US20040115218A1 - Ubiquitin fusion-based vaccine system - Google Patents

Ubiquitin fusion-based vaccine system Download PDF

Info

Publication number
US20040115218A1
US20040115218A1 US10/681,389 US68138903A US2004115218A1 US 20040115218 A1 US20040115218 A1 US 20040115218A1 US 68138903 A US68138903 A US 68138903A US 2004115218 A1 US2004115218 A1 US 2004115218A1
Authority
US
United States
Prior art keywords
ubiquitin
fusion protein
epitope
epitopes
protein
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
US10/681,389
Inventor
John Kenten
Alfonso Tramontano
Aprile Pilon
Gerald Lohnas
Steven Roberts
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.)
Wellstat Biocatalysis LLC
Original Assignee
Proteinex Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Proteinex Inc filed Critical Proteinex Inc
Priority to US10/681,389 priority Critical patent/US20040115218A1/en
Publication of US20040115218A1 publication Critical patent/US20040115218A1/en
Assigned to WELLSTAT BIOCATALYSIS, LLC reassignment WELLSTAT BIOCATALYSIS, LLC ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: PROTIENIX CORPORATION
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
    • C07K14/575Hormones
    • C07K14/61Growth hormones [GH] (Somatotropin)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/35Fusion polypeptide containing a fusion for enhanced stability/folding during expression, e.g. fusions with chaperones or thioredoxin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/40Fusion polypeptide containing a tag for immunodetection, or an epitope for immunisation
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/70Fusion polypeptide containing domain for protein-protein interaction
    • C07K2319/74Fusion polypeptide containing domain for protein-protein interaction containing a fusion for binding to a cell surface receptor
    • C07K2319/75Fusion polypeptide containing domain for protein-protein interaction containing a fusion for binding to a cell surface receptor containing a fusion for activation of a cell surface receptor, e.g. thrombopoeitin, NPY and other peptide hormones
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/95Fusion polypeptide containing a motif/fusion for degradation (ubiquitin fusions, PEST sequence)

Definitions

  • immunogenic peptides or peptide conjugates are an active and ongoing research pursuit.
  • the goals include production of reagent antibodies for research, for example in neurobiology, and production of synthetic vaccines for human or veterinary application.
  • Small synthetic peptides are poor antigens and typically require covalent association with macromolecular carriers and administration with adjuvant in order to elicit an immune response.
  • Carriers also provide T-cell epitopes necessary for cell-mediated response and for helper functions in the humoral response. When presented appropriately, synthetic peptides can elicit antibodies against large proteins which display the same peptide epitope within their sequence.
  • Vaccine research further seeks to define those synthetic immunogens capable of inducing an antibody response that is also able to neutralize the infectious activity of a virus or other pathogen from which the protein is derived.
  • a similar structure may be induced in a small peptide by intramolecular covalent modification between two residues that constrains its conformational freedom. These ordered structures can be important as B-cell determinants. However, they are insufficient immunogens in the absence of helper T-cell determinants.
  • Recently developed peptide vaccine models have incorporated T-cell epitopes in association with the B-cell epitope. Designs include simple tandem linear synthesis of peptides as well as increased epitope valency through coupling T-cell and B-cell peptides to a branched polylysine oligomer. The latter assemblies, referred to as multiple antigenic peptides, have shown promise as vaccines against various pathogens.
  • sequences recognized by T-cells undergo extensive processing to short linear peptide fragments before they are bound to a major histocompatibility complex (MHC) for recognition at the surface of an antigen-presenting cell.
  • MHC major histocompatibility complex
  • This elaborate processing mechanism depends on intracellular proteolytic activity and translocation of the products to the cell membrane.
  • Synthetic peptide immunogens may not effectively participate in this process, despite the presence of the T-cell epitope.
  • the immunogenicity of the molecule can be expected to correlate with the efficiency of natural processing of the T-cell epitope.
  • T-cell helper function Further limitations to the use of synthetic peptides as vaccines result from the genetic restriction to T-cell helper function.
  • Multiple MHC class II molecules encoded within the genome of a species are subject to allelic exclusion.
  • a specific T-cell epitope may interact with only one or a few alleles of the MHC. Therefore individuals may respond differently to the immunogen despite inclusion of T-cell helper epitopes.
  • Vaccine development must overcome the MHC-restricted response to provide the broadest possible response in an outbred population.
  • the T-helper cell responses are MHC-restricted and major haplotypes H-2b, H-2k, and H-2d are represented in several inbred strains.
  • T-cell epitopes are known to be recognized in the context of multiple MHC class II alleles and can thereby provide “promiscuous” T-helper stimulation.
  • a number of epitopes, such as from tetanus toxin, measles virus, and Mycobacterium tuberculosis have been reported to be universally immunogenic. These may have significant benefit for subunit vaccine design.
  • the present invention relates to a variety of epitope-containing heat shock fusion proteins.
  • the heat shock protein ubiquitin is fused to a variety of eptiopes or epitope-containing segments.
  • the specific fusion architecture is described in detail below.
  • the epitope-containing segments of the ubiquitin fusion protein comprise either a single epitope or a group of identical or non-identical epitopes.
  • the present invention also relates to DNA constructs which encode an epitope-containing heat shock fusion protein of the type described above, and to cells transformed with such expression constructs.
  • the present invention relates to methods for stimulating an immune response in an animal, the immune response being directed toward a heat shock fusion protein of the type described above.
  • the heat shock fusion protein is administered to an animal under conditions appropriate for the stimulation of an immune response.
  • a DNA construct encoding the heat shock fusion protein is introduced into cells, rather than the fusion protein directly.
  • the present invention also relates to methods for inducing the production of antibodies to endogenous biomolecules using a heat shock fusion protein as described above where the said peptide epitopes are, or are not, related to the endogenous biomolecules in chemical composition but are so called “mimics” of the biomolecular structure and as such have the ability to elicite antibodies to the said biomolecules.
  • Such peptide (epitope) mimics are isolated typically by using phage peptide libraries.
  • the present invention also relates to methods for reducing levels of a predetermined protein (e.g., hormone(s)) in an animal relative to base-line levels, to methods for reducing endogenous TNF levels relative to those in a disease state, to methods for reducing the sperm count in males or inactivating sperm in men and women, to methods for reducing the allergic response, to methods for increasing the growth rate of an animal and to methods for the production and identification of antibodies for use in experimental or diagnostic samples.
  • a predetermined protein e.g., hormone(s)
  • the present invention is based, in one aspect, on the discovery that a fusion protein comprising a heat shock protein (e.g., ubiquitin), fused to an epitope or epitopes in a defined manner, is useful for the stimulation of a highly specific immune response when administered to an animal.
  • a heat shock protein e.g., ubiquitin
  • Heat shock proteins are proteins which are induced during a heat shock. Other stress stimuli are also known to induce a similar response. These proteins are produced to enable the cell to better stand the heat shock or stress. Under these conditions the pattern of gene expression changes and cells overproduce a characteristic set of proteins commonly referred to as heat-shock proteins (hsps).
  • hsps heat-shock proteins
  • One factor in the induction of the heat shock response is the proteolysis of abnormal proteins.
  • ubiquitin is one of these proteins.
  • Ubiquitin is known to be involved in an ATP-dependent pathway of proteolysis. It is also known that proteolysis is an important step in the production of peptides which function in development of the immune response to antigens. Thus it is recognized that hsps are ideal candidates for use in connection with vaccine development.
  • ubiquitin is used as a scaffold to stabilize and display recombinant immunologically active heterologous antigens (referred to herein as epitopes), presumably in a form which generally approximates the native conformation of the epitope.
  • This technique is sometimes referred to as conformational mimicry.
  • the preferred size of the amino acid segments referred to in items a) and b) of the preceding paragraph range from about 5 to about 70 amino acid residues, although larger segments are not intended to be excluded by this statement of the preferred size range.
  • Ubiquitin is a small 76-residue single domain protein which does not induce an appreciable immune response when administered to an animal. Presumably, this “immunological silence” is based on the fact that ubiquitin is expressed in nearly identical form in all eukaryotic systems. Ubiquitin has a variety of other characteristics which make it an ideal “carrier” for the conformational mimicry approach. For example, ubiquitin is highly resistant to protease digestion and is extremely stable both in vivo and when stored for extended periods of time in vitro. The three-dimensional structure of ubiquitin has been determined by X-ray crystallography and its small size makes it amenable to molecular modeling.
  • ubiquitin fusions can be overexpressed in prokaryotic systems such as E. coli , in soluble form, and purified.
  • prokaryotic systems such as E. coli
  • soluble form and purified.
  • protein scaffold system available which offer the benefits of the ubiquitin system.
  • epitopes are to be fused to the ubiquitin framework as outlined above, whether at a single location or non-contiguous locations, it is important to determine what types of ubiquitin modifications are tolerated.
  • “tolerated” can have at least two meanings, systemic tolerance and functional tolerance.
  • the expression “fused”, as used herein, means covalent bound by an amide linkage. This expression encompasses insertion, as well as substitution. At times, these expressions may be used interchangeably herein.
  • Systemic tolerance (i.e., tolerance by the immune system) is important to ensure that the immune response is directed to the epitopes, and not to the ubiquitin carrier.
  • epitope insertion should be designed such that changes to the secondary and tertiary structure of ubiquitin are minimized.
  • Ubiquitin functional tolerance refers to the ability of the ubiquitin protein to behave functionally in a manner analogous to wild-type ubiquitin. This functional tolerance can also be important in a variety of contexts. This property should also be maintained, at least in connection with certain applications, by the ubiquitin fusion carrier constructs of the present invention.
  • ubiquitin as an immunogenic display scaffold, particularly for an internally-fused epitope or epitopes, resides in its ability to maintain the secondary structure found in the epitopes native protein. These secondary structures include, for example, ⁇ -turns and ⁇ -helices.
  • the conformation of the epitope can be further modified by introducing intramolecular bonds between two residues of the epitope which results in conformational constraints on the overall structure of the epitope.
  • the fusion of an epitope or epitopes to terminal regions of ubiquitin also offers advantages in connection with immune-stimulatory activities.
  • the present invention relates to a ubiquitin fusion protein comprising ubiquitin fused to a single epitope-containing segment comprising two or more identical or non-identical epitopes, the epitope-containing segments being fused to ubiquitin at fusion sites selected from the group consisting of the N-terminus and an internal fusion site.
  • a variety of considerations are taken into account when selecting an epitope of use in connection with the fusion proteins of the present invention.
  • an epitope which can stimulate an immune response which protects against an infectious disease, an auto-immune disease or allergic reactions are candidate epitopes for use in connection with the present invention. Epitopes which do not fall into one of these categories can also be useful, and non-limiting examples are discussed more fully below.
  • fusions at a single internal location in the ubiquitin moiety must be designed rationally to minimize, for example, adverse consequences with respect to ubiquitin structure and function.
  • fused epitopes must be “seen” by the immune surveillance system, it is also important that internally fused epitopes are exposed in the folded fusion protein, not buried within a hydrophobic domain.
  • the plurality of epitopes can also be fused to ubiquitin at the N-terminus of the molecule.
  • the epitopes can be identical or non-identical.
  • the epitopes can be B cell epitopes, T cell epitopes or a mixture of B and T cell epitopes.
  • preferred epitopes are B-cell epitopes which are known to be a target for neutralizing antibodies.
  • a second embodiment of the present invention relates to a ubiquitin fusion protein comprising ubiquitin fused to two or more non-contiguous epitope-containing segments, each epitope-containing segment comprising one or more identical or non-identical epitopes.
  • the non-contiguous locations where fusion is appropriate are internal locations within the ubiquitin moiety, or at the N- or C-terminus of the ubiquitin molecule.
  • epitope-containing segment refers to a sequence of amino acids containing one or more epitopes.
  • the epitopes within any particular epitope-containing segment can be identical, or non-identical.
  • the epitopes in a particular epitope-containing segment can be B cell epitopes, T cell epitopes or a mixture of B and T cell epitopes. As discussed above, B-cell epitopes targeted by neutralizing antibodies are preferred in some contexts.
  • an ⁇ -helix fits within the concavity formed by the mixed ⁇ -sheet.
  • loop regions can be defined as domains of ubiquitin which link either two strands within a ⁇ -sheet or a strand of a ⁇ -sheet and an ⁇ -helix. Insertions and substitutions can be made within these loop regions without disrupting the integrity of the ubiquitin molecule or abolishing the features which make ubiquitin a useful carrier for the display of constrained epitopes. Insertions and substitutions within these loop regions tend not to alter the relationships between the prominent structural features defined in the preceding paragraph. Rather, the epitopes introduced into these loop regions tend to protrude from the compact globular ubiquitin structure thereby exposing these epitope residues such that they are easily recognizable by lymphocytes, for example.
  • Epitopes can also be joined to ubiquitin as extensions of the C-terminus. Epitopes fused to the C-terminus of ubiquitin can be cleaved off by ubiquitin-specific proteases in vivo or in vitro. This allows the peptide to be administered to a cell as part of a larger fusion protein which is both easier to purify and handle as compared to free epitope. Following cellular uptake, the epitope attached to the ubiquitin can be cleaved from the C-terminus of ubiquitin and associated with a surface protein such as the MHC complex for expression on the cellular membrane.
  • a surface protein such as the MHC complex for expression on the cellular membrane.
  • the subject invention relates to a ubiquitin fusion protein comprising ubiquitin fused to a single epitope-containing segment, the epitope-containing segment comprising two or more identical or non-identical epitopes.
  • the epitope-containing segment can be fused to ubiquitin at its N-terminus, terminus or internally.
  • the invention relates to yet another fusion protein embodiment comprising ubiquitin fused to a single epitope-containing segment comprising one or more identical or non-identical epitopes.
  • the epitope-containing segment is fused to the ubiquitin moiety at the N-terminus of ubiquitin.
  • the ubiquitin fusion protein comprises an internally inserted B-cell epitope and a T-cell epitope joined to the C-terminus of ubiquitin.
  • B-cell epitopes which have the ability to drive a strong humoral immune response following administration to an animal.
  • the B-cell epitope which is selected will depend upon the intended use of the ubiquitin fusion protein. For instance, if the ubiquitin fusion protein is to be used as a vaccine, the B-cell epitope can be derived from a protein which is expressed by a virus, bacteria or other infectious organism associated with causing a disease.
  • the protein which is selected should be one which contains epitopes which elicit strong antibody responses. These responses are associated with protection of the animal species from the symptomology caused by the infectious organism.
  • the B-cell epitope which is selected is derived from a portion of the protein from the infectious organism known to be both highly immunogenic and to which protective antibodies can be produced. In general, this will include proteins found on the surface of the infectious organism which are involved in binding and to which antibodies have a high degree of access.
  • V3 loop of the HIV gp120 glycoprotein One example of a B-cell epitope which fulfills the requirements set forth above is the V3 loop of the HIV gp120 glycoprotein.
  • an epitope derived from the V3 loop of the HIV gp120 glycoprotein when internally inserted within a ubiquitin fusion protein, is able to drive a strong humoral response.
  • the antibody response was stronger than that found when the epitope was administered independently as a peptide antigen.
  • the selection of this epitope was based on extensive data showing it is a target of neutralizing antibodies.
  • the selection of the B-cell epitope is not limited to proteins associated with infectious organisms. For instance, as shown in Example 2 below, when the ubiquitin fusion protein contains an internally inserted epitope from a prostate-specific antigen, a strong antibody recognition was detected. Examples of other epitopes useful in connection with the present invention include those from proteins which are commonly used as immunological “carriers” as part of experimental studies. These include hen egg lysozyme, keyhole limpet hemocyanin and ovalbumin. One of skill in the art will recognize that any protein containing a B-cell epitope which is capable of driving a humoral immune response can be included in a fusion protein of the present invention. Many such epitopes are known and others can be determined through routine experimentation.
  • a T-cell epitope is joined to the C-terminus of the ubiquitin fusion protein. This epitope is selected based on its ability to enhance the humoral immune response directed to the internally inserted B-cell epitope when both are placed in their proper orientation with respect to ubiquitin.
  • the preferred T-cell epitope is selected from a group of T-cell epitopes which are able to elicit “promiscuus” T-cell help. This type of T-cell epitope is commonly referred to as a universal epitope.
  • Universal T-cell epitopes function by stimulating helper T-cells specific to the B-cells responsive to the ubiquitin fusion protein containing the internally inserted B-cell epitope. They do this regardless of the subject's MHC haplotype or whether the specific “target” protein is different than the protein the universal T-cell epitope is derived from.
  • Examples of universal T-cell epitopes include epitopes from tetanus toxin, measles virus and Mycobacterium tuberculosis . This list of universal T-cell epitopes is not intended to be comprehensive, others which are known or can be determined through application of routine experimentation are also included.
  • T-cell epitopes are preferably inserted internally within the ubiquitin moiety.
  • the T-cell epitopes are selected on the basis of their ability to ensure stimulation of cytotoxic T-cells specific for the particular epitope.
  • a universal T-cell epitope can be attached to the C-terminus of the ubiquitin fusion protein to enhance the stimulation of the specific T-cell response.
  • Cytotoxic T-cells play an important role in the surveillance and control of viral infections, bacterial infections, parasitic infections and cancer, for example.
  • Vaccination with synthetic epitopes, or with epitope pulsed cells has been shown to induce specific cytotoxic T-cell responses directed against immunogenic viral or tumor-derived epitopes.
  • Alternative protocols of T-cell activation allow the triggering of more selective cytotoxic T-cell responses with greater therapeutic effectiveness.
  • the fusion of peptides to the C-terminus of ubiquitin generates a construct which is cleavable, in vivo, by ubiquitin-specific proteases. It is well-established that such ubiquitin-specific proteases cleave ubiquitin fusions after a C-terminal residue (residue 76), thereby releasing the C-terminal peptide.
  • the present invention also encompasses ubiquitin fusion proteins which have been modified such that the fusion is not efficiently cleaved by ubiquitin-specific proteases.
  • ubiquitin can be made resistant to ubiquitin-specific proteases by altering residues at the C-terminus of ubiquitin. For example, by altering the identity of the amino acid at position 76 of ubiquitin (e.g., from glycine to valine or cysteine), the rate of cleavage of a C-terminal ubiquitin fusion can be substantially reduced to the point where cleavage can not be detected using the assays typically employed for monitoring such cleavage.
  • residues at the C-terminus of ubiquitin For example, by altering the identity of the amino acid at position 76 of ubiquitin (e.g., from glycine to valine or cysteine), the rate of cleavage of a C-terminal ubiquitin fusion can be substantially reduced to the point where cleavage can not be detected using the assays typically employed for monitoring such cleavage.
  • the present invention also encompasses fusions to the N-terminus of ubiquitin. It is noted that fusion proteins in which an epitope or epitopes are attached to the N-terminus ubiquitin must be designed such that the N-terminal residue of the encoded fusion protein is methionine. If the N-terminal residue is a residue other than methionine, initiation of translation does not occur efficiently. Unfortunately, however, previous studies have demonstrated that fusions of this type can reduce antibody binding or elicit the production of antibodies of low affinity for the epitope fused to the N-terminus of ubiquitin.
  • tandem ubiquitin fusion is created by attaching a second ubiquitin protein to the N-terminus of the epitope or epitopes attached to the N-terminus of the first ubiquitin moiety.
  • two ubiquitin molecules flank an epitope or epitopes.
  • the C-terminus of the N terminal ubiquitin protein is of the wild-type sequence such that it is cleavable by ubiquitin specific proteases.
  • DNA encoding this tandem ubiquitin fusion is used to transform either prokaryotic or eukaryotic cells. Previous work has shown that the tandem ubiquitin fusion is produced at an equivalent or increased level as compared to the single ubiquitin fusion proteins.
  • tandem ubiquitin fusion can be cleaved, in vitro, by a ubiquitin specific protease thereby releasing the N terminal ubiquitin protein from the core fusion protein which is comprised of an epitope attached to the N-terminus of the C terminal ubiquitin protein.
  • the C terminal ubiquitin protein in the tandem ubiquitin fusion protein can be modified by the inclusion of other epitopes in a manner consistent with the description of the invention above.
  • fusion can be made within the C terminal ubiquitin molecule or to the C-terminus of the C terminal ubiquitin molecule.
  • an alternative tandem ubiquitin construct suitable for use in connection with all embodiments of the present invention is encompassed within the scope of the present invention. More specifically, in the alternative tandem ubiquitin construct, the two ubiquitin moieties are contiguous (i.e., the N-terminus of a first ubiquitin moiety is joined to the C-terminus of a second ubiquitin moiety). In this embodiment, the N-terminal residue of the first ubiquitin moiety is a residue other than methionine. This first ubiquitin moiety is then fused to a wild-type ubiquitin moiety which is cleavable by a ubiquitin-specific protease.
  • the second ubiquitin moiety in this tandem construct need not be a complete ubiquitin moiety. Rather, a C-terminal subdomain of ubiquitin competent to direct cleavage by a ubiquitin-specific protease, is sufficient.
  • ubiquitin or portions thereof may be modified to inhibit cleavage by a ubiquitin-specific protease.
  • a variant of one of the two major embodiments of the present invention is a ubiquitin fusion comprising a first and a second epitope-containing segment inserted internally, and a third epitope-containing segment fused to the C-terminus of ubiquitin.
  • the subject invention encompasses a wide range of such variant embodiments. There is no theoretical limit on the number of epitopes which can be inserted within or fused to the N and C-terminus of a ubiquitin fusion protein.
  • internally fused epitopes are fused as single epitopes, non-contiguously. This design ensures that antibodies produced following vaccination are specific for a single epitope and do not cross-react with other epitopes which have also been internally fused to ubiquitin. Thus, each epitope elicits a specific antibody response by producing antibodies which do not cross-react with other epitopes contained within the same ubiquitin fusion protein.
  • the use of an epitope-containing segment in which two or more distinct epitopes are displayed is preferred when attempting to create bifunctional antibodies for experimental, diagnostic or therapeutic uses.
  • an epitope-containing ubiquitin fusion protein is modified by conjugation to a carrier protein such as ovalbumin (OVA) or keyhole limpet hemocyanin (KLH).
  • OVA ovalbumin
  • KLH keyhole limpet hemocyanin
  • Ubiquitin fusion proteins of the type described above can be modified post-translationally by the addition of fatty acids to enhance immunogenicity.
  • fatty acids for example, palmatic acid (C 16 ) can be added using appropriate chemistry for this purpose.
  • the discussion above has focused on a wide variety of epitope-containing ubiquitin fusion proteins.
  • the invention also relates to DNA expression constructs which encode such epitope-containing ubiquitin fusion proteins.
  • These constructs can be based on prokaryotic expression vectors or eukaryotic expression vectors. Many examples of such expression vectors are known in the art.
  • Prokaryotic expression vectors are useful, for example, for the preparation of large quantities (e.g., up to milligram quantities) of the ubiquitin fusion protein.
  • Eukaryotic expression vectors are useful, for example, when the addition of carbohydrate side chains (i.e., glycosylation) is important.
  • the carbohydrate side chains can affect the properties of a protein in a variety of ways including, for example, the ability of the protein to function in vivo or in vitro; the ability of the protein to form a complex and associate with other proteins or nucleic acids; and the ability of the protein to bind to an antibody or other molecules specific for the protein of interest.
  • the present invention relates to methods of vaccination.
  • the vaccine can be used to drive a cellular and/or humoral immune response depending on the type of epitopes fused to the ubiquitin fusion protein.
  • the therapeutic amount of the ubiquitin fusion protein given to an animal species will be determined as that amount deemed effective in eliciting the desired immune response.
  • the ubiquitin fusion protein is administered in a pharmaceutically acceptable or compatible carrier or adjuvant.
  • the present invention also encompasses pharmaceutical compositions for the administration of ubiquitin fusion proteins.
  • specific diseases which can be treated in this manner include, for example, gastrointestinal diseases, pulmonary infections, respiratory infections and infection with HIV.
  • the pharmaceutical compositions are prepared by methods known to one of skill in the art.
  • the ubiquitin fusion protein is admixed with a carrier and other necessary diluents which are known in the art to aid in producing a product which is stable and administrable.
  • Administration of the pharmaceutical composition can be accomplished by several means known to those of skill in the art. These include oral, intradermal, subcutaneous, intranasal, intravenous or intramuscular.
  • DNA vaccination Conventional vaccination methods involve the administration of an epitope-containing protein. Recently, and alternative to conventional vaccination methods, referred to as DNA vaccination, has been developed. In this method, DNA encoding the epitope-containing protein is introduced into the cells of an organism. Within these cells, the epitope-containing protein is directly expressed. Direct expression of the ubiquitin fusion proteins of the present invention by endogenous cells of a vaccinated animal allows for the continual stimulation of humoral and cellular immune responses over an extended period of time. This is in contrast to standard immunization protocols whereby the vaccine is injected at a single site one or more times. Following injection, the vaccine is disseminated to lymphoid organs where a single immune response occurs.
  • Direct expression can be accomplished by introducing DNA constructs which encode the desired ubiquitin fusion protein into the cells of an animal.
  • the constructs typically contain promoter elements and other transcriptional control elements which direct the expression of the ubiquitin fusion protein.
  • Introduction of the DNA construct can be by any conventional means including direct injection.
  • the preferred administration site is muscle tissue or tissues rich in antigen presenting cells.
  • a ubiquitin fusion protein as described above can also induce a tolerizing effect on the humoral or cellular immune response in an animal.
  • Tolerization occurs following delivery of the ubiquitin fusion proteins to T-cells.
  • the induction of a tolerization response is useful, for example, in connection with the treatment of allergic or autoimmune disorders.
  • epitopes which can be used in a therapeutic regimen designed to induce tolerization include the Fel d 1 peptides, which are the major allergens found in cat pelts. These peptides can be internally inserted, for example, and fused to a cleavable C-terminus of ubiquitin.
  • ubiquitin fusion proteins typically are dosed subcutaneously with the ubiquitin fusion proteins once per week for several weeks. However, dosing can also be done orally or intranasally over a similar length of time. The result is a reduction of the allergic and/or autoimmune responses. These ubiquitin fusion proteins can also be given orally.
  • ubiquitin as a scaffold for the presentation and stimulation of immune responses also allows the stimulation and generation of anti-self responses.
  • An example of a potentially valuable anti-self response is the generation of anti-GnRH (gonadotrophin releasing hormone) antibodies.
  • GnRH gonadotrophin releasing hormone
  • efforts have been made to generate immunogens which stimulate a strong anti-GnRH response which results in the suppression of luteinizing hormone (LH) and follicle stimulating hormone (FSH) and indirectly suppresses the production of the steroidogenesis and gamete maturation in both males and females.
  • LH luteinizing hormone
  • FSH follicle stimulating hormone
  • the value of this type of anti-self response in humans lies in the treatment of prostate cancer and breast cancer.
  • Immunocastration of pigs is especially valuable since it does not result in the detrimental side-effects associated with physical castration. More specifically, physical castration of pigs typically results in animals which do not grow as well as normal animals. In addition, physically castrated pigs tend to have a higher fat percentage than non-castrated pigs. Since, immunocastrated animals are not castrated as early as those which undergo normal physical castration, a farmer can take advantage of the growth rates found with non-castrated animals. Finally, by immunocastrating, the farmer avoids the production of unpalatable meat found with uncastrated male pigs.
  • a further use of ubiquitin as a scaffold is as part of a vaccine to enhance the growth rate and thereby the final weight of livestock prior to shipment to market.
  • This type of vaccine offers a cost effective means to increase the value of livestock such as pigs, cattle and other commonly raised animals.
  • livestock such as pigs, cattle and other commonly raised animals.
  • Currently, to increase the weight of livestock several methods are being utilized. Amongst these methods, the addition of antibiotics to the feed has become very common. However, while addition of antibiotics to feed is cost effective, it has limitations. For instance, it has been blamed for an increase in the creation of antibiotic resistant strains of bacteria.
  • An alternative means to increase the growth rate of livestock which does not result in detrimental side-effects is through vaccination of the animals with an epitope of growth hormone which is part of a ubiquitin fusion protein.
  • the result of this vaccination is an increase in the activity of the animals endogenous growth hormone.
  • the vaccine is created by inserting an epitope from the growth hormone protein (e.g., amino acids 54-95) into the ubiquitin protein thus creating a ubiquitin-growth hormone fusion protein.
  • the effective use of this type of vaccine is described in Example 7 below.
  • the growth rate of pigs was improved when compared to control pigs which received either adjuvant only or adjuvant and ubiquitin only.
  • the ubiquitin fusion proteins of the present invention can be used for the identification of antibodies from experimental or clinical samples.
  • Antibodies to be assayed can be found, for example, in blood, fecal material, the linings of mucosal associated lymphoid tissues, cellular biopsies and other sources known by one of skill in the art to contain at least a minute quantity of antibody.
  • Assays for which the ubiquitin fusion proteins are well suited include ELISAs, radioimmunoassay as well as other commonly used competition assays. These types of assays are useful in identifying antibodies from an experimental or clinical sample which have specificity to at least one epitope of a known protein.
  • the assays involve mixing a predetermined aliquot of the ubiquitin fusion protein with a series of dilutions of the experimental or clinical antibody sample. This is followed by detection of the antibodies which bind to the ubiquitin fusion protein.
  • Detection can be accomplished by various means, but for the present invention, a labeled detection antibody is preferable.
  • the detection antibody can be a polyclonal antibody with specificity for the human heavy chain portion of the sample antibody.
  • the detection antibody is attached to either an enzymatic label, a radioactive label or a fluorochromatic label. Examples of commonly used enzymatic labels are horse radish peroxidase and alkaline phosphatase.
  • a standard radioactive label includes iodine 131.
  • Fluorochromatic labels include fluorescein and Texas red. The method of visualization of the complex containing the ubiquitin fusion protein, the sample antibody, and the labeled antibody depends on the label attached to the antibody and would be known to those of skill in the art.
  • Fusion proteins consisting of short-peptide sequences inserted within a ubiquitin scaffold were developed and tested as immunogens for eliciting a targeted immune response. Preparation and testing of the fusion proteins required several steps including: (I) the design and construction of plasmids encoding peptide sequences fused to the ubiquitin polypeptide at two permissive sites which could be useful for epitope display; (II) the high-level expression in E. coli , purification and characterization of the expected ubiquitin fusions; and (III) evaluation of the immune response in mice with regard to immunogenicity, antibody specificity, cross-reactivity and helper T cell response. Fusion proteins were developed to display the sequence of the HIV-1 gp120 V3 loop, the principal recognition determinant of virus neutralizing antibodies.
  • mice at 6-8 weeks of age were obtained from Charles River (Frederick, Md.) or Jackson Labs. Animals were housed and cared for in an IACUC supervised facility in accordance with an NIH approved Animal Welfare Assurance.
  • Bovine ubiquitin (Sigma, St. Louis Mo.) was dissolved in PBS and filtered through a 0.2 micron filter. Goat anti-mouse peroxidase conjugate was used as obtained from Promega.
  • Mouse monoclonal antibodies against V3 peptides of gp120MN (HG-1) and gp1201IIB (# 13-105-100) were purchased from Biodesign International (Kennebunk, Me.) and from Advanced Biotechnologies Inc. (Guilford, Md.) respectively.
  • Recombinant proteins gp1201IIB and gp120MN and a 36 residue “universal” V3 peptide CTRPNNNTRKSIHIGPGRAFYTTGEIIGDIRQAHC (SEQ ID NO:1) were obtained from Intracel Corporation (Cambridge, Mass.).
  • Synthetic peptides KRIHIGPGRAFYTTK (L-V3) (SEQ ID NO:2) and CKSIHIGPGRAFYTTGC (C-V3) (SEQ ID NO:3) were obtained through the AIDS Research and Reference Reagent Program, Division of AIDS, NIAID, NIH: from Peptide & Protein Research Consultants. (Exeter, Devon, UK).
  • Oligonucleotides were obtained by custom synthesis through Bioserve Biotechnologies. Sequences shown below were designed to encode peptide sequences and to provide appropriate complementary overhangs for ligation to restriction sites in plasmids. Oligonucleotides were treated with T4 polynucleotide kinase and complementary pairs were annealed by heating to 65EC for 5 minutes, then cooling: to room temperature over 30 minutes.
  • Ubiquitin fusions were cloned and expressed in the Proteinix proprietary E. coli strain DH5 ⁇ F′ IQTM (Life Technologies). Vector constructs for expression of fusion proteins are derivatives of pDSUb (provided by M. Rechsteiner, Univ. of Utah), which is a derivative of pDS78/RBSII. The ubiquitin codon usage was optimized for expression in E. coli . Transformants containing pDSUb or its derivatives were selected for ampicillin resistance. Ubiquitin fusion expression, under control of the lac promoter, is inducible by addition of IPTG.
  • Cloning at the ubiquitin codon 35 was initially performed in pRSETUb, derived from PRSET (Invitrogen, Inc.) by subcloning of the ubiquitin gene from pDSUb to PRSET utilizing the Nde I and Hind III restriction sites. Restriction sites for insertions at codon 35 of ubiquitin were created by in vitro mutagenesis with a synthetic oligonucleotide using the MORPH mutagenesis kit (5 Prime-3 Prime, Inc.) to obtain pPX153. Digested vector fragments were treated with calf intestinal alkaline phosphatase and purified from agarose gel slices after electrophoresis using the Geneclean kit (Bio 101).
  • E. coli strain DHSaF′ IQTM Life Technologies. All restriction digests, ligations and transformation of E. coli were done according to standard manipulations using commercial DNA modifying enzymes as instructed by the manufacturer (New England Biolabs). Correct cloning of the oligonucleotide insertions was confirmed by DNA sequencing using the Sequenase Version 2.0 kit (USB, Cleveland, Ohio).
  • lysis buffer 5 ml/g wet paste
  • General purpose buffer consisted of 20 mM MES pH 5.5, supplemented with 1 mM PMSF, 0.5 mM ZnCl2.
  • Cultures expressing certain fusion proteins were lysed in 50 mM acetate pH 4.0 (UbaMT) or pH 4.5 (UbgMT). Cells were disrupted by sonication on ice 2 ⁇ 4 min with a 50% duty cycle. Homogenates were centrifuged at 15,000 ⁇ g, 45 min, 4EC.
  • the supernatants were further diluted with 3 volumes of lysis buffer, filtered through a 0.45 Fm filter, and loaded onto a 30 mL SP sepharose HP ion exchange column at a linear flow rate of 1.4 cm/min.
  • the fusion protein was eluted by a NaCl gradient (0-0.5 M) over 16 column volumes. Fractions were assayed by SDS-PAGE on 10-20% tricine gels (Novex, San Diego Calif.) and protein concentration was determined by the BCA method (Pierce Co.) using a ubiquitin standard curve.
  • Peak fractions were pooled and evaluated further by C18 reversed phase HPLC on a Vydac 218TP54 column using a gradient of 30-45% acetonitrile in water containing 0.1% TFA over 7 min at 1.00 mL/min, with UV detector set at 214 nm.
  • Immunoblots were prepared by electroblotting of samples from SDS-PAGE gels onto Immobilon-P membrane (Millipore Corp.) using an X-Cell II Blot Module (Novex). The membranes were incubated in PBS containing 4% dry milk overnight and then washed with PBS. Incubations with primary antibody were done in PBS, 0.1% Tween 20 for 2 hr at room temperature. After 3 washes with the dilution buffer, membranes were incubated with goat anti-mouse IgG HRP conjugate at 1:5000 dilution in the same buffer. The wash steps were repeated and the membrane was developed with ECL Western detection reagents kit (Amersham, Waltham, Mass.) according to the manufacturer's instructions.
  • the product peptide peak was purified by semi-preparative C18 HPLC (Vydac 218TP510 1.0 cm diameter column) and lyophilized to yield the V3 peptide IHIGPGRAFYTT (SEQ ID NO:16). Identity was verified by FAB-MS.
  • the MT peptide DQVHFQPLPPAVVKLSDAL (SEQ ID NO:17) was obtained by a similar procedure from ion exchange purified UbgMT (4.82 mg/ml).
  • the peptide product was isolated from semi-preparative C18 HPLC using a 20-40% gradient of acetonitrile in 20 mM potassium phosphate, pH 6.0 over 8 min. The peptide content in the lyophilized product was estimated at 10% by weight based on HPLC peak area.
  • mice were inoculated in groups of 3 to 5 per antigen by an initial subcutaneous (s.c.) injection of 100 Fg of proteins in PBS emulsified in an equal volume of complete Freund's adjuvant (CFA) or incomplete Freund's adjuvant (IFA).
  • CFA complete Freund's adjuvant
  • IFA incomplete Freund's adjuvant
  • Booster injections of proteins (100 Fg) were delivered intraperitoneal in IFA 3 weeks and 7 weeks later. Blood samples were collected at 4 weeks, 6 weeks, 8 weeks and 10 weeks from the initial injection. Serum was separated and diluted in PBS.
  • mice were immunized in groups of 3 by injection s.c. in footpads with 50 Fg of proteins in PBS emulsified in IFA.d Eight to ten days later mice were sacrificed and popliteal lymph nodes (LN) were removed aseptically. LN cell suspensions in RPMI 1640 were prepared as described and dispensed into 96-well plates at 5 ⁇ 105 cells per well. Antigens or peptides were added in triplicate wells and plates were kept in a CO 2 incubator at 37EC for 4 days. Wells were then pulsed with 3H-deoxythymidine (1 FCi/well) and cells were harvested on filter pads 24 hours later using an automated collecting device.
  • LN cell suspensions in RPMI 1640 were prepared as described and dispensed into 96-well plates at 5 ⁇ 105 cells per well. Antigens or peptides were added in triplicate wells and plates were kept in a CO 2 incubator at 37EC for 4 days. Wells were then pulsed with 3H-deoxythym
  • Filter sheets were allowed to dry and then counts were read on a Wallac microbeta plate reader (Wallac, Inc., Gaithersburg Md.). Stimulation was expressed as the average signal of duplicate or triplicate wells corrected for mean background counts (c.p.m. with protein or peptide—c.p.m. with buffer added).
  • Fusion proteins 50 Fg/ml or synthetic peptides (10 Fg/ml) in PBS were dispensed into immunosorbent 96-well plates (0.1 ml/well, Corning high binding flat bottom plates) and incubated for 1 hour at 37EC. Excess antigen was shaken out and nonspecific sites were blocked by addition of PBS containing 10 mg/ml of BSA (0.1 ml/well, incubated 30 min at 37EC). Plates were washed three times (Tris buffer 10 mM, 0.1% Tween 20, pH 8) and serially diluted serum samples (1/103-1/104 in PBS supplemented with 1% BSA) were added (0.1 ml/well).
  • Solution phase binding assays were performed by incubating peptides or proteins at concentrations ranging from 0.5-50 Fg/ml with antiserum in 0.1 M potassium phosphate, 2 mM EDTA, 10 mg/ml BSA, pH 7.8 at a fixed concentration of 2-fold greater than the previously determined titer dilution. Samples were incubated at 37EC for 2 hours, then applied to antigen-coated ELISA plates. The standard ELISA procedure was followed to determine the concentration of unbound antibody relative to samples containing no added ligand.
  • the new vector pRSETUbV3 was digested with Bgl II and Afl II, and the smaller fragment encoding UbV3 was gel purified. Ligation of this fragment to pDSUb, pDSUbgMT or pDSUbaMT, each digested with Bgl II and Afl II, generated expression vectors pDSUbV3, pDSUbV3gMT, and pDSUbV3aMT respectively.
  • the enzymatic digest of ubiquitin fusions is a practical bioprocess for production of peptides.
  • Antibodies that neutralize HIV-1 infectivity are directed against the V3 loop or principal neutralizing determinant (PND) of gp120.
  • PND principal neutralizing determinant
  • the PND is implicated in several viral mechanisms known to evade the humoral immune response.
  • Subunit vaccines based on the native structure in gp120 indicate only weak immunogenicity toward this region.
  • Variability of the V3 loop in divergent viral isolates and depletion of B-cells producing antibodies to the PND in infected individuals could also allow for HIV-1 escape from immune surveillance.
  • the gp120 V3 loop has been studied in the context of hybrid protein scaffolds or peptide fusions as a means of producing immunogens that could induce neutralizing antibodies against HIV-1. These materials could be important as components of an effective vaccine for combating the virus.
  • Monoclonal antibodies raised against synthetic V3 peptides which are known to react with gp120 were tested for binding to the recombinant UbV3 by ELISA.
  • the antibody HG-1 specific for gp120 from the HIV-1 strain MN bound to UbV3 and to a 36-residue synthetic V3 loop “universal” peptide with similar efficiency.
  • the binding to UbV3 was inhibited by preincubating the antibody with the 36-residue V3 peptide.
  • Absorbance was reduced by 50% in the presence of 0.5 Fg/ml peptide.
  • Antibody specific for gp120 of the 111B strain did not bind-UbV3.
  • Ubiquitin fusion proteins analyzed by SDS-PAGE were transferred to nitrocellulose membranes for Western blot assays. Strong staining was seen when membranes were developed with mAb HG-1 which is specific for the V3 sequence of gp120 from HIV-1 strain MN. No staining occurred with the mAb derived against the V3 peptide of gp120 of strain IIB.
  • the antibody in these sera was shown to be primarily IgG.
  • the V3 insert was the dominant B-cell target in all three immunogens.
  • the sera bound only weakly or not at all to native ubiquitin or the UbMT fusion.
  • Animals receiving. UbgMT or UbaMT developed measurable antibody against UbgMT and ubiquitin. This response was directed partly to the C-terminal peptide as suggested by the higher ELISA signals obtained against the fusion protein compared with those against ubiquitin (Table 1).
  • Fusion proteins were purified from E. coli lysates and purified by ion exchange chromatography. Solutions in Tris buffer, pH 7.4 were emulsified with Freund's complete adjuvant and injected as described.
  • Antigens used to coat microtiter plates for ELISA were prepared from recombinant E. coli extracts and purified by ion exchange. Three antigens used displayed a single (V3 or MT) or no (Ub) heterologous epitope. Immunoreactivity is indicated by the relative scale from no reaction ( ⁇ ) to strongest reaction++++). Borderline reactivity (signal at the highest concentration of antisera used) is scored as +/ ⁇ .
  • Proteins with C-terminal fusions were produced with the native (rgg) and a mutated (rga) ubiquitin C-terminal sequence to test for stability to or assisted processing of the T-cell epitope by endogenous ubiquitin C-terminal hydrolases that may be present in antigen processing cells.
  • Double epitope fusions had either the native ubiquitin C-terminus (UbV3gMT), retaining the processing site (RGG) for cleavage by cellular ubiquitin-specific proteases (UBPs), or a single residue mutation expressing the sequence RGA at the ubiquitin C-terminus (UbV3aMT), that is resistant to processing.
  • Immunizations with the two types of double-epitope fusions were compared to determine if processing by UBPs in antigen presenting cells could influence a T-helper response. Since the antibody response to the V3 site was independent of the MT epitope in the BALB/c mouse, the effect of the processing mutation could not be observed in this strain.
  • the immunogenicity in C57BL/6 (H-2b) and C3H (H-2k) mouse strains was determined after similar treatment.
  • Ubiquitin has been used as a scaffold for displaying multiple epitopes. These ubiquitin fusion proteins have been successfully recognized by antibodies of the appropriate specificity and analyzed using Origen technology. An immediate technological application for these ubiquitin fusion proteins that display a structurally defined antigenic epitope which is known to be a target for neutralizing antibodies is in production of diagnostic antibodies for clinical or research use.
  • the advantages conferred by the use of ubiquitin as a scaffold include: 1) its ability to stabilize the epitopes in storage, and in in vitro assays, and 2) its ability to constrain the peptide at one: or both ends, thereby conferring the appropriate conformation for: antibody recognition.
  • a commonly used sandwich immunoassay requires two distinct monoclonal antibodies against a single macromolecular analyte, in the present case the ubiquitin fusion proteins. These antibodies are directed to nonoverlapping epitopes on the ubiquitin fusion proteins, allowing the formation of a trimeric complex, or sandwich. Typically, many monoclonal antibodies must be screened in order to identify an appropriate pair for the immunoassay. The technology described here permits the production of monoclonal or polyclonal antibodies against structurally defined epitopes in a macromolecule. Thus, a pair of reagent antibodies for a sandwich immunoassay may be produced by design rather than by random screening. Such reagents may be employed in a wide variety of important immunoassays utilizing any number of formats that are standard in the industry.
  • Example 2 In Example 2, several ubiquitin fusions were constructed. These ubiquitin fusions contain a peptide epitope loop derived from the human PSA (prostate specific antigen). As shown in Table 3 below, ubiquitin fusion proteins contained either single or double epitope insertions on the ubiquitin scaffold. Four different versions of an antigenic PSA peptide loop from human prostate specific antigen were inserted into the ubiquitin scaffold at amino acid position 35. In Table 3 below they are labeled as Fus 5, Fus 5M, Fus 7 and Fus 7M. Four C-terminal fusions were also constructed, both individually and in various combinations with the fusions inserted into the ubiquitin scaffold at amino acid position 35.
  • the peptides joined to the C-terminus of the ubiquitin fusions are shown below in Table 3. They include ATNAT, FLAG, PSA conpep and Fus 7 conpep.
  • the “PSA conpep” fusions contain a modification at amino acid 76 of the ubiquitin to prevent cleavage by a ubiquitin specific protease. These fusions were produced for testing by growth and isolation from bacterial cultures transformed with DNA constructs that encode the ubiquitin fusion proteins. The bacterial lysate from these transformed cultures is nearly homogeneous with respect to protein content. Thus, dilution was sufficient for the following experimentation.
  • the polyclonal antibody raised to a KLH conjugated peptide bound to the PSA peptide in the ubiquitin fusion.
  • This PSA ubiquitin fusion was able to act like the native PSA by competing with native PSA for binding to this polyclonal antibody in an elisa assay.
  • the structural authenticity of the ubiquitin scaffold in maintaining the secondary structure of the PSA peptide inserted internally at position 35 was probed by an in vivo assay.
  • the assay involved the cleavage of the C-terminal PSA peptide from the double epitope ubiquitin fusion protein by a ubiquitin specific protease.
  • Bacteria were transformed with DNA expression constructs for a double epitope ubiquitin fusion protein, UbFus7-conpep or UbFus7-ATNAT shown above in Table 3 and a ubiquitin containing plasmid.
  • UbFus7-conpep fusion protein which was modified at the C-terminus to prevent cleavage by a ubiquitin specific protease remained intact. No ubiquitin specific protease cleavage fragments were identified with UbFus7-conpep, as expected.
  • a fusion protein which contained a C-terminal extension to ubiquitin with the following sequence of the GnRH dimer; QHWSYGLRPGQHWSYGLRPG (SEQ ID NO:26) followed by a T cell epitope, DDPKTGQFLQQINAYARPSEV (corona virus T cell epitope) (SEQ ID NO:27) or DQVHFQPLPPAVVKLSDAL (MT epitope)(SEQ ID NO:17).
  • the ubiquitin used in these constructs was also modified so that its last amino acid was replaced by a valine to render the fusions noncleavable by ubiquitin-specific proteases.
  • These ubiquitin fusions were expressed as described in Example 1 above and purified by ion exchange chromatography and HPLC (when necessary) following standard protocols. The ubiquitin fusion protein were purified to greater than 90% purity.
  • the purified ubiquitin fusion proteins were then formulated with adjuvants as described in Example 1 above and used to immunize mice.
  • the mice were re-immunized about 0.25-30 days following the initial immunization.
  • Sera prepared following bleeds from the mice were tested to determine the level of epitope specific antibodies which were induced by the specific ubiquitin fusion proteins. The results demonstrated that the immunizations resulted in the induction of high levels of anti-GnRH antibodies.
  • Other immunogens which were constructed include ubiquitin with an internal GnRH epitope as a single and as a double epitope at position 35 of ubiquitin and T cell epitopes attached at the C-terminus as described above.
  • the ubiquitin fusion protein with the internal GnRH dimer at position 35 is also fused at its C-terminus with a dimer of GnRH followed by a T cell epitope or with MT at the C-terminus followed by GnRH.
  • the T cell epitope was attached at the C-terminus of ubiquitin which is then fused to the dimer or monomer sequence of GnRH.
  • the GnRH monomer can consist of EHWSYGLRPG (SEQ ID NO:28) with a corresponding dimer of EHWSYGLRPGEHWSYGLRPG (SEQ ID NO:29) or a mixed dimer of EHWSYGLRPGQHWSYGLRPG (SEQ ID NO:30) or QHWSYGLRPGEHWSYGLRPG(SEQ ID NO:31).
  • different GnRH monomers could be used provided such monomers are able to induce antibodies to GNRH.
  • Novel constructs are prepared by placing an epitope at the N-terminus of a first ubiquitin protein to create a fusion protein which can elicit a desired immune response. These constructs differ from those in the prior art by allowing placement of any epitope at the N-terminus of the first ubiquitin protein in order to produce an effective vaccine conjugate.
  • N-terminal epitope presentation is in the generation of an anti-self antibody response.
  • Expression vectors capable of this include those generated for immunocastration. These vectors are based on the vaccine constructs described above in Example 3. However, in the present example, the vaccine constructs include a N terminal ubiquitin protein fused to the N-terminus of the epitope attached to the N-terminus of the C terminal ubiquitin protein. Linkage of the N terminal ubiquitin protein to the N-terminal epitope is through a ubiquitin specific cleavable C-terminus.
  • the sequence coding for (QHWSYGLRPG) n (SEQ ID NO:32), where n is from 1-8, is fused to the 3′ end of the second ubiquitin protein using synthetic oligonucleotides and methods known to one of skill in the art.
  • the resultant gene sequence contains a fusion protein comprised of an epitope flanked on its C-terminus by a C terminal ubiquitin protein and on its N-terminus by a N terminal ubiquitin protein.
  • the ubiquitin proteins joined to GnRH can be used to generate a number of possible combinations of fusion proteins containing multiple GRIRH sequences and T cell epitopes.
  • the fusion between the N terminal ubiquitin protein and an N-terminal epitope can occur via an RGG native ubiquitin C-terminus and a Q at the N-terminus of the GnRH sequence.
  • the GnRH epitope sequence is comprised of from 1 to 8 copies of the following sequence QHWSYGLRPG (SEQ ID NO:32).
  • the fusion protein comprising the GnRH epitope flanked on both its N- and C-terminal ends may be fused to at least one T cell epitope such as DQVHFQPLPPAVVKLSDAL (SEQ ID NO:33) at its C-terminus via a non-native ubiquitin C-terminal sequence such as RGV to render it non-cleavable by the ubiquitin specific proteases.
  • T cell epitope such as DQVHFQPLPPAVVKLSDAL (SEQ ID NO:33)
  • a non-native ubiquitin C-terminal sequence such as RGV to render it non-cleavable by the ubiquitin specific proteases.
  • Variations on the basic construct described above are made using different C terminal ubiquitin fusion proteins. Examples of these can be found in Example 3 above.
  • the C terminal ubiquitin protein can be further modified to include GnRH epitopes (from 1 to 8 epitopes) inserted at position
  • N-terminal epitope ubiquitin fusion proteins these gene constructs are placed within the expression vector described in Example 1 and used to transform E. coli for protein expression.
  • the expressed protein is isolated from the E. coli cells by sonication followed by ion exchange purification to give a preparation which can then be subjected to the action of a ubiquitin specific protease (UBP). Digestion of the fusion protein with the UBP results in the release of the N-terminal ubiquitin protein from the N-terminus of the N-terminally fused epitope.
  • UBP ubiquitin specific protease
  • This cleavage reaction is then subjected to a further ion exchange purification to yield a fusion protein with a GnRH epitope(s) fused to the N-terminus of the COOH terminal ubiquitin protein.
  • the purified ubiquitin fusion protein, with its N-terminal and or C-terminal epitopes, can now be formulated to generate the vaccine for study as described in Example 6.
  • Ubiquitin fusion proteins containing peptide epitopes can be efficiently coupled directly to another protein.
  • two ubiquitin-GnRH fusion proteins are created which are site specifically coupled to ovalbumin.
  • These ubiquitin-GnRH fusion proteins are-constructed using synthetic oligonucleotides, which encode the GnRH sequence; QHWSYGLRPGQHWSYGLRPGQHWSYGLRPGQHWSYGLRPGQHWSYGLRPGQHWSYGLRPG (SEQ ID NO:35) for the second construct.
  • oligonucleotides are cloned into the UBP-cleavable C-terminal site in the coding sequence of ubiquitin as described above in Example 1.
  • the resulting fusion constructs consist of the coding sequence for ubiquitin fused to four copies of the GnRH epitope, with both containing either a C-terminal Cys or not.
  • the coding sequence was placed under the control of a lac promoter which when induced elicits high levels of expressed fusion protein.
  • the resulting construct is used to transform E. coli as described above in Example 1 and the cells were cultured followed by induction of expression of the fusion protein.
  • the fusion protein was isolated from the E. coli cell pellet by first subjecting the cells to sonication, followed by purification of the fusion protein by ion exchange chromatography.
  • Ovalbumin is activated by reaction with N-succinimidyl-3-(2-pyridyldithio)-propionate (SPDP) or by succinimidyl 4-(N-maleimido-methyl) cyclohexane-1-carboxylate (SMCC) followed by gel filtration to remove unreacted cross linking agents.
  • SPDP N-succinimidyl-3-(2-pyridyldithio)-propionate
  • SMCC succinimidyl 4-(N-maleimido-methyl) cyclohexane-1-carboxylate
  • Ubiquitin fusion protein is coupled to ovalbumin by reacting it with activated ovalbumin.
  • the resultant reaction between a free SH group present on the ubiquitin fusion protein and the activated ovalbumin results in a covalent linkage through either a thiol ether linkage with the SMCC activated ovalbumin or a disulfide bond from the reaction with the SPDP activated ovalbumin.
  • the resultant conjugate is formulated with an adjuvant such as Quil-A, complete Freunds adjuvant (CFA) or incomplete Freunds adjuvant (IFA) and then used for immunization as described in Example 6 below.
  • the ubiquitin fusion protein is coupled to ovalbumin by reacting it with ovalbumin in the presence of a cross linking agent such as disuccinimidyl suberate or gluteraldehyde (Pierce).
  • a cross linking agent such as disuccinimidyl suberate or gluteraldehyde (Pierce).
  • the resultant reaction the ubiquitin fusion protein and ovalbumin results in a covalent linkage through amino groups on the ubiquitin fusion protein and ovalbumin.
  • the resultant conjugate is formulated with an adjuvant such as Quil-A, complete Freunds adjuvant (CFA) or incomplete Freunds adjuvant (IFA) and then used for immunization as described in Example 6 below.
  • an adjuvant such as Quil-A, complete Freunds adjuvant (CFA) or incomplete Freunds adjuvant (IFA)
  • the ubiquitin fusion protein immunogens constructed as described above in Examples 3, 4 and 5 were tested in piglets. Male piglets which were between the age of 9-10 weeks were immunized with 1-10 mg of the ubiquitin GnRH immunogens in complete Freund's adjuvant (CFA) intramuscularly. Immunizations were repeated 8 weeks following the initial CFA immunization, with IFA. The piglets were slaughtered 16 weeks after the initial immunization at which time the testicles were excised and weighed. In addition, the serum testosterone levels of the piglets was determined along with the androstenone levels in fat.
  • CFA complete Freund's adjuvant
  • a vaccine was constructed which included the insertion of a growth hormone epitpope into ubiquitin.
  • a growth hormone epitpope into ubiquitin.
  • Prior studies have shown that the epitope encoded by amino acids 54-95 of growth hormone can be used to make a vaccine which improves the growth rate of pigs by increasing the activity of the endogenous growth hormone.
  • ubiquitin fusion proteins containing this growth hormone epitope novel vaccines can be generated which offer the advantages of enhanced hormone activity and lower costs.
  • the growth hormone epitope was inserted into ubiquitin as described in Examples 3, 4 and 5 above, with the growth hormone epitope inserted at the same sites described above for the GnRH epitopes.
  • the growth hormone epitope can be inserted as a multimer, with up to four contiguous repeats to enhance its immunogenicity. Constructs encoding the growth hormone-ubiquitin fusion proteins were transformed into and expressed in E. coli , followed by purification of the fusion protein by methods described above.
  • the purified growth hormone-ubiquitin fusion proteins were formulated with adjuvant and used to immunize pigs weighing 15-20 kg. CFA was used for the first immunization followed by two subsequent booster injections with IPA. These subsequent booster injections were each given at 4 week intervals following the initial injection. Pigs were monitored until they reached a weight of 110-120 Kg at which time the animals were killed. The resultant weight gain by immunized pigs when compared to control animals receiving only adjuvant or ubiquitin only and adjuvant demonstrated the improved growth rates of the immunized pigs.

Abstract

Disclosed are epitope-containing heat shock fusion proteins, DNA constructs encoding such fusion proteins, and methods of use. More specifically, disclosed are ubiquitin fusion proteins comprising ubiquitin fused to a plurality of identical or non-identical epitopes at specified locations.

Description

    BACKGROUND OF THE INVENTION
  • The construction of immunogenic peptides or peptide conjugates is an active and ongoing research pursuit. The goals include production of reagent antibodies for research, for example in neurobiology, and production of synthetic vaccines for human or veterinary application. Small synthetic peptides are poor antigens and typically require covalent association with macromolecular carriers and administration with adjuvant in order to elicit an immune response. Carriers also provide T-cell epitopes necessary for cell-mediated response and for helper functions in the humoral response. When presented appropriately, synthetic peptides can elicit antibodies against large proteins which display the same peptide epitope within their sequence. Vaccine research further seeks to define those synthetic immunogens capable of inducing an antibody response that is also able to neutralize the infectious activity of a virus or other pathogen from which the protein is derived. [0001]
  • A significant effort has been devoted to discovery of general rules which govern the selection of protein epitopes by the immune system and development of methodology for mimicry of such epitopes with synthetic immunogens. Evidence has emerged suggesting that linear or discontinuous epitopes may be recognized and that these may adopt defined conformational states that are not readily duplicated in a synthetic peptide. Attempts to devise conformationally restricted peptides as superior antigens have also been given serious attention. In such approaches the structural context of a peptide sequence within a protein antigen is considered in producing a suitable mimic. Typically, the epitope may adopt a secondary structure such as a β-turn or a-helix. A similar structure may be induced in a small peptide by intramolecular covalent modification between two residues that constrains its conformational freedom. These ordered structures can be important as B-cell determinants. However, they are insufficient immunogens in the absence of helper T-cell determinants. Recently developed peptide vaccine models have incorporated T-cell epitopes in association with the B-cell epitope. Designs include simple tandem linear synthesis of peptides as well as increased epitope valency through coupling T-cell and B-cell peptides to a branched polylysine oligomer. The latter assemblies, referred to as multiple antigenic peptides, have shown promise as vaccines against various pathogens. [0002]
  • Unlike the conformationally defined B-cell epitopes, sequences recognized by T-cells undergo extensive processing to short linear peptide fragments before they are bound to a major histocompatibility complex (MHC) for recognition at the surface of an antigen-presenting cell. This elaborate processing mechanism depends on intracellular proteolytic activity and translocation of the products to the cell membrane. Synthetic peptide immunogens may not effectively participate in this process, despite the presence of the T-cell epitope. The immunogenicity of the molecule can be expected to correlate with the efficiency of natural processing of the T-cell epitope. Studies with linear synthetic peptides indicated that chimeric peptides containing T-cell and B-cell epitopes were superior immunogens when the B-cell epitope was amino-terminal. However, the reverse orientation has also been reported to produce a stronger immune response. A general rule may not be obvious since natural antigen processing probably accepts various orientations, including internal epitopes that require multiple processing steps to release the peptide. Also the efficiency of a construct may depend on many other factors, such as molecular context and flanking sequences that affect processing or presentation and the overall nature of the immunogen which can affect the functional pairing between several available T-cell and B-cell epitopes. [0003]
  • Further limitations to the use of synthetic peptides as vaccines result from the genetic restriction to T-cell helper function. Multiple MHC class II molecules encoded within the genome of a species are subject to allelic exclusion. A specific T-cell epitope may interact with only one or a few alleles of the MHC. Therefore individuals may respond differently to the immunogen despite inclusion of T-cell helper epitopes. Vaccine development must overcome the MHC-restricted response to provide the broadest possible response in an outbred population. In the murine model the T-helper cell responses are MHC-restricted and major haplotypes H-2b, H-2k, and H-2d are represented in several inbred strains. Certain T-cell epitopes are known to be recognized in the context of multiple MHC class II alleles and can thereby provide “promiscuous” T-helper stimulation. A number of epitopes, such as from tetanus toxin, measles virus, and [0004] Mycobacterium tuberculosis have been reported to be universally immunogenic. These may have significant benefit for subunit vaccine design.
  • As an alternative to chemical synthesis, molecular biological techniques can provide significant advantages for production of polypeptides that display both B-cell and T-cell epitopes. Expression of proteins from cloned genes obviates burdensome peptide synthesis, purification and conjugation chemistry typically used in production of immunogenic materials. Furthermore, the stochastic chemistry for preparation of peptide-carrier conjugates is replaced by the defined chemical structure provided by the genetic fusion. Therefore the epitopes can be introduced in ordered structures that have optimal and reproducible immunogenic properties. Considerations that arise in the development of optimal designs can be addressed at the genetic level. Thus, definition of the target epitopes and their flanking sequences, relative orientation and conformations of these sequences within the larger polypeptide, and epitope copy frequency can be established in the gene design. [0005]
  • Several recombinant host proteins have been successfully utilized for immune presentation of peptide epitopes. The [0006] E. coli maltose-binding protein (MalE) has been used to study the influence of location and orientation of inserted T-cell epitopes. The major coat protein (pVIII) of filamentous bacteriophage fd has been used for display of HIV B-cell epitopes at the N-terminus. The recombinant phage particles evoked a strong antibody response in mice, which cross-reacted with HIV strains and which is also capable of neutralizing the virus. These approaches promise to enhance the potential of subunit or synthetic vaccine models.
  • SUMMARY OF THE INVENTION
  • The present invention relates to a variety of epitope-containing heat shock fusion proteins. In one embodiment, the heat shock protein ubiquitin is fused to a variety of eptiopes or epitope-containing segments. The specific fusion architecture is described in detail below. The epitope-containing segments of the ubiquitin fusion protein comprise either a single epitope or a group of identical or non-identical epitopes. [0007]
  • The present invention also relates to DNA constructs which encode an epitope-containing heat shock fusion protein of the type described above, and to cells transformed with such expression constructs. [0008]
  • In other aspects, the present invention relates to methods for stimulating an immune response in an animal, the immune response being directed toward a heat shock fusion protein of the type described above. The heat shock fusion protein is administered to an animal under conditions appropriate for the stimulation of an immune response. In an alternative embodiment, a DNA construct encoding the heat shock fusion protein is introduced into cells, rather than the fusion protein directly. [0009]
  • The present invention also relates to methods for inducing the production of antibodies to endogenous biomolecules using a heat shock fusion protein as described above where the said peptide epitopes are, or are not, related to the endogenous biomolecules in chemical composition but are so called “mimics” of the biomolecular structure and as such have the ability to elicite antibodies to the said biomolecules. Such peptide (epitope) mimics are isolated typically by using phage peptide libraries. [0010]
  • The present invention also relates to methods for reducing levels of a predetermined protein (e.g., hormone(s)) in an animal relative to base-line levels, to methods for reducing endogenous TNF levels relative to those in a disease state, to methods for reducing the sperm count in males or inactivating sperm in men and women, to methods for reducing the allergic response, to methods for increasing the growth rate of an animal and to methods for the production and identification of antibodies for use in experimental or diagnostic samples. [0011]
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention is based, in one aspect, on the discovery that a fusion protein comprising a heat shock protein (e.g., ubiquitin), fused to an epitope or epitopes in a defined manner, is useful for the stimulation of a highly specific immune response when administered to an animal. The specific fusion architecture encompassed by the invention will be discussed in greater detail below. [0012]
  • Heat shock proteins are proteins which are induced during a heat shock. Other stress stimuli are also known to induce a similar response. These proteins are produced to enable the cell to better stand the heat shock or stress. Under these conditions the pattern of gene expression changes and cells overproduce a characteristic set of proteins commonly referred to as heat-shock proteins (hsps). One factor in the induction of the heat shock response is the proteolysis of abnormal proteins. [0013]
  • Various examples of heat shock proteins-exist. In yeast, mammals and other eukaryotes, ubiquitin is one of these proteins. Ubiquitin is known to be involved in an ATP-dependent pathway of proteolysis. It is also known that proteolysis is an important step in the production of peptides which function in development of the immune response to antigens. Thus it is recognized that hsps are ideal candidates for use in connection with vaccine development. [0014]
  • In connection with the present invention, ubiquitin is used as a scaffold to stabilize and display recombinant immunologically active heterologous antigens (referred to herein as epitopes), presumably in a form which generally approximates the native conformation of the epitope. This technique is sometimes referred to as conformational mimicry. Generally, the preferred size of the amino acid segments referred to in items a) and b) of the preceding paragraph range from about 5 to about 70 amino acid residues, although larger segments are not intended to be excluded by this statement of the preferred size range. [0015]
  • Ubiquitin is a small 76-residue single domain protein which does not induce an appreciable immune response when administered to an animal. Presumably, this “immunological silence” is based on the fact that ubiquitin is expressed in nearly identical form in all eukaryotic systems. Ubiquitin has a variety of other characteristics which make it an ideal “carrier” for the conformational mimicry approach. For example, ubiquitin is highly resistant to protease digestion and is extremely stable both in vivo and when stored for extended periods of time in vitro. The three-dimensional structure of ubiquitin has been determined by X-ray crystallography and its small size makes it amenable to molecular modeling. Additionally, ubiquitin fusions can be overexpressed in prokaryotic systems such as [0016] E. coli, in soluble form, and purified. One of skill in the art will recognize that for particular applications some of the aforementioned properties are unimportant and dispensable. At the present time, there is no comparable protein scaffold system available which offer the benefits of the ubiquitin system.
  • If epitopes are to be fused to the ubiquitin framework as outlined above, whether at a single location or non-contiguous locations, it is important to determine what types of ubiquitin modifications are tolerated. In this context, “tolerated” can have at least two meanings, systemic tolerance and functional tolerance. It should also be noted that the expression “fused”, as used herein, means covalent bound by an amide linkage. This expression encompasses insertion, as well as substitution. At times, these expressions may be used interchangeably herein. [0017]
  • Systemic tolerance, (i.e., tolerance by the immune system) is important to ensure that the immune response is directed to the epitopes, and not to the ubiquitin carrier. Thus, epitope insertion should be designed such that changes to the secondary and tertiary structure of ubiquitin are minimized. [0018]
  • Ubiquitin functional tolerance refers to the ability of the ubiquitin protein to behave functionally in a manner analogous to wild-type ubiquitin. This functional tolerance can also be important in a variety of contexts. This property should also be maintained, at least in connection with certain applications, by the ubiquitin fusion carrier constructs of the present invention. [0019]
  • As disclosed herein, insertions at particular internal sites in ubiquitin are “tolerated”, as this term is defined above. One advantage of using ubiquitin as an immunogenic display scaffold, particularly for an internally-fused epitope or epitopes, resides in its ability to maintain the secondary structure found in the epitopes native protein. These secondary structures include, for example, β-turns and α-helices. The conformation of the epitope can be further modified by introducing intramolecular bonds between two residues of the epitope which results in conformational constraints on the overall structure of the epitope. The fusion of an epitope or epitopes to terminal regions of ubiquitin also offers advantages in connection with immune-stimulatory activities. [0020]
  • In a first embodiment, the present invention relates to a ubiquitin fusion protein comprising ubiquitin fused to a single epitope-containing segment comprising two or more identical or non-identical epitopes, the epitope-containing segments being fused to ubiquitin at fusion sites selected from the group consisting of the N-terminus and an internal fusion site. As discussed in greater detail below, a variety of considerations are taken into account when selecting an epitope of use in connection with the fusion proteins of the present invention. Generally speaking, an epitope which can stimulate an immune response which protects against an infectious disease, an auto-immune disease or allergic reactions are candidate epitopes for use in connection with the present invention. Epitopes which do not fall into one of these categories can also be useful, and non-limiting examples are discussed more fully below. [0021]
  • With respect to the first embodiment, fusions at a single internal location in the ubiquitin moiety must be designed rationally to minimize, for example, adverse consequences with respect to ubiquitin structure and function. In light of the fact that fused epitopes must be “seen” by the immune surveillance system, it is also important that internally fused epitopes are exposed in the folded fusion protein, not buried within a hydrophobic domain. The plurality of epitopes can also be fused to ubiquitin at the N-terminus of the molecule. The epitopes can be identical or non-identical. In addition, the epitopes can be B cell epitopes, T cell epitopes or a mixture of B and T cell epitopes. For many applications, preferred epitopes are B-cell epitopes which are known to be a target for neutralizing antibodies. [0022]
  • A second embodiment of the present invention relates to a ubiquitin fusion protein comprising ubiquitin fused to two or more non-contiguous epitope-containing segments, each epitope-containing segment comprising one or more identical or non-identical epitopes. The non-contiguous locations where fusion is appropriate are internal locations within the ubiquitin moiety, or at the N- or C-terminus of the ubiquitin molecule. [0023]
  • As used herein in connection with the second embodiment, the term “epitope-containing segment” refers to a sequence of amino acids containing one or more epitopes. The epitopes within any particular epitope-containing segment can be identical, or non-identical. In addition, the epitopes in a particular epitope-containing segment can be B cell epitopes, T cell epitopes or a mixture of B and T cell epitopes. As discussed above, B-cell epitopes targeted by neutralizing antibodies are preferred in some contexts. [0024]
  • When considering the insertion of epitopes within the ubiquitin molecule, the structure of the ubiquitin molecule must be considered. The prominent structural features of ubiquitin, as determined by X-ray crystallography (see, e.g., Vijay-Kumar et al., [0025] Proc. Natl. Acad. Sci. USA 82: 3582 (1985); Vijay-Kumar et al., J. Mol. Biol. 194: 525 (1987); and Vijay-Kumar et al., J. Biol. Chem. 262: 6396 (1987)) include a mixed β-sheet comprising two parallel inner strands (residues 1-7 and. 64-72), as well as two antiparallel strands (residues 10-17 and 40-45). In addition, an α-helix (residues 23-34) fits within the concavity formed by the mixed β-sheet.
  • The amino acid sequences which link the structural elements defined in the preceding paragraph are referred to herein as “loop regions”. Thus, loop regions can be defined as domains of ubiquitin which link either two strands within a β-sheet or a strand of a β-sheet and an α-helix. Insertions and substitutions can be made within these loop regions without disrupting the integrity of the ubiquitin molecule or abolishing the features which make ubiquitin a useful carrier for the display of constrained epitopes. Insertions and substitutions within these loop regions tend not to alter the relationships between the prominent structural features defined in the preceding paragraph. Rather, the epitopes introduced into these loop regions tend to protrude from the compact globular ubiquitin structure thereby exposing these epitope residues such that they are easily recognizable by lymphocytes, for example. [0026]
  • As discussed above, internal modification sites are selected such that the ubiquitin secondary structure is maintained and the conformation of the inserted epitope is constrained. Epitopes can also be joined to ubiquitin as extensions of the C-terminus. Epitopes fused to the C-terminus of ubiquitin can be cleaved off by ubiquitin-specific proteases in vivo or in vitro. This allows the peptide to be administered to a cell as part of a larger fusion protein which is both easier to purify and handle as compared to free epitope. Following cellular uptake, the epitope attached to the ubiquitin can be cleaved from the C-terminus of ubiquitin and associated with a surface protein such as the MHC complex for expression on the cellular membrane. [0027]
  • In another embodiment, the subject invention relates to a ubiquitin fusion protein comprising ubiquitin fused to a single epitope-containing segment, the epitope-containing segment comprising two or more identical or non-identical epitopes. The epitope-containing segment can be fused to ubiquitin at its N-terminus, terminus or internally. [0028]
  • The invention relates to yet another fusion protein embodiment comprising ubiquitin fused to a single epitope-containing segment comprising one or more identical or non-identical epitopes. In this embodiment, the epitope-containing segment is fused to the ubiquitin moiety at the N-terminus of ubiquitin. [0029]
  • The use of ubiquitin fusion proteins to initiate a humoral response is described in more detail in the following Exemplification section. More, specifically, these experiments demonstrate, for example, that the B-cell and T-cell epitopes expressed in the ubiquitin fusion protein stimulated targeted immune responses. Further, the experiments demonstrate that a humoral immune response to an internally inserted B-cell epitope was enhanced by the addition of a T-cell epitope to the C-terminus of the ubiquitin fusion protein. Although the bulk of the in vivo data reported herein were generated in experiments employing murine indicator assays for the generation of antibodies against the ubiquitin fusion proteins, the fundamental principles are applicable to humans as well as other animals. Given the disclosure of the subject application it is a matter of routine experimentation to select epitopes of interest and incorporate such epitopes of interest into a ubiquitin fusion protein for use as an immunogen. [0030]
  • Thus, in a preferred embodiment, the ubiquitin fusion protein comprises an internally inserted B-cell epitope and a T-cell epitope joined to the C-terminus of ubiquitin. One of skill in the art can identify B-cell epitopes which have the ability to drive a strong humoral immune response following administration to an animal. The B-cell epitope which is selected will depend upon the intended use of the ubiquitin fusion protein. For instance, if the ubiquitin fusion protein is to be used as a vaccine, the B-cell epitope can be derived from a protein which is expressed by a virus, bacteria or other infectious organism associated with causing a disease. The protein which is selected should be one which contains epitopes which elicit strong antibody responses. These responses are associated with protection of the animal species from the symptomology caused by the infectious organism. Preferably, the B-cell epitope which is selected is derived from a portion of the protein from the infectious organism known to be both highly immunogenic and to which protective antibodies can be produced. In general, this will include proteins found on the surface of the infectious organism which are involved in binding and to which antibodies have a high degree of access. [0031]
  • One example of a B-cell epitope which fulfills the requirements set forth above is the V3 loop of the HIV gp120 glycoprotein. As described in the following section, an epitope derived from the V3 loop of the HIV gp120 glycoprotein, when internally inserted within a ubiquitin fusion protein, is able to drive a strong humoral response. In fact, the antibody response was stronger than that found when the epitope was administered independently as a peptide antigen. The selection of this epitope was based on extensive data showing it is a target of neutralizing antibodies. [0032]
  • The selection of the B-cell epitope is not limited to proteins associated with infectious organisms. For instance, as shown in Example 2 below, when the ubiquitin fusion protein contains an internally inserted epitope from a prostate-specific antigen, a strong antibody recognition was detected. Examples of other epitopes useful in connection with the present invention include those from proteins which are commonly used as immunological “carriers” as part of experimental studies. These include hen egg lysozyme, keyhole limpet hemocyanin and ovalbumin. One of skill in the art will recognize that any protein containing a B-cell epitope which is capable of driving a humoral immune response can be included in a fusion protein of the present invention. Many such epitopes are known and others can be determined through routine experimentation. [0033]
  • In a preferred embodiment, a T-cell epitope is joined to the C-terminus of the ubiquitin fusion protein. This epitope is selected based on its ability to enhance the humoral immune response directed to the internally inserted B-cell epitope when both are placed in their proper orientation with respect to ubiquitin. The preferred T-cell epitope is selected from a group of T-cell epitopes which are able to elicit “promiscuus” T-cell help. This type of T-cell epitope is commonly referred to as a universal epitope. Universal T-cell epitopes function by stimulating helper T-cells specific to the B-cells responsive to the ubiquitin fusion protein containing the internally inserted B-cell epitope. They do this regardless of the subject's MHC haplotype or whether the specific “target” protein is different than the protein the universal T-cell epitope is derived from. Examples of universal T-cell epitopes include epitopes from tetanus toxin, measles virus and [0034] Mycobacterium tuberculosis. This list of universal T-cell epitopes is not intended to be comprehensive, others which are known or can be determined through application of routine experimentation are also included.
  • To stimulate cytotoxic T-cells as part of a cellular immune response, T-cell epitopes are preferably inserted internally within the ubiquitin moiety. In addition, it is preferable to fuse at least one T-cell epitope to the C-terminus of the ubiquitin fusion protein. In this case, the T-cell epitopes are selected on the basis of their ability to ensure stimulation of cytotoxic T-cells specific for the particular epitope. However, a universal T-cell epitope can be attached to the C-terminus of the ubiquitin fusion protein to enhance the stimulation of the specific T-cell response. [0035]
  • Cytotoxic T-cells play an important role in the surveillance and control of viral infections, bacterial infections, parasitic infections and cancer, for example. Vaccination with synthetic epitopes, or with epitope pulsed cells, has been shown to induce specific cytotoxic T-cell responses directed against immunogenic viral or tumor-derived epitopes. Alternative protocols of T-cell activation allow the triggering of more selective cytotoxic T-cell responses with greater therapeutic effectiveness. [0036]
  • Generally, the fusion of peptides to the C-terminus of ubiquitin generates a construct which is cleavable, in vivo, by ubiquitin-specific proteases. It is well-established that such ubiquitin-specific proteases cleave ubiquitin fusions after a C-terminal residue (residue 76), thereby releasing the C-terminal peptide. The present invention also encompasses ubiquitin fusion proteins which have been modified such that the fusion is not efficiently cleaved by ubiquitin-specific proteases. As is well known to those of skill in the art, ubiquitin can be made resistant to ubiquitin-specific proteases by altering residues at the C-terminus of ubiquitin. For example, by altering the identity of the amino acid at position 76 of ubiquitin (e.g., from glycine to valine or cysteine), the rate of cleavage of a C-terminal ubiquitin fusion can be substantially reduced to the point where cleavage can not be detected using the assays typically employed for monitoring such cleavage. [0037]
  • As mentioned previously, the present invention also encompasses fusions to the N-terminus of ubiquitin. It is noted that fusion proteins in which an epitope or epitopes are attached to the N-terminus ubiquitin must be designed such that the N-terminal residue of the encoded fusion protein is methionine. If the N-terminal residue is a residue other than methionine, initiation of translation does not occur efficiently. Unfortunately, however, previous studies have demonstrated that fusions of this type can reduce antibody binding or elicit the production of antibodies of low affinity for the epitope fused to the N-terminus of ubiquitin. [0038]
  • To overcome this problem, a tandem ubiquitin fusion is created by attaching a second ubiquitin protein to the N-terminus of the epitope or epitopes attached to the N-terminus of the first ubiquitin moiety. Thus, in this embodiment of the present invention, two ubiquitin molecules flank an epitope or epitopes. The C-terminus of the N terminal ubiquitin protein is of the wild-type sequence such that it is cleavable by ubiquitin specific proteases. DNA encoding this tandem ubiquitin fusion is used to transform either prokaryotic or eukaryotic cells. Previous work has shown that the tandem ubiquitin fusion is produced at an equivalent or increased level as compared to the single ubiquitin fusion proteins. [0039]
  • Following purification, the tandem ubiquitin fusion can be cleaved, in vitro, by a ubiquitin specific protease thereby releasing the N terminal ubiquitin protein from the core fusion protein which is comprised of an epitope attached to the N-terminus of the C terminal ubiquitin protein. [0040]
  • The C terminal ubiquitin protein in the tandem ubiquitin fusion protein can be modified by the inclusion of other epitopes in a manner consistent with the description of the invention above. Thus, fusion can be made within the C terminal ubiquitin molecule or to the C-terminus of the C terminal ubiquitin molecule. [0041]
  • An alternative tandem ubiquitin construct suitable for use in connection with all embodiments of the present invention is encompassed within the scope of the present invention. More specifically, in the alternative tandem ubiquitin construct, the two ubiquitin moieties are contiguous (i.e., the N-terminus of a first ubiquitin moiety is joined to the C-terminus of a second ubiquitin moiety). In this embodiment, the N-terminal residue of the first ubiquitin moiety is a residue other than methionine. This first ubiquitin moiety is then fused to a wild-type ubiquitin moiety which is cleavable by a ubiquitin-specific protease. [0042]
  • Alternatively, the second ubiquitin moiety in this tandem construct need not be a complete ubiquitin moiety. Rather, a C-terminal subdomain of ubiquitin competent to direct cleavage by a ubiquitin-specific protease, is sufficient. In addition, in connection with this and any other embodiment of the present invention, ubiquitin (or portions thereof) may be modified to inhibit cleavage by a ubiquitin-specific protease. [0043]
  • A variant of one of the two major embodiments of the present invention is a ubiquitin fusion comprising a first and a second epitope-containing segment inserted internally, and a third epitope-containing segment fused to the C-terminus of ubiquitin. The subject invention encompasses a wide range of such variant embodiments. There is no theoretical limit on the number of epitopes which can be inserted within or fused to the N and C-terminus of a ubiquitin fusion protein. [0044]
  • In preferred embodiments, internally fused epitopes are fused as single epitopes, non-contiguously. This design ensures that antibodies produced following vaccination are specific for a single epitope and do not cross-react with other epitopes which have also been internally fused to ubiquitin. Thus, each epitope elicits a specific antibody response by producing antibodies which do not cross-react with other epitopes contained within the same ubiquitin fusion protein. The use of an epitope-containing segment in which two or more distinct epitopes are displayed is preferred when attempting to create bifunctional antibodies for experimental, diagnostic or therapeutic uses. [0045]
  • In another embodiment of the present invention, an epitope-containing ubiquitin fusion protein is modified by conjugation to a carrier protein such as ovalbumin (OVA) or keyhole limpet hemocyanin (KLH). Example 5, presented below, exemplifies such an embodiment. [0046]
  • Ubiquitin fusion proteins of the type described above can be modified post-translationally by the addition of fatty acids to enhance immunogenicity. For example, palmatic acid (C[0047] 16) can be added using appropriate chemistry for this purpose.
  • The discussion above has focused on a wide variety of epitope-containing ubiquitin fusion proteins. The invention also relates to DNA expression constructs which encode such epitope-containing ubiquitin fusion proteins. These constructs can be based on prokaryotic expression vectors or eukaryotic expression vectors. Many examples of such expression vectors are known in the art. Prokaryotic expression vectors are useful, for example, for the preparation of large quantities (e.g., up to milligram quantities) of the ubiquitin fusion protein. Eukaryotic expression vectors are useful, for example, when the addition of carbohydrate side chains (i.e., glycosylation) is important. The carbohydrate side chains can affect the properties of a protein in a variety of ways including, for example, the ability of the protein to function in vivo or in vitro; the ability of the protein to form a complex and associate with other proteins or nucleic acids; and the ability of the protein to bind to an antibody or other molecules specific for the protein of interest. [0048]
  • In another aspect, the present invention relates to methods of vaccination. The vaccine can be used to drive a cellular and/or humoral immune response depending on the type of epitopes fused to the ubiquitin fusion protein. The therapeutic amount of the ubiquitin fusion protein given to an animal species will be determined as that amount deemed effective in eliciting the desired immune response. The ubiquitin fusion protein is administered in a pharmaceutically acceptable or compatible carrier or adjuvant. [0049]
  • Thus, the present invention also encompasses pharmaceutical compositions for the administration of ubiquitin fusion proteins. Examples of specific diseases which can be treated in this manner include, for example, gastrointestinal diseases, pulmonary infections, respiratory infections and infection with HIV. The pharmaceutical compositions are prepared by methods known to one of skill in the art. In general, the ubiquitin fusion protein is admixed with a carrier and other necessary diluents which are known in the art to aid in producing a product which is stable and administrable. Administration of the pharmaceutical composition can be accomplished by several means known to those of skill in the art. These include oral, intradermal, subcutaneous, intranasal, intravenous or intramuscular. [0050]
  • Conventional vaccination methods involve the administration of an epitope-containing protein. Recently, and alternative to conventional vaccination methods, referred to as DNA vaccination, has been developed. In this method, DNA encoding the epitope-containing protein is introduced into the cells of an organism. Within these cells, the epitope-containing protein is directly expressed. Direct expression of the ubiquitin fusion proteins of the present invention by endogenous cells of a vaccinated animal allows for the continual stimulation of humoral and cellular immune responses over an extended period of time. This is in contrast to standard immunization protocols whereby the vaccine is injected at a single site one or more times. Following injection, the vaccine is disseminated to lymphoid organs where a single immune response occurs. [0051]
  • Direct expression can be accomplished by introducing DNA constructs which encode the desired ubiquitin fusion protein into the cells of an animal. The constructs typically contain promoter elements and other transcriptional control elements which direct the expression of the ubiquitin fusion protein. Introduction of the DNA construct can be by any conventional means including direct injection. The preferred administration site is muscle tissue or tissues rich in antigen presenting cells. [0052]
  • The introduction of a ubiquitin fusion protein as described above can also induce a tolerizing effect on the humoral or cellular immune response in an animal. Tolerization occurs following delivery of the ubiquitin fusion proteins to T-cells. The induction of a tolerization response is useful, for example, in connection with the treatment of allergic or autoimmune disorders. Examples of epitopes which can be used in a therapeutic regimen designed to induce tolerization include the Fel d 1 peptides, which are the major allergens found in cat pelts. These peptides can be internally inserted, for example, and fused to a cleavable C-terminus of ubiquitin. Typically patients to be treated are dosed subcutaneously with the ubiquitin fusion proteins once per week for several weeks. However, dosing can also be done orally or intranasally over a similar length of time. The result is a reduction of the allergic and/or autoimmune responses. These ubiquitin fusion proteins can also be given orally. [0053]
  • The use of ubiquitin as a scaffold for the presentation and stimulation of immune responses also allows the stimulation and generation of anti-self responses. An example of a potentially valuable anti-self response is the generation of anti-GnRH (gonadotrophin releasing hormone) antibodies. As described in Example 3 below, efforts have been made to generate immunogens which stimulate a strong anti-GnRH response which results in the suppression of luteinizing hormone (LH) and follicle stimulating hormone (FSH) and indirectly suppresses the production of the steroidogenesis and gamete maturation in both males and females. The value of this type of anti-self response in humans lies in the treatment of prostate cancer and breast cancer. [0054]
  • In livestock and pets, the ability to stimulate an anti-self response provides a simple alternative to physical castration. Previous work employing complex immunogens has demonstrated varying degrees of success in immunological castration. However, the production of complex immunogens is cumbersome, typically involving a combination of synthetic chemistry, expensive HPLC purifications, and chemical coupling methods. The use of ubiquitin fusion proteins containing GnRH epitopes facilitates the production of inexpensive and potent immunogens for use in connection with immunocastration. In particular, ubiquitin fusions which include the peptide QHWSYGLRPGQHWSYGLRPGQHWSYGLRPGQHWSYGLRPGC are of interest. [0055]
  • Immunocastration of pigs is especially valuable since it does not result in the detrimental side-effects associated with physical castration. More specifically, physical castration of pigs typically results in animals which do not grow as well as normal animals. In addition, physically castrated pigs tend to have a higher fat percentage than non-castrated pigs. Since, immunocastrated animals are not castrated as early as those which undergo normal physical castration, a farmer can take advantage of the growth rates found with non-castrated animals. Finally, by immunocastrating, the farmer avoids the production of unpalatable meat found with uncastrated male pigs. [0056]
  • Other examples of self proteins which could be used with ubiquitin to generate vaccines able to modulate the hormones, cytokines and physiology of humans and animals are: growth hormone and its peptides to modulate growth both negatively and positively; TNF and its epitopes to modulate septic shock, arthritis, inflammatory bowel disease, crohn's disease, and ulcerative colitis; immunoglobulin epsilon heavy chain for the control of allergic reactions; chorionic gonadotrophin for fertility control; inhibit for fertility control; and Sperm proteins such as sp17 (for example amino acids 4-19 and 118-127) and the 71 kd sperm protein for control of fertility both in men and in women. [0057]
  • A further use of ubiquitin as a scaffold is as part of a vaccine to enhance the growth rate and thereby the final weight of livestock prior to shipment to market. This type of vaccine offers a cost effective means to increase the value of livestock such as pigs, cattle and other commonly raised animals. Presently, to increase the weight of livestock several methods are being utilized. Amongst these methods, the addition of antibiotics to the feed has become very common. However, while addition of antibiotics to feed is cost effective, it has limitations. For instance, it has been blamed for an increase in the creation of antibiotic resistant strains of bacteria. [0058]
  • An alternative means to increase the growth rate of livestock which does not result in detrimental side-effects is through vaccination of the animals with an epitope of growth hormone which is part of a ubiquitin fusion protein. The result of this vaccination is an increase in the activity of the animals endogenous growth hormone. The vaccine is created by inserting an epitope from the growth hormone protein (e.g., amino acids 54-95) into the ubiquitin protein thus creating a ubiquitin-growth hormone fusion protein. The effective use of this type of vaccine is described in Example 7 below. More specifically, following the injection of a vaccine comprised of adjuvant and a ubiquitin fusion protein containing the growth hormone protein epitope, the growth rate of pigs was improved when compared to control pigs which received either adjuvant only or adjuvant and ubiquitin only. [0059]
  • In addition to the uses described above, the ubiquitin fusion proteins of the present invention can be used for the identification of antibodies from experimental or clinical samples. Antibodies to be assayed can be found, for example, in blood, fecal material, the linings of mucosal associated lymphoid tissues, cellular biopsies and other sources known by one of skill in the art to contain at least a minute quantity of antibody. Assays for which the ubiquitin fusion proteins are well suited include ELISAs, radioimmunoassay as well as other commonly used competition assays. These types of assays are useful in identifying antibodies from an experimental or clinical sample which have specificity to at least one epitope of a known protein. In general, the assays involve mixing a predetermined aliquot of the ubiquitin fusion protein with a series of dilutions of the experimental or clinical antibody sample. This is followed by detection of the antibodies which bind to the ubiquitin fusion protein. [0060]
  • Detection can be accomplished by various means, but for the present invention, a labeled detection antibody is preferable. For example, if the experimental or clinical antibody sample is from a human, the detection antibody can be a polyclonal antibody with specificity for the human heavy chain portion of the sample antibody. The detection antibody is attached to either an enzymatic label, a radioactive label or a fluorochromatic label. Examples of commonly used enzymatic labels are horse radish peroxidase and alkaline phosphatase. A standard radioactive label includes iodine 131. Fluorochromatic labels include fluorescein and Texas red. The method of visualization of the complex containing the ubiquitin fusion protein, the sample antibody, and the labeled antibody depends on the label attached to the antibody and would be known to those of skill in the art.[0061]
  • EXAMPLES
  • No. 1 [0062]
  • Fusion proteins consisting of short-peptide sequences inserted within a ubiquitin scaffold were developed and tested as immunogens for eliciting a targeted immune response. Preparation and testing of the fusion proteins required several steps including: (I) the design and construction of plasmids encoding peptide sequences fused to the ubiquitin polypeptide at two permissive sites which could be useful for epitope display; (II) the high-level expression in [0063] E. coli, purification and characterization of the expected ubiquitin fusions; and (III) evaluation of the immune response in mice with regard to immunogenicity, antibody specificity, cross-reactivity and helper T cell response. Fusion proteins were developed to display the sequence of the HIV-1 gp120 V3 loop, the principal recognition determinant of virus neutralizing antibodies.
  • Materials and Methods [0064]
  • Mice [0065]
  • BALB/c, C57BL/6 and C3H/HeN female mice at 6-8 weeks of age were obtained from Charles River (Frederick, Md.) or Jackson Labs. Animals were housed and cared for in an IACUC supervised facility in accordance with an NIH approved Animal Welfare Assurance. [0066]
  • Peptides, Proteins and Antibodies [0067]
  • Bovine ubiquitin (Sigma, St. Louis Mo.) was dissolved in PBS and filtered through a 0.2 micron filter. Goat anti-mouse peroxidase conjugate was used as obtained from Promega. Mouse monoclonal antibodies against V3 peptides of gp120MN (HG-1) and gp1201IIB (# 13-105-100) were purchased from Biodesign International (Kennebunk, Me.) and from Advanced Biotechnologies Inc. (Guilford, Md.) respectively. Recombinant proteins gp1201IIB and gp120MN and a 36 residue “universal” V3 peptide CTRPNNNTRKSIHIGPGRAFYTTGEIIGDIRQAHC (SEQ ID NO:1) were obtained from Intracel Corporation (Cambridge, Mass.). Synthetic peptides KRIHIGPGRAFYTTK (L-V3) (SEQ ID NO:2) and CKSIHIGPGRAFYTTGC (C-V3) (SEQ ID NO:3) were obtained through the AIDS Research and Reference Reagent Program, Division of AIDS, NIAID, NIH: from Peptide & Protein Research Consultants. (Exeter, Devon, UK). [0068]
  • Oligonucleotide Design [0069]
  • Oligonucleotides were obtained by custom synthesis through Bioserve Biotechnologies. Sequences shown below were designed to encode peptide sequences and to provide appropriate complementary overhangs for ligation to restriction sites in plasmids. Oligonucleotides were treated with T4 polynucleotide kinase and complementary pairs were annealed by heating to 65EC for 5 minutes, then cooling: to room temperature over 30 minutes. [0070]
    (SEQ ID NO:4)
    A 5′-TTAAGACTGCGTGGCGGCGACCA
    GGTTCACTTCCAGCCGCTGCCGCCGGC-3′
    (SEQ ID NO:5)
    B 5′-TGTTGTTAAACTGTCTGACGCTC
    TGTAAGCTTCTGCA-3′
    (SEQ ID NO:6)
    C 5′-GAAGCTTACAGAGCGTCAGACAGTTTA
    ACAACAGCCGGCGGCA-3′
    (SEQ ID NO:7)
    D 5′-GCGGCTGGAAGTGAACCTGGTCGCC
    GCCACGCAGTC-3′
    (SEQ ID NO:8)
    E 5′-TTAAGACTGCGTGGCGCTGACCAGGTTCACT
    TCCAGCCGCTGCCGCCGGC-3′
    (SEQ ID NO:9)
    F 5′-GCGGCTGGAAGTGAACCTGGTCA
    GCGCCACGCAGTC-3′
    (SEQ ID NO:10)
    G 5′-AAGAAATCCACATCGGTCCGGGTCGTGCTTT
    CTACACCACCATCCCGCCGGATCA-3′
    (SEQ ID NO:11)
    H 5′-ATCCGGCGGGATGGTGGTGTAGAAAGCACGA
    CCCGGACCGATGTGGATTTCTTT-3′
    (SEQ ID NO:12)
    I 5′-TTAAGACTGCGTGGCGG
    CATCCACATCGGTCCG-3′
    (SEQ ID NO:13)
    J 5′-GGTCGTGCTTTCTACAC
    CACCTAACTGCA-3′
    (SEQ ID NO:14)
    K 5′-GTTAGGTGGTGTAGAAAGC
    ACGACCCGGACCGAT-3′
    (SEQ ID NO:15)
    L 5′-GTGGATGCCGCCACGCAGTC-3′
  • Strains and Vector Construction [0071]
  • Ubiquitin fusions were cloned and expressed in the Proteinix proprietary [0072] E. coli strain DH5α F′ IQ™ (Life Technologies). Vector constructs for expression of fusion proteins are derivatives of pDSUb (provided by M. Rechsteiner, Univ. of Utah), which is a derivative of pDS78/RBSII. The ubiquitin codon usage was optimized for expression in E. coli. Transformants containing pDSUb or its derivatives were selected for ampicillin resistance. Ubiquitin fusion expression, under control of the lac promoter, is inducible by addition of IPTG. Cloning at the ubiquitin codon 35 was initially performed in pRSETUb, derived from PRSET (Invitrogen, Inc.) by subcloning of the ubiquitin gene from pDSUb to PRSET utilizing the Nde I and Hind III restriction sites. Restriction sites for insertions at codon 35 of ubiquitin were created by in vitro mutagenesis with a synthetic oligonucleotide using the MORPH mutagenesis kit (5 Prime-3 Prime, Inc.) to obtain pPX153. Digested vector fragments were treated with calf intestinal alkaline phosphatase and purified from agarose gel slices after electrophoresis using the Geneclean kit (Bio 101). Vector fragments and double stranded oligos were ligated and products were used for cloning in E. coli strain DHSaF′ IQ™ (Life Technologies. All restriction digests, ligations and transformation of E. coli were done according to standard manipulations using commercial DNA modifying enzymes as instructed by the manufacturer (New England Biolabs). Correct cloning of the oligonucleotide insertions was confirmed by DNA sequencing using the Sequenase Version 2.0 kit (USB, Cleveland, Ohio).
  • Expression, Purification and Characterization of fusion Proteins [0073]
  • Cell paste from bacterial fermentation was resuspended in lysis buffer (5 ml/g wet paste) with vortexing. General purpose buffer consisted of 20 mM MES pH 5.5, supplemented with 1 mM PMSF, 0.5 mM ZnCl2. Cultures expressing certain fusion proteins were lysed in 50 mM acetate pH 4.0 (UbaMT) or pH 4.5 (UbgMT). Cells were disrupted by sonication on ice 2×4 min with a 50% duty cycle. Homogenates were centrifuged at 15,000×g, 45 min, 4EC. The supernatants were further diluted with 3 volumes of lysis buffer, filtered through a 0.45 Fm filter, and loaded onto a 30 mL SP sepharose HP ion exchange column at a linear flow rate of 1.4 cm/min. The fusion protein was eluted by a NaCl gradient (0-0.5 M) over 16 column volumes. Fractions were assayed by SDS-PAGE on 10-20% tricine gels (Novex, San Diego Calif.) and protein concentration was determined by the BCA method (Pierce Co.) using a ubiquitin standard curve. Peak fractions were pooled and evaluated further by C18 reversed phase HPLC on a Vydac 218TP54 column using a gradient of 30-45% acetonitrile in water containing 0.1% TFA over 7 min at 1.00 mL/min, with UV detector set at 214 nm. [0074]
  • Immunoblots were prepared by electroblotting of samples from SDS-PAGE gels onto Immobilon-P membrane (Millipore Corp.) using an X-Cell II Blot Module (Novex). The membranes were incubated in PBS containing 4% dry milk overnight and then washed with PBS. Incubations with primary antibody were done in PBS, 0.1% Tween 20 for 2 hr at room temperature. After 3 washes with the dilution buffer, membranes were incubated with goat anti-mouse IgG HRP conjugate at 1:5000 dilution in the same buffer. The wash steps were repeated and the membrane was developed with ECL Western detection reagents kit (Amersham, Waltham, Mass.) according to the manufacturer's instructions. [0075]
  • In Vitro cleavage of Ubicuitin Fusion Proteins by UBP [0076]
  • Reactions of UbV3gMT, UbgMT and UbaMT were monitored by C18 reverse phase HPLC on a Vydac 218TP54 column with detection at 214 nm using a 10 min gradienty of 20-50% acetonitrile in water containing 0.1% TFA, and by SDS-PAGE on 10-20% tricine gels. Aliquots of fusion protein (100-200 Fg) were diluted into 200 FL of 50 mM Tris pH 8.0, 5 mM DTT, 1 mM EDTA. A 1 Fl aliquot of UCH-L3 (2 ug) or reaction buffer (control) was added and the samples were incubated at 37EC and monitored over 1 hr by HPLC. SDS-PAGE samples were run after 2 hrs reaction time. [0077]
  • For large scale digests, purified UbgV3 from SP sepharose HP ion exchange was concentrated 3-fold by Centriplus 3 membrane concentrators. The final concentration of fusion protein was 2.1 mg/ml. The fusion protein in 12 ml of buffer was buffered with 631 Fl of 1 M Tris pH 8.0, 64.8 uL 1 M DTT, 24.0 uL 0.5 M EDTA. UCH-L3 (0.24 mg in 120 Fl) was added and the reaction was incubated at room temperature and monitored by HPLC. The product peptide peak was purified by semi-preparative C18 HPLC (Vydac 218TP510 1.0 cm diameter column) and lyophilized to yield the V3 peptide IHIGPGRAFYTT (SEQ ID NO:16). Identity was verified by FAB-MS. The MT peptide DQVHFQPLPPAVVKLSDAL (SEQ ID NO:17) was obtained by a similar procedure from ion exchange purified UbgMT (4.82 mg/ml). The peptide product was isolated from semi-preparative C18 HPLC using a 20-40% gradient of acetonitrile in 20 mM potassium phosphate, pH 6.0 over 8 min. The peptide content in the lyophilized product was estimated at 10% by weight based on HPLC peak area. [0078]
  • Immunization Protocols and Mouse Lymphocyte Proliferation Assay [0079]
  • Mice were inoculated in groups of 3 to 5 per antigen by an initial subcutaneous (s.c.) injection of 100 Fg of proteins in PBS emulsified in an equal volume of complete Freund's adjuvant (CFA) or incomplete Freund's adjuvant (IFA). Booster injections of proteins (100 Fg) were delivered intraperitoneal in IFA 3 weeks and 7 weeks later. Blood samples were collected at 4 weeks, 6 weeks, 8 weeks and 10 weeks from the initial injection. Serum was separated and diluted in PBS. [0080]
  • For preparation of sensitized T cells, mice were immunized in groups of 3 by injection s.c. in footpads with 50 Fg of proteins in PBS emulsified in IFA.d Eight to ten days later mice were sacrificed and popliteal lymph nodes (LN) were removed aseptically. LN cell suspensions in RPMI 1640 were prepared as described and dispensed into 96-well plates at 5×105 cells per well. Antigens or peptides were added in triplicate wells and plates were kept in a CO[0081] 2 incubator at 37EC for 4 days. Wells were then pulsed with 3H-deoxythymidine (1 FCi/well) and cells were harvested on filter pads 24 hours later using an automated collecting device. Filter sheets were allowed to dry and then counts were read on a Wallac microbeta plate reader (Wallac, Inc., Gaithersburg Md.). Stimulation was expressed as the average signal of duplicate or triplicate wells corrected for mean background counts (c.p.m. with protein or peptide—c.p.m. with buffer added).
  • Immunoassays [0082]
  • Fusion proteins (50 Fg/ml) or synthetic peptides (10 Fg/ml) in PBS were dispensed into immunosorbent 96-well plates (0.1 ml/well, Corning high binding flat bottom plates) and incubated for 1 hour at 37EC. Excess antigen was shaken out and nonspecific sites were blocked by addition of PBS containing 10 mg/ml of BSA (0.1 ml/well, incubated 30 min at 37EC). Plates were washed three times (Tris buffer 10 mM, 0.1% Tween 20, pH 8) and serially diluted serum samples (1/103-1/104 in PBS supplemented with 1% BSA) were added (0.1 ml/well). After 1 hour at 37EC plates were washed as before and developed with affinity purified goat anti-mouse IgG-horse radish peroxidase conjugate (0.5 Fg/ml in PBS supplemented with 1% BSA, 0.1 ml/well). After washing, bound enzyme was detected with o-phenylenediamine (1 mg/ml in phosphate-citrate buffer, 0.05M, 0.02% H[0083] 2O2, pH 5). Plates were read at 450 nm on a 96-well plate reader (Titertek). Titers are expressed as the dilution of serum giving an absorbance reading of 0.3 or 15% of the maximum reading.
  • Solution phase binding assays were performed by incubating peptides or proteins at concentrations ranging from 0.5-50 Fg/ml with antiserum in 0.1 M potassium phosphate, 2 mM EDTA, 10 mg/ml BSA, pH 7.8 at a fixed concentration of 2-fold greater than the previously determined titer dilution. Samples were incubated at 37EC for 2 hours, then applied to antigen-coated ELISA plates. The standard ELISA procedure was followed to determine the concentration of unbound antibody relative to samples containing no added ligand. [0084]
  • Results [0085]
  • Plasmid Construction and Sequences [0086]
  • Sequences of the oligonucleotides encoding for peptide inserts and their letter designations are given above. Linearized DNA from pDSUb restriction digested with Afl II and Pst I was ligated to double-stranded oligos A/D and B/C to obtain pDSUbgMT. Similar ligation to oligos E/F and B/C provided pDSUbaMT. Double stranded oligos G/H were ligated to the 1.2 kb and 1.6 kb fragments obtained from digest of pPX153 with restriction enzymes Xcm I and Bpm I to provide the V3 insertion at codon 35 of ubiquitin. [0087]
  • The new vector pRSETUbV3 was digested with Bgl II and Afl II, and the smaller fragment encoding UbV3 was gel purified. Ligation of this fragment to pDSUb, pDSUbgMT or pDSUbaMT, each digested with Bgl II and Afl II, generated expression vectors pDSUbV3, pDSUbV3gMT, and pDSUbV3aMT respectively. These were used to express the internal UbV3 fusion and double epitope fusions with a 19-residue C-terminal extension DQVHFQPLPPAVVKLSDAL (MT sequence)(SEQ ID NO:17) and either native (RGG) or mutant (RGA) protease recognition site at the ubiquitin C-terminus. A vector encoding ubiquitin fusion with a C-terminal 12-residue V3 sequence was assembled as follows: pDSUb was digested with Afl II and Pst I and the product ligated to paired oligos I/L and J/K to obtain pDSUbV3c. [0088]
  • Expression Yield and Purification [0089]
  • Fermentations in 10 L batches provided 150-175 g of wet cell paste. Fusion protein yields after a single ion exchange chromatography step ranged from 185 mg to 336 mg per 15 g of cell paste, equivalent to 1 L of culture. Product was isolated from 1-2 L shaker flask cultures (UbgV3) with similar results and yields. Fusion proteins UbgMT and UbaMT required chromatography with 50 mM acetic acid pH 4.0 and pH 4.5, respectively for retention on the ion exchange matrix. Ubiquitin fusions purified by ion exchange were greater than 98% pure as judged by SDS-PAGE. [0090]
  • In Vitro Cleavage of C-terminal Ubiquitin Fusions with UCH-L3 [0091]
  • Processing of ubiquitin fusion polypeptides by the ubiquitin C-terminal hydrolase UCH-L3 is presumed to depend on recognition of the native structure of the ubiquitin domain. Analytical digests of UbV3gMT and UbgMT by UCH-L3 proceeded with similar efficiency as shown by the HPLC and SDS-PAGE results. Cleavage of the UbV3gMT having an internal fusion in the ubiquitin sequence suggests that an insert in the 34-40 loop does not interfere with folding of ubiquitin or recognition by the enzyme. The reaction can be followed by the formation of peptide product detected by HPLC analysis. SDS-PAGE analysis is useful for determining completion by depletion of substrate and formation of a band corresponding in size with ubiquitin. [0092]
  • The enzymatic digest of ubiquitin fusions is a practical bioprocess for production of peptides. Two short peptides, 1HIGPGRAFYTT (SEQ ID NO:16) and DQVHFQPLPPAVVKLSDAL (SEQ ID NO:17), useful in this study, were conveniently prepared by processing of recombinant C-terminal ubiquitin fusions. [0093]
  • Binding and Specificity of Anti-V3 Deptide Antibodies to UbV3 [0094]
  • Antibodies that neutralize HIV-1 infectivity are directed against the V3 loop or principal neutralizing determinant (PND) of gp120. The PND is implicated in several viral mechanisms known to evade the humoral immune response. Subunit vaccines based on the native structure in gp120 indicate only weak immunogenicity toward this region. Variability of the V3 loop in divergent viral isolates and depletion of B-cells producing antibodies to the PND in infected individuals could also allow for HIV-1 escape from immune surveillance. The gp120 V3 loop has been studied in the context of hybrid protein scaffolds or peptide fusions as a means of producing immunogens that could induce neutralizing antibodies against HIV-1. These materials could be important as components of an effective vaccine for combating the virus. [0095]
  • Monoclonal antibodies raised against synthetic V3 peptides which are known to react with gp120 were tested for binding to the recombinant UbV3 by ELISA. The antibody HG-1 specific for gp120 from the HIV-1 strain MN bound to UbV3 and to a 36-residue synthetic V3 loop “universal” peptide with similar efficiency. The binding to UbV3 was inhibited by preincubating the antibody with the 36-residue V3 peptide. Absorbance was reduced by 50% in the presence of 0.5 Fg/ml peptide. Antibody specific for gp120 of the 111B strain did not bind-UbV3. [0096]
  • Ubiquitin fusion proteins analyzed by SDS-PAGE were transferred to nitrocellulose membranes for Western blot assays. Strong staining was seen when membranes were developed with mAb HG-1 which is specific for the V3 sequence of gp120 from HIV-1 strain MN. No staining occurred with the mAb derived against the V3 peptide of gp120 of strain IIB. [0097]
  • Immunogenicitv of Ubicuitin and Ubicuitin Fusion Proteins [0098]
  • Immune responses in mice were evaluated from antiserum titers and from comparison at fixed dilutions of antisera taken at different times after immunizations. Reactivity against ubiquitin, UbV3 and UbgMT proteins, displaying either native ubiquitin epitopes alone, the “V3” epitope at the internal site or the MT sequence at the C-terminal site were assessed by ELISA with the proteins adsorbed to the solid support. [0099]
  • In the BALB/c (H-2d) strain a strong immune response was observed after an initial inoculation and one boost of either UbV3, UbV3aMT, or UbV3gMT fusion proteins. In contrast ubiquitin, given by an analogous protocol, was a poor immunogen (Table 1). Mice receiving two or more injections of ubiquitin over 4 weeks generally showed decreasing anti-ubiquitin reactive antibody. Mice immunized with fusion proteins UbV3, UbV3aMT and UbV3gMT produced antiserum specific for UbV3. The anti-UbV3 response continued to increase after an additional boost. Average titers of the final sera collected were in excess of 1:105. The antibody in these sera was shown to be primarily IgG. The V3 insert was the dominant B-cell target in all three immunogens. The sera bound only weakly or not at all to native ubiquitin or the UbMT fusion. Animals receiving. UbgMT or UbaMT developed measurable antibody against UbgMT and ubiquitin. This response was directed partly to the C-terminal peptide as suggested by the higher ELISA signals obtained against the fusion protein compared with those against ubiquitin (Table 1). [0100]
    TABLE 1
    Summary of Immune Response Specificity to Antigens Displaying
    Heterologous and Native Ubiquitin Epitopes
    Strain: BALB/c Adjuvant: CFA, or
    PN222/IFA
    Routes: SQ, IP
    9 Immunogena // Antigenb6 Ub-V3 Ub-MT Ub
    Ub-V3-rgg-MT + + + +/−
    Ub-V3-rga-MTc + + + +/−
    Ub-V3-rgg + +
    Ub-rgg-MT +/− + + + +
    Ub-rga-MTc + + +
  • (a) Fusion proteins were purified from [0101] E. coli lysates and purified by ion exchange chromatography. Solutions in Tris buffer, pH 7.4 were emulsified with Freund's complete adjuvant and injected as described.
  • (b) Antigens used to coat microtiter plates for ELISA were prepared from recombinant [0102] E. coli extracts and purified by ion exchange. Three antigens used displayed a single (V3 or MT) or no (Ub) heterologous epitope. Immunoreactivity is indicated by the relative scale from no reaction (−) to strongest reaction++++). Borderline reactivity (signal at the highest concentration of antisera used) is scored as +/−.
  • (c) Proteins with C-terminal fusions were produced with the native (rgg) and a mutated (rga) ubiquitin C-terminal sequence to test for stability to or assisted processing of the T-cell epitope by endogenous ubiquitin C-terminal hydrolases that may be present in antigen processing cells. [0103]
  • Immune Responses to Cleavable and Noncleavable C-Terminal Fusions [0104]
  • Double epitope fusions had either the native ubiquitin C-terminus (UbV3gMT), retaining the processing site (RGG) for cleavage by cellular ubiquitin-specific proteases (UBPs), or a single residue mutation expressing the sequence RGA at the ubiquitin C-terminus (UbV3aMT), that is resistant to processing. Immunizations with the two types of double-epitope fusions were compared to determine if processing by UBPs in antigen presenting cells could influence a T-helper response. Since the antibody response to the V3 site was independent of the MT epitope in the BALB/c mouse, the effect of the processing mutation could not be observed in this strain. The immunogenicity in C57BL/6 (H-2b) and C3H (H-2k) mouse strains was determined after similar treatment. [0105]
  • In both strains a specific anti-UbV3 or UbV3gMT. Responses against UbV3aMT were comparable in the two mouse strains when evaluated after two injections given two weeks apart. [0106]
  • Immunoassays against ubiquitin and UbgMT, determined at 1/4000 serum dilution, indicated significantly lower antigenicity of these proteins. Signals were 16 and 35% of the values against UbV3 in the H-2b and H-2k mice, respectively. A second boost of the H-2b mice with double epitope fusion UbV3aMT produced improvements in the anti-UbV3 response similar to those seen in the BALB/c mice. The anti-UbV3 antiserum persisted at 3 weeks from the boost, although the signal at {fraction (1/4000)} dilution was diminished by about 50% from sera collected in the previous bleed. [0107]
  • Moreover, the anti-ubiquitin response was negligible in the mice that received the additional boosts. Immunizations with UbaMT produced antiserum of significant titer against all three antigens. A weaker, but similarly non-specific reaction was observed in mice inoculated with UbgMT. The anti-UbMT antisera reacted equivalently with native bovine ubiquitin and with recombinant ubiquitin but not with BSA. The anti-ubiquitin antibody persisted with additional immunizations. [0108]
  • Epitope Specificity of Antisera [0109]
  • Differences in antigenicity of the three proteins could suggest the epitope specificity of antisera for the V3 insert or the MT tag or specificity for discontinuous determinants composed of insert sequences as well as ubiquitin residues. Antiserum specific for UbV3 was incubated with short peptides analogous to the V3 sequence at varying concentrations, and residual unbound antibody measured by indirect ELISA. Reduction in signal was apparent in the 1-30 FM range of peptide concentration. By contrast, no competition was seen using ubiquitin at 10-100 FM as a competitor. [0110]
  • Anti-UbV3 sera reacted similarly with peptide L-V3 or a disulfide-bridged cyclic peptide C-V3 as mimics of the epitope. Although the peptide C-V3 could mimic the conformation of the V3 loop in the fusion protein this assay did not discriminate widely between the two peptides in solution. [0111]
  • Similar conformations of the V3 insert in the UbV3 and the V3 loop structure presented in gp120 could be suggested by better cross-reactivity of the antiserum with gp120. Adsorption of recombinant gp120 to ELISA plates appears to mask the V3 epitope such that detection of antibodies is inefficient. Binding to other epitopes on gp120 was detectable by direct or sandwich capture ELISA, but this technique was less successful with anti-V3 loop antibodies. Cross-reactivity of the anti-UbV3 antiserum for gp120 was demonstrated by Western blot analysis. Solution-phase competition binding as described for the V3 peptides using accessible concentrations of gp120 (# 0.8 FM) did not produce measurable changes in the indirect ELISA. [0112]
  • T-cell Proliferative Responses to Peptides and Ubicuitin Fusions [0113]
  • LN cells from BALB/c mice immunized with fusion proteins UbV3 or UbV3MT showed a proliferative response when cultured in the presence of UbV3 fusion. No obvious stimulation was provided by V3 or MT peptides at 100 Fg/ml, suggesting that the major T-helper epitope is not found in these sequences. No proliferation was observed in the presence of native bovine ubiquitin. In order to control for possible mitogenic contaminants contributed from the [0114] E. coli extracts, mice were immunized with recombinant ubiquitin or with buffer. LN cells from these mice were incubated in the presence of ubiquitin or UbV3. No proliferation was observed in any case. Potential T-helper epitopes in UbV3 could include sequences spanning the inserted residues and flanking ubiquitin sequences. Peptides representing these regions should be prepared and tested in the assay to address this point.
  • LN cells from C3H mice immunized with UbV3aMT were marginally stimulated by UbV3. However, incubation in the presence of increasing concentration of MT peptide, but not the V3 peptide, produced a significant proliferation signal. No stimulation was observed in the same experiment when C3H mice were immunized with UbV3. These preliminary experiments did not allow quantitation of the relative efficiency of T-cell stimulation by peptide and intact fusion proteins used as immunogen. Positive stimulation values were in the same range as those observed in the presence of concanavalin A added at 2-20 Fg/ml. [0115]
  • No. 2 [0116]
  • Results [0117]
  • Production of Ubicuitin Fusion Proteins [0118]
  • Ubiquitin has been used as a scaffold for displaying multiple epitopes. These ubiquitin fusion proteins have been successfully recognized by antibodies of the appropriate specificity and analyzed using Origen technology. An immediate technological application for these ubiquitin fusion proteins that display a structurally defined antigenic epitope which is known to be a target for neutralizing antibodies is in production of diagnostic antibodies for clinical or research use. The advantages conferred by the use of ubiquitin as a scaffold include: 1) its ability to stabilize the epitopes in storage, and in in vitro assays, and 2) its ability to constrain the peptide at one: or both ends, thereby conferring the appropriate conformation for: antibody recognition. [0119]
  • A commonly used sandwich immunoassay requires two distinct monoclonal antibodies against a single macromolecular analyte, in the present case the ubiquitin fusion proteins. These antibodies are directed to nonoverlapping epitopes on the ubiquitin fusion proteins, allowing the formation of a trimeric complex, or sandwich. Typically, many monoclonal antibodies must be screened in order to identify an appropriate pair for the immunoassay. The technology described here permits the production of monoclonal or polyclonal antibodies against structurally defined epitopes in a macromolecule. Thus, a pair of reagent antibodies for a sandwich immunoassay may be produced by design rather than by random screening. Such reagents may be employed in a wide variety of important immunoassays utilizing any number of formats that are standard in the industry. [0120]
  • Construction of Ubiquitin Fusion Proteins [0121]
  • In Example 2, several ubiquitin fusions were constructed. These ubiquitin fusions contain a peptide epitope loop derived from the human PSA (prostate specific antigen). As shown in Table 3 below, ubiquitin fusion proteins contained either single or double epitope insertions on the ubiquitin scaffold. Four different versions of an antigenic PSA peptide loop from human prostate specific antigen were inserted into the ubiquitin scaffold at amino acid position 35. In Table 3 below they are labeled as Fus 5, Fus 5M, Fus 7 and Fus 7M. Four C-terminal fusions were also constructed, both individually and in various combinations with the fusions inserted into the ubiquitin scaffold at amino acid position 35. The peptides joined to the C-terminus of the ubiquitin fusions are shown below in Table 3. They include ATNAT, FLAG, PSA conpep and Fus 7 conpep. The “PSA conpep” fusions contain a modification at amino acid 76 of the ubiquitin to prevent cleavage by a ubiquitin specific protease. These fusions were produced for testing by growth and isolation from bacterial cultures transformed with DNA constructs that encode the ubiquitin fusion proteins. The bacterial lysate from these transformed cultures is nearly homogeneous with respect to protein content. Thus, dilution was sufficient for the following experimentation. [0122]
    TABLE 2
    Single and Double Insertions on the Ubipuitin Scaffold
    Fus 5  92 AA 10.44 k pI 8.20 KE-DVCAQVHPQKVTKFMLC-IPP (SEQ ID NO:18)
    Fus 5M  90 AA 10.24 k pI 8.63 KE-DVCAQVHPQKVTKFMLC-MPP (SEQ ID NO:19)
    Fus 7  90 AA 10.22 k pI 8.81 KE-CAQVHPQKVTKFMLC-IPP (SEQ ID NO:20)
    Fus 7M  87 AA 98.93 k pI 9.26 KE-CAQVHPQKVTKFM-PP (SEQ ID NO:21)
    ATNAT 104 AA 11.63 k pI 9.65 RGG-SLRRSSCFGGRMDRIGAQSGLGCNSFRY (SEQ ID NO:22)
    FLAG  98 AA 11.22 k pI 6.12 RGG-DYKDD DDK (SEQ ID NO:23)
    PSA conpep (W/O FUS 7)  96 AA 10.99 k pI 9.71 RGA-LYTKVVHYRKWIKDTIVANP (SEQ ID NO:24)
    Fus 7 conpep 110 AA 12.65 k pI 9.67 RGA-LYTKVVHYRKWIKDTIVANP (SEQ ID NO:25)
  • Competition Analysis of Single Epitope Ubicuitin [0123]
  • The polyclonal antibody raised to a KLH conjugated peptide bound to the PSA peptide in the ubiquitin fusion. This PSA ubiquitin fusion was able to act like the native PSA by competing with native PSA for binding to this polyclonal antibody in an elisa assay. [0124]
  • In Vivo Cleavage of Double Epitope Ubicuitin Fusions [0125]
  • The structural authenticity of the ubiquitin scaffold in maintaining the secondary structure of the PSA peptide inserted internally at position 35 was probed by an in vivo assay. The assay involved the cleavage of the C-terminal PSA peptide from the double epitope ubiquitin fusion protein by a ubiquitin specific protease. Bacteria were transformed with DNA expression constructs for a double epitope ubiquitin fusion protein, UbFus7-conpep or UbFus7-ATNAT shown above in Table 3 and a ubiquitin containing plasmid. SDS-PAGE analysis of whole bacterial cell lysates showed that the UBFus7-ATNAT ubiquitin double epitope fusion protein was cleaved in vivo, liberating the C-terminal peptide from the main body of the ubiquitin fusion protein. Following the successful cleavage of the double epitope fusion UbFus7-ATNAT by in vivo ubiquitin specific proteases, the portion of the ubiquitin fusion protein which contained the internal insertion was analyzed. Based on this cleavage activity, it was deduced that a ubiquitin fusion protein with an internal insert would maintain its basic structure despite the 15 amino acid insertion of the PSA peptide at position 35. On the other hand, UbFus7-conpep fusion protein, which was modified at the C-terminus to prevent cleavage by a ubiquitin specific protease remained intact. No ubiquitin specific protease cleavage fragments were identified with UbFus7-conpep, as expected. [0126]
  • Direct Binding of Double Epitope Ubicuitin Fusion [0127]
  • The proof of concept that a double epitope ubiquitin fusion could be utilized in a sandwich assay was shown by two different direct-binding immunoassays using methods known to those skilled in the art. In one of these immunoassays, the quantity of the double epitope ubiquitin fusion protein UbFus7-Flag used in the immunoassay was kept constant, while the concentration of the anti-PSA specific polyclonal serum was diluted out from 1×10−2 to 1×10−5. The intensity of signal for the anti-PSA specific polyclonal serum for the ubiquitin fusion protein as measured by ECL (IGEN, Inc.) decreased in a linear manner as the quantity of antibody was diluted out. A similar result was found in a second direct-binding immunoassay. In this assay, the double epitope ubiquitin fusion protein was titered out from 10,000 ng/ml UbFus7-Flag to 0.1 ng/ml UbFus7-Flag, while the quantity of anti-PSA polyclonal serum was kept constant. As seen for the first experimental assay, the intensity of signal for the anti-PSA polyclonal serum as measured by ECL intensity decreased in a linear manner as the quantity of UbFus7-Flag was reduced. Thus, the anti-PSA polyclonal serum antibody showed a high degree of binding specificity for the double epitope ubiquitin fusions. This dual recognition supports the notion that the double epitope ubiquitin fusions can serve as calibrators in a sandwich immunoassay. [0128]
  • No. 3 [0129]
  • Results [0130]
  • Ubicuitin GnRH Immunogens [0131]
  • In order to generate an ubiquitin fusion protein which is able to stimulate the production of self antibodies, a fusion protein was constructed which contained a C-terminal extension to ubiquitin with the following sequence of the GnRH dimer; QHWSYGLRPGQHWSYGLRPG (SEQ ID NO:26) followed by a T cell epitope, DDPKTGQFLQQINAYARPSEV (corona virus T cell epitope) (SEQ ID NO:27) or DQVHFQPLPPAVVKLSDAL (MT epitope)(SEQ ID NO:17). This was constructed using standard methods known to one of skill in the art with sets of synthetic oligonucleotides. The ubiquitin used in these constructs was also modified so that its last amino acid was replaced by a valine to render the fusions noncleavable by ubiquitin-specific proteases. These ubiquitin fusions were expressed as described in Example 1 above and purified by ion exchange chromatography and HPLC (when necessary) following standard protocols. The ubiquitin fusion protein were purified to greater than 90% purity. [0132]
  • The purified ubiquitin fusion proteins were then formulated with adjuvants as described in Example 1 above and used to immunize mice. The mice were re-immunized about 0.25-30 days following the initial immunization. Sera prepared following bleeds from the mice were tested to determine the level of epitope specific antibodies which were induced by the specific ubiquitin fusion proteins. The results demonstrated that the immunizations resulted in the induction of high levels of anti-GnRH antibodies. [0133]
  • Other immunogens which were constructed include ubiquitin with an internal GnRH epitope as a single and as a double epitope at position 35 of ubiquitin and T cell epitopes attached at the C-terminus as described above. To further increase the epitope density, the ubiquitin fusion protein with the internal GnRH dimer at position 35 is also fused at its C-terminus with a dimer of GnRH followed by a T cell epitope or with MT at the C-terminus followed by GnRH. [0134]
  • In further variations of the ubiquitin fusion protein design which have been described above, the T cell epitope was attached at the C-terminus of ubiquitin which is then fused to the dimer or monomer sequence of GnRH. [0135]
  • The GnRH monomer can consist of EHWSYGLRPG (SEQ ID NO:28) with a corresponding dimer of EHWSYGLRPGEHWSYGLRPG (SEQ ID NO:29) or a mixed dimer of EHWSYGLRPGQHWSYGLRPG (SEQ ID NO:30) or QHWSYGLRPGEHWSYGLRPG(SEQ ID NO:31). Alternatively, different GnRH monomers could be used provided such monomers are able to induce antibodies to GNRH. [0136]
  • No. 4 [0137]
  • N-terminal Fusions of GnRH to Ubicuitin for Immunocastration [0138]
  • Novel constructs are prepared by placing an epitope at the N-terminus of a first ubiquitin protein to create a fusion protein which can elicit a desired immune response. These constructs differ from those in the prior art by allowing placement of any epitope at the N-terminus of the first ubiquitin protein in order to produce an effective vaccine conjugate. [0139]
  • One use for this type of novel N-terminal epitope presentation is in the generation of an anti-self antibody response. Expression vectors capable of this include those generated for immunocastration. These vectors are based on the vaccine constructs described above in Example 3. However, in the present example, the vaccine constructs include a N terminal ubiquitin protein fused to the N-terminus of the epitope attached to the N-terminus of the C terminal ubiquitin protein. Linkage of the N terminal ubiquitin protein to the N-terminal epitope is through a ubiquitin specific cleavable C-terminus. For example, in the case of GnRH, the sequence coding for (QHWSYGLRPG)[0140] n (SEQ ID NO:32), where n is from 1-8, is fused to the 3′ end of the second ubiquitin protein using synthetic oligonucleotides and methods known to one of skill in the art. The resultant gene sequence contains a fusion protein comprised of an epitope flanked on its C-terminus by a C terminal ubiquitin protein and on its N-terminus by a N terminal ubiquitin protein. The ubiquitin proteins joined to GnRH can be used to generate a number of possible combinations of fusion proteins containing multiple GRIRH sequences and T cell epitopes.
  • The fusion between the N terminal ubiquitin protein and an N-terminal epitope can occur via an RGG native ubiquitin C-terminus and a Q at the N-terminus of the GnRH sequence. In this example, the GnRH epitope sequence is comprised of from 1 to 8 copies of the following sequence QHWSYGLRPG (SEQ ID NO:32). The fusion protein comprising the GnRH epitope flanked on both its N- and C-terminal ends may be fused to at least one T cell epitope such as DQVHFQPLPPAVVKLSDAL (SEQ ID NO:33) at its C-terminus via a non-native ubiquitin C-terminal sequence such as RGV to render it non-cleavable by the ubiquitin specific proteases. Variations on the basic construct described above are made using different C terminal ubiquitin fusion proteins. Examples of these can be found in Example 3 above. For instance, the C terminal ubiquitin protein can be further modified to include GnRH epitopes (from 1 to 8 epitopes) inserted at position 35. In addition, the T cell epitope(s) fused to the C-terminus of the C terminal ubiquitin protein can be varied. [0141]
  • To prepare the N-terminal epitope ubiquitin fusion proteins, these gene constructs are placed within the expression vector described in Example 1 and used to transform [0142] E. coli for protein expression. The expressed protein is isolated from the E. coli cells by sonication followed by ion exchange purification to give a preparation which can then be subjected to the action of a ubiquitin specific protease (UBP). Digestion of the fusion protein with the UBP results in the release of the N-terminal ubiquitin protein from the N-terminus of the N-terminally fused epitope. This cleavage reaction is then subjected to a further ion exchange purification to yield a fusion protein with a GnRH epitope(s) fused to the N-terminus of the COOH terminal ubiquitin protein. The purified ubiquitin fusion protein, with its N-terminal and or C-terminal epitopes, can now be formulated to generate the vaccine for study as described in Example 6.
  • No. 5 [0143]
  • Conjugation of Ubiquitin GnRH Fusion Proteins to carrier Proteins [0144]
  • Ubiquitin fusion proteins containing peptide epitopes can be efficiently coupled directly to another protein. In the present example, two ubiquitin-GnRH fusion proteins are created which are site specifically coupled to ovalbumin. These ubiquitin-GnRH fusion proteins are-constructed using synthetic oligonucleotides, which encode the GnRH sequence; QHWSYGLRPGQHWSYGLRPGQHWSYGLRPGQHWSYGLRPGC (SEQ ID NO:34) for one construct and QHWSYGLRPGQHWSYGLRPGQHWSYGLRPGQHWSYGLRPG (SEQ ID NO:35) for the second construct. [0145]
  • These oligonucleotides are cloned into the UBP-cleavable C-terminal site in the coding sequence of ubiquitin as described above in Example 1. The resulting fusion constructs consist of the coding sequence for ubiquitin fused to four copies of the GnRH epitope, with both containing either a C-terminal Cys or not. To regulate expression, the coding sequence was placed under the control of a lac promoter which when induced elicits high levels of expressed fusion protein. The resulting construct is used to transform [0146] E. coli as described above in Example 1 and the cells were cultured followed by induction of expression of the fusion protein. The fusion protein was isolated from the E. coli cell pellet by first subjecting the cells to sonication, followed by purification of the fusion protein by ion exchange chromatography.
  • Conjugation of Ubicuitin Fusions with the C terminal Cys [0147]
  • Ovalbumin is activated by reaction with N-succinimidyl-3-(2-pyridyldithio)-propionate (SPDP) or by succinimidyl 4-(N-maleimido-methyl) cyclohexane-1-carboxylate (SMCC) followed by gel filtration to remove unreacted cross linking agents. Ubiquitin fusion protein is coupled to ovalbumin by reacting it with activated ovalbumin. The resultant reaction between a free SH group present on the ubiquitin fusion protein and the activated ovalbumin results in a covalent linkage through either a thiol ether linkage with the SMCC activated ovalbumin or a disulfide bond from the reaction with the SPDP activated ovalbumin. The resultant conjugate is formulated with an adjuvant such as Quil-A, complete Freunds adjuvant (CFA) or incomplete Freunds adjuvant (IFA) and then used for immunization as described in Example 6 below. [0148]
  • Conjugation of Ubicuitin Fusions Without the C Terminal Cys [0149]
  • The ubiquitin fusion protein is coupled to ovalbumin by reacting it with ovalbumin in the presence of a cross linking agent such as disuccinimidyl suberate or gluteraldehyde (Pierce). The resultant reaction the ubiquitin fusion protein and ovalbumin results in a covalent linkage through amino groups on the ubiquitin fusion protein and ovalbumin. The resultant conjugate is formulated with an adjuvant such as Quil-A, complete Freunds adjuvant (CFA) or incomplete Freunds adjuvant (IFA) and then used for immunization as described in Example 6 below. [0150]
  • No. 6 [0151]
  • Immunocastration of Pigs with Ubicuitin GnRH Immunogens [0152]
  • The ubiquitin fusion protein immunogens constructed as described above in Examples 3, 4 and 5 were tested in piglets. Male piglets which were between the age of 9-10 weeks were immunized with 1-10 mg of the ubiquitin GnRH immunogens in complete Freund's adjuvant (CFA) intramuscularly. Immunizations were repeated 8 weeks following the initial CFA immunization, with IFA. The piglets were slaughtered 16 weeks after the initial immunization at which time the testicles were excised and weighed. In addition, the serum testosterone levels of the piglets was determined along with the androstenone levels in fat. All the animals immunized with the ubiquitin GnRH immunogens showed significant reduction in testicular weight, along with significantly reduced levels of testosterone in the serum. The levels of androstenone in fat were below 0.1 Fg/ml. These experiments have demonstrated the potential of the ubiquitin fusion proteins to act as an immunogen for the generation of self immune responses known more specifically as immunocastration. [0153]
  • No. 7 [0154]
  • Growth Hormone Vaccine to Enhance the Growth Rate [0155]
  • In order to improve the growth rate of pigs, a vaccine was constructed which included the insertion of a growth hormone epitpope into ubiquitin. Prior studies have shown that the epitope encoded by amino acids 54-95 of growth hormone can be used to make a vaccine which improves the growth rate of pigs by increasing the activity of the endogenous growth hormone. By using ubiquitin fusion proteins containing this growth hormone epitope, novel vaccines can be generated which offer the advantages of enhanced hormone activity and lower costs. [0156]
  • In the present example, the growth hormone epitope was inserted into ubiquitin as described in Examples 3, 4 and 5 above, with the growth hormone epitope inserted at the same sites described above for the GnRH epitopes. The growth hormone epitope can be inserted as a multimer, with up to four contiguous repeats to enhance its immunogenicity. Constructs encoding the growth hormone-ubiquitin fusion proteins were transformed into and expressed in [0157] E. coli, followed by purification of the fusion protein by methods described above.
  • The purified growth hormone-ubiquitin fusion proteins were formulated with adjuvant and used to immunize pigs weighing 15-20 kg. CFA was used for the first immunization followed by two subsequent booster injections with IPA. These subsequent booster injections were each given at 4 week intervals following the initial injection. Pigs were monitored until they reached a weight of 110-120 Kg at which time the animals were killed. The resultant weight gain by immunized pigs when compared to control animals receiving only adjuvant or ubiquitin only and adjuvant demonstrated the improved growth rates of the immunized pigs. [0158]
  • 1 35 1 35 PRT Artificial Sequence Description of Artificial Sequence cloning oligo 1 Cys Thr Arg Pro Asn Asn Asn Thr Arg Lys Ser Ile His Ile Gly Pro 1 5 10 15 Gly Arg Ala Phe Tyr Thr Thr Gly Glu Ile Ile Gly Asp Ile Arg Gln 20 25 30 Ala His Cys 35 2 15 PRT Artificial Sequence Description of Artificial Sequence cloning oligo 2 Lys Arg Ile His Ile Gly Pro Gly Arg Ala Phe Tyr Thr Thr Lys 1 5 10 15 3 17 PRT Artificial Sequence Description of Artificial Sequence cloning oligo 3 Cys Lys Ser Ile His Ile Gly Pro Gly Arg Ala Phe Tyr Thr Thr Gly 1 5 10 15 Cys 4 50 DNA Artificial Sequence Description of Artificial Sequence cloning oligo 4 ttaagactgc gtggcggcga ccaggttcac ttccagccgc tgccgccggc 50 5 37 DNA Artificial Sequence Description of Artificial Sequence cloning oligo 5 tgttgttaaa ctgtctgacg ctctgtaagc ttctgca 37 6 43 DNA Artificial Sequence Description of Artificial Sequence cloning oligo 6 gaagcttaca gagcgtcaga cagtttaaca acagccggcg gca 43 7 36 DNA Artificial Sequence Description of Artificial Sequence cloning oligo 7 gcggctggaa gtgaacctgg tcgccgccac gcagtc 36 8 50 DNA Artificial Sequence Description of Artificial Sequence cloning oligo 8 ttaagactgc gtggcgctga ccaggttcac ttccagccgc tgccgccggc 50 9 36 DNA Artificial Sequence Description of Artificial Sequence cloning oligo 9 gcggctggaa gtgaacctgg tcagcgccac gcagtc 36 10 55 DNA Artificial Sequence Description of Artificial Sequence cloning oligo 10 aagaaatcca catcggtccg ggtcgtgctt tctacaccac catcccgccg gatca 55 11 54 DNA Artificial Sequence Description of Artificial Sequence cloning oligo 11 atccggcggg atggtggtgt agaaagcacg acccggaccg atgtggattt cttt 54 12 33 DNA Artificial Sequence Description of Artificial Sequence cloning oligo 12 ttaagactgc gtggcggcat ccacatcggt ccg 33 13 29 DNA Artificial Sequence Description of Artificial Sequence cloning oligo 13 ggtcgtgctt tctacaccac ctaactgca 29 14 34 DNA Artificial Sequence Description of Artificial Sequence cloning oligo 14 gttaggtggt gtagaaagca cgacccggac cgat 34 15 20 DNA Artificial Sequence Description of Artificial Sequence cloning oligo 15 gtggatgccg ccacgcagtc 20 16 12 PRT HIV-1 16 Ile His Ile Gly Pro Gly Arg Ala Phe Tyr Thr Thr 1 5 10 17 19 PRT Mycobacterium tuberculosis 17 Asp Gln Val His Phe Gln Pro Leu Pro Pro Ala Val Val Lys Leu Ser 1 5 10 15 Asp Ala Leu 18 22 PRT Homo sapiens 18 Lys Glu Asp Val Cys Ala Gln Val His Pro Gln Lys Val Thr Lys Phe 1 5 10 15 Met Leu Cys Ile Pro Pro 20 19 22 PRT Homo sapiens 19 Lys Glu Asp Val Cys Ala Gln Val His Pro Gln Lys Val Thr Lys Phe 1 5 10 15 Met Leu Cys Met Pro Pro 20 20 20 PRT Homo sapiens 20 Lys Glu Cys Ala Gln Val His Pro Gln Lys Val Thr Lys Phe Met Leu 1 5 10 15 Cys Ile Pro Pro 20 21 17 PRT Homo sapiens 21 Lys Glu Cys Ala Gln Val His Pro Gln Lys Val Thr Lys Phe Met Pro 1 5 10 15 Pro 22 31 PRT Homo sapiens 22 Arg Gly Gly Ser Leu Arg Arg Ser Ser Cys Phe Gly Gly Arg Met Asp 1 5 10 15 Arg Ile Gly Ala Gln Ser Gly Leu Gly Cys Asn Ser Phe Arg Tyr 20 25 30 23 11 PRT Homo sapiens 23 Arg Gly Gly Asp Tyr Lys Asp Asp Asp Asp Lys 1 5 10 24 23 PRT Homo sapiens 24 Arg Gly Ala Leu Tyr Thr Lys Val Val His Tyr Arg Lys Trp Ile Lys 1 5 10 15 Asp Thr Ile Val Ala Asn Pro 20 25 25 000 26 20 PRT Porcine 26 Gln His Trp Ser Tyr Gly Leu Arg Pro Gly Gln His Trp Ser Tyr Gly 1 5 10 15 Leu Arg Pro Gly 20 27 21 PRT corona virus 27 Asp Asp Pro Lys Thr Gly Gln Phe Leu Gln Gln Ile Asn Ala Tyr Ala 1 5 10 15 Arg Pro Ser Glu Val 20 28 10 PRT Porcine 28 Glu His Trp Ser Tyr Gly Leu Arg Pro Gly 1 5 10 29 20 PRT Porcine 29 Glu His Trp Ser Tyr Gly Leu Arg Pro Gly Glu His Trp Ser Tyr Gly 1 5 10 15 Leu Arg Pro Gly 20 30 20 PRT Porcine 30 Glu His Trp Ser Tyr Gly Leu Arg Pro Gly Gln His Trp Ser Tyr Gly 1 5 10 15 Leu Arg Pro Gly 20 31 20 PRT Porcine 31 Gln His Trp Ser Tyr Gly Leu Arg Pro Gly Glu His Trp Ser Tyr Gly 1 5 10 15 Leu Arg Pro Gly 20 32 10 PRT Porcine 32 Gln His Trp Ser Tyr Gly Leu Arg Pro Gly 1 5 10 33 33 000 34 41 PRT Porcine 34 Gln His Trp Ser Tyr Gly Leu Arg Pro Gly Gln His Trp Ser Tyr Gly 1 5 10 15 Leu Arg Pro Gly Gln His Trp Ser Tyr Gly Leu Arg Pro Gly Gln His 20 25 30 Trp Ser Tyr Gly Leu Arg Pro Gly Cys 35 40 35 40 PRT Porcine 35 Gln His Trp Ser Tyr Gly Leu Arg Pro Gly Gln His Trp Ser Tyr Gly 1 5 10 15 Leu Arg Pro Gly Gln His Trp Ser Tyr Gly Leu Arg Pro Gly Gln His 20 25 30 Trp Ser Tyr Gly Leu Arg Pro Gly 35 40

Claims (100)

1. A fusion protein comprising a heat shock protein fused to a single epitope-containing segment, the epitope-containing segment comprising two or more identical epitopes.
2. The fusion protein of claim 1 wherein the heat shock protein is ubiquitin and the fusion protein is a ubiquitin fusion protein.
3. The ubiquitin fusion protein of claim 2 wherein the epitope-containing segment is fused to ubiquitin at a fusion site selected from the group consisting of the N-terminus, the C-terminus and an internal fusion site.
4. The ubiquitin fusion protein of claim 2 wherein the N-terminal residue of ubiquitin is a residue other than methionine, and the N-terminal residue other than methionine is fused to the C-terminal residue of a second, unmodified ubiquitin protein.
5. The ubiquitin fusion protein of claim 2 wherein the N-terminal residue of ubiquitin is a residue other than methionine, and the N-terminal residue other than methionine is fused to the C-terminal residue of a C-terminal ubiquitin subdomain competent to specify cleavage by a ubiquitin-specific protease between the C-terminal residue of the C-terminal ubiquitin subdomain and the N-terminal residue other than methionine.
6. The ubiquitin fusion protein of claim 5 wherein at least one epitope-containing segment is positioned between the C-terminal residue of the C-terminal ubiquitin subdomain and the N-terminal residue other than methionine, and the C-terminus of the C-terminal subdomain is modified to inhibit cleavage by a ubiquitin-specific protease.
7. The ubiquitin fusion protein of claim 2 which is post-translationally modified by the addition of fatty acids to enhance immunogenicity.
8. The ubiquitin fusion protein of claim 2 wherein the epitope-containing segment contains from about 2 to about 30 epitopes.
9. The ubiquitin fusion protein of claim 2 wherein the identical epitopes are B cell epitopes.
10. The ubiquitin fusion protein of claim 2 wherein the identical epitopes are T cell epitopes.
11. The ubiquitin fusion protein of claim 2 wherein the identical epitopes are structural mimics of biomolecules.
12. The ubiquitin fusion protein of claim 2 wherein the identical epitopes are microbial epitopes.
13. The ubiquitin fusion protein of claim 2 wherein the identical epitopes are self epitopes.
14. The ubiquitin fusion protein of claim 2 wherein the identical epitopes represent epitopes from the proteins selected from the group consisting of gonadotropin releasing hormone, tumor necrosis factor, immunoglobulins, human immunodeficiency virus proteins, chorionic gonadotrophin, inhibin, growth hormones and sperm proteins.
15. The ubiquitin fusion protein of claim 2 wherein the identical epitopes which comprise the plurality of identical epitopes are gonadotropin releasing hormone epitopes.
16. The ubiquitin fusion protein of claim 2 wherein internal fusion sites comprise regions of ubiquitin linking two domain of secondary structure, the two domains of secondary structure being selected from the group consisting β-strand and α-helix.
17. The ubiquitin fusion protein of claim 2 wherein the epitope-containing segment is fused to the C-terminus of ubiquitin and the C-terminus of ubiquitin is modified to inhibit cleavage of the ubiquitin fusion protein by a ubiquitin-specific protease.
18. The ubiquitin fusion protein of claim 17 wherein the C-terminus of ubiquitin is modified at amino acid 76.
19. The ubiquitin fusion protein of claim 18 wherein the modification at amino acid 76 of ubiquitin is a substitution of an amino acid selected from the group consisting of: alanine, valine, and cysteine for the wild-type glycine amino acid residue.
20. A fusion protein comprising a heat shock protein fused to two or more non-contiguous epitope-containing segments, each epitope-containing segment comprising one or more identical or non-identical epitopes.
21. The fusion protein of claim 20 wherein the heat shock protein is ubiquitin and the fusion protein is a ubiquitin fusion protein.
22. The ubiquitin fusion protein of claim 21 wherein the non-contiguous epitope-containing segments are fused to ubiquitin at fusion sites selected from the group consisting of the N-terminus, the C-terminus and internal fusion sites.
23. The ubiquitin fusion protein of claim 21 wherein the N-terminal residue of ubiquitin is a residue other than methionine, and the N-terminal residue other than methionine is fused to the C-terminal residue of a second, unmodified ubiquitin protein.
24. The ubiquitin fusion protein of claim 21 wherein the N-terminal residue of ubiquitin is a residue other than methionine, and the N-terminal residue other than methionine is fused to the C-terminal residue of a C-terminal ubiquitin subdomain competent to specify cleavage by a ubiquitin-specific protease between the C-terminal residue of the C-terminal ubiquitin subdomain and the N-terminal residue other than methionine.
25. The ubiquitin fusion protein of claim 24 wherein at least one epitope-containing segment is positioned between the C-terminal residue of the C-terminal ubiquitin subdomain and the N-terminal residue other than methionine, and the C-terminus of the C-terminal subdomain is modified to inhibit cleavage by a ubiquitin-specific protease.
26. The ubiquitin fusion protein of claim 21 which is post-translationally modified by the addition of fatty acids to enhance immunogenicity.
27. The ubiquitin fusion protein of claim 21 wherein the epitope-containing segments contain from about 1 to about 30 epitopes.
28. The ubiquitin fusion protein of claim 21 wherein one epitope-containing segment contains at least one B cell epitope and one T cell epitope.
29. The ubiquitin fusion protein of claim 21 wherein one epitope-containing segments contains at least two B cell epitopes.
30. The ubiquitin fusion protein of claim 21 wherein one epitope-containing segments contains at least two T cell epitopes.
31. The ubiquitin fusion protein of claim 21 wherein one or more epitopes contained within the epitope-containing segments are structural mimics of biomolecules.
32. The ubiquitin fusion protein of claim 21 wherein one or more epitopes contained within the epitope-containing segments are microbial epitopes.
33. The ubiquitin fusion protein of claim 21 wherein one or more epitopes contained within the epitope-containing segments are self epitopes.
34. The ubiquitin fusion protein of claim 21 wherein one or more epitopes contained within the epitope-containing segments represent epitopes from the group of proteins selected from the group consisting of gonadotropin releasing hormone, tumor necrosis factor, immunoglobulins, human immunodeficiency proteins, chorionic gonadotrophin, inhibin, growth hormones and sperm proteins.
35. The ubiquitin fusion protein of claim 21 wherein one or more epitopes contained within the epitope-containing segments represent epitopes from gonadotropin releasing hormone.
36. The ubiquitin fusion protein of claim 21 wherein the internal fusion sites comprise regions of ubiquitin linking two domain of secondary structure, the two domains of secondary structure being selected from the group consisting β-strand and α-helix.
37. The ubiquitin fusion protein of claim 21 wherein one epitope-containing segment comprises a single B-cell epitope or a plurality of identical or non-identical B-cell epitopes and a second epitope-containing segment comprises a single T-cell epitope or a plurality of identical or non-identical T-cell epitopes.
38. The ubiquitin fusion protein of claim 21 wherein at least one epitope-containing segment is fused to the C-terminus of ubiquitin and the C-terminus of ubiquitin is modified to inhibit cleavage of the ubiquitin fusion protein by a ubiquitin-specific protease.
39. The ubiquitin fusion protein of claim 38 wherein the C-terminus of ubiquitin is modified at amino acid 76.
40. The ubiquitin fusion protein of claim 21 wherein the modification at amino acid 76 of ubiquitin is a substitution of an amino acid selected from the group consisting of: alanine, valine, and cysteine for the wild-type glycine amino acid residue.
41. A fusion protein comprising a heat shock protein fused to a single epitope-containing segment comprising two or more identical or non-identical epitopes, the epitope-containing segments being fused to the heat shock protein at fusion sites selected from the group consisting of the N-terminus and an internal fusion site.
42. The fusion protein of claim 41 wherein the heat shock protein is ubiquitin and the fusion protein is a ubiquitin fusion protein.
43. The ubiquitin fusion protein of claim 42 wherein single epitope containing segment contains from about 2 to about 30 identical or non-identical epitopes.
44. The ubiquitin fusion protein of claim 42 wherein the N-terminal residue of ubiquitin is a residue other than methionine, and the N-terminal residue other than methionine is fused to the C-terminal residue of a second, unmodified ubiquitin protein.
45. The ubiquitin fusion protein of claim 42 wherein the N-terminal residue of ubiquitin is a residue other than methionine, and the N-terminal residue other than methionine is fused to the C-terminal residue of a C-terminal ubiquitin subdomain competent to specify cleavage by a ubiquitin-specific protease between the C-terminal residue of the C-terminal ubiquitin subdomain and the N-terminal residue other than methionine.
46. The ubiquitin fusion protein of claim 45 wherein at least one epitope-containing segment is positioned between the C-terminal residue of the C-terminal ubiquitin subdomain and the N-terminal residue other than methionine, and the C-terminus of the C-terminal subdomain is modified to inhibit cleavage by a ubiquitin-specific protease.
47. The ubiquitin fusion protein of claim 42 which is post-translationally modified by the addition of fatty acids to enhance immunogenicity.
48. The ubiquitin fusion protein of claim 42 wherein the epitope-containing segment contains at least one B cell and one T cell epitope.
49. The ubiquitin fusion protein of claim 42 wherein the epitope-containing segment contains at least two B cell epitopes.
50. The ubiquitin fusion protein of claim 42 wherein the epitope-containing segment contains at least two T cell epitopes.
51. The ubiquitin fusion protein of claim 42 wherein the epitope-containing segment contains epitopes which are structural mimics of biomolecules.
52. The ubiquitin fusion protein of claim 42 wherein the epitope-containing segment contains epitopes which are microbial epitopes.
53. The ubiquitin fusion protein of claim 42 wherein the epitope-containing segment contains epitopes which are self epitopes.
54. The ubiquitin fusion protein of claim 42 wherein the epitope-containing segment contains at least one epitope from proteins selected from the group consisting of gonadotrophin releasing hormone, tumor necrosis factor, immunoglobulins, human immunodeficiency virus proteins, chorionic gonadotrophin, inhibin, growth hormones and sperm proteins.
55. The ubiquitin fusion protein of claim 42 wherein the epitope-containing segment contains epitopes from gonadotropin releasing hormone.
56. The ubiquitin fusion protein of claim 42 wherein the internal fusion site comprises a region of ubiquitin linking two regions of secondary structure selected from the group consisting of β-strand and α-helix.
57. The ubiquitin fusion protein of claim 42 wherein the epitope-containing segment contains a single B-cell epitope-or a plurality of identical or non-identical B-cell epitopes and a second epitope-containing segment comprises a single T-cell epitope or a plurality of identical or non-identical T-cell epitopes.
58. A fusion protein comprising a heat shock protein fused to a single epitope-containing segment comprising one or more identical or non-identical epitopes, the epitope-containing segment being fused to the heat shock protein at the N-terminus of the heat shock protein.
59. The fusion protein of claim 58 wherein the heat shock protein is ubiquitin and the fusion protein is a ubiquitin fusion protein.
60. The ubiquitin fusion protein of claim 59 wherein the epitope-containing segment contains from about 1 to about 30 epitopes.
61. The ubiquitin fusion protein of claim 59 wherein the N-terminal residue of ubiquitin is a residue other than methionine, and the N-terminal residue other than methionine is fused to the C-terminal residue of a second, unmodified ubiquitin protein.
62. The ubiquitin fusion protein of claim 59 wherein the N-terminal residue of ubiquitin is a residue other than methionine, and the N-terminal residue other than methionine is fused to the C-terminal residue of a C-terminal ubiquitin subdomain competent to specify cleavage by a ubiquitin-specific protease between the C-terminal residue of the C-terminal ubiquitin subdomain and the N-terminal residue other than methionine.
63. The ubiquitin fusion protein of claim 62 wherein at least one epitope-containing segment is positioned between the C-terminal residue of the C-terminal ubiquitin subdomain and the N-terminal residue other than methionine, and the C-terminus of the C-terminal subdomain is modified to inhibit cleavage by a ubiquitin-specific protease.
64. The ubiquitin fusion protein of claim 59 which is post-translationally modified by the addition of fatty acids to enhance immunogenicity.
65. The ubiquitin fusion protein of claim 59 wherein the epitope-containing segment contains at least one B cell epitope and one T cell epitope.
66. The ubiquitin fusion protein of claim 59 wherein the epitope-containing segment contains at least two B cell epitopes.
67. The ubiquitin fusion protein of claim 59 wherein the epitope-containing segment contains at least two T cell epitopes.
68. The ubiquitin fusion protein of claim 59 wherein one or more epitopes contained within the epitope-containing segment is a structural mimic of a biomolecule.
69. The ubiquitin fusion protein of claim 59 wherein one or more epitopes contained within the epitope-containing segment is a microbial epitope.
70. The ubiquitin fusion protein of claim 59 wherein one or more epitopes contained within the epitope-containing segment is a self epitope.
71. The ubiquitin fusion protein of claim 59 wherein one or more epitopes contained within the epitope-containing segment represents an epitope from the group of proteins selected from the group consisting of gonadotropin releasing hormone, tumor necrosis factor, immunoglobulins, human immunodeficiency proteins, chorionic gonadotrophin, inhibin, growth hormones and sperm proteins.
72. The ubiquitin fusion protein of claim 59 wherein one or more epitopes contained within the epitope-containing segment represents an epitope from gonadotropin releasing hormone.
73. The ubiquitin fusion protein of claim 59 wherein the epitope-containing segment contains a single B-cell epitope or a plurality of identical or non-identical B-cell epitopes and a second epitope-containing segment comprises a single T-cell epitope or a plurality of identical or non-identical T-cell epitopes.
74. A DNA construct encoding a fusion protein of claims 20, 41, or 58.
75. A cell containing a DNA construct encoding a fusion protein of claims 1, 20, 41 or 58.
76. A method for stimulating an immune response in an animal, the immune response being directed toward a ubiquitin fusion protein, the method comprising:
a) providing a ubiquitin fusion protein comprising ubiquitin fused to a single epitope-containing segment, the epitope-containing segment comprising two or more identical epitopes; and.
b) administering the fusion protein of step a) to an animal under conditions appropriate for the stimulation of an immune response.
77. A method for stimulating an immune response in an animal, the immune response being directed toward a ubiquitin fusion protein, the method comprising:
(a) providing a ubiquitin fusion protein comprising ubiquitin fused to two or more non-contiguous epitope-containing segments, each epitope-containing segment comprising one or more identical or non-identical epitopes; and
(b) administering the fusion protein of step a) to an animal under conditions appropriate for the stimulation of an immune response.
78. A method for stimulating an immune response in an animal, the immune response being directed toward a ubiquitin fusion protein, the method comprising:
(a) providing a ubiquitin fusion protein comprising ubiquitin fused to a single epitope-containing segment comprising two or more identical or non-identical epitopes, the epitope-containing segments being fused to ubiquitin at fusion sites selected from the group consisting of the N-terminus and an internal fusion site;
(b) administering the fusion protein of step a) to an animal under conditions appropriate for the stimulation of an immune response.
79. A method for stimulating an immune response in an animal, the immune response being directed toward a ubiquitin fusion protein, the method comprising:
(a) providing a ubiquitin fusion protein comprising ubiquitin fused to a single epitope-containing segment comprising one or more identical or non-identical epitopes, the epitope-containing segment being fused to ubiquitin at N-terminus of ubiquitin;
(b) administering the fusion protein of step a) to an animal under conditions appropriate for the stimulation of an immune response.
80. A method for stimulating an immune response in an animal, the immune response being directed toward a fusion protein, the method comprising:
a) providing a DNA construct encoding a fusion protein of claims 1, 20, 41 or 58;
b) introducing the DNA construct of step a) into the cells of the animal under conditions appropriate for expression.
81. A ubiquitin fusion protein comprising ubiquitin having the peptide QHWSYGLRPGQHWSYGLRPGQHWSYGLRPGQHWSYGLRPGC fused via its N terminus to the C-terminal residue of ubiquitin, the ubiquitin fusion protein being cleavable by a ubiquitin-specific protease.
82. The fusion protein of claim 81 conjugated to an immunogenic carrier protein.
83. A ubiquitin fusion protein comprising ubiquitin having the peptide QHWSYGLRPGQHWSYGLRPGQHWSYGLRPGQHWSYGLRPG fused via its N terminus to the C-terminal residue of ubiquitin, the ubiquitin moiety being modified such that the ubiquitin fusion protein is non-cleavable by a ubiquitin-specific protease.
84. A method for stimulating an immune response in an animal, the immune response being directed toward a ubiquitin fusion protein, the method comprising:
(a) providing a ubiquitin fusion protein comprising ubiquitin having the peptide QHWSYGLRPGQHWSYGLRPGQHWSYGLRPGQHWSYGLRPGC fused via its N terminus to the C-terminal residue of ubiquitin; and
(b) administering the conjugate of step (a) to an animal under conditions appropriate for the stimulation of an immune response.
85. The method of claim 84 wherein thephysiological consequences of administration to the animal are substantially similar to the consequences of surgical castration.
86. A method for the identification of antibodies in experimental or diagnostic samples, comprising:
a) providing a ubiquitin fusion protein of selected from the group consisting of ubiquint fusion proteins described in claims 1, 20, 41 and 58;
b) providing antibodies from an experimental or clinical source;
c) forming an incubation mixture comprising the ubiquitin fusion protein of step a) and the antibodies of step b); and
d) detecting binding of the antibodies of step b) to the ubiquitin fusion protein of step a).
87. A method for reducing levels of a predetermined protein in an animal relative to base-line levels, comprising:
a) providing a ubiquitin fusion protein of selected from the group consisting of ubiquitin fusion proteins described in claims 1, 20, 41 and 58 which contain at least one epitope representing an epitope from the predetermined protein and
b) administering the fusion protein of step a) to the animal under, conditions appropriate for the stimulation of an immune response.
88. The method of claim 87 wherein the predetermined protein is a peptide hormone.
89. The method of claim 88 wherein the predetermined peptide hormone is a male-specific or female-specific peptide hormone.
90. The method of claim 89 wherein the predetermined peptide hormone is gonadotropin releasing hormone.
91. The method of claim 87 wherein the predetermined protein is tumor necrosis factor.
92. The method of claim 87 wherein the predetermined protein is a growth hormone protein.
93. The method of claim 87 wherein the fusion protein is conjugated to a non-ubiquitin carrier protein.
94. A method for reducing levels of a predetermined protein in an animal relative to base-line levels, comprising:
a) providing a DNA construct encoding a ubiquitin fusion protein of selected from the group consisting of ubiquitin fusion proteins described in claims 1, 20, 41 and 58 which contain at least one epitope representing an epitope from the predetermined protein; and
b) introducing the DNA construct of step a) into the cells of an animal under conditions appropriate for the expression and stimulation of an immune responses.
95. The method of claim 94 wherein the predetermined protein is a peptide hormone.
96. The method of claim 95 wherein the predetermined peptide hormone is a male-specific or female-specific peptide hormone.
97. The method of claim 96 wherein the predetermined peptide hormone is gonadotropin releasing hormone.
98. The method of claim 94 wherein the predetermined protein is tumor necrosis factor.
99. The method of claim 94 wherein the predetermined protein is a growth hormone protein.
100. The method of claim 94 wherein the fusion protein is conjugated to a non-ubiquitin carrier protein.
US10/681,389 1998-02-19 2003-10-07 Ubiquitin fusion-based vaccine system Abandoned US20040115218A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US10/681,389 US20040115218A1 (en) 1998-02-19 2003-10-07 Ubiquitin fusion-based vaccine system

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US09/026,276 US6319503B1 (en) 1998-02-19 1998-02-19 Heat shock fusion-based vaccine system
US09/964,201 US6660271B2 (en) 1998-02-19 2001-09-26 Ubiquitin fusion-based vaccine system
US10/681,389 US20040115218A1 (en) 1998-02-19 2003-10-07 Ubiquitin fusion-based vaccine system

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US09/964,201 Division US6660271B2 (en) 1998-02-19 2001-09-26 Ubiquitin fusion-based vaccine system

Publications (1)

Publication Number Publication Date
US20040115218A1 true US20040115218A1 (en) 2004-06-17

Family

ID=21830878

Family Applications (4)

Application Number Title Priority Date Filing Date
US09/026,276 Expired - Fee Related US6319503B1 (en) 1997-05-12 1998-02-19 Heat shock fusion-based vaccine system
US09/964,201 Expired - Fee Related US6660271B2 (en) 1998-02-19 2001-09-26 Ubiquitin fusion-based vaccine system
US10/681,389 Abandoned US20040115218A1 (en) 1998-02-19 2003-10-07 Ubiquitin fusion-based vaccine system
US10/681,388 Expired - Fee Related US7662642B2 (en) 1998-02-19 2003-10-07 Method for detection of specific antibodies using ubiquitin fusion proteins

Family Applications Before (2)

Application Number Title Priority Date Filing Date
US09/026,276 Expired - Fee Related US6319503B1 (en) 1997-05-12 1998-02-19 Heat shock fusion-based vaccine system
US09/964,201 Expired - Fee Related US6660271B2 (en) 1998-02-19 2001-09-26 Ubiquitin fusion-based vaccine system

Family Applications After (1)

Application Number Title Priority Date Filing Date
US10/681,388 Expired - Fee Related US7662642B2 (en) 1998-02-19 2003-10-07 Method for detection of specific antibodies using ubiquitin fusion proteins

Country Status (3)

Country Link
US (4) US6319503B1 (en)
AU (1) AU2470499A (en)
WO (1) WO1999042472A1 (en)

Families Citing this family (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7157089B1 (en) * 1996-11-26 2007-01-02 Stressgen Biotechnologies Corporation Immune responses using compositions containing stress proteins
NL1006164C2 (en) * 1997-05-29 1998-12-01 Univ Leiden Antimicrobial peptides.
AUPQ233799A0 (en) * 1999-08-19 1999-09-09 Minister For Agriculture, Minister For Land And Water Conservation For And On Behalf Of The State Of New South Wales Recombinant sub-unit vaccine
JP2004500047A (en) * 1999-10-20 2004-01-08 ザ ジョンズ ホプキンス ユニバーシティー スクール オブ メディシン Chimeric immunogenic compositions and nucleic acids encoding them
US8128922B2 (en) 1999-10-20 2012-03-06 Johns Hopkins University Superior molecular vaccine linking the translocation domain of a bacterial toxin to an antigen
EP1284992A2 (en) * 2000-05-30 2003-02-26 Viridis Biotech Inc. Polyubiquitin based hydrogel and uses thereof
ES2454640T3 (en) 2000-08-03 2014-04-11 Johns Hopkins University Molecular vaccine that carries a chaperone polypeptide from the endoplasmic reticulum to an antigen
IT1319277B1 (en) * 2000-10-24 2003-09-26 Chiesi Farma Spa MELTING PROTEINS USEFUL FOR ALZHEIMER'S MILK IMMUNIZATION TREATMENT.
AUPR446801A0 (en) * 2001-04-18 2001-05-17 University Of Queensland, The Novel compositions and uses therefor
WO2003089590A2 (en) * 2002-04-16 2003-10-30 Vaxin, Inc. TRANSIENT AND/OR PERMANENT MODIFICATION OF SEXUAL BEHAVIOR AND/OR FERTILITY USING RECOMBINANT CHIMERIC GnRH
PL201419B1 (en) * 2002-12-04 2009-04-30 Inst Biotechnologii I Antybiot Chimeric protein, sequence, construction, vegetable cell, method of obtaining chimeric protein and transgenic plant, transgenic plant, application of transgenic plant and chimeric protein
PL201420B1 (en) * 2002-12-04 2009-04-30 Inst Biotechnologii I Antybiot Chimeric protein, sequence, construction, vegetable cell, method of obtaining chimeric protein and transgenic plant, application of transgenic plant and chimeric protein
US20080206264A1 (en) * 2003-02-04 2008-08-28 New York University Constrained Hiv V3 Loop Peptides as Novel Immunogens and Receptor Antagonists
CA2519025A1 (en) * 2003-03-28 2004-10-07 The Government Of The United States Of America As Represented By The Sec Retary Of The Department Of Health And Human Services, Centers For Disea Immunogenic hiv-1 multi-clade, multivalent constructs and methods of their use
EP1644048B1 (en) 2003-05-05 2015-04-29 Johns Hopkins University Anti-cancer dna vaccine employing plasmids encoding signal sequence, mutant oncoprotein antigen, and heat shock protein
WO2005002512A2 (en) * 2003-06-19 2005-01-13 University Of Rochester Immunotherapeutic vaccine strategy
RU2251699C1 (en) * 2003-09-25 2005-05-10 Киселев Всеволод Иванович Method for early and preclinical diagnostics of cervical cancer
WO2006073970A2 (en) 2005-01-06 2006-07-13 The Johns Hopkins University Rna interference that blocks expression of pro-apoptotic proteins potentiates immunity induced by dna and transfected dendritic cell vaccines
US20090075392A1 (en) * 2005-07-19 2009-03-19 John Colyer Agents for and method of quanitifying multiple related components in a biological system
WO2007084568A2 (en) 2006-01-17 2007-07-26 Health Research, Inc. Heteroduplex tracking assay
US9085638B2 (en) 2007-03-07 2015-07-21 The Johns Hopkins University DNA vaccine enhancement with MHC class II activators
KR101643761B1 (en) * 2009-12-04 2016-07-29 삼성전자주식회사 Fusion protein comprising ubiquitin or ubiquitin-like protein, membrane transfer domain and biologically active molecule and uses thereof
CA3056433A1 (en) * 2017-03-22 2018-09-27 Pharmain Corporation Npra agonists, compositions, and uses thereof
EP3965825A4 (en) 2019-04-12 2023-09-13 Therapten Biosciences Inc. Compositions and methods to manufacture phosphatase and tensin homolog (pten) fusions
EP4183417A1 (en) * 2019-09-12 2023-05-24 Hexamer Therapeutics, Inc. An immunotherapeutic for prostate cancer treatment

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5262322A (en) * 1988-06-24 1993-11-16 Genentech, Inc. Host transformed with yeast gene and ubiquitin/polypeptide fusions
US5459051A (en) * 1993-03-26 1995-10-17 Celtrix Pharmaceuticals, Inc. Methods and vectors for over-expression of ubiquitin fusion proteins in host cells
US5703057A (en) * 1995-04-07 1997-12-30 Board Of Regents The University Of Texas System Expression library immunization
US7157089B1 (en) * 1996-11-26 2007-01-02 Stressgen Biotechnologies Corporation Immune responses using compositions containing stress proteins

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5196321A (en) 1986-10-02 1993-03-23 Massachusetts Institute Of Technology Methods for in vitro cleavage of ubiquitin fusion proteins
CA1339684C (en) 1988-05-17 1998-02-24 Peter H. Quail Plant ubquitin promoter system
WO1994029459A1 (en) 1993-06-04 1994-12-22 Whitehead Institute For Biomedical Research Stress proteins and uses therefor
US5620923A (en) * 1989-10-12 1997-04-15 The University Of Utah Synthesis of peptides as cloned ubiquitin extensions
JPH07147987A (en) * 1993-05-28 1995-06-13 Wisconsin Alumni Res Found Ubiqutin conjugative enzyme (e2) fusion protein
DK96493D0 (en) * 1993-08-26 1993-08-26 Mouritsen Og Elsner Aps PROCEDURE FOR INDUCING ANTIBODY RESPONSE TO SELF-PROTEINS AND AUTOVACCINE PROCESSED BY THE PROCEDURE
WO1997001627A1 (en) 1995-06-27 1997-01-16 Igen International, Inc. High-level expression and efficient recovery of ubiquitin fusion proteins from escherichia coli
US5840866A (en) 1996-07-10 1998-11-24 Incyte Pharmaceuticals, Inc. Human ubiquitin-conjugating enzyme
DE19651443A1 (en) 1996-12-11 1998-06-18 Hoechst Ag Self-reinforcing, pharmacologically controllable expression systems
US6007821A (en) 1997-10-16 1999-12-28 Fordham University Method and compositions for the treatment of autoimmune disease using heat shock proteins

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5262322A (en) * 1988-06-24 1993-11-16 Genentech, Inc. Host transformed with yeast gene and ubiquitin/polypeptide fusions
US5459051A (en) * 1993-03-26 1995-10-17 Celtrix Pharmaceuticals, Inc. Methods and vectors for over-expression of ubiquitin fusion proteins in host cells
US5703057A (en) * 1995-04-07 1997-12-30 Board Of Regents The University Of Texas System Expression library immunization
US7157089B1 (en) * 1996-11-26 2007-01-02 Stressgen Biotechnologies Corporation Immune responses using compositions containing stress proteins

Also Published As

Publication number Publication date
US7662642B2 (en) 2010-02-16
US20030091575A1 (en) 2003-05-15
US6660271B2 (en) 2003-12-09
US20040170643A1 (en) 2004-09-02
WO1999042472A1 (en) 1999-08-26
US6319503B1 (en) 2001-11-20
AU2470499A (en) 1999-09-06

Similar Documents

Publication Publication Date Title
US6660271B2 (en) Ubiquitin fusion-based vaccine system
JP3993605B2 (en) Artificial T helper cell epitopes as immunostimulators for synthetic peptide immunogens
Kaumaya et al. Peptide vaccines incorporating a ‘promiscuous’ T‐cell epitope bypass certain haplotype restricted immune responses and provide broad spectrum immunogenicity
AU708472B2 (en) Heterodimer polypeptide immunogen carrier composition and method
JPH0762031B2 (en) Synthetic peptides that induce cell-mediated immunity to AIDS virus and AIDS virus proteins
EP0787139B1 (en) Synthetic peptides and vaccines comprising same
Dalum et al. Induction of cross-reactive antibodies against a self protein by immunization with a modified self protein containing a foreign T helper epitope
US20060121051A1 (en) Heat shock fusion-based vaccine system
Kaul et al. Expression of bonnet monkey (Macaca radiata) zona pellucida‐3 (ZP3) in a prokaryotic system and its immunogenicity
US20070161545A1 (en) Triple polypeptide complexes
US9657070B2 (en) Recombinant allergen
EP0446313B1 (en) Fusion proteins comprising TraTp and at least one LHRH analogue
EP0706574B1 (en) Methods for peptide synthesis and purification
EP0912195B1 (en) Vaccine comprising antigens bound to carriers through labile bonds
TWI285111B (en) VP1 of foot-and-mouth disease virus
He et al. Immunogenic comparison for two different recombinant chimeric peptides (CP12 and CP22) containing one or two copies of three linear B cell epitopes from β-hCG subunit
WO2001012216A1 (en) Heat shock fusion-based vaccine system
TW528760B (en) Peptide repeat immunogens
Lohnas et al. Epitope-specific antibody and suppression of autoantibody responses against a hybrid self protein
AU631350B2 (en) Peptides
US5864008A (en) Peptides derived from foot-and-mouth disease virus, pharmaceutical compositions, and methods for using the peptides
Subramanian et al. Helix stabilization in the C-terminal peptide of chicken riboflavin carrier protein enhances immunogenicity and prolongs contraceptive potential as an epitope-based vaccine in female rats
CA2259237A1 (en) Immunocontrapceptive methods and peptide or polypeptides for use in these methods
JP3457004B6 (en) Peptide derived from hPTH (1-37) sequence
JP3457004B2 (en) Peptide derived from hPTH (1-37) sequence

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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

AS Assignment

Owner name: WELLSTAT BIOCATALYSIS, LLC, MARYLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:PROTIENIX CORPORATION;REEL/FRAME:023379/0176

Effective date: 20041224

Owner name: WELLSTAT BIOCATALYSIS, LLC,MARYLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:PROTIENIX CORPORATION;REEL/FRAME:023379/0176

Effective date: 20041224