FMLVGICLSI 128.242 SEQ ID NO:34
    B2702 Nonamer 7 ARFSYSVIW 500.000 SEQ ID NO:35
    B2702 Nonamer 3 CRKMARFSY 200.000 SEQ ID NO:36
    B2702 Decamer 7 ARFSYSVIWI 300.000 SEQ ID NO:37
    B2702 Decamer 37 ARPSRGYLAF 200.000 SEQ ID NO:38
    B2705 Nonamer 7 ARFSYSVIW 1000.000 SEQ ID NO:35
    B2705 Nonamer 3 CRKMARFSY 1000.000 SEQ ID NO:36
    B2705 Nonamer 170 ARTTTFMLV 600.000 SEQ ID NO:39
    B2705 Nonamer 37 ARPSRGYLA 200.000 SEQ ID NO:40
    B2705 Decamer 7 ARFSYSVIWI 3000.000 SEQ ID NO:37
    B2705 Decamer 37 ARPSRGYLAF 1000.000 SEQ ID NO:38
    B2705 Decamer 3 CRKMARFSYS 200.000 SEQ ID NO:41
    B4403 Decamer 34 QEFARPSRGY 120.000 SEQ ID NO:42
    B5101 Nonamer 6 MARFSYSVI 286.000 SEQ ID NO:43
    B5101 Decamer 169 SARTTTFMLV 110.000 SEQ ID NO:44
    B5102 Nonamer 17 MAISKVFEL 150.000 SEQ ID NO:45
    B5102 Nonamer 6 MARFSYSVI 100.000 SEQ ID NO:43
    B5103 Nonamer 6 MARFSYSVI 100.000 SEQ ID NO:43
    B5103 Decamer 169 SARTTTFMLV 121.000 SEQ ID NO:44
    B7 Nonamer 36 FARPSRGYL 180.000 SEQ ID NO:46
  • [0337]
    A-3 Cripto-3 specific class I epitopes.
    Start
    HLA allele Epitope size position Sequence Score SEQ ID NO:
    A0201 Nonamer 5 KMVRFSYSV 668.086 SEQ ID NO:47
    A0201 Nonamer 89 CMLESFCAC 103.417 SEQ ID NO:48
    A0201 Decamer 16 IMAISKAFEL 152.124 SEQ ID NO:49
    A0201 Decamer 175 FMLAGICLSI 128.242 SEQ ID NO:50
    B2702 Nonamer 7 VRFSYSVIW 500.000 SEQ ID NO:51
    B2702 Nonamer 3 CRKMVRFSY 200.000 SEQ ID NO:52
    B2702 Decamer 7 VRFSYSVIWI 300.000 SEQ ID NO:53
    B2702 Decamer 37 ARPSRGDLAF 200.000 SEQ ID NO:54
    B2705 Nonamer 3 CRKMVRFSY 1000.000 SEQ ID NO:52
    B2705 Nonamer 7 VRFSYSVIW 1000.000 SEQ ID NO:51
    B2705 Nonamer 37 ARPSRGDLA 200.000 SEQ ID NO:55
    B2705 Nonamer 170 ARTTTFMLA 200.000 SEQ ID NO:56
    B2705 Decamer 7 VRFSYSVIWI 3000.000 SEQ ID NO:53
    B2705 Decamer 37 ARPSRGDLAF 1000.000 SEQ ID NO:54
    B2705 Decamer 59 IRPRSSQRVL 600.000 SEQ ID NO:57
    B2705 Decamer 3 CRKMVRFSYS 200.000 SEQ ID NO:58
    B2705 Decamer 65 QRVLPMGIQH 200.000 SEQ ID NO:59
    B5101 Nonamer 60 RPRSSQRVL 120.000 SEQ ID NO:60
    B5102 Nonamer 17 MAISKAFEL 150.000 SEQ ID NO:61
    B7 Nonamer 60 RPRSSQRVL 800.000 SEQ ID NO:60
    B7 Nonamer 36 FARPSRGDL 180.000 SEQ ID NO:62
  • B—Prediction of Class I Epitopes Using the Rammensee Method. [0338]
    B-1 Cripto-1 and -3 Class I epitopes.
    Start
    HLA allele Epitope size position Sequence Score SEQ ID NO:
    A26 Nonamer 179 GICLSIQSY 27 SEQ ID NO:63
    A26 Decamer 27 LVAGLGHQEF 25 SEQ ID NO:64
    A26 Decamer 121 DTWLPKKCSL 25 SEQ ID NO:65
    A3 Nonamer 58 AIRPRSSQR 29 SEQ ID NO:66
    A3 Nonamer 13 VIWIMAISK 24 SEQ ID NO:67
    A3 Decamer 12 SVIWIMAISK 29 SEQ ID NO:13
    A3 Decamer 117 SVPHDTWLPK 24 SEQ ID NO:14
    B2705 Nonamer 103 GRNCEHDVR 25 SEQ ID NO:18
    B2705 Nonamer 138 LRCFPQAFL 24 SEQ ID NO:20
  • [0339]
    B-2 Cripto-1 specific class I epitopes.
    Start
    HLA allele Epitope size position Sequence Score SEQ ID NO:
    A0201 Nonamer 83 CLNGGTCML 27 SEQ ID NO:68
    A0201 Nonamer 5 KMARFSYSV 25 SEQ ID NO:31
    A0201 Decamer 16 IMAISKVFEL 28 SEQ ID NO:32
    A0201 Decamer 13 VIWIMAISKV 26 SEQ ID NO:33
    A26 Nonamer 35 EFARPSRGY 24 SEQ ID NO:69
    A3 Nonamer 66 RVPPMGIQH 27 SEQ ID NO:70
    A3 Nonamer 21 KVFELGLVA 24 SEQ ID NO:71
    B08 Nonamer 17 MAISKVFEL 26 SEQ ID NO:45
    B5101 Nonamer 6 MARFSYSVI 25 SEQ ID NO:43
  • [0340]
    B-3 Cripto-3 specific class I epitopes.
    Start
    HLA allele Epitope size position Sequence Score SEQ ID NO:
    A0201 Nonamer 83 CLNGGTCML 27 SEQ ID NO:68
    A0201 Nonamer 176 MLAGICLSI 25 SEQ ID NO:72
    A0201 Decamer 16 IMAISKAFEL 24 SEQ ID NO:49
    A3 Nonamer 66 RVLPMGIQH 29 SEQ ID NO:73
    B0702 Nonamer 60 RPRSSQRVL 24 SEQ ID NO:60
    B08 Nonamer 17 MAISKAFEL 25 SEQ ID NO:61
  • C—Prediction of Class II Epitopes Using the Tepitope Method. [0341]
    C-1 Cripto-1 and -3 Class II epitopes.
    Start
    HLA allele Epitope size position Sequence Score SEQ ID NO:
    DRB1*0102 Nonamer 70 MGIQHSKEL 1.8 SEQ ID NO:74
    DRB1*0301 Nonamer 152 LVMDEHLVA 5.9 SEQ ID NO:75
    DRB1*0301 Nonamer 158 LVASRTPEL 4.3 SEQ ID NO:76
    DRB1*0401 Nonamer 152 LVMDEHLVA 3.2 SEQ ID NO:75
    DRB1*0402 Nonamer 59 IRPRSSQRV 4.9 SEQ ID NO:16
    DRB1*0703 Nonamer 11 YSVIWIMAI 6 SEQ ID NO:77
    DRB1*0703 Nonamer 158 LVASRTPEL 5.7 SEQ ID NO:76
    DRB1*0802 Nonamer 59 IRPRSSQRV 1.8 SEQ ID NO:16
    DRB1*0802 Nonamer 123 WLPKKCSLC 2.3 SEQ ID NO:78
    DRB1*0804 Nonamer 59 IRPRSSQRV 2.8 SEQ ID NO:16
    DRB1*0806 Nonamer 59 IRPRSSQRV 3.1 SEQ ID NO:16
    DRB1*1101 Nonamer 11 YSVIWIMAI 2.8 SEQ ID NO:77
    DRB1*1101 Nonamer 152 LVMDEHLVA 2.2 SEQ ID NO:75
    DRB1*1102 Nonamer 152 LVMDEHLVA 2.4 SEQ ID NO:75
    DRB1*1104 Nonamer 25 LGLVAGLGH 2.8 SEQ ID NO:79
    DRB1*1104 Nonamer 152 LVMDEHLVA 3.2 SEQ ID NO:75
    DRB1*1106 Nonamer 25 LGLVAGLGH 2.8 SEQ ID NO:79
    DRB1*1106 Nonamer 152 LVMDEHLVA 3.2 SEQ ID NO:75
    DRB1*1107 Nonamer 46 FRDDSIWPQ 2.8 SEQ ID NO:80
    DRB1*1107 Nonamer 152 LVMDEHLVA 5.9 SEQ ID NO:75
    DRB1*1107 Nonamer 158 LVASRTPEL 3.3 SEQ ID NO:76
    DRB1*1305 Nonamer 11 YSVIWIMAI 3.7 SEQ ID NO:77
    DRB1*1307 Nonamer 11 YSVIWIMAI 1.2 SEQ ID NO:77
    DRB1*1501 Nonamer 138 LRCFPQAFL 4.3 SEQ ID NO:20
    DRB1*1501 Nonamer 152 LVMDEHLVA 4.5 SEQ ID NO:75
    DRB1*1502 Nonamer 152 LVMDEHLVA 3.5 SEQ ID NO:75
    DRB5*0101 Nonamer 13 VIWIMAISK 5.3 SEQ ID NO:67
  • [0342]
    C-2 Cripto-1 specific class II epitopes.
    Start
    HLA allele Epitope size position Sequence Score SEQ ID NO:
    DRB1*0101 Nonamer 15 WIMAISKVF 1.3 SEQ ID NO:81
    DRB1*0102 Nonamer 176 MLVGICLSI 1.8 SEQ ID NO:82
    DRB1*0102 Nonamer 178 VGICLSIQS 1.7 SEQ ID NO:83
    DRB1*0301 Nonamer 17 MAISKVFEL 3.9 SEQ ID NO:45
    DRB1*0401 Nonamer 178 VGICLSIQS 2.8 SEQ ID NO:83
    DRB1*0402 Nonamer 178 VGICLSIQS 4.2 SEQ ID NO:83
    DRB1*0404 Nonamer 14 IWIMAISKV 2.9 SEQ ID NO:84
    DRB1*0404 Nonamer 177 LVGICLSIQ 3.3 SEQ ID NO:85
    DRB1*0404 Nonamer 178 VGICLSIQS 3.8 SEQ ID NO:83
    DRB1*0405 Nonamer 175 FMLVGICLS 3.2 SEQ ID NO:86
    DRB1*0405 Nonamer 177 LVGICLSIQ 3.1 SEQ ID NO:85
    DRB1*0405 Nonamer 178 VGICLSIQS 2.8 SEQ ID NO:83
    DRB1*0703 Nonamer 15 WIMAISKVF 5.7 SEQ ID NO:81
    DRB1*0703 Nonamer 17 MAISKVFEL 7.6 SEQ ID NO:45
    DRB1*0703 Nonamer 176 MLVGICLSI 5 SEQ ID NO:82
    DRB1*0801 Nonamer 175 FMLVGICLS 3.8 SEQ ID NO:86
    DRB1*0802 Nonamer 175 FMLVGICLS 3.8 SEQ ID NO:86
    DRB1*0804 Nonamer 175 FMLVGICLS 2.8 SEQ ID NO:86
    DRB1*1101 Nonamer 175 FMLVGICLS 3.9 SEQ ID NO:86
    DRB1*1101 Nonamer 178 VGICLSIQS 2.4 SEQ ID NO:83
    DRB1*1104 Nonamer 175 FMLVGICLS 2.9 SEQ ID NO:86
    DRB1*1104 Nonamer 177 LVGICLSIQ 2.6 SEQ ID NO:85
    DRB1*1104 Nonamer 178 VGICLSIQS 3.4 SEQ ID NO:83
    DRB1*1106 Nonamer 175 FMLVGICLS 2.9 SEQ ID NO:86
    DRB1*1106 Nonamer 177 LVGICLSIQ 2.6 SEQ ID NO:85
    DRB1*1106 Nonamer 178 VGICLSIQS 3.4 SEQ ID NO:83
    DRB1*1107 Nonamer 17 MAISKVFEL 2.9 SEQ ID NO:45
    DRB1*1107 Nonamer 177 LVGICLSIQ 3.6 SEQ ID NO:85
    DRB1*1107 Nonamer 178 VGICLSIQS 3 SEQ ID NO:83
    DRB1*1302 Nonamer 15 WIMAISKVF 3.3 SEQ ID NO:81
    DRB1*1302 Nonamer 175 FMLVGICLS 3 SEQ ID NO:86
    DRB1*1305 Nonamer 15 WIMAISKVF 3.1 SEQ ID NO:81
    DRB1*1305 Nonamer 175 FMLVGICLS 4.3 SEQ ID NO:86
    DRB1*1307 Nonamer 175 FMLVGICLS 3.9 SEQ ID NO:86
    DRB1*1307 Nonamer 177 LVGICLSIQ 1.6 SEQ ID NO:85
    DRB1*1501 Nonamer 6 MARFSYSVI 4.5 SEQ ID NO:43
    DRB1*1501 Nonamer 176 MLVGICLSI 4.1 SEQ ID NO:82
    DRB1*1502 Nonamer 6 MARFSYSVI 3.5 SEQ ID NO:43
    DRB5*0101 Nonamer 15 WIMAISKVF 4.1 SEQ ID NO:81
  • [0343]
    C-2 Cripto-3 specific class II epitopes.
    Start
    HLA allele Epitope size position Sequence Score SEQ ID NO:
    DRB1*0101 Nonamer 15 WIMAISKAF 1.3 SEQ ID NO:87
    DRB1*0102 Nonamer 6 MVRFSYSVI 1.4 SEQ ID NO:88
    DRB1*0102 Nonamer 17 MAISKAFEL 1.6 SEQ ID NO:61
    DRB1*0401 Nonamer 7 VRFSYSVIW 2.9 SEQ ID NO:51
    DRB1*0401 Nonamer 175 FMLAGICLS 2.7 SEQ ID NO:89
    DRB1*0402 Nonamer 67 VLPMGIQHS 3.6 SEQ ID NO:90
    DRB1*0404 Nonamer 6 MVRFSYSVI 2.5 SEQ ID NO:88
    DRB1*0404 Nonamer 14 IWIMAISKA 3.6 SEQ ID NO:91
    DRB1*0404 Nonamer 67 VLPMGIQHS 3.5 SEQ ID NO:90
    DRB1*0405 Nonamer 175 FMLAGICLS 2.7 SEQ ID NO:89
    DRB1*0703 Nonamer 7 VRFSYSVIW 6.5 SEQ ID NO:51
    DRB1*0703 Nonamer 15 WIMAISKAF 5.7 SEQ ID NO:87
    DRB1*0703 Nonamer 17 MAISKAFEL 7.5 SEQ ID NO:61
    DRB1*0801 Nonamer 16 IMAISKAFE 3 SEQ ID NO:92
    DRB1*0801 Nonamer 175 FMLAGICLS 3.5 SEQ ID NO:89
    DRB1*0802 Nonamer 67 VLPMGIQHS 1.9 SEQ ID NO:90
    DRB1*0802 Nonamer 175 FMLAGICLS 3.5 SEQ ID NO:89
    DRB1*0804 Nonamer 67 VLPMGIQRS 2.9 SEQ ID NO:90
    DRB1*0804 Nonamer 175 FMLAGICLS 2.5 SEQ ID NO:89
    DRB1*0806 Nonamer 16 IMAISKAFE 4 SEQ ID NO:92
    DRB1*1101 Nonamer 175 FMLAGICLS 3.5 SEQ ID NO:89
    DRB1*1102 Nonamer 67 VLPMGIQHS 3.1 SEQ ID NO:90
    DRB1*1102 Nonamer 175 FMLAGICLS 2.5 SEQ ID NO:89
    DRB1*1107 Nonamer 7 VRFSYSVIW 2.8 SEQ ID NO:51
    DRB1*1301 Nonamer 67 VLPMGIQHS 3.5 SEQ ID NO:90
    DRB1*1302 Nonamer 15 WIMAISKAF 3.3 SEQ ID NO:87
    DRB1*1302 Nonamer 175 FMLAGICLS 3.9 SEQ ID NO:89
    DRB1*1305 Nonamer 15 WIMAISKAF 3.1 SEQ ID NO:87
    DRB1*1305 Nonamer 175 FMLAGICLS 3.9 SEQ ID NO:89
    DRB1*1307 Nonamer 67 VLPMGIQHS 1.5 SEQ ID NO:90
    DRB1*1307 Nonamer 175 FMLAGICLS 3.5 SEQ ID NO:89
    DRB1*1501 Nonamer 6 MVRFSYSVI 6.6 SEQ ID NO:88
    DRB1*1502 Nonamer 6 MVRFSYSVI 5.6 SEQ ID NO:88
    DRB5*0101 Nonamer 15 WIMAISKAF 4.1 SEQ ID NO:87
  • Example 5 Anti-Tumor Potential of the Humoral Response Induced by Cripto Vaccines
  • A series of experiments aimed at assessing the inhibitory effect of the Cripto-specific humoral response on the pathophysiological activities of Cripto in cancer could be carried out by using various standards in vitro assays. The in vitro assays could be, but not restricted to, growth inhibition assay, cell motility inhibition assay, chemotaxis inhibition assay, inhibition of the invasion through extracellular matrix protein (ECM), and growth signal transduction pathway inhibition. The effects of the sera of immunized animals with Cripto peptides or protein in adjuvant, or with a plasmid DNA or a viral delivery system, e.g. adenoviral vector, encoding the Cripto protein could be assessed in these in vitro assays that gauge Cripto-mediated biological effects on Cripto-expressing cell lines. In parallel, the effect of the pre-immune sera from the same animals will be tested as negative control. The cell lines used in these assays could be, but not limited to, human tumor cells expressing Cripto such as GEO cells, NTERA2 cells, the CRL-5815 cell line, or murine tumor cell lines that naturally over-express Cripto. As example, the inhibition of the in vitro growth of both GEO and NTERA2 cells has previously been demonstrated by treatment with anti-sense oligonucleotides designed to prevent the translation of Cripto mRNA (Baldassarre, et al. [0344] Int J Cancer 66: 538-543 (1996); Ciardiello, et al. Oncogene 9:291-298 (1996); Alpe, et al. Int J Cancer 88: 566-574 (2000))
  • 5.1: Immunization Protocol: [0345]
  • Mice or rabbits would be immunized on day 0, 14, and 21 by intra-footpad injections of either peptides or protein in adjuvant, intra-dermal injections of a plasmid DNA encoding Cripto using gene-gun devices, or intra-dermal injections of a viral vector delivery system, e.g. adenoviral vector, encoding Cripto. [0346]
  • 5.2: Cell Proliferation Inhibition Assay: [0347]
  • The sera from immunized animals will be collected and added at different dilutions to the culture medium of cells platted in 96-well plates. Similarly, pre-immune sera of these animals will also be added to cells as negative control. The cells will be treated for 3 to 7 days. The cell growth will be measured by standard methods such as [0348] 3H-thymidine incorporation assay, MTT assay, crystal violet staining, or colony counting for proliferation in soft agar-medium.
  • 5.3: Cell Invasion, Motility, and Chemotaxis Inhibition Assays [0349]
  • The sera from immunized animals will be collected pre- and post-immunization and added at different dilutions to cell suspensions used for invasion, motility, and chemotaxis standards assays. The inhibition of Cripto-mediated invasion, motility, and chemotaxis could be assessed with the use of, for instance, but limited to, commercially available Falcon chambers with matrigel inserts (Collaborative Research), agarose droplet motility assay (Yamamoto et al. [0350] Ophthalmology 97: 1204-1210 (1994)), and commercially available Boyden apparatus (Neuro Probe), respectively.
  • 5.4: Signal Transduction Inhibition Assays: [0351]
  • The sera from immunized animals will be collected pre- and post-immunization and added at different dilutions to Cripto-expressing tumor cells in culture. Exogenous Cripto protein or human sera or milk containing the highest concentration of Cripto detected by ELISA (Bianco, et al. [0352] Breast Cancer Res Treat 66:1-7 (2001)) will be added to the culture medium of the cells. After various incubation times, the cells will be harvested and processed by standard methods in order to perform immunoblot analysis. The phosphorylation status of various key signal transduction pathways involved in cell proliferation will be assessed in these Cripto-simulated cells in the presence of pre- or post-immune sera. For instance, the tyrosine phosphorylation status of erb B-4 and mitogen-activated protein (MAP) kinase family such as ERK1, ERK-2, and P38 will be assessed by immunoblotting with antibodies that recognize the phosphorylated, active form of these enzymes as previous described (Bianco, et al. J Biol Chem 274: 8624-8629 (1999); Paine, et al. J Biol Chem 275: 11284-11290 (2000)).
  • Example 6 In Vivo Anti-Tumor Effect of the Immune Response Induced by Cripto Vaccines in Animal Models
  • The prophylactic or therapeutic potential of vaccines containing the human Cripto protein, Cripto peptides, or Cripto gene can be evaluated in mice challenged with syngeneic murine tumor cell line that express Cripto. The tumor cell lines could be a murine tumor cell line transfected with the human Cripto gene. For instance the transfected cell lines could be the TC1 cell line transfected with human Cripto gene. On the other hand, the high level of homology between human Cripto and its murine homologue suggests that cross-reactive immune responses induced by the human vaccine can protect against mouse Cripto-expressing tumors. Indeed, Cripto gene (TDGF-1) encodes a 171-amino acid protein which has 93% identity with its human counterpart (Liguori, et al [0353] Mamm Genome 7:344-348 (1996)). Therefore, mice could be protected by immunization with a human Critpo vaccine from murine tumor challenge or spontaneously arising tumor known to endogenously over-express the Cripto protein. With this respect, spontaneous tumors in transgenic mice designed to over-express several different oncogenes such as MMTV-Polyoma virus middle T antigen, MMTV-c-ErbB2, and MT-hTGF alpha, have been shown also to over-express Cripto. (Kenney, et al. Mol Carcinog 15: 44-56 (1996); Niemeye, et al. Int. J Cancer 81:588-591 (1999)).
  • Alternatively, in order to vaccinate with the syngeneic gene, the tumor-bearing mice could be vaccinated with a plasmid DNA or a viral delivery system, e.g. adenoviral vector, encoding the murine Cripto protein to protect from murine tumor growth. [0354]
  • 6.1: Prophylactic Experimental Design: [0355]
  • Mice would be vaccinated on day 0 and 14, prior tumor challenge, by either intra footpad injections of 5 μg of Cripto protein in adjuvant, intra-dermal injections of DNA plasmid encoding Cripto using gene-gun technology, or intra-dermal injections of a viral vector, e.g. adenoviral vector, encoding Cripto. Then, 10[0356] 6 TC1-CR-1 cells, human Cripto-expressing tumor cells, could be injected subcutaneously in the flank of C57BL/6 immunocompetent mice 1 or 2 weeks post vaccination. The tumor growth should be monitored in vivo by measuring individual tumors twice a week for several weeks post-tumor challenge.
  • On the other hand, the efficacy of prophylactic vaccination could be assessed by inhibiting the development of spontaneous Cripto-expressing tumors in transgenic mice (Niemeye, et al. [0357] Int J Cancer 81:588-591 (1999)) immunized multiple times before onset of the palpable tumor with either form of Cripto vaccines.
  • 6.2: Therapeutic Experimental Design: [0358]
  • 10[0359] 6 TC1-CR-1 cells, human-Critpo expressing tumor cells, would be injected subcutaneously in the flank of C57BL/6 immunocompetent mice. Mice could be vaccinated on days 7 and 14, post-tumor challenge, by either intra-footpad injections of 5 μg Cripto protein in adjuvant, intra-dermal injections of DNA plasmid encoding Cripto using gene-gun technology, or intra-dermal injections of a viral vector, e.g. adenoviral vector, encoding Cripto. The tumor growth should be monitored in vivo by measuring individual tumors twice a week. One to 4 weeks after the second immunization, several mice per group will be sacrificed to harvest spleen cells, draining lymph nodes, and sera for analysis of the immune responses to establish a correlation between the induction of a Cripto-specific immune response and the antitumor effect. The analysis of the Cripto-specific immune response induced by immunization could be assessed by measuring the antibody titers, antibody isotypic profile, the CD4+ T-cell proliferation response, and CTL CD8+ T-cell responses including cytokines production and lysis activity against Cripto-expressing target cells. All assays would be performed according to standards protocols.
  • A similar kind of experiment could be carried out by challenging parental mice with transplantable murine mammary tumor lines over-expressing Cripto. These cells would be established from spontaneous tumor arising in various oncogene transgenic mouse strains such as MMTV-Polyoma virus middle T antigen, MMTV-c-ErbB2, and MT-hTGF alpha transgenic mice (Niemeye, et al. [0360] Int J Cancer 81:588-591 (1999)).
  • Example 7 Demonstration of Cripto-1 Immunogenicity and Recognition of Cripto Protein
  • 7.1 Objectives [0361]
  • This experiment demonstrates that two selected polypeptide fragments of Cripto (SEQ ID NO:11 and SEQ ID NO:12) mixed with a vaccine adjuvant induces Cripto peptide specific antibody response in immunized animals. Furthermore, the assessment of the recognition of Cripto protein shows that antibodies recognize the tumour-associated antigen itself. Finally, an immunostaining study on cancer tissues using anti-Cripto antibodies shows Cripto-specific immune response to target cripto naturally expressed in cancer cells. [0362]
  • 7.2 Selection of Cripto-1 Derived Peptides [0363]
  • Two peptides were selected and prepared from the amino acid sequence of human Cripto-1: a 13 mer synthetic peptide (GHQEFARPSRGYL—SEQ ID NO:11) that corresponds to amino acids 32 to 44 and an 18 mer synthetic peptide (QEEPAIRPRSSQRVPPMG—SEQ ID NO:12) that corresponds to amino acids 54 to 71 of human Cripto-1 sequence. These peptides were coupled to KLH using gluteraldehyde. [0364]
  • 7.3 Immunisation Protocol of Rabbits [0365]
  • The KLH-coupled synthetic peptides were used to immunize two rabbits for each synthetic peptide. Antibodies were produced in rabbits during 3 months according to the protocol described in section 3.1 of the specification. Antisera were tested by ELISA against the peptide and the carrier protein. [0366]
  • 7.4 Test of Rabbits Sera on Extracts of Recombinant Full-Length Cripto-1 Expressed in [0367] E. coli
  • [0368] E. coli recombinant protein Cripto-1 (18 Kd) was detected with the sera of rabbits injected with the synthetic peptides aa32->44 (SEQ ID NO:11) or aa54->71 (SEQ ID NO:12). Western blots using monoclonal antibodies to Cripto-1 and rabbit antisera from rabbits injected with SEQ ID NO:11 or SEQ ID NO:12 indicated the presence of a single intense polypeptide band at approximately 18 kD in cell extract as well as cell pellets. In addition, Coommassie Blue stained polyacrylamide gel indicated the presence of a polypeptide (˜18 kD). No polypeptide having a molecular weight of 18 kD was detected by antibodies, antisera, or Coommassie Blue stain in the negative control or cellular extract supernatant.
  • Example 8 Demonstration of the Recognition of Cripto in Tumour Tissue by Immunostaining Studies Using Anti-Cripto Polyclonal Antibodies
  • SCID mice subcutaneous xenograft tumors (mRNA cripto-expressing CRL5815 cell line—human small cell lung carcinoma—expression determined by real time PCR methods) were incubated in buffered formalin fixative for 24 h at room temperature and paraffin embedded. Five micron thick sections were attached to superfrost glass slides and deparaffinized. Prior to antibody binding, the sections were heated in glyca buffer (Biogenex) (microwave for 10 min), then saturated with PBS buffer containing 0.5% blocking reagent (Boërhinger) for 30 min. Two rabbit anti-human CR-1 peptides (32-44; 54-71) were separately added (dil. {fraction (1/50)}-{fraction (1/200)}) and incubated at 4° C. overnight. The sections were rinsed three times in PBS, incubated with mouse-anti-rabbit IgG biotinylated secondary antibody (Sigma) at 1:500 in PBS/0.5% blocking reagent for 30 min at room temp. The sections were then rinsed three times in PBS, treated with H[0369] 2O2 1% to quench endogenous peroxidases, incubated in HRP-Streptavidin (Zymed) at 1:500 in PBS and, finally, after PBS rinsing, HRP activity was revealed using DAB (Zymed) solution for 5 min. The reaction was stopped with extensive rinsing in deionised water. The sections were counterstained for 2 minutes in hematoxylin, brought to xylene and mounted with DPX solution.
  • 8.1 Results [0370]
  • Cripto-1 peptide was detected using rabbit anti-human CR-1 peptide 54-71 (dil. {fraction (1/50)}) (SEQ ID NO:12) by immunostaining. The immunostaining specificity was confirmed by preabsorption of the anti-CR-1 peptide (54-71) antibody with the corresponding purified Cripto-1 peptide (50 μg/ml). [0371]
  • Cripto peptides formulated in vaccine adjuvant are immunogenic in animals. Antibodies raised in immunized rabbits may recognize not only the peptide used as immunogen, but also a recombinant Cripto protein expressed in [0372] E. coli and Cripto naturally expressed in tumour tissue. Cripto is a suitable target for active immunotherapy.
  • Example 9 Demonstration of the Immunogenicity of Recombinant Cripto-1 Protein
  • 9.1. Immunogenicity of Recombinant Cripto-1 Protein Mixed with Adjuvant [0373]
  • Recombinant Cripto protein purified from [0374] E. Coli was used to immunize rabbits as described in Example 3.
  • 9.2. ELISA Protocol to Monitor Antibody Response [0375]
  • Quantification of anti-Cripto antibodies in the sera were performed by ELISA using the Cripto protein as coating antigen and expressed as the titer at midpoint dilution. For example, antigen and antibodies solutions were used at 100 μl/well. Antigen was diluted to a final concentration of 1 μg/ml in PBS and was absorbed overnight at 4° C. to the wells of 96 wells microtiter plates. The plates were then incubated for 1 hr at 37° C. with PBS containing 1% bovine serum albumin and 0.1% tween 20 (saturation buffer). Two-fold dilutions of sera (starting {fraction (1/100)}) in the saturation buffer were added to the Cripto coated plates and incubated for 1 hr 30 min at 37° C. The plates were washed four times with PBS 0.1% Tween 20 and biotin-conjugated anti-rabbit Ig diluted {fraction (1/5000)} in saturation buffer were added to each well. The plates were incubated for 1 hour 30 minutes at 37° C. After washing step, streptavidin biotinylated peroxydase complex reagent diluted {fraction (1/5000)} in saturation buffer was added. Plates were kept for an additional 30 min at 37° C. [0376]
  • The plates were washed as above and incubated for 15 minutes with TMB. The reaction was stopped with 0.2 M H[0377] 2SO4 and the optical densities were measured using a microplate reader connected to a computer. Data were captured with SoftMaxPro software and analyzed.
  • 9.3 Results: [0378]
  • No anti-Cripto antibodies were detected pre-vaccination as shown in Table 2. At day 21 after the second vaccinations all the three animals showed anti-Cripto antibodies, and 21 days after 4 vaccinations antibody titers were increased as shown in Table 2. [0379]
    TABLE 2
    Mid Point Dilution
    Pre- 21 days 21 days 21 days 21 days
    vaccination Post 1 Post 2 Post 3 Post 4
    rabbit 777 232 50 2686 2858 1127
    rabbit 778 58 67 3074 4848 7645
    rabbit 779 132 119 8548 8315 11523
  • This experiment shows that repeated administration of the purified Cripto adjuvanted with QS21, 3D-MPL and oil-in-water emulsion induces specific antibodies. This experiment also demonstrates that recombinant Cripto antigen delivered with a vaccine adjuvant is immunogenic. [0380]
  • Example 10 Immunogenicity of Recombinant Cripto-1 Gene by Genetic DNA Immunisation
  • This experiment demonstrates that recombinant Cripto when delivered as a nucleic acid sequence (genetic immunization) encoding Cripto protein induces a Cripto-specific immune response in mice. [0381]
  • 10.1. Genetic Construct Design for In Vivo Expression & Mouse Immunization—[0382]
  • Genetic construct, in vivo expression and mouse immunization were performed as described in Example 3. [0383]
  • 10.2. Determination of Serum Antibodies by Western Blot on Crude Extracts of [0384] E. coli Recombinant Cripto-1 Protein
  • Serum from vaccinated mice was taken at day 0 and 42, for Western blot analyses for the presence of anti-cripto antibodies. Proteins contained in the solubilized pellet of an acellular extract, were separated by SDS-PAGE and used to test the sera of mice by Western blot. The pre-immune and post-immune serum (42 days post-immunisation) of eight mice, and a pool of the negative control group sera were tested at the dilution {fraction (1/500)}. [0385]
  • The sera of all immunized mice reacted with the mature Cripto-1 recombinant protein present in the extract as demonstrated by Western blot analysis. [0386]
  • This experiment shows that repeated administration of cripto DNA sequence can induce Cripto specific antibodies. Therefore, recombinant Cripto protein delivered as the genetic vaccine is immunogenic in mice. [0387]
  • Sequence Listing
  • [0388]
    GGAGAATCCCCGGAAAGGCTGAGTCTCCAGCTCAAGGTCAAAACGTCCAAGGCCGAA SEQ ID NO:1
    AGCCCTCCAGTTTCCCCTGGACGCCTTGCTCCTGCTTCTGCTACGACCTTCTGGGGA
    AAACGAATTTCTCATTTTCTTCTTAAATTGCCATTTTCGCTTTAGGAGATGAATGTT
    TTCCTTTGGCTGTTTTGGCAATGACTCTGAATTAAAGCGATGCTAACGCCTCTTTTC
    CCCCTAATTGTTAAAAGCTATGGACTGCAGGAAGATGGCCCGCTTCTCTTACAGTGT
    GATTTGGATCATGGCCATTTCTAAAGTCTTTGAACTGGGATTAGTTGCCGGGCTGGG
    CCATCAGGAATTTGCTCGTCCATCTCGGGGATACCTGGCCTTCAGAGATGACAGCAT
    TTGGCCCCAGGAGGAGCCTGCAATTCGGCCTCGGTCTTCCCAGCGTGTGCCGCCCAT
    GGGGATACAGCACAGTAAGGAGCTAAACAGAACCTGCTGCCTGAATGGGGGAACCTG
    CATGCTGGGGTCCTTTTGTGCCTGCCCTCCCTCCTTCTACGGACGGAACTGTGAGCA
    CGATGTGCGCAAAGAGAACTGTGGGTCTGTGCCCCATGACACCTGGCTGCCCAAGAA
    GTGTTCCCTGTGTAAATGCTGGCACGGTCAGCTCCGCTGCTTTCCTCAGGCATTTCT
    ACCCGGCTGTGATGGCCTTGTGATGGATGAGCACCTCGTGGCTTCCAGGACTCCAGA
    ACTACCACCGTCTGCACGTACTACCACTTTTATGCTAGTTGGCATCTGCCTTTCTAT
    ACAAAGCTACTATTAATCGACATTGACCTATTTCCAGAAATACAATTTTAGATATCA
    TGCAAATTTCATGACCAGTAAAGGCTGCTGCTACAATGTCCTAACTGAAAGATGATC
    ATTTGTAGTTGCCTTAAAATAATGAATACAATTTCCAAAATGGTCTCTAACATTTCC
    TTACAGAACTACTTCTTACTTCTTTGCCCTGCCCTCTCCCAAAAAACTACTTCTTTT
    TTCAAAAGAAAGTCAGCCATATCTCCATTGTGCCTAAGTCCAGTGTTTCTTTTTTTT
    TTTTTTTTTGAGACGGAGTCTCACTCTGTCACCCAGGCTGGACTGCAATGACGCGAT
    CTTGGTTCACTGCAACCTCCGCATCCGGGGTTCAAGCCATTCTCCTGCCTAAGCCTC
    CCAAGTAACTGGGATTACAGGCATGTGTCACCATGCCCAGCTAATTTTTTTGTATTT
    TAGTAGAGATGGGGGTTTCACCATATTGGCCAGTCTGGTCTCGAACTCTGACCTTGT
    GATCCATCGATCAGCCTCTCGAGTGCTGAGATTACACACGTGAGCAACTGTGCAAGG
    CCTGGTGTTTCTTGATACATGTAATTCTACCAAGGTCTTCTTAATATGTTCTTTTAA
    ATGATTGAATTATATGTTCAGATTATTGGAGACTAATTCTAATGTGGACCTTAGAAT
    ACAGTTTTGAGTAGAGTTGATCAAAATCAATTAAAATAGTCTCTTTAAAAGGAAAGA
    AAACATCTTTAAGGGGAGGAACCAGAGTGCTGAAGGAATGGAAGTCCATCTGCGTGT
    GTGCAGGGAGACTGGGTAGGAAAGAGGAAGCAAATAGAAGAGAGAGGTTGAAAAACA
    AAATGGGTTACTTGATTGGTGATTAGGTGGTGGTAGAGAAGCAAGTAAAAAGGCTAA
    ATGGAAGGGCAAGTTTCCATCATCTATAGAAAGCTATATAAGACAAGAACTCCCCTT
    TTTTTCCCAAAGGCATTATAAAAAGAATGAAGCCTCCTTAGAAAAAAAATTATACCT
    CAATGTCCCCAACAAGATTGCTTAATAAATTGTGTTTCCTCCAAGCTATTCAATTCT
    TTTAACTGTTGTAGAAGACAAAATGTTCACAATATATTTAGTTGTAAACCAAGTGAT
    CAAACTACATATTGTAAAGCCCATTTTTAAAATACATTGTATATATGTGTATGCACA
    GTAAAAATGGAAACTATATTGACCTAAAAAAAAAAAAA
  • [0389]
    AAGCTTGCGCGCCATGTAAGGTAAAGTGACTGATTCTATAGCAATCCAATTGTTCCT SEQ ID NO:2
    TTGTCTGCCCGTTTACATATAACAATGTTGTCAATGTTTGATTGAAAATACCTAGCA
    GGCGACACACACACACCTAGCTCCTCAGGCGGAGAGCACCCCTTTCTTGGCCACCCG
    GGTATCCCCCAGGGAGTACGGGGCTCAAAACACCCTTTTGGAGAACAAGGTGGAAGC
    AAATTTCAGGAAGTAAAACTTCCTGAAATAAAATAAAATATCGAATGCCTTGAGACC
    CATACATTTTCAGGTTTTCCTAATTAAAGCAATTACTTTCCACCACCCCTCCAACCT
    GGAATCACCAACTTGGTTAGAGAAACTGATTTTTCTTTTTTCTTTTTTTTTCCCAAA
    AGAGTACATCTGATCATTTTAGCCTGCAACTAATGATAGAGATATTAGGGCTAGTTA
    ACCACAGTTTTACAAGACTCCTCTCCCGCGTGTGGGCCATTGTCATGCTGTCGGTCC
    CGCCCACCTGAAAGGTCTCCCCGCCCCGACTGGGGTTTGTTGTTGAAGAAGGAGAAT
    CCCCGGAAAGGCTGAGTCTCCAGCTCAAGGTCAAAACGTCCAAGGCCGAAAGCCCTC
    CACTTTCCCCTGGACACCTTGCTCCTGCTTCTGCTACGACCTTCTGGGAACGCGAAT
    TTCTCATTTTCTTCTTAAATTGCCATTTTCGCTTTAGGAGATGAATGTTTTCCTTTG
    GCTGTTTTGGCAATGACTCTGAATTAAAGCGATGCTAACGCCTCTTTTCCCCCTAAT
    TGTTAAAAGCTATGGACTGCAGGAAGATGGTCCGCTTCTCTTACAGTGTGATTTGGA
    TCATGGCCATTTCTAAAGCCTTTGAACTGGGATTAGTTGCCCGGCTGGGCCATCAGG
    AATTTGCTCGTCCATCTCGGGGAGACCTGGCCTTCAGAGATGACAGCATTTGGCCCC
    AGGAGGAGCCTGCAATTCGGCCTCGGTCTTCCCAGCGTGTGCTGCCCATGGGAATAC
    AGCACAGTAAGGAGCTAAACAGAACCTGCTGCCTGAATGGGGGAACCTGCATGCTGG
    AGTCCTTTTGTGCCTGCCCTCCCTCCTTCTACGGACGGAACTGTGAGCACGATGTGC
    GCAAAGAGAACTGTGGGTCTGTGCCCCATGACACCTGGCTGCCCAAGAAGTGTTCCC
    TGTGTAAATGCTGGCACGGTCAGCTCCGCTGCTTTCCTCAGGCATTTCTACCCGGCT
    GTGATGGCCTTGTGATGGATGAGCACCTCGTGGCTTCCAGGACTCCAGAACTACCAC
    CGTCTGCACGTACTACCACTTTTATGCTAGCTGGCATCTGCCTTTCTATACAAAGCT
    ACTATTAATCGACATTGACCTATTTCCAGAAATACAATTTTAGATATTATGCAAATT
    TCATGACCCGTAAAGGCTGCTGCTACAATGTCCTAACTGAAAGATGATCATTTGTAG
    TTGCCTTAAAATAATGAATACAATTTCCAAAACGGTCTCTAACATTTCCTTACAGAA
    CTAACTACTTCTTACCTCTTTGCCCTGCCCTCTCCCAAAAAACTACTTCTTTTTTCA
    AAAGAAAGTCAGCCATATCTCCATTGTGCCCAAGTCCAGTGTTTCTTTTTTTTTTTT
    GAGACGGAGTCTCACTCTGTCACCCAGGCTGGACTGCAATGACGCGATCTCGGTTCA
    CTGCAACCTCCGCATCCGGGGTTCAAGCCATTCTCCTGCCTCAGCCTCCCAAGTAGC
    TGGGATTACAGGCATGTGTCACCATGCCGGCTAATTTTTTTGTATTTTAGTAGAGAC
    GGGGGTTTCACCATATTGGCCAGCTGGTCTCGAACTCTGACCTTGTGATCCATCGCT
    CGCCTCTCGAGTGCTGAGATTACACACGTGAGCAACTGTGCAAGGCCTGGTGTTTCT
    TGATACATGTAATTCTACCAAGGTCTTCTTAATATGTTCTTTTAAATGATTGAATTA
    TACACTCAGATTATTGGAGACTAAGTCTAATGTGGACCTTAGAATACAGTTTTGAGT
    AGAGTTGATCAAAATCAATTAAAATAGTCTCTTTAAAAGGAAAGAAAACATCTTTAA
    GGGGAGGAACCAGAGTGCTGAAGGAATGGAAGTCCATCTGCGTGTGTGCAGGGAGAC
    TGGGTAGGAAAGAGGAAGCAAATAGAAGAGAGAGGTTGAAAAACAAAATGGGTTACT
    TGATTGGTGATTAGGTGGTGGTAGAGAAGCAAGTAAAAAGGCTAAATGGAAGGGCAA
    GTTTCCATCATCTATAGAAAGCTATGTAAGACAAGGACTCCCCTTTTTTTCCCAAAG
    GCATTGTAAAAAGAATGAAGTCTCCTTAGAAAAAAAATTATACCTCAATGTCCCCAA
    CAAGATTGCTTAATAAATTGTGTTTCCTCCAAGCTATTCAATTCTTTTAACTGTTGT
    AGAAGAGAAAATGTTCACAATATATTTAGTTGTAAACCAAGTGATCAAACTACATAT
    TGTAAAGCCCATTTTTAAAATACATTGTATATATGTGTATGCACAGTAAAAATGGAA
    ACTATATTGACCTAAAAAAAAAAAAAGGAAACCACCCTTAGGCAGGCAGGACATGCT
    CTTCAGAACTCTGCTCTTCAGAGTTCCAAAGAAGGGATAAAACATCTTTTATNNCCA
    TCAAATAGC
  • [0390]
    MDCRKMARFSYSVIWIMAISKVFELGLVAGLGHQEFARPSRGYLAFRDDSIWPQEEP SEQ ID NO:3
    AIRPRSSQRVPPMGIQHSKELNRTCCLNGGTCMLGSFCACPPSFYGRNCEHDVRKEN
    CGSVPHDTWLPKKCSLCKCWHGQLRCFPQAFLPGCDGLVMDEHLVASRTPELPPSAR
    TTTFMLVGICLSIQSYY
  • [0391]
    MDCRKMVRFSYSVIWIMAISKAFELGLVAGLGHQEFARPSRGDLAFRDDSIWPQEEP SEQ ID NO:4
    AIRPRSSQRVLPMGIQHSKELNRTCCLNGGTCMLESFCACPPSFYGRNCEHDVRKEN
    CGSVPHDTWLPKKCSLCKCWHGQLRCFPQAFLPGCDGLVMDEHLVASRTPELPPSAR
    TTTFMLAGICLSIQSYY
  • [0392]
    GGAGAATCCCCGGAAAGGCTGAGTCTCCAGCTCAAGGTCAAAACGTCCAAGGCCGAA SEQ ID NO:5
    AGCCCTCCAGTTTCCCCTGGACGCCTTGCTCCTGCTTCTGCTACGACCTTCTGGGGA
    AAACGAATTTCTCATTTTCTTCTTAAATTGCCATTTTCGCTTTAGGAGATGAATGTT
    TTCCTTTGGCTGTTTTGGCAATGACTCTGAATTAAAGCGATGCTAACGCCTCTTTTC
    CCCCTAATTGTTAAAAGCTATGGACTGCAGGAAGATGGCCCGCTTCTCTTACAGTGT
    GATTTGGATCATGGCCATTTCTAAAGTCTTTGAACTGGGATTAGTTGCCGGGCTGGG
    CCATCAGGAATTTGCTCGTCCATCTCGGGGATACCTGGCCTTCAGAGATGACAGCAT
    TTGGCCCCAGGAGGAGCCTGCAATTCGGCCTCGGTCTTCCCAGCGTGTGCCGCCCAT
    GGGGATACAGCACAGTAAGGAGCTAAACAGAACCTGCTGCCTGAATGGGGGAACCTG
    CATGCTGGGGTCCTTTTGTGCCTGCCCTCCCTCCTTCTACGGACGGAACTGTGAGCA
    CGATGTGCGCAAAGAGAACTGTGGGTCTGTGCCCCATGACACCTGGCTGCCCAAGAA
    GTGTTCCCTGTGTAAATGCTGGCACGGTCAGCTCCGCTGCTTTCCTCAGGCATTTCT
    ACCCGGCTGTGATGGCCTTGTGATGGATGAGCACCTCGTGGCTTCCAGGACTCCAGA
    ACTACCACCGTCTGCACGTACTACCACTTTTATGCTAGTTGGCATCTGCCTTTCTAT
    ACAAAGCTACTATTAATCGACATTGACCTATTTCCAGAAATACAATTTTAGATATCA
    TGCAAATTTCATGACCAGTAAAGGCTGCTGCTACAATGTCCTAACTGAAAGATGATC
    ATTTGTAGTTGCCTTAAAATAATGAATACAATTTCCAAAATGGTCTCTAACATTTCC
    TTACAGAACTACTTCTTACTTCTTTGCCCTGCCCTCTCCCAAAAAACTACTTCTTTT
    TTCAAAAGAAAGTCAGCCATATCTCCATTGTGCCTAAGTCCAGTGTTTCTTTTTTTT
    TTTTTTTTTGAGACGGAGTCTCACTCTGTCACCCAGGCTGGACTGCAATGACGCGAT
    CTTGGTTCACTGCAACCTCCGCATCCGGGGTTCAAGCCATTCTCCTGCCTAAGCCTC
    CCAAGTAACTGGGATTACAGGCATGTGTCACCATGCCCAGCTAATTTTTTTGTATTT
    TAGTAGAGATGGGGGTTTCACCATATTGGCCAGTCTGGTCTCGAACTCTGACCTTGT
    GATCCATCGATCAGCCTCTCGAGTGCTGAGATTACACACGTGAGCAACTGTGCAAGG
    CCTGGTGTTTCTTGATACATGTAATTCTACCAAGGTCTTCTTAATATGTTCTTTTAA
    ATGATTGAATTATATGTTCAGATTATTGGAGACTAATTCTAATGTGGACCTTAGAAT
    ACAGTTTTGAGTAGAGTTGATCAAAATCAATTAAAATAGTCTCTTTAAAAGGAAAGA
    AAACATCTTTAAGGGGAGGAACCAGAGTGCTGAAGGAATGGAAGTCCATCTGCGTGT
    GTGCAGGGAGACTGGGTAGGAAAGAGGAAGCAAATAGAAGAGAGAGGTTGAAAAACA
    AAATGGGTTACTTGATTGGTGATTAGGTGGTGGTAGAGAAGCAAGTAAAAAGGCTAA
    ATGGAAGGGCAAGTTTCCATCATCTATAGAAAGCTATATAAGACAAGAACTCCCCTT
    TTTTTCCCAAAGGCATTATAAAAAGAATGAAGCCTCCTTAGAAAAAAAATTATACCT
    CAATGTCCCCAACAAGATTGCTTAATAAATTGTGTTTCCTCCAAGCTATTCAATTCT
    TTTAACTGTTGTAGAAGACAAAATGTTCACAATATATTTAGTTGTAAACCAAGTGAT
    CAAACTACATATTGTAAAGCCCATTTTTAAAATACATTGTATATATGTGTATGCACA
    GTAAAAATGGAAACTATATTGACCTAAAAAAAAAAAAA
  • [0393]
    MDCRKMARFSYSVIWIMAISKVFELGLVAGLGHQEFARPSRGYLAFRDDSIWPQEEP SEQ ID NO:6
    AIRPRSSQRVPPMGIQHSKELNRTCCLNGGTCMLGSFCACPPSFYGRNCEHDVRKEN
    CGSVPHDTWLPKKCSLCKCWHGQLRCFPQAFLPGCDGLVMDEHLVASRTPELPPSAR
    TTTFMLVGICLSIQSYY
  • [0394]
    TGGGTAGGAAAGAGGAAGCAAAT SEQ ID NO:7
  • [0395]
    TGCTTCTCTACCACCACCTAATCA SEQ ID NO:8
  • [0396]
    CGTCCAAGGCCGAAAGCCCTCCAGTT SEQ ID NO:9
  • [0397]
    TTGGGAGAGGGCAGGGCAAAGAAGTAAGAA SEQ ID NO:10
  • [0398]
    GHQEFARPSRGYL SEQ ID NO:11
  • [0399]
    QEEPAIRPRSSQRVPPMG SEQ ID NO:12
  • [0400]
    SVIWIMAISK SEQ ID NO:13
  • [0401]
    SVPHDTWLPK SEQ ID NO:14
  • [0402]
    HQEFARPSR SEQ ID NO:15
  • [0403]
    IRPRSSQRV SEQ ID NO:16
  • [0404]
    IQHSKELNR SED ID NO:17
  • [0405]
    GRNCEHDVR SEQ ID NO:18
  • [0406]
    VRKENCGSV SEQ ID NO:19
  • [0407]
    LRCFPQAFL SEQ ID NO:20
  • [0408]
    SRTPELPPS SEQ ID NO:21
  • [0409]
    QRVPPMGIQH SEQ ID NO:22
  • [0410]
    NRTCCLNGGT SEQ ID NO:23
  • [0411]
    GQLRCFPQAF SEQ ID NO:24
  • [0412]
    QAFLPGCDGL SEQ ID NO:25
  • [0413]
    DGLVMDEHLV SEQ ID NO:26
  • [0414]
    FELGLVAGL SEQ ID NO:27
  • [0415]
    KELNRTCCL SEQ ID NO:28
  • [0416]
    QLRCFPQAF SEQ ID NO:29
  • [0417]
    SARTTTFML SEQ ID NO:30
  • [0418]
    KMARFSYSV SEQ ID NO:31
  • [0419]
    IMAISKVFEL SEQ ID NO:32
  • [0420]
    VIWIMAISKV SEQ ID NO:33
  • [0421]
    FMLVGICLSI SEQ ID NO:34
  • [0422]
    ARFSYSVIW SEQ ID NO:35
  • [0423]
    CRKMARFSY SEQ ID NO:36
  • [0424]
    ARFSYSVIWI SEQ ID NO:37
  • [0425]
    ARPSRGYLAF SEQ ID NO:38
  • [0426]
    ARTTTFMLV SEQ ID NO:39
  • [0427]
    ARPSRGYLA SEQ ID NO:40
  • [0428]
    CRKMARFSYS SEQ ID NO:41
  • [0429]
    QEFARPSRGY SEQ ID NO:42
  • [0430]
    MARFSYSVI SEQ ID NO:43
  • [0431]
    SARTTTFMLV SEQ ID NO:44
  • [0432]
    MAISKVFEL SEQ ID NO:45
  • [0433]
    FARPSRGYL SEQ ID NO:46
  • [0434]
    KMVRFSYSV SEQ ID NO:47
  • [0435]
    CMLESFCAC SEQ ID NO:48
  • [0436]
    IMAISKAFEL SEQ ID NO:49
  • [0437]
    FMLAGICLSI SEQ ID NO:50
  • [0438]
    VRFSYSVIW SEQ ID NO:51
  • [0439]
    CRKMVRFSY SEQ ID NO:52
  • [0440]
    VRFSYSVIWI SEQ ID NO:53
  • [0441]
    ARPSRGDLAF SEQ ID NO:54
  • [0442]
    ARPSRGDLA SEQ ID NO:55
  • [0443]
    ARTTTFMLA SEQ ID NO:56
  • [0444]
    IRPRSSQRVL SEQ ID NO:57
  • [0445]
    CRKMVRFSYS SEQ ID NO:58
  • [0446]
    QRVLPMGIQH SEQ ID NO:59
  • [0447]
    RPRSSQRVL SEQ ID NO:60
  • [0448]
    MAISKAFEL SEQ ID NO:61
  • [0449]
    FARPSRGDL SEQ ID NO:62
  • [0450]
    GICLSIQSY SEQ ID NO:63
  • [0451]
    LVAGLGHQEF SEQ ID NO:64
  • [0452]
    DTWLPKKCSL SEQ ID NO:65
  • [0453]
    AIRPRSSQR SEQ ID NO:66
  • [0454]
    VIWIMAISK SEQ ID NO:67
  • [0455]
    CLNGGTCML SEQ ID NO:68
  • [0456]
    EFARPSRGY SEQ ID NO:69
  • [0457]
    RVPPMGIQH SEQ ID NO:70
  • [0458]
    KVFELGLVA SEQ ID NO:71
  • [0459]
    MLAGICLSI SEQ ID NO:72
  • [0460]
    RVLPMGIQH SEQ ID NO:73
  • [0461]
    MGIQHSKEL SEQ ID NO:74
  • [0462]
    LVMDEHLVA SEQ ID NO:75
  • [0463]
    LVASRTPEL SEQ ID NO:76
  • [0464]
    YSVIWIMAI SEQ ID NO:77
  • [0465]
    WLPKKCSLC SEQ ID NO:78
  • [0466]
    LCLVAGLGH SEQ ID NO:79
  • [0467]
    FRDDSIWPQ SEQ ID NO:80
  • [0468]
    WIMAISKVF SEQ ID NO:81
  • [0469]
    MLVGICLSI SEQ ID NO:82
  • [0470]
    VGICLSIQS SEQ ID NO:83
  • [0471]
    IWIMAISKV SEQ ID NO:84
  • [0472]
    LVGICLSIQ SEQ ID NO:85
  • [0473]
    FMLVGICLS SEQ ID NO:86
  • [0474]
    WIMAISKAF SEQ ID NO:87
  • [0475]
    MVRFSYSVI SEQ ID NO:88
  • [0476]
    FMLAGICLS SEQ ID NO:89
  • [0477]
    VLPMGIQHS SEQ ID NO:90
  • [0478]
    IWIMAISKA SEQ ID NO:91
  • [0479]
    IMAISKAFE SEQ ID NO:92
  • [0480]
    GRNCEHDVRK SEQ ID NO:93
  • [0481]
    SRTPELPPSA SEQ ID NO:94
  • [0482]
    CGTCCAAGGCCGAAAGCCCTCCAGTTTCCCCTGGACGCCTTGCTCCTGCTTCTGCTA SEQ ID NO:95
    CGACCTTCTGGGGAAAACGAATTTCTCATTTTCTTCTTAAATTGCCATTTTCGCTTT
    AGGAGATGAATGTTTTCCTTTGGCTGTTTTGGCAATGACTCTGAATTAAAGCGATGC
    TAACGCCTCTTTTCCCCCTAATTGTTAAAAGCTATGGACTGCAGGAAGATGGCCCGC
    TTCTCTTACAGTGTGATTTGGATCATGGCCATTTCTAAAGcCTTTGAACTGGGATTA
    GTTGCCGGGCTGGGCCATCAGGAATTTGCTCGTCCATCTCGGGGATACCTGGCCTTC
    AGAGATGACAGCATTTGGCCCCAGGAGGAGCCTGCAATTCGGCCTCGGTCTTCCCAG
    CGTGTGCCGCCCATGGGGATACAGCACAGTAAGGAGCTAAACAGAACCTGCTGCCTG
    AATGGGGGAACCTGCATGCTGGGGTCCTTTTGTGCCTGCCCTCCCTCCTTCTACGGA
    CGGAACTGTGAGCACGATGTGCGCAAAGAGAACTGTGGGTCTGTGCCCCATGACACC
    TGGCTGCCCAAGAAGTGTTCCCTGTGTAAATGCTGGCACGGTCAGCTCCGCTGCTTT
    CCTCAGGCATTTCTACCCGGCTGTGATGGCCTTGTGATGGATGAGCACCTCGTGGCT
    TCCAGGACTCCAGAACTACCACCGTCTGCACGTACTACCACTTTTATGCTAGTTGGC
    ATCTGCCTTTCTATACAAAGCTACTATTAATCGACATTGACCTATTTCCAGAAATAC
    AATTTTAGATATCATGCAAATTTCATGACCAGTAAAGGCTGCTGCTACAATGTCCTA
    ACTGAAAGATGATCATTTGTAGTTGCCTTAAAATAATGAATACAATTTCCAAAATGG
    TCTCTAACATTTCCTTACAGAACTACTTCTTACTTCTTTGCCCTGCCCTCTCCCAA
  • [0483]
    MDCRKMARFSYSVIWIMAISKAFELGLVAGLGHQEFARPSRGYLAFRDDSIWPQEEP SEQ ID NO:96
    AIRPRSSQRVPPMGIQHSKELNRTCCLNGGTCMLGSFCACPPSKYGRNCEHDVRKEN
    CGSVPHDTWLPKKCSLCKCWHGQLRCFPQAFLPGCDGLVMDEHLVASRTPELPPSAR
    TTTFMLVGICLSIQSYY
  • [0484]
    Figure US20040138112A1-20040715-P00001
    Figure US20040138112A1-20040715-P00002
    Figure US20040138112A1-20040715-P00003
    Figure US20040138112A1-20040715-P00004
    Figure US20040138112A1-20040715-P00005

Claims (10)

1. A method for inhibiting development of a cancer in a patient, comprising the steps of:
(e) incubating CD4+ and/or CD8+T cells isolated from a patient with at least one component selected from the group consisting of SEQ ID NO:11 or SEQ ID NO:12; and
(f) administering to the patient an effective amount of the T cells, and thereby inhibiting development of a cancer in the patient.
2. The method of claim 1, further comprising allowing the T cells to proliferate.
3. A method for producing an immunogenic response to a tumor-associated antigen in an animal comprising administering a first component comprising a polynucleotide encoding a fragment of the tumor-associated antigen to the animal.
4. The method of claim 3, wherein the tumor-associated antigen is Cripto.
5. The method of claim 3, wherein the polynucleotide encodes SEQ ID NO:11 and does not encode SEQ ID NO:3 or SEQ ID NO:4.
6. The method of claim 3, wherein the polynucleotide encodes SEQ ID NO:12 and does not encode SEQ ID NO:3 or SEQ ID NO:4.
7. The method of claim 3, wherein the polynucleotide is recombinant DNA.
8. The method of claim 3, further comprising admixing the polynucleotide and a second component comprising an immunostimulant, adjuvant and physiologically acceptable carrier.
9. A method of inducing an immunoresponse to Cripto in an animal comprising repeatedly administering a composition comprising a first component comprising SEQ ID NO:11 or SEQ ID NO:12.
10. The method of claim 9 further comprising admixing the first component and a second component comprising a physiologically acceptable carrier, adjuvant and immunostimulant prior to administering the first component to the animal.
US10/407,481 2000-08-24 2003-04-04 Vaccines Abandoned US20040138112A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/407,481 US20040138112A1 (en) 2000-08-24 2003-04-04 Vaccines

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
GBGB0020953.6A GB0020953D0 (en) 2000-08-24 2000-08-24 Vaccine
US10/362,597 US7439320B2 (en) 2000-08-24 2001-08-20 Cripto tumour polypeptide
PCT/EP2001/009646 WO2002016413A2 (en) 2000-08-24 2001-08-20 Cripto tumour polypeptide
US10/407,481 US20040138112A1 (en) 2000-08-24 2003-04-04 Vaccines

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
US10/362,597 Continuation-In-Part US7439320B2 (en) 2000-08-24 2001-08-20 Cripto tumour polypeptide
PCT/EP2001/009646 Continuation-In-Part WO2002016413A2 (en) 2000-08-24 2001-08-20 Cripto tumour polypeptide

Publications (1)

Publication Number Publication Date
US20040138112A1 true US20040138112A1 (en) 2004-07-15

Family

ID=9898284

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/362,597 Expired - Fee Related US7439320B2 (en) 2000-08-24 2001-08-20 Cripto tumour polypeptide
US10/407,481 Abandoned US20040138112A1 (en) 2000-08-24 2003-04-04 Vaccines

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/362,597 Expired - Fee Related US7439320B2 (en) 2000-08-24 2001-08-20 Cripto tumour polypeptide

Country Status (19)

Country Link
US (2) US7439320B2 (en)
EP (1) EP1311678A2 (en)
JP (1) JP2004506441A (en)
KR (1) KR20030045041A (en)
CN (1) CN1471579A (en)
AR (1) AR032173A1 (en)
AU (1) AU2001295498A1 (en)
BR (1) BR0113491A (en)
CA (1) CA2420087A1 (en)
CZ (1) CZ2003537A3 (en)
GB (1) GB0020953D0 (en)
HU (1) HUP0302824A2 (en)
IL (1) IL154533A0 (en)
MX (1) MXPA03001634A (en)
NO (1) NO20030823L (en)
NZ (1) NZ524344A (en)
PL (1) PL363005A1 (en)
WO (1) WO2002016413A2 (en)
ZA (1) ZA200301436B (en)

Cited By (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8645817B1 (en) 2006-12-29 2014-02-04 Monster Worldwide, Inc. Apparatuses, methods and systems for enhanced posted listing generation and distribution management
US9779390B1 (en) 2008-04-21 2017-10-03 Monster Worldwide, Inc. Apparatuses, methods and systems for advancement path benchmarking
US9959525B2 (en) 2005-05-23 2018-05-01 Monster Worldwide, Inc. Intelligent job matching system and method
US10181116B1 (en) 2006-01-09 2019-01-15 Monster Worldwide, Inc. Apparatuses, systems and methods for data entry correlation
US10387839B2 (en) 2006-03-31 2019-08-20 Monster Worldwide, Inc. Apparatuses, methods and systems for automated online data submission

Families Citing this family (83)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AUPR395801A0 (en) * 2001-03-26 2001-04-26 Austin Research Institute, The Antibodies against cancer
EP1390389B1 (en) 2001-04-26 2009-01-14 Biogen Idec MA Inc. Cripto blocking antibodies and uses thereof
US7582299B2 (en) 2001-04-26 2009-09-01 Biogen Idec Ma Inc. Cripto-specific antibodies
AU2002334799B2 (en) * 2001-04-26 2009-05-07 Biogen Ma Inc. Cripto-specific antibodies
US20060194265A1 (en) * 2001-10-23 2006-08-31 Morris David W Novel therapeutic targets in cancer
NZ544923A (en) 2003-06-27 2009-02-28 Biogen Idec Inc Use of hydrophobic-interaction-chromatography or hinge-region modifications for the production of homogeneous anti-body solutions
ZA200603619B (en) 2003-11-06 2008-10-29 Seattle Genetics Inc Monomethylvaline compounds capable of conjugation to ligands
CN102973947A (en) 2004-06-01 2013-03-20 健泰科生物技术公司 Antibody-drug conjugates and methods
KR101270829B1 (en) 2004-09-23 2013-06-07 제넨테크, 인크. Cystein engineered antibodies and conjugates
US20100111856A1 (en) 2004-09-23 2010-05-06 Herman Gill Zirconium-radiolabeled, cysteine engineered antibody conjugates
CN101137673B (en) 2005-01-05 2013-12-04 比奥根艾迪克Ma公司 Cripto binding molecules
US20100041032A1 (en) * 2006-04-28 2010-02-18 Biogen Idec Ma Inc. Composition and methods for the detection of cripto-3
WO2008040759A1 (en) * 2006-10-03 2008-04-10 Pharmexa A/S Method for down-regulation of cripto
IN2012DN03025A (en) 2009-09-09 2015-07-31 Ct Se Llc
MA34277B1 (en) 2010-04-15 2013-06-01 Spirogen Developments Sarl PYRROLOBENZODIAZEPINES AND CONJUGATES THEREOF
CN114246952A (en) 2010-06-08 2022-03-29 基因泰克公司 Cysteine engineered antibodies and conjugates
JP5889912B2 (en) 2010-11-17 2016-03-22 ジェネンテック, インコーポレイテッド Alaninyl maytansinol antibody conjugate
ES2567276T3 (en) 2011-05-12 2016-04-21 Genentech, Inc. LC-MS / MS method of monitoring multiple reactions to detect therapeutic antibodies in animal samples using frame-changing peptides
EA026827B1 (en) 2011-10-14 2017-05-31 Медимьюн Лимитед Pyrrolobenzodiazepines and conjugates thereof
WO2013130093A1 (en) 2012-03-02 2013-09-06 Genentech, Inc. Biomarkers for treatment with anti-tubulin chemotherapeutic compounds
PT2906253T (en) 2012-10-12 2018-11-05 Medimmune Ltd Pyrrolobenzodiazepine - anti-psma antibody conjugates
JP6392763B2 (en) 2012-10-12 2018-09-19 エイディーシー・セラピューティクス・エス・アーAdc Therapeutics Sa Pyrrolobenzodiazepine-antibody conjugate
EP2906297B1 (en) 2012-10-12 2017-12-06 ADC Therapeutics SA Pyrrolobenzodiazepine-antibody conjugates
WO2014057114A1 (en) 2012-10-12 2014-04-17 Adc Therapeutics Sàrl Pyrrolobenzodiazepine-anti-psma antibody conjugates
CN104837502B (en) 2012-10-12 2018-08-10 麦迪穆有限责任公司 Pyrrolobenzodiazepines Zhuo and its conjugate
CA2887897C (en) 2012-10-12 2020-02-18 Adc Therapeutics Sarl Pyrrolobenzodiazepine-anti-cd25 antibody conjugates
HUE035694T2 (en) 2012-10-12 2018-05-28 Adc Therapeutics Sa Pyrrolobenzodiazepine-anti-cd22 antibody conjugates
WO2014096368A1 (en) 2012-12-21 2014-06-26 Spirogen Sàrl Pyrrolobenzodiazepines and conjugates thereof
CA2894959C (en) 2012-12-21 2022-01-11 Spirogen Sarl Unsymmetrical pyrrolobenzodiazepines-dimers for use in the treatment of proliferative and autoimmune diseases
SG11201507214SA (en) 2013-03-13 2015-10-29 Medimmune Ltd Pyrrolobenzodiazepines and conjugates thereof
ES2687439T3 (en) 2013-03-13 2018-10-25 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
CA2904044C (en) 2013-03-13 2020-03-31 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
US10442836B2 (en) 2013-08-12 2019-10-15 Genentech, Inc. 1-(chloromethyl)-2,3-dihydro-1H-benzo[E]indole dimer antibody-drug conjugate compounds, and methods of use and treatment
US9950078B2 (en) 2013-10-11 2018-04-24 Medimmune Limited Pyrrolobenzodiazepine-antibody conjugates
GB201317982D0 (en) 2013-10-11 2013-11-27 Spirogen Sarl Pyrrolobenzodiazepines and conjugates thereof
WO2015052535A1 (en) 2013-10-11 2015-04-16 Spirogen Sàrl Pyrrolobenzodiazepine-antibody conjugates
WO2015052532A1 (en) 2013-10-11 2015-04-16 Spirogen Sàrl Pyrrolobenzodiazepine-antibody conjugates
BR112016013258A2 (en) 2013-12-16 2018-01-16 Genentech Inc antibody-drug conjugate, pharmaceutical composition, method for treating cancer and kit
KR102354207B1 (en) 2013-12-16 2022-01-20 제넨테크, 인크. Peptidomimetic compounds and antibody-drug conjugates thereof
AU2014364927A1 (en) 2013-12-16 2016-07-07 Genentech, Inc. Peptidomimetic compounds and antibody-drug conjugates thereof
US10188746B2 (en) 2014-09-10 2019-01-29 Medimmune Limited Pyrrolobenzodiazepines and conjugates thereof
GB201416112D0 (en) 2014-09-12 2014-10-29 Medimmune Ltd Pyrrolobenzodiazepines and conjugates thereof
JP2017531620A (en) 2014-09-12 2017-10-26 ジェネンテック, インコーポレイテッド Cysteine engineered antibodies and conjugates
EP3191134B1 (en) 2014-09-12 2019-11-20 Genentech, Inc. Anthracycline disulfide intermediates, antibody-drug conjugates and methods
AU2015317653A1 (en) 2014-09-17 2017-04-06 Genentech, Inc. Pyrrolobenzodiazepines and antibody disulfide conjugates thereof
US9616114B1 (en) 2014-09-18 2017-04-11 David Gordon Bermudes Modified bacteria having improved pharmacokinetics and tumor colonization enhancing antitumor activity
KR20170101895A (en) 2014-11-25 2017-09-06 에이디씨 테라퓨틱스 에스에이 Pyrrolobenzodiazepine-antibody conjugates
KR20170086121A (en) 2014-12-03 2017-07-25 제넨테크, 인크. Quaternary amine compounds and antibody-drug conjugates thereof
GB201506411D0 (en) 2015-04-15 2015-05-27 Bergenbio As Humanized anti-axl antibodies
GB201506402D0 (en) 2015-04-15 2015-05-27 Berkel Patricius H C Van And Howard Philip W Site-specific antibody-drug conjugates
MA43345A (en) 2015-10-02 2018-08-08 Hoffmann La Roche PYRROLOBENZODIAZEPINE ANTIBODY-DRUG CONJUGATES AND METHODS OF USE
MA43354A (en) 2015-10-16 2018-08-22 Genentech Inc CONJUGATE DRUG CONJUGATES WITH CLOUDY DISULPHIDE
MA45326A (en) 2015-10-20 2018-08-29 Genentech Inc CALICHEAMICIN-ANTIBODY-DRUG CONJUGATES AND METHODS OF USE
US10691773B2 (en) 2015-12-30 2020-06-23 General Electric Company Cell processing techniques
GB201601431D0 (en) 2016-01-26 2016-03-09 Medimmune Ltd Pyrrolobenzodiazepines
GB201602359D0 (en) 2016-02-10 2016-03-23 Medimmune Ltd Pyrrolobenzodiazepine Conjugates
GB201602356D0 (en) 2016-02-10 2016-03-23 Medimmune Ltd Pyrrolobenzodiazepine Conjugates
JP6943872B2 (en) 2016-03-25 2021-10-06 ジェネンテック, インコーポレイテッド Multiple whole antibody and antibody complex drug quantification assay
GB201607478D0 (en) 2016-04-29 2016-06-15 Medimmune Ltd Pyrrolobenzodiazepine Conjugates
EP3458101B1 (en) 2016-05-20 2020-12-30 H. Hoffnabb-La Roche Ag Protac antibody conjugates and methods of use
JP7022080B2 (en) 2016-05-27 2022-02-17 ジェネンテック, インコーポレイテッド Biochemical analytical methods for the characterization of site-specific antibody-drug conjugates
CN109476648B (en) 2016-06-06 2022-09-13 豪夫迈·罗氏有限公司 Sevelamer antibody-drug conjugates and methods of use
JP7093767B2 (en) 2016-08-11 2022-06-30 ジェネンテック, インコーポレイテッド Pyrrolobenzodiazepine prodrug and its antibody conjugate
JP7050770B2 (en) 2016-10-05 2022-04-08 エフ・ホフマン-ラ・ロシュ・アクチェンゲゼルシャフト Method for preparing antibody drug conjugate
GB201617466D0 (en) 2016-10-14 2016-11-30 Medimmune Ltd Pyrrolobenzodiazepine conjugates
US11180535B1 (en) 2016-12-07 2021-11-23 David Gordon Bermudes Saccharide binding, tumor penetration, and cytotoxic antitumor chimeric peptides from therapeutic bacteria
US11129906B1 (en) 2016-12-07 2021-09-28 David Gordon Bermudes Chimeric protein toxins for expression by therapeutic bacteria
GB201702031D0 (en) 2017-02-08 2017-03-22 Medlmmune Ltd Pyrrolobenzodiazepine-antibody conjugates
DK3544636T3 (en) 2017-02-08 2021-05-10 Adc Therapeutics Sa Pyrrolobenzodiazepine antibody conjugates
HRP20221063T1 (en) 2017-04-18 2022-11-11 Medimmune Limited Pyrrolobenzodiazepine conjugates
CN110536703A (en) 2017-04-20 2019-12-03 Adc治疗有限公司 Use Anti-AXL antibodies-drug conjugate combination treatment
UA127900C2 (en) 2017-06-14 2024-02-07 Ейдісі Терапьютікс Са Dosage regimes for the administration of an anti-cd19 adc
PL3668874T3 (en) 2017-08-18 2022-03-28 Medimmune Limited Pyrrolobenzodiazepine conjugates
AU2018337815A1 (en) 2017-09-20 2020-03-12 Ph Pharma Co., Ltd. Thailanstatin analogs
GB201803342D0 (en) 2018-03-01 2018-04-18 Medimmune Ltd Methods
GB201806022D0 (en) 2018-04-12 2018-05-30 Medimmune Ltd Pyrrolobenzodiazepines and conjugates thereof
GB201814281D0 (en) 2018-09-03 2018-10-17 Femtogenix Ltd Cytotoxic agents
EP3870235A1 (en) 2018-10-24 2021-09-01 F. Hoffmann-La Roche AG Conjugated chemical inducers of degradation and methods of use
CN113227119A (en) 2018-12-10 2021-08-06 基因泰克公司 Photocrosslinked peptides for site-specific conjugation to Fc-containing proteins
GB201901197D0 (en) 2019-01-29 2019-03-20 Femtogenix Ltd G-A Crosslinking cytotoxic agents
US11471497B1 (en) 2019-03-13 2022-10-18 David Gordon Bermudes Copper chelation therapeutics
US10973908B1 (en) 2020-05-14 2021-04-13 David Gordon Bermudes Expression of SARS-CoV-2 spike protein receptor binding domain in attenuated salmonella as a vaccine
GB2597532A (en) 2020-07-28 2022-02-02 Femtogenix Ltd Cytotoxic compounds

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5256643A (en) * 1990-05-29 1993-10-26 The Government Of The United States Human cripto protein
US5633147A (en) * 1994-03-08 1997-05-27 Human Genome Sciences, Inc. Transforming growth factor αH1
US5834399A (en) * 1997-12-22 1998-11-10 Eastman Kodak Company Subbing layer for dye-donor element used in thermal dye transfer
US6270777B1 (en) * 1996-12-20 2001-08-07 University Technologies International Inc. Conserved metalloprotease epitopes
US6387888B1 (en) * 1998-09-30 2002-05-14 American Foundation For Biological Research, Inc. Immunotherapy of cancer through expression of truncated tumor or tumor-associated antigen
US6537552B1 (en) * 1999-10-19 2003-03-25 Iowa State University Research Foundation Vaccine adjuvant
US20060147478A1 (en) * 2002-08-24 2006-07-06 Terrett Jonathan A Protein involved in carcinoma

Family Cites Families (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5264557A (en) * 1991-08-23 1993-11-23 The United States Of America As Represented By The Department Of Health And Human Services Polypeptide of a human cripto-related gene, CR-3
IL125608A0 (en) 1998-07-30 1999-03-12 Yeda Res & Dev Tumor associated antigen peptides and use of same as anti-tumor vaccines
EP1358349A2 (en) 2000-06-05 2003-11-05 Avalon Pharmaceuticals Cancer gene determination and therapeutic screening using signature gene sets
AUPR395801A0 (en) 2001-03-26 2001-04-26 Austin Research Institute, The Antibodies against cancer

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5256643A (en) * 1990-05-29 1993-10-26 The Government Of The United States Human cripto protein
US5633147A (en) * 1994-03-08 1997-05-27 Human Genome Sciences, Inc. Transforming growth factor αH1
US6270777B1 (en) * 1996-12-20 2001-08-07 University Technologies International Inc. Conserved metalloprotease epitopes
US5834399A (en) * 1997-12-22 1998-11-10 Eastman Kodak Company Subbing layer for dye-donor element used in thermal dye transfer
US6387888B1 (en) * 1998-09-30 2002-05-14 American Foundation For Biological Research, Inc. Immunotherapy of cancer through expression of truncated tumor or tumor-associated antigen
US6537552B1 (en) * 1999-10-19 2003-03-25 Iowa State University Research Foundation Vaccine adjuvant
US20060147478A1 (en) * 2002-08-24 2006-07-06 Terrett Jonathan A Protein involved in carcinoma

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9959525B2 (en) 2005-05-23 2018-05-01 Monster Worldwide, Inc. Intelligent job matching system and method
US10181116B1 (en) 2006-01-09 2019-01-15 Monster Worldwide, Inc. Apparatuses, systems and methods for data entry correlation
US10387839B2 (en) 2006-03-31 2019-08-20 Monster Worldwide, Inc. Apparatuses, methods and systems for automated online data submission
US8645817B1 (en) 2006-12-29 2014-02-04 Monster Worldwide, Inc. Apparatuses, methods and systems for enhanced posted listing generation and distribution management
US20140188743A1 (en) * 2006-12-29 2014-07-03 Monster Worldwide, Inc. Apparatuses, methods and systems for enhanced posted listing generation and distribution management
US9779390B1 (en) 2008-04-21 2017-10-03 Monster Worldwide, Inc. Apparatuses, methods and systems for advancement path benchmarking
US9830575B1 (en) 2008-04-21 2017-11-28 Monster Worldwide, Inc. Apparatuses, methods and systems for advancement path taxonomy
US10387837B1 (en) 2008-04-21 2019-08-20 Monster Worldwide, Inc. Apparatuses, methods and systems for career path advancement structuring

Also Published As

Publication number Publication date
PL363005A1 (en) 2004-11-15
EP1311678A2 (en) 2003-05-21
AR032173A1 (en) 2003-10-29
CN1471579A (en) 2004-01-28
WO2002016413A2 (en) 2002-02-28
AU2001295498A1 (en) 2002-03-04
HUP0302824A2 (en) 2005-05-30
NZ524344A (en) 2004-07-30
CZ2003537A3 (en) 2003-09-17
MXPA03001634A (en) 2004-09-10
GB0020953D0 (en) 2000-10-11
NO20030823D0 (en) 2003-02-21
KR20030045041A (en) 2003-06-09
CA2420087A1 (en) 2002-02-28
IL154533A0 (en) 2003-09-17
WO2002016413A8 (en) 2003-02-06
WO2002016413A3 (en) 2002-12-05
JP2004506441A (en) 2004-03-04
BR0113491A (en) 2003-06-24
ZA200301436B (en) 2004-05-21
US7439320B2 (en) 2008-10-21
NO20030823L (en) 2003-04-23
US20040054142A1 (en) 2004-03-18

Similar Documents

Publication Publication Date Title
US7439320B2 (en) Cripto tumour polypeptide
US8182823B2 (en) Compositions and methods for the therapy and diagnosis of breast cancer
US7563880B2 (en) Compositions and methods for the therapy and diagnosis of breast cancer
US20020193329A1 (en) Compositions and methods for the therapy and diagnosis of Her-2/neu-associated malignancies
US20080249013A1 (en) Vaccines
WO2002092001A2 (en) Compositions and methods for the therapy and diagnosis of lung cancer
US20070292415A1 (en) Compositions and methods for the therapy and diagnosis of breast cancer
EP1212354A2 (en) Ovarian tumor sequences and methods of use therefor
WO2001077168A2 (en) Compositions and methods for the therapy and diagnosis of lung cancer
EP1696028A2 (en) Compositions and methods for the therapy and diagnosis of breast cancer
EP1349485A2 (en) Compositions and methods for the therapy and diagnosis of ovarian cancer
US6958361B2 (en) Compositions and methods for the therapy and diagnosis of breast cancer
US20110150919A1 (en) Compositions and methods for the therapy and diagnosis of breast cancer
US7598226B2 (en) Compositions and methods for the therapy and diagnosis of breast cancer
WO2001098339A2 (en) Compositions and methods for the therapy and diagnosis of breast cancer
WO2002024057A2 (en) Compositions and methods for the therapy and diagnosis of lung cancer

Legal Events

Date Code Title Description
AS Assignment

Owner name: GLAXOSMITHKLINE BIOLOGICALS S.A., BELGIUM

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:CASSART, JEAN-POL;COCHE, THIERRY;PALMANTIER, REMI M.;AND OTHERS;REEL/FRAME:013883/0875;SIGNING DATES FROM 20030605 TO 20030610

STCB Information on status: application discontinuation

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