WO2014144024A1 - Hyr1 compositions and methods for treating fungal and bacterial pathogens - Google Patents

Hyr1 compositions and methods for treating fungal and bacterial pathogens Download PDF

Info

Publication number
WO2014144024A1
WO2014144024A1 PCT/US2014/028256 US2014028256W WO2014144024A1 WO 2014144024 A1 WO2014144024 A1 WO 2014144024A1 US 2014028256 W US2014028256 W US 2014028256W WO 2014144024 A1 WO2014144024 A1 WO 2014144024A1
Authority
WO
WIPO (PCT)
Prior art keywords
polypeptide
candidiasis
vaccine
candida
mammal
Prior art date
Application number
PCT/US2014/028256
Other languages
French (fr)
Inventor
Ashraf S. Ibrahim
Michael R. Yeaman
Scott G. Filler
John E. Edwards, Jr.
John P. Hennessey, Jr.
Original Assignee
Los Angeles Biomedical Research Institute At Harbor-Ucla Medical Center
Novadigm Therapeutics, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Los Angeles Biomedical Research Institute At Harbor-Ucla Medical Center, Novadigm Therapeutics, Inc. filed Critical Los Angeles Biomedical Research Institute At Harbor-Ucla Medical Center
Publication of WO2014144024A1 publication Critical patent/WO2014144024A1/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0002Fungal antigens, e.g. Trichophyton, Aspergillus, Candida
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/10Antimycotics

Definitions

  • the present invention relates generally to compositions and methods for detecting, treating and preventing infectious diseases in a subject.
  • the fungus Candida the third most common cause of healthcare- associated bloodstream infections, causes approximately 60,000 cases of hematogenously disseminated candidiasis per year in the United States, resulting in billions of dollars of healthcare expenditures. Despite current antifungal therapy, mortality remains unacceptably high. Because of the rising incidence of life-threatening candidiasis and high treatment failure rates, more effective prophylactic and therapeutic strategies are needed.
  • Acinetobacter is a genus of gram negative bacteria belonging to the Gammaproteobacteria . Acinetobacter species contribute to the mineralization of aromatic compounds in the soil. Unfortunately, no technology presently exists that prevents
  • Acinetobacter infections aside from standard hand washing and other infection control practices in hospital settings.
  • the invention features fragments of Hyrl.
  • the amino acid sequence of native C. albicans SC5314 Hyrl polypeptide is as follows :
  • the invention features an isolated polypeptide optionally fused to a heterologous fusion partner, wherein the amino acid sequence of the polypeptide consists of an amino acid sequence havinq at least 95% identity to
  • the invention features an isolated polypeptide optionally fused to a heterologous fusion partner, wherein the amino acid sequence of the polypeptide consists of an amino acid sequence having at least 95% identity to
  • the invention features an isolated polypeptide optionally fused to a heterologous fusion partner, wherein the amino acid sequence of the polypeptide consists of an amino acid sequence having at least 95% identity to
  • the invention features an isolated polypeptide optionally fused to a heterologous fusion partner, wherein the amino acid sequence of said polypeptide consists of an amino acid sequence having at least 95% identity to
  • the polypeptide is SEQ ID NO: 6; SEQ ID NO: 7; SEQ ID NO: 9; or SEQ ID NO: 33.
  • the invention features an isolated nucleic acid molecule including a nucleic acid seguence which is substantially identical to any of the isolated nucleic acid molecule which encodes any of the polypeptides described herein.
  • the invention further features a vector including any of the nucleic acid molecules encoding the polypeptides described herein.
  • the invention accordingly also provides vectors containing the nucleic acids of the invention.
  • Suitable expression vectors are well-known in the art and include vectors capable of expressing a nucleic acid operatively linked to a regulatory seguence or element such as a promoter region or enhancer region that is capable of regulating expression of the nucleic acid.
  • Appropriate expression vectors include vectors that are replicable in eukaryotic cells and/or prokaryotic cells and vectors that remain episomal or integrate into the host cell genome.
  • the invention also provides a method for expression of a
  • polypeptide as disclosed herein by culturing cells containing a nucleic acid that encodes the polypeptide under conditions suitable for expression of polypeptide.
  • a method for the recombinant production of a polypeptide of the invention by expressing the nucleic acid seguences encoding the polypeptide in suitable host cells.
  • Recombinant DNA expression systems that are suitable for production of polypeptides are described herein and are well-known in the art.
  • the above-described nucleotide seguences can be incorporated into vectors for further manipulation.
  • Vectors can include a recombinant DNA or RNA plasmid or virus
  • the invention features a cell including any of the nucleic acid molecules encoding polypeptides described herein.
  • the invention features a method of producing a recombinant polypeptide, the method including the steps of: (a) providing a cell transformed with the nucleic acid molecule of encoding an polypeptide described herein positioned for expression in the cell; (b) culturing the transformed cell under conditions for expressing the nucleic acid molecule, wherein the culturing results in expression of the recombinant polypeptide; and (c) isolating the recombinant polypeptide.
  • cell is a bacterium (e.g., E. coli) .
  • the cell is a yeast cell (e.g., Saccharomyces cerevisae) .
  • the invention features a recombinant polypeptide produced according to this
  • the invention features a substantially pure antibody that specifically recognizes and binds to any one of the polypeptides described herein.
  • the invention features an antigenic composition including the aforementioned polypeptides and a
  • composition further includes an adjuvant.
  • the invention features a method of inducing an immune response in a mammal against an antigen including administering any of the aforementioned polypeptides, or the aforementioned
  • the mammal e.g., a human
  • the polypeptide or the composition induces an immune response against the antigen in the mammal.
  • the mammal is administered a single dose of the polypeptide or the composition.
  • the mammal is administered a plurality of doses of the polypeptide or the composition.
  • the plurality of doses are
  • the composition is administered twice.
  • the invention features a vaccine including an immunogenic amount of any of the aforementioned polypeptides, and a pharmaceutically acceptable excipient.
  • the vaccine includes a mixture of distinct polypeptides of any one of the aforementioned polypeptides.
  • the vaccine further includes an adjuvant (Alhydrogel) .
  • the vaccine of the invention is useful for vaccination of a mammal (e.g., a human) against candidiasis or a bacterial infection such as one caused by Acinetobacter or both.
  • the vaccine is to be administered by intramuscular, subcutaneous, or intradermal administration.
  • the vaccine may also be administered by intramuscular administration.
  • Vaccination may further includes administering a booster dose.
  • Candidiasis may take many forms such as disseminated candidiasis (e.g., hematogenously
  • candidiasis disseminated candidiasis or mucosal candidiasis.
  • Candidiasis is caused, for example, by Candida albicans, Candida glabrata, Candida krusei, Candida parapsilosis, or Candida tropicalis .
  • vaccination is against Acinetobacter.
  • the invention features a method of producing a chimeric vaccine including the steps of: (a) providing a phage, yeast, or virus; (b) inserting into the phage, yeast, or virus a nucleic acid molecule that encodes any of the aforementioned polypeptides; (c) allowing expression of the polypeptide in the phage, yeast, or virus; (d) isolating the phage, yeast, or virus of step (c) including the expressed polypeptide; and (e) adding a pharmaceutically acceptable excipient to the isolated phage, yeast, or virus of step (d) .
  • the polypeptide is displayed on the surface of the phage, yeast, or virus following step (c).
  • the invention features an isolated monoclonal antibody that binds to any of the aforementioned polypeptides .
  • the antibody is human or humanized.
  • the antibody may also be chimeric.
  • the antibody may also be produced recombinantly .
  • a diagnostic composition including these antibodies is within the invention .
  • compositions including any of the aforementioned antibodies alone or in combination, and a pharmaceutically acceptable excipient.
  • the pharmaceutical composition includes a mixture of antibodies with a plurality of distinct specificities.
  • the invention features a pharmaceutical composition including polyclonal antibodies that bind to the any of the polypeptides described herein, or that bind to a mixture of distinct polypeptides of such described polypeptides.
  • the pharmaceutical composition is for use in the passive immunization of a mammal (e.g., a mammal) against candidiasis or a bacterial infection.
  • the pharmaceutical composition is administered by intramuscular, subcutaneous, or intradermal
  • the pharmaceutical composition is administered by intramuscular administration.
  • the candidiasis is disseminated candidiasis, for example,
  • candidiasis is mucosal candidiasis.
  • candidiasis is caused by Candida albicans, Candida glabrata, Candida krusei,
  • passive immunization is against Acinetobacter.
  • the invention features a method of passive immunization of a mammal (e.g., a human) against candidiasis or a bacterial infection such as one caused Acinetobacter including administering to the mammal an effective amount of any of the mammal (e.g., a human) against candidiasis or a bacterial infection such as one caused Acinetobacter including administering to the mammal an effective amount of any of the mammal (e.g., a human) against candidiasis or a bacterial infection such as one caused Acinetobacter) including administering to the mammal an effective amount of any of the mammal (e.g., a human) against candidiasis or a bacterial infection such as one caused Acinetobacter) including administering to the mammal an effective amount of any of the mammal (e.g., a human) against candidiasis or a bacterial infection such as one caused Acinetobacter) including administering to the mammal an effective amount
  • the pharmaceutical composition is administered by intramuscular, subcutaneous, or intradermal administration. In other embodiments, the pharmaceutical composition is administered by intramuscular administration.
  • the candidiasis is disseminated candidiasis, for example, hematogenously disseminated candidiasis. In some embodiments, the candidiasis is mucosal candidiasis. In some embodiments, the candidiasis is caused by Candida albicans, Candida glabrata, Candida krusei, Candida parapsilosis, or Candida tropicalis .
  • compositions and methods as disclosed herein that are based, at least in part, on the proposition that an immune response, such as antibodies and other mechanisms, that target the a Candida Hyrl polypeptide and confer protection from Acinetobacter infection such as Acinetobacter
  • compositions and methods disclosed herein include passive vaccination of acutely at-risk patients with a dose of anti-Hyrl antibody to prevent the acquisition of Acinetobacter baumannii infection. Additionally, patients with active Acinetobacter baumannii infection can be treated with the antibody alone or combined with other antibacterial agents . Alternatively, patients who are at risk of developing such infections, such as, for example, military personnel, can be actively vaccinated with a Hyrl polypeptides disclosed herein to prevent such infections.
  • the vaccine compositions of the present invention can also be used to treat,
  • vaccines that contain one or more of the polypeptides and/or antibody compositions described herein in combination with adjuvants, can act for the purposes of prophylactic or therapeutic treatment of infections from gram negative bacteria.
  • vaccines of the present invention will induce the body's own immune system to seek out and inhibit gram negative bacteria or Candida or both.
  • Vaccines according to the invention refer to a composition that can be administered to an individual to protect the individual against an infectious disease. Vaccines protect against diseases by inducing or increasing an immune response in an animal against the infectious disease.
  • An exemplary infectious disease amenable to treatment with the vaccines of the invention include severe pneumonia, infections of the urinary tract, infections of the bloodstream and infections of other parts of the body.
  • the vaccine-mediated protection can be humoral and/or cell mediated immunity induced in host when a subject is challenged with, for example, or an immunogenic portion of a polypeptide or protein described herein.
  • the invention provides a method of treating or preventing an infection from gram negative bacteria in a subject in need thereof by administering a therapeutically effective amount of a pharmaceutical composition as disclosed herein or a vaccine composition as disclosed herein.
  • the invention provides methods of treating or preventing infections caused by one or more gram negative bacteria including bacteria of the Acinetobacter genus, such as A. baumannii, A. iwoffii, A. haemolyticus, A.
  • H. aphrophilus H. avium, H. ducreyi, H. felis, H. haemolyticus, H.
  • bacteria of the Bordetella genus such as B. ansorpii, B avium, B. bronchiseptica, B. hinzii, B. holmesii, B. parapertussis, B. pertussis, B. petrii, and B. trematum
  • bacteria of the Salmonella genus such as S. typhimurium, S. bongori , S. enterica subsp . enterica, S. enterica subsp. salamae, S. arizonae, S. enterica subsp .
  • bacteria of the Yersina genus such as Yersina pseudotuber, Y. aldovae, Y. aleksiciae, Y. bercovieri, Y. enterocolitica, Y.
  • frederiksenii Y. intermedia, Y. kristensenii, Y. mollaretii, Y.
  • bacteria of the Escherichia genus such as E. albertii, E. blattae, E. coli, E. fergusonii, E. hermannii and E. vulneris
  • bacteria of the Pedobacter genus such as P. heparinus, P. roseus sp. nov. and P. aquatilis sp. nov
  • bacteria of the Pseudomonas genus such as P. aeruginosa, P.
  • Candida species as disclosed herein may be treated or prevented.
  • adjuvant is meant one or more substances that cause stimulation of the immune system.
  • an adjuvant is used to enhance an immune response to one or more vaccine antigens or antibodies.
  • An adjuvant may be administered to a subject before, in combination with, or after administration of the vaccine or antibody.
  • Examples of chemical compounds used as adjuvants include, but are not limited to, aluminum compounds (e.g., alum, Alhydrogel), oils, block polymers, immune stimulating complexes, vitamins and minerals (e.g., vitamin E, vitamin A, selenium, and vitamin B12) , Quil A (saponins), bacterial and fungal cell wall components (e.g., lipopolysaccarides , lipoproteins, and glycoproteins), hormones, cytokines, and co- stimulatory factors.
  • aluminum compounds e.g., alum, Alhydrogel
  • oils block polymers
  • immune stimulating complexes e.g., vitamins and minerals (e.g., vitamin E, vitamin A, selenium, and vitamin B12) , Quil A (saponins), bacterial and fungal cell wall components (e.g., lipopolysaccarides , lipoproteins, and glycoproteins), hormones, cytokines, and co- stimulatory factors.
  • antibody is meant whole antibodies, immunoglobulins, or any antigen-binding fragment or single chains thereof.
  • Antibodies can be mammalian (e.g., human or mouse), humanized, chimeric, recombinant, synthetically produced, or naturally isolated, and can be, e.g., monoclonal or polyclonal. In most mammals, including humans, whole antibodies have at least two heavy (H) chains and two light (L) chains connected by disulfide bonds. Each heavy chain consists of a heavy chain variable region (abbreviated herein as V H ) and a heavy chain constant region.
  • V H heavy chain variable region
  • the heavy chain constant region consists of three domains, C H 1, C H 2, and C H 3 and a hinge region between C H 1 and C H 2.
  • Each light chain consists of a light chain variable region (abbreviated herein as V L ) and a light chain constant region.
  • the light chain constant region consists of one domain, C L .
  • the V H and V L regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR) , interspersed with regions that are more conserved, termed
  • CDR complementarity determining regions
  • Each V H and V L is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4.
  • the variable regions of the heavy and light chains contain a binding domain that interacts with an antigen.
  • the constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (Clg) of the classical complement system.
  • Antibodies of the present invention include all known forms of antibodies and other protein scaffolds with antibody-like properties.
  • the antibody can be a human antibody, a humanized antibody, a bispecific antibody, a chimeric antibody, or a protein scaffold with antibody-like properties, such as fibronectin or ankyrin repeats.
  • the antibody also can be a Fab, Fab'2, scFv, SMIP, diabody, nanobody, aptamers, or a domain antibody.
  • the antibody can have any of the following isotypes: IgG (e.g., IgGl, IgG2, IgG3, and IgG4), IgM, IgA (e.g., IgAl, IgA2, and IgAsec) , IgD, or IgE.
  • IgG e.g., IgGl, IgG2, IgG3, and IgG4
  • IgM e.g., IgAl, IgA2, and IgAsec
  • IgD e.gAl, IgA2, and IgAsec
  • antibody fragment refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen.
  • the antigen-binding function of an antibody can be performed by fragments of a full-length antibody, which include but are not limited to: (i) a Fab fragment, a monovalent fragment consisting of the V L , V H , C L , and C H 1 domains; (ii) a F(ab')2 fragment, a bivalent fragment
  • CDR complementarity determining region
  • V L and V H complementarity determining region
  • antibody fragments are obtained using conventional technigues known to those with skill in the art, and the fragments are screened for utility in the same manner as are intact antibodies.
  • Antibody fragments can be produced by recombinant DNA technigues, or by enzymatic or chemical cleavage of intact
  • antibody By “antigen” is meant a molecule to which an antibody can
  • the target antigen may be a protein (e.g., an
  • antigenic peptide carbohydrate, nucleic acid, lipid, hapten, or other naturally occurring or synthetic compound.
  • the target antigen may be a polypeptide or peptide mimic.
  • An antigen may also be administered to an animal to generate an immune response in the animal .
  • carrier in the context of a conjugate is meant a moiety or particle, e.g., KLH, CRM197, tetanus toxoid, a phage, a yeast, a virus, a virosome, or a recombinant virus-like particle, that is suitable for being linked to or displaying a polypeptide as described herein .
  • chimeric antibody is meant an immunoglobulin or antibody whose variable regions derive from a first species and whose constant regions derive from a second species.
  • Chimeric antibodies can be constructed, for example, by genetic engineering, from immunoglobulin gene segments belonging to different species (e.g., from a mouse and a human) .
  • chimeric vaccine is meant a vaccine that includes at least two distinct antigens, e.g., joined covalently.
  • An example of a chimeric vaccine is a composition that includes a polypeptide
  • a particle such as a phage, virus, yeast, virosome, or recombinant virus-like particle.
  • conjugate is meant a compound that includes a polypeptide of the invention linked to another moiety or particle, e.g., KLH, CRM197, tetanus toxoid, a phage, a yeast, a virus, a virosome, or a
  • conservative substitution in an amino acid seguence is meant replacement of an amino acid for another within a family of amino acids that are related in the chemical nature of their side chains.
  • acidic aspartate, glutamate
  • basic lysine, arginine, histidine
  • nonpolar alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan
  • uncharged polar glycine, asparagine, glutamine, cysteine, serine, threonine, tyrosine
  • Phenylalanine, tryptophan, and tyrosine are sometimes grouped as aromatic amino acids.
  • the amino acids can also be separated into the following groups: acidic (aspartate, glutamate); basic (lysine, arginine, histidine); alipathic (glycine, alanine, valine, leucine, isoleucine, serine, threonine), with serine and threonine optionally grouped separately as alipathic-hydroxyl ;
  • aromatic phenylalanine, tyrosine, tryptophan
  • amide asparagine, glutamine
  • sulfur-containing cyste, methionine
  • Whether a change in the amino acid seguence results in a functional variant can be determined by assessing the ability of the variant polypeptide to function in a fashion similar to the wild-type polypeptide using standard methods such as those described herein.
  • diagnostic composition a composition containing a polypeptide, conjugate, vaccine, or antibody of the invention, formulated for use in conjunction with a diagnostic method.
  • an effective amount in the context of passive immunization using a pharmaceutical composition, e.g., comprising an antibody, is meant the amount of the pharmaceutical composition reguired to passively immunize in a clinically relevant manner.
  • An effective amount of pharmaceutical composition used to practice the methods of passive immunization described herein varies depending upon the manner of administration, the age, body weight, and general health of the subject. Ultimately, the prescribers will decide the appropriate amount and dosage regimen.
  • flanking amino acid is meant an amino acid in a polypeptide seguence that is immediately adjacent to the N- or C-terminus of a particular defined seguence.
  • a flanking amino acid is present on the N- and/or C-terminus of the amino acid seguence of SC5314 Hyrl or rHyrl or a fragment thereof; and more desirably, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 flanking amino acids are present at the N- and/or C-terminus of the amino acid seguence of SC5314 Hyrl or rHyrl, or fragment thereof .
  • fusion protein is meant a protein that includes a
  • polypeptide of the invention e.g., a peptide fragment or variant, and a fusion partner.
  • fusion partner is meant a heterologous seguence that can be fused to a polypeptide or peptide of the invention , e.g., one or more of Peptide 3-11 or variants thereof.
  • fusion partners are described herein and include detection markers, stabilizing domains, sequences which aid in production or purification of the protein, or domains which increase the antigenicity of the polypeptide.
  • immunogenic is meant any substance that is capable of inducing an immune response in a subject.
  • immunogenic amount in the context of a vaccine is meant an amount of the vaccine required to induce an immune response in a subject in a clinically relevant manner.
  • An immunogenic amount of vaccine used to practice the methods of vaccination as described herein varies depending upon the manner of administration, the age, body weight, and general health of the subject. Ultimately, the prescribers will decide the appropriate amount and dosage regimen.
  • isolated or “purified” is meant separated from other naturally accompanying components.
  • a compound e.g., nucleic acid, polypeptide, antibody, or small molecule
  • the definition also extends, e.g., to a polypeptide or nucleic acid molecule separated from its flanking sequences (e.g., for an amino acid sequence, isolated refers to a sequence that is free from the flanking amino acids with which the sequence is naturally associated in a polypeptide) .
  • the compound is at least 75%, more preferably at least 90%, and most preferably at least 99%, by weight, isolated.
  • An isolated compound e.g., polypeptide
  • conjugated to in the context of a conjugate is meant a covalent or non-covalent interaction between the
  • interactions include, but are not limited to, hydrogen bonding, ionic interactions among charged groups, electrostatic binding, van der Waals interactions, hydrophobic interactions among non-polar groups, lipophobic interactions, and LogP-based attractions.
  • monoclonal antibody an antibody obtained from a population of substantially homogeneous antibodies, i.e., the
  • Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to conventional (polyclonal) antibody preparations which typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen. Monoclonal antibodies can be prepared using any art recognized technigue and those described herein such as, for example, a hybridoma method, as described by Kohler et al . , Nature 256:495 (1975), a transgenic animal (e.g., Lonberg et al . , Nature 368 (6474) : 856-859 (1994)), recombinant DNA methods (e.g., U.S. Pat.
  • nucleic acid molecule is meant a molecule, e.g., RNA or DNA, having a seguence of two or more covalently bonded, naturally
  • the nucleic acid molecule may be, e.g., single or double stranded, and may include modified or
  • patient or “subject” is meant a mammal, including, but not limited to, a human or non-human mammal, such as a bovine, eguine, canine, ovine, or feline.
  • a human or non-human mammal such as a bovine, eguine, canine, ovine, or feline.
  • peptide refers to any chain of two or more natural or unnatural amino acids, regardless of post-translational modification (e.g., glycosylation or phosphorylation), constituting all or part of a naturally-occurring or non-naturally occurring polypeptide or peptide, as is described herein.
  • polypeptides typically are continuous and unbranched peptide.
  • a peptide is a short polymer of amino acid monomers.
  • Proteins are intended to include one or more polypeptides arranged in a biologically functional way.
  • the amino acids comprising the polypeptides of the invention may be linked by peptide bonds or other bonds, for example, ester or ether bonds.
  • the amino acids comprising the polypeptides of the invention can include non-genetically coded amino acids that either occur naturally or are chemically synthesized.
  • a polypeptide of the invention can also encompass one or more conservative substitutions .
  • Conservative substitutions of encoded amino acids include, for example, amino acids that belong within the following groups: (1) non-polar amino acids (Gly, Ala, Val, Leu, and lie); (2) polar neutral amino acids (Cys, Met, Ser, Thr, Asn, and Gin); (3) polar acidic amino acids (Asp and Glu) ; (4) polar basic amino acids (Lys, Arg and His); and (5) aromatic amino acids (Phe, Trp, Tyr, and His) .
  • Other minor modifications are also included within polypeptides of the invention so long as the polypeptide retains some or all of its function as described herein.
  • polypeptides can also include derivatives, analogues and functional mimetics thereof, provided that such
  • polypeptide retains some or all of its function as disclosed herein.
  • derivatives can include chemical modifications of the polypeptide such as alkylation, acylation, carbamylation, iodination, or any modification that derivatizes the polypeptide.
  • derivatized molecules include, for example, those molecules in which free amino groups have been derivatized to form amine hydrochlorides, p-toluene sulfonyl groups, carbobenzoxy groups, t-butyloxycarbonyl groups, chloroacetyl groups or formyl groups.
  • Free carboxyl groups can be derivatized to form salts, methyl and ethyl esters or other types of esters or hydrazides. Free hydroxyl groups can be
  • O-acyl or O-alkyl derivatives The imidazole nitrogen of histidine can be derivatized to form N-im-benzylhistidine .
  • derivatives or analogues those peptides which contain one or more naturally occurring amino acid derivatives of the twenty standard amino acids, for example, 4-hydroxyproline, 5-hydroxylysine, 3-methylhistidine, homoserine, ornithine or carboxyglutamate, and can include amino acids that are not linked by peptide bonds.
  • Polypeptides of the present invention also include any polypeptide having one or more additions and/or deletions of residues, relative to the seguence of a polypeptide whose seguence is shown herein, so long as immunogenic activity as disclosed herein is maintained .
  • polypeptides can be isolated by a variety of methods well-known in the art, for example, recombinant expression systems, precipitation, gel filtration, ion-exchange, reverse-phase and affinity chromatography, and the like. Other well-known methods are described in Deutscher et al . , Guide to Protein Purification:
  • the isolated polypeptides of the present invention can be obtained using well-known recombinant methods (see, for example, Ausubel et al . , "Immunology,” Short Protocols in Molecular Biology, John Wiley & Sons, Inc. Chapter 11. Page 11.1-11.29 (1999); Sambrook and Russell, “Molecular Cloning: A Laboratory Manual,” Cold Spring Harbor Laboratory (2001)).
  • the methods and conditions for biochemical purification of a polypeptide of the invention can be chosen by those skilled in the art, and purification monitored, for example, by an immunological assay or a functional assay.
  • An example of the means for preparing an invention polypeptide is to express nucleic acids encoding a polypeptide of the invention in a suitable host cell, such as a bacterial cell, a yeast cell, an amphibian cell such as an oocyte, or a mammalian cell, using methods well known in the art, and recovering the expressed polypeptide, again using well-known purification methods, so described herein.
  • a suitable host cell such as a bacterial cell, a yeast cell, an amphibian cell such as an oocyte, or a mammalian cell
  • polypeptides can also be produced by chemical synthesis.
  • Recombinantly expressed polypeptides of the invention can also be expressed as fusion proteins with appropriate fusion partners.
  • An appropriate fusion partner can be an amino acid sequence that is not normally connected to the amino acid sequence such as an heterologous sequence, which serves a particular function or provides additional characteristic to the polypeptides of the invention.
  • suitable heterologous seguences include a detectable marker, a stabilizing domain, a carrier protein for the generation of antibodies, a linker seguence and a seguence that aids in the
  • polypeptides that can aid in the purification of the invention polypeptides include affinity tags, such as glutathione S transferase (GST) or poly His.
  • affinity tags such as glutathione S transferase (GST) or poly His.
  • GST glutathione S transferase
  • the invention provide a fusion protein having a polypeptide as disclosed herein fused to a heterologous seguence, a carrier protein, an affinity tag or a linker seguence or other polypeptides as disclosed herein.
  • a natural amino acid is a natural -amino acid having the L-configuration, such as those normally occurring in natural polypeptides.
  • Unnatural amino acid refers to an amino acid that normally does not occur in polypeptides, e.g., an epimer of a natural a-amino acid having the L configuration, that is to say an amino acid having the unnatural D-configuration; or a (D, L) -isomeric mixture thereof; or a homolog of such an amino acid, for example, a ⁇ - amino acid, an a, -disubstituted amino acid, or an a-amino acid wherein the amino acid side chain has been shortened by one or two methylene groups or lengthened to up to 10 carbon atoms, such as an a- amino alkanoic acid with 5 up to and including 10 carbon atoms in a linear chain, an unsubstituted or substituted aromatic (a-aryl or a- aryl lower alkyl), for example,
  • pharmaceutically acceptable excipient are used interchangeably and mean a carrier or excipient that is physiologically acceptable to the treated patient while retaining the therapeutic properties of the compound with which it is administered.
  • physiologically acceptable carrier substance is physiological saline.
  • physiologically acceptable carriers and their formulations are known to those skilled in the art and described, for example, in
  • composition a composition containing a polypeptide, conjugate, vaccine, or antibody of the invention, formulated with a pharmaceutically acceptable excipient, and
  • compositions can be formulated, for example, for intravenous
  • administration e.g., as a sterile solution free of particulate emboli and in a solvent system suitable for intravenous use
  • oral administration e.g., a tablet, capsule, caplet, gelcap, or syrup
  • any other formulation described herein e.g., in unit dosage form.
  • binding moiety e.g., an antibody, antibody fragment, receptor, ligand, or small molecule portion of an agent as described herein
  • a target molecule e.g., a polypeptide or conjugate including same
  • a cell or tissue bearing the target molecule e.g., a cell surface antigen, such as a receptor or ligand
  • binding moiety e.g., an antibody
  • target molecule e.g., a polypeptide or conjugate including same
  • Binding moieties bind to the target molecule or marker with a dissociation constant of e.g., less than 1CT 6 M, less than 1CT 7 M, 1CT 8 M, 1CT 9 M, 1CT 10 M, 10 _11 , or 1CT 12 M, or even less than 1CT 13 M, 1CT 14 M, or 10 ⁇ 15 M.
  • binding moiety that is selected for its specificity for that particular protein.
  • a variety of assay formats are appropriate for selecting binding moieties (e.g., antibodies) capable of specifically binding to a particular target molecule. For example, solid-phase ELISA
  • immunoassays are routinely used to select monoclonal antibodies specifically immunoreactive with a protein. See Harlow & Lane,
  • substantially identical is meant an amino acid sequence or nucleic acid sequence that exhibits at least 50% identity to a reference sequence. Such a sequence is generally at least, e.g., 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identical at the amino acid level or nucleic acid level to a reference sequence.
  • the length of comparison sequences can be at least five amino acids, e.g., 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 125, 150, 175, 200, 250, 300, or more amino acids, up to the entire length of the polypeptide.
  • the length of comparison sequences can generally be at least 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 125, 150, 175, 200, 250, 300, 400, 500, 600, 700, 800, 900, or more nucleotides, up to the entire length of the nucleic acid molecule. It is understood that for the purposes of determining sequence identity when comparing a DNA sequence to an RNA sequence, a thymine nucleotide is equivalent to a uracil nucleotide.
  • polypeptide or nucleic acid sequence is referred to as having "at least X% sequence identity" to a reference sequence, it is meant that at least X percent of the amino acids or nucleotides in the polypeptide or nucleic acid are identical to those of the reference sequence when the sequences are optimally aligned.
  • An optimal alignment of sequences can be determined in various ways that are within the skill in the art, for instance, the Smith Waterman alignment algorithm (Smith et al . , J. Mol . Biol. 147:195-7, 1981) and BLAST (Basic Local Alignment Search Tool; Altschul et al . , J. Mol. Biol. 215: 403-10, 1990). These and other alignment algorithms are accessible using publicly available computer software such as “Best Fit” (Smith and Waterman, Advances in Applied Mathematics, 482-489, 1981) as incorporated into GeneMatcher PlusTM (Schwarz and Dayhof,
  • target molecule or “target cell” is meant a molecule (e.g., a polypeptide, epitope, antigen, receptor, or ligand) or cell to which a binding moiety (e.g., an antibody) can specifically bind.
  • a binding moiety e.g., an antibody
  • target molecules are exposed on the exterior of a target cell (e.g., a cell surface or secreted protein), but target molecules may alternately or also be present in the interior of a target cell.
  • treating or “treatment” is meant the medical management of a patient with the intent to cure, ameliorate, stabilize, reduce the likelihood of, or prevent a disease, pathological condition, disorder, or event, by administering a pharmaceutical composition.
  • This term includes active treatment, that is, treatment directed specifically toward the improvement or associated with the cure of a disease, pathological condition, disorder, or event, and also includes causal treatment, that is, treatment directed toward removal of the cause of the associated disease, pathological condition, disorder, or event.
  • palliative treatment that is, treatment designed for the relief of symptoms rather than the curing of the disease, pathological condition, disorder, or event;
  • symptomatic treatment that is, treatment directed toward
  • preventative treatment that is, treatment directed to minimizing or partially or completely inhibiting the development of the associated disease, pathological condition, disorder, or event, e.g., in a patient who is not yet ill, but who is susceptible to, or otherwise at risk of, a particular disease, pathological condition, disorder, or event
  • supportive treatment that is, treatment employed to supplement another specific therapy directed toward the improvement of the associated disease
  • pathological condition disorder, or event.
  • vaccine as used herein, is meant a composition that elicits an immune response in a subject to which it is administered.
  • vaccine is meant to treat a patient by administering a vaccine, e.g., to prevent or ameliorate a disease, pathological condition, disorder, or event.
  • variants in the context of a polypeptide or portion thereof as described herein, or a nucleic acid molecule encoding same, is meant to include substitutions or alterations in the amino acid seguence or nucleic acid seguence, e.g., resulting in a substantially identical seguence.
  • a polypeptide having a variant seguence may maintain at least one biological activity of the original polypeptide, e.g., immunogenic activity.
  • variant includes, e.g., amino acid insertional derivatives such as amino and/or carboxylterminal fusions, as well as intraseguence insertions of single or multiple amino acids. Insertional amino acid variants are those in which one or more amino acid residues are introduced into a predetermined site in the protein. Random insertion is also possible with suitable screening of the resulting product. Deletional variants are
  • substitutional amino acid variants are those in which at least one residue inserted in its place.
  • amino acids are generally replaced by conservative substitutions, e.g., other amino acids having similar physical chemical properties such as hydrophobicity, hydrophilicity, electronegativity, bulky sidechains and the like.
  • variants also include single or multiple substitutions, deletions and/or additions of any component (s) naturally or artificially associated with the portion of a naturally occurring protein from which the polypeptide may be derived, such as carbohydrate, lipid and/or other proteinaceous moieties. All such molecules are encompassed by the term "variant.”
  • variant sequence is meant the amino acid or nucleic acid sequence of a variant as defined herein.
  • Hyrl polypeptide fragments and other compositions described herein allow, e.g., for the effective treatment of and vaccination against candidiasis or bacterial infections such as those caused by any pathogen disclosed herein.
  • the invention provides polypeptides, e.g., derived from Hyrl, conjugates, vaccines, antibodies, compositions, methods of vaccination using same, and methods of production of same, as described in further detail below.
  • the invention features polypeptides derived from SC5413 Hyrl or rHyrl : Hyrl
  • the amino acid sequence of native C. albicans SC5314 Hyrl polypeptide is as follows:
  • the amino acid sequence of rHyrl polypeptide is:
  • the amino acid sequence of the rHyrl protein is as follows:
  • a modification to a polypeptide as described herein does not substantially reduce the biological activity, e.g., immunogenic activity, of the polypeptide.
  • the modified polypeptide may have or may optimize a characteristic of a polypeptide, such as in vivo stability, bioavailability, toxicity, immunological activity, immunological identity, or conjugation properties.
  • Modifications include those by natural processes, such as posttranslational processing, or by chemical modification techniques known in the art. Modifications may occur anywhere in a polypeptide including the polypeptide backbone, the amino acid side chains, and the amino- or carboxy-terminus . The same type of modification may be present in the same or varying degrees at several sites in a given polypeptide, and a polypeptide may contain more than one type of modification .
  • a variant or otherwise modified polypeptide can also include one or more amino acid insertions, deletions, or substitutions, either conservative or non-conservative (e.g., D-amino acids, desamino acids) in the polypeptide sequence.
  • conservative or non-conservative e.g., D-amino acids, desamino acids
  • the addition of one or more cysteine residues to the amino or carboxy terminus of any of the polypeptides of the invention can facilitate conjugation of these polypeptides.
  • Exemplary polypeptides having an N- or C-terminal cysteine having an N- or C-terminal cysteine .
  • Amino acid substitutions can be conservative (i.e., wherein a residue is replaced by another of the same general type or group) or non-conservative (i.e., wherein a residue is replaced by an ammo acid of another type) .
  • a non-naturally occurring amino acid can be substituted for a naturally occurring amino acid (i.e., non- naturally occurring conservative amino acid substitution or a non- naturally occurring non-conservative amino acid substitution) .
  • Polypeptides made synthetically, e.g., using methods known in the art, can include substitutions of amino acids not naturally encoded by DNA (e.g., non-naturally occurring or unnatural amino acid).
  • non-naturally occurring amino acids include D-amino acids, an amino acid having an acetylaminomethyl group attached to a sulfur atom of a cysteine, a pegylated amino acid, the omega amino acids of the formula NH 2 (CH 2 ) n COOH wherein n is 2-6, neutral nonpolar amino acids, such as sarcosine, t-butyl alanine, t-butyl glycine, N-methyl isoleucine, and norleucine.
  • Phenylglycine may substitute for Trp, Tyr, or Phe;
  • citrulline and methionine sulfoxide are neutral nonpolar, cysteic acid is acidic, and ornithine is basic.
  • Proline may be substituted with hydroxyproline and retain the conformation conferring properties.
  • Variants may be generated by substitutional mutagenesis and retain or even increase the biological activity, e.g., immunogenic activity, of the original polypeptide.
  • polypeptides described herein can be obtained, e.g., by chemical synthesis using a commercially available automated peptide synthesizer.
  • the synthesized protein or polypeptide can be obtained, e.g., by chemical synthesis using a commercially available automated peptide synthesizer.
  • the synthesized protein or polypeptide can be obtained, e.g., by chemical synthesis using a commercially available automated peptide synthesizer.
  • the synthesized protein or polypeptide can be obtained, e.g., by chemical synthesis using a commercially available automated peptide synthesizer.
  • the synthesized protein or polypeptide can be obtained, e.g., by chemical synthesis using a commercially available automated peptide synthesizer.
  • the synthesized protein or polypeptide can be obtained, e.g., by chemical synthesis using a commercially available automated peptide synthesizer.
  • the synthesized protein or polypeptide can be obtained, e.
  • proteins and polypeptides can be obtained by recombinant methods, e.g., that are well-known in the art.
  • Polypeptides of the invention may be conjugated to another moiety or particle.
  • polypeptide it may be useful to conjugate the polypeptide to a protein that is immunogenic in the species to be immunized, e.g., keyhole limpet hemocyanin (KLH) , CRM197, tetanus toxoid, diptheria toxoid, serum albumin, bovine thyroglobulin, soybean trypsin inhibitor, or a polycation (poly-L-Lysine or poly-L-arginine) , e.g., using a bifunctional or derivatizing agent as known in the art, for example, maleimidobenzoyl sulfosuccinimide ester (conjugation through cysteine residues), N-hydroxysuccinimide (through lysine residues), glutaraldehyde, or succinic anhydride.
  • KLH keyhole limpet hemocyanin
  • CRM197 tetanus toxoid
  • diptheria toxoid
  • the conjugate may be a recombinant fusion protein, e.g., to facilitate expression and purification of the polypeptide .
  • polypeptides are conjugated to or displayed on a particle, e.g., a phage, a yeast, a virus, a virosome, or a
  • one or more polypeptides may be conjugated to a phage, a yeast, or a virus particle, e.g., to the surface of the particle.
  • a nucleic acid molecule encoding the polypeptide is inserted into the phage, yeast, or virus particle, resulting in expression of the polypeptide in the phage, yeast, or virus, e.g., at the surface of the particle.
  • the phage, yeast, or virus population containing the polypeptide may then be isolated and prepared, e.g., as a vaccine, by adding a pharmaceutically acceptable excipient .
  • polypeptides as described herein are conjugated to a virosome or virus-like particle (VLP) .
  • VLPs generally contain one or more proteins from a virus optionally combined or formulated with a phospholipid. They are generally nonpathogenic, non-replicating and generally do not contain any of the native viral genome. The viral proteins may be recombinantly produced or isolated from whole viruses .
  • Viral proteins suitable for use in virosomes or VLPs include proteins derived from influenza virus (such as HA or NA) , Hepatitis B virus (such as core or capsid proteins), Hepatitis E virus, measles virus, Sindbis virus, Rotavirus, Foot-and- Mouth Disease virus, Retrovirus, Norwalk virus, human Papilloma virus, HIV, RNA-phages, ⁇ -phage (such as coat proteins), GA-phage, fr-phage, AP205 phage, and Ty (such as retrotransposon Ty protein p 1) .
  • Virosomes are discussed further in, e.g., Gluck et al . (2002), Vaccine 20:B10-B16, which is incorporated by reference in its entirety.
  • VLPs are discussed further, e.g., in Niikura et al . (2002), Virology 293:273-280; Lenz et al . (2001), J Immunol 166:5346-5355; Pinto et al . (2003), J Infect Dis 188:327-338; Gerber et al . (2001), Viral 75:4752-4760; WO03/024480; and WO03/024481, each of which is incorporated by reference in its entirety.
  • the invention features monoclonal and polyclonal antibodies that bind to the polypeptides or conjugates described herein.
  • Monoclonal antibodies may be made, e.g., using the hybridoma method first described by Kohler et al., Nature 256:495, 1975, or may be made by recombinant DNA methods (see, e.g., U.S. Patent No.
  • a mouse or other appropriate host animal such as a hamster or macague monkey, is immunized, e.g., using a polypeptide or conjugate described herein, to elicit a polypeptide or conjugate described herein.
  • lymphocytes that produce or are capable of producing antibodies that will specifically bind to the polypeptide or conjugate used for immunization.
  • lymphocytes may be immunized in vitro. Lymphocytes then are fused with myeloma cells using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell (Goding, Monoclonal Antibodies: Principles and Practice, pp. 59-103, Academic Press, 1986) .
  • the hybridoma cells thus prepared are seeded and grown in a suitable culture medium that can contain one or more substances that inhibit the growth or survival of the unfused, parental myeloma cells .
  • a suitable culture medium that can contain one or more substances that inhibit the growth or survival of the unfused, parental myeloma cells .
  • the parental myeloma cells lack the enzyme
  • hypoxanthine guanine phosphoribosyl transferase HGPRT or HPRT
  • the culture medium for the hybridomas typically will include hypoxanthine, aminopterin, and thymidine (HAT medium) , which substances prevent the growth of HGPRT-deficient cells .
  • HAT medium thymidine
  • Exemplary myeloma cells are those that fuse efficiently, support stable high-level production of antibody by the selected antibody- producing cells, and are sensitive to a medium such as HAT medium.
  • particular myeloma cell lines that may be considered for use are murine myeloma lines, such as those derived from MOPC-21 and MPC-11 mouse tumors available from the Salk Institute Cell
  • Culture medium in which hybridoma cells are growing is assayed for production of monoclonal antibodies directed against the antigen.
  • the binding specificity of monoclonal antibodies produced by hybridoma cells can be determined by immunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or enzyme-linked immunoabsorbent assay (ELISA) .
  • RIA radioimmunoassay
  • ELISA enzyme-linked immunoabsorbent assay
  • hybridoma cells After hybridoma cells are identified that produce antibodies of the desired specificity, affinity, and/or activity, clones may be subcloned by limiting dilution procedures and grown by standard methods (Goding, Monoclonal Antibodies: Principles and Practice, pp. 59-103, Academic Press, 1986). Suitable culture media for this purpose include, for example, D-MEM or RPMI-1640 medium. In addition, the hybridoma cells may be grown in vivo as ascites tumors in an animal .
  • the monoclonal antibodies secreted by the subclones are suitably separated from the culture medium, ascites fluid, or serum by
  • immunoglobulin purification procedures such as, for example, protein A-Sepharose, hydroxylapatite chromatography, gel electrophoresis, dialysis, or affinity chromatography.
  • DNA encoding the monoclonal antibodies is readily isolated and seguenced using conventional procedures (e.g., by using
  • oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of the monoclonal antibodies
  • the hybridoma cells serve as a source of such DNA.
  • the DNA may be placed into expression vectors, which are then transfected into host cells such as E. coli cells, simian COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells. Recombinant production of antibodies will be described in more detail below.
  • antibodies or antibody fragments can be isolated from antibody phage libraries generated using the technigues described, for example, in McCafferty et al . , Nature 348:552-554, 1990.
  • the DNA also may be modified, for example, by substituting the coding seguence for human heavy- and light-chain constant domains in place of the homologous murine seguences (U.S. Patent No. 4,816,567; Morrison et al . , Proc. Natl. Acad. Sci. U.S.A. 81:6851, 1984), or by covalently joining to the immunoglobulin coding seguence all or part of the coding seguence for a non-immunoglobulin polypeptide.
  • non-immunoglobulin polypeptides are substituted for the constant domains of an antibody, or they are substituted for the variable domains of one antigen-combining site of an antibody to create a chimeric bivalent antibody comprising one antigen-combining site having specificity for an antigen and another antigen-combining site having specificity for a different antigen.
  • Polyclonal antibodies are typically raised in animals by multiple injections, e.g., subcutaneous or intraperitoneal injections, of the relevant antigen and an adjuvant.
  • KLH keyhole limpet hemocyanin
  • CRM197 tetanus toxoid, diptheria toxoid, serum albumin, bovine thyroglobulin, soybean trypsin inhibitor, or a polycation (poly-L- Lysine or poly-L-arginine) , e.g., using a bifunctional or derivatizing agent as known in the art, for example, maleimidobenzoyl
  • compositions for vaccines and antibody-containing pharmaceutical compositions (collectively "compositions") as described herein can be prepared using standard pharmaceutical formulation chemistries and methodologies that are readily available to the reasonably skilled artisan.
  • polypeptides, conjugates, or antibodies as described herein can be combined with one or more pharmaceutically acceptable excipients or vehicles.
  • auxiliary substances such as wetting or emulsifying agents, pH buffering substances and the like, may be present in the excipient or vehicle.
  • excipients, vehicles and auxiliary substances are generally pharmaceutical agents that do not induce an immune response in the individual receiving the composition, and which may be administered without undue toxicity.
  • Pharmaceutically acceptable excipients include, but are not limited to, liguids such as water, saline, polyethyleneglycol, hyaluronic acid, glycerol and ethanol .
  • Pharmaceutically acceptable salts can also be included therein, for example, mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulfates, and the like; and the salts of organic acids such as acetates, propionates, malonates, benzoates, and the like.
  • mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulfates, and the like
  • organic acids such as acetates, propionates, malonates, benzoates, and the like.
  • compositions may be prepared, packaged, or sold in a form suitable for bolus administration or for continuous administration.
  • injectable compositions may be prepared, packaged, or sold in unit dosage form, such as in ampoules or in multi-dose containers
  • compositions may include, but are not limited to, suspensions, solutions, emulsions in oily or agueous vehicles, pastes, and implantable sustained-release or biodegradable formulations. Such compositions may further comprise one or more additional ingredients including, but not limited to, suspending, stabilizing, or dispersing agents.
  • the active ingredient is provided in dry (for e.g., a powder or granules) form for reconstitution with a suitable vehicle (e.g., sterile pyrogen-free water) prior to
  • compositions may be prepared, packaged, or sold in the form of a sterile injectable agueous or oily suspension or solution.
  • This suspension or solution may be formulated according to the known art, and may comprise, in addition to the active ingredient, additional ingredients such as the dispersing agents, wetting agents, or
  • formulations may be prepared using a non-toxic parenterally-acceptable diluent or solvent, such as water or 1,3-butane diol, for example.
  • a non-toxic parenterally-acceptable diluent or solvent such as water or 1,3-butane diol, for example.
  • Other acceptable diluents and solvents include, but are not limited to, Ringer's solution, isotonic sodium chloride solution, and fixed oils such as synthetic mono- or di-glycerides .
  • compositions for sustained release or implantation may comprise pharmaceutically acceptable polymeric or hydrophobic materials such as an emulsion, an ion exchange resin, a sparingly soluble polymer, or a sparingly soluble salt.
  • pharmaceutically acceptable polymeric or hydrophobic materials such as an emulsion, an ion exchange resin, a sparingly soluble polymer, or a sparingly soluble salt.
  • the polypeptides, conjugates, and antibodies described herein may be encapsulated, adsorbed to, or associated with particulate carriers. Suitable particulate carriers include those derived from polymethyl methacrylate polymers, as well as PLG
  • microparticles derived from poly ( lactides ) and poly ( lactide-co- glycolides) See, e.g., Jeffery et al . (1993) Pharm. Res. 10:362-368.
  • Other particulate systems and polymers can also be used, for example, polymers such as polylysine, polyarginine, polyornithine, spermine, spermidine, as well as conjugates of these molecules.
  • compositions will include an amount of one or more polypeptides or conjugates described herein that is sufficient to mount an immunological response.
  • An immunogenic amount can be readily determined by one of skill in the art. Such an amount will fall in a relatively broad range that can be determined through routine trials.
  • the compositions may contain from about 0.1% to about 99.9% of the polypeptides, conjugates, or antibodies, and can be administered directly to the subject or, alternatively, delivered ex vivo, to cells derived from the subject, using methods known to those skilled in the art .
  • compositions can include a mixture of distinct polypeptides, conjugates, or antibodies as described herein.
  • vaccines may include, e.g., 2, 3, or 4, or even 5, or more distinct
  • polypeptides or conjugates as described herein e.g., containing or consisting of the amino acid seguences disclosed herein, or a variant seguence thereof having up to three substitutions, deletions, or additions to the amino acid seguence of any one of amino acid
  • a vaccine includes eight distinct polypeptides, wherein the amino acid seguence of the eight polypeptides consist of the seguence of the amino acid seguences disclosed herein.
  • antibody-containing polypeptides consist of the seguence of the amino acid seguences disclosed herein.
  • compositions may include a mixture of monoclonal or polyclonal antibodies, e.g., having distinct specificities to
  • Substances that stimulate the immune response may be included in the compositions, e.g., in vaccines.
  • chemical compounds used as adjuvants include, but are not limited to, aluminum compounds (e.g., alum, Alhydrogel) , oils, block polymers, immune stimulating complexes, vitamins and minerals (e.g., vitamin E, vitamin A, selenium, and vitamin B12), Quil A (saponins), bacterial and fungal cell wall components (e.g., lipopolysaccarides ,
  • lipoproteins and glycoproteins
  • hormones include IL-1, IL-2, and others.
  • the invention features methods of vaccinating a mammal against candidiasis including administering to the animal a vaccine as described herein, thereby vaccinating the mammal against candidiasis. Additionally, the invention features methods of passive immunization of a mammal against candidiasis including administering to the mammal an effective amount of a pharmaceutical composition as described herein, thereby passively immunizing the mammal against candidiasis.
  • Candidiasis may include, e.g., disseminated candidiasis, e.g., hematogenously disseminated candidiasis, or mucosal candidiasis.
  • the candidiasis is caused, e.g., by Candida albicans, Candida glabrata, Candida krusei, Candida parapsilosis, or Candida tropicalis .
  • Other Candida species include Candida lusitaniae and Candida stellatoidea .
  • compositions can be administered prophylactically or therapeutically on their own or in combination with other art-known compositions that induce protective responses against pathogens (e.g., viral, bacterial, fungal, or parasitic pathogens), tumors or cancers, allergens, autoimmune disorders, or graft rejection.
  • pathogens e.g., viral, bacterial, fungal, or parasitic pathogens
  • the compositions can be administered simultaneously, separately, or seguentially, e.g., with another immunization vaccine, such as a vaccine for, e.g., influenza, malaria, tuberculosis, smallpox, measles, rubella, mumps, or any other vaccines known in the art .
  • compositions as described herein can be delivered to a mammalian subject (e.g., a human or other mammal described herein) using a variety of known routes and techniques.
  • a composition can be provided as an injectable solution, suspension, or emulsion, and administered via intramuscular, subcutaneous, intradermal, intracavity, parenteral, epidermal, intraarterial, intraperitoneal, or intravenous injection using a conventional needle and syringe, or using a liquid jet injection system.
  • Compositions can also be administered topically to skin or mucosal tissue, such as nasally, intratracheally, intestinal, rectally or vaginally, or provided as a finely divided spray suitable for respiratory or pulmonary
  • compositions described herein can be administered to a mammalian subject (e.g., a human or other mammal described herein) in an amount that is compatible with the dosage formulation and that will be prophylactically and/or therapeutically effective.
  • a mammalian subject e.g., a human or other mammal described herein
  • An appropriate effective amount will fall in a relatively broad range but can be readily determined by one of skill in the art by routine trials.
  • Pharmacological Basis of Therapeutics are useful for the purpose of determining the amount needed.
  • Prophylaxis or therapy can be accomplished by a single direct administration at a single time point or by multiple administrations, optionally at multiple time points. Administration can also be delivered to a single or to multiple sites. Those skilled in the art can adjust the dosage and concentration to suit the particular route of delivery.
  • a single dose is administered on a single occasion.
  • a number of doses are administered to a subject on the same occasion but, for example, at different sites.
  • multiple doses are
  • Such multiple doses may be administered in batches, i.e. with multiple administrations at different sites on the same occasion, or may be administered
  • administration regimes may be used.
  • compositions of the invention may be administered at different sites or on different occasions as part of the same treatment regime.
  • Different administrations may be performed on the same occasion, on the same day, one, two, three, four, five or six days apart, or one, two, three, four or more weeks apart.
  • administrations are 1 to 5 weeks apart, e.g., 2 to 4 weeks apart, such as 2 weeks, 3 weeks or 4 weeks apart.
  • the schedule and timing of such multiple administrations can be optimised for a particular vaccine or pharmaceutical composition by one of skill in the art by routine trials .
  • compositions described herein may vary depending on such factors as preparation method, administration method, age, body weight and sex of the patient, severity of symptoms, administration time, administration route, rate of excretion, and responsivity .
  • a physician of ordinary skill in the art will easily determine and diagnose the administration dose effective for treatment.
  • compositions may be prepared into unit-dose or multiple-dose preparations by those skilled in the art using a pharmaceutically acceptable carrier and/or excipient according to a method known in the art .
  • the invention also provides vectors containing the nucleic acids encoding the polypeptides disclosed herein.
  • Suitable expression vectors are well-known in the art and include vectors capable of expressing a nucleic acid operatively linked to a regulatory seguence or element such as a promoter region or enhancer region that is capable of regulating expression of the nucleic acid.
  • Appropriate expression vectors include vectors that are replicable in eukaryotic cells and/or prokaryotic cells and vectors that remain episomal or integrate into the host cell genome.
  • vector means the vehicle by which a nucleic acid can be introduced into a host cell.
  • the vector can be used for propagation or harboring a nucleic acid or for polypeptide expression of an encoded sequence.
  • vectors include, for example, plasmids, phages and viruses. Exemplary vectors can be found
  • sequences or regulatory elements are operatively linked to a nucleic acid of the invention such that the physical and functional
  • nucleic acid and the regulatory sequence allows transcription of the nucleic acid.
  • Vectors useful for expression in eukaryotic cells can include, for example, regulatory elements including the SV40 early promoter, the cytomegalovirus (CMV) promoter, the mouse mammary tumor virus (MMTV) steroid-inducible promoter, Moloney murine leukemia virus (MMLV) promoter, and the like.
  • the vectors of the invention are useful for subcloning and amplifying a nucleic acid molecule and for recombinantly expressing a polypeptide as disclosed herein.
  • a vector of the invention can include, for example, viral vectors such as a bacteriophage, a baculovirus or a retrovirus; cosmids or plasmids; and, particularly for cloning large nucleic acid molecules, bacterial artificial chromosome vectors (BACs) and yeast artificial chromosome vectors (YACs).
  • viral vectors such as a bacteriophage, a baculovirus or a retrovirus
  • cosmids or plasmids and, particularly for cloning large nucleic acid molecules, bacterial artificial chromosome vectors (BACs) and yeast artificial chromosome vectors (YACs).
  • BACs bacterial artificial chromosome vectors
  • YACs yeast artificial chromosome vectors
  • the invention additionally provides recombinant cells containing nucleic acids of the invention.
  • the recombinant cells are generated by introducing into a host cell a vector containing a nucleic acid molecule.
  • the recombinant cells are transducted, transfected or otherwise genetically modified.
  • Exemplary host cells that can be used to express recombinant molecules include mammalian primary cells; established mammalian cell lines, such as COS, CHO, HeLa, NIH3T3, HEK 293 and PC12 cells; amphibian cells, such as Xenopus embryos and oocytes; and other vertebrate cells.
  • Exemplary host cells also include insect cells such as Drosophila, yeast cells such as
  • Saccharomyces cerevisiae Saccharomyces pombe, or Pichia pastoris
  • prokaryotic cells such as Escherichia coli.
  • Embodiments of the present invention also provide specific Hyrl polypeptides that can act as antigens for generating an immune response to Candida, gram negative rod bacteria including bacteria of the Acinetobacter genus, for example, Acinetobacter baumannii .
  • the polypeptides of the invention include substantially the same amino acid seguence set forth in any one of the amino acid seguences described herein.
  • the amino acid seguence can have at least 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98% or 99% seguence identity to any one of polypeptides described herein.
  • such polypeptides are immunogenic and capable of eliciting production of an anti-Hyrl antibody or immunogenic response in a subject.
  • polypeptides of the invention can encompass substantially similar amino acid seguences having at least about 65% identity with respect to the reference amino acid seguence, and retaining comparable functional and biological activity
  • polypeptides having substantially the same amino acid seguence will have at least 50% or 60% identity, at least 65% identity, at least 70% identity, at least 75% identity, at least 80% identity, at least 85% identity, at least 90% identity, at least 95% identity, at least 98% identity, or at least 99% identity. It is recognized, however, that polypeptides, or encoding nucleic acids, containing less than the described levels of seguence identity arising as splice variants or that are modified by conservative amino acid substitutions, or by substitution of degenerate codons are also encompassed within the scope of the present invention.
  • C. albicans 15663, C. glabrata 31028, C. parapsilosis 22019 and C. tropicalis 4243 are clinical bloodstream isolates collected from
  • C. krusei 91-1159 was generously provided by Michael Rinaldi, San Antonio, TX.
  • C. albicans strains CAAH-31 and THE31 are as described in the literature. All tested strains were routinely grown in YPD (2% Bacto Peptone, 1% yeast extract, 2% dextrose) . Cell densities were determined by counting in a
  • Recombinant polypeptides are generated according to standard methods.
  • the peptides may be purified and conjugated to keyhole limpet hemocyanin (KLH) before raising rabbit antiserum individually using a standard immunization protocol.
  • KLH keyhole limpet hemocyanin
  • Total IgG from pooled serum is affinity purified using Pierce Protein A plus Agarose (Thermo Scientific, Rockford, IL) prior to administering in passive immunization studies.
  • mice are vaccinated with alum alone.
  • mice are vaccinated as above prior to inducing neutropenia by
  • immune IgG is administered
  • mice are given isotype matching IgG (Innovative Research, USA) . IgG doses are repeated 3 days after infection, and survival of mice was monitored twice daily.
  • mice are infected through tail veins. Kidneys are harvested 3 day post infection, homogenized, serially diluted in 0.85% saline, and guantitatively cultured on YPD that contained 50 g/ L chloramphenicol. Colonies are counted after incubation of the plates at 37°C for 24 to 48 h, and results are expressed as log CFU per gram of infected organ.
  • kidneys are removed aseptically from two mice per group for histopathological examination. Kidneys are immersed in zinc formalin fixative until examination. Fixed organs are dehydrated in graded alcohol solutions, embedded in paraffin, and cut into 6- m-thick sections. Mounted sections are stained with Gomori methenamine silver and examined by light microscopy (Davis et al . (2000) Infect Immun 68: 5953-5959).
  • antibody titers of serum samples are collected from vaccinated and control mice are determined by ELISA in 96-well plates as previously described (Ibrahim et al . (2005) Infect Immun 73: 999-1005). Wells are coated at 100 ⁇ per well with a peptide (e.g., one of more of peptide 2-11) at 5 g/ml in PBS. Mouse sera are incubated for 1 h at room temperature following a blocking step with Tris-buffered saline (TBS; 0.01 M Tris HC1 [pH 7.4], 0.15 M NaCl) containing 3% bovine serum albumin.
  • TBS Tris-buffered saline
  • the color is allowed to develop for 30 min, after which the reaction is terminated by addition of 10% H 2 S0 4 and the optical density (OD) at 490 nm is determined in a microtiter plate reader.
  • Negative control wells received only diluent, and background absorbance is subtracted from the test wells to obtain final OD readings.
  • the ELISA titer is taken as the reciprocal of the last serum dilution that gave a positive OD reading (i.e., more than the mean OD of negative control samples plus 2 standard deviations).
  • anti-polypetides e.g., one described herein
  • phagocyte-mediated killing of C. albicans HL-60 cells that have been differentiated to
  • neutrophil-like phenotype neutrophil-like phenotype are employed (Luo et al . , (2010) J Infect Dis 201: 1718-1728) .
  • a killing assay is conducted in the presence of anti-peptide IgG or F(ab ' ) 2 fragments as described before (Luo (2010) J Infect Dis 201: 1718-1728).
  • HL-60 cells are induced with 2.5 ⁇ of retinoic acid and 1.3% DMSO for three days at 37°C with 5%C0 2 .
  • Immune anti-Hyrl polypeptide sera are, if desired, pooled and total IgG is isolated using protein A agarose (Thermo Scientific) .
  • Serum collected from the same rabbits prior to immunization with the polypeptides serves as control serum.
  • the F(ab ' ) 2 fragments from immune or control IgG is purified with Pierce F(ab ' ) 2 Preparation Kit according to the manufacturer's instruction. SDS-PAGE analysis is utilized to indicate >95% of Fc fragment is digested. Next, C.
  • albicans cells overexpressing or suppressing Hyrl is incubated with 50 ⁇ g/ml of vaccinated or control F(ab ' ) 2 fragments on ice for 45 min.
  • C. albicans cocultured with the F(ab ' ) 2 fragments is incubated with HL-60 derived neutrophils for 1 h at 37°C with 5% C0 2 prior to sonication and guantitative culturing on YPD plates.
  • % killing is calculated by dividing the number of CFU after coculturing with HL-60 derived neutrophils by the number of CFU from C. albicans incubated with media without neutrophil-like cells.
  • the nonparametric log rank test is used to determine differences in the survival times of the mice. Neutrophil killing assay, titers of antibody, and tissue fungal burden is compared by the Mann-Whitney U test for unpaired comparisons. Correlations are calculated with the Spearman rank sum test. P values of ⁇ 0.05 are considered significant.
  • Polypeptides that significantly improved survival and decreased fungal burden in immunocompetent mice challenged i.v. with C. albicans are taken as being useful in the invention.
  • polypeptides that statistically protect immunocompromised mice against candidiasis are useful in the invention.
  • Mice protected from fungal infection after receiving purified IgG targeting a polypeptide disclosed herein in a dose specific manner are not only taken as an indication of the usefulness of passive immunization strategies for treating candidiasis but also for the usefulness of the polypeptide antigen used to raise an immune response.
  • Polypeptide vaccines that substantially reduce tissue fungal burden in BALB/c mice challenged with several non- albicans species of Candida are likewise taken as being useful in the invention .
  • overall passive immunization against Acinetobacter baumannii infection may also be assayed in diabetic mice.
  • Purified IgG from the eight different polyclonal antibodies are given to diabetic mice 2 hours prior to infection.
  • Commercially available unrelated rabbit IgG is given to diabetic control mice.
  • the mice are then infected with a lethal dose of Acinetobacter baumannii via tail vein injection.
  • Mice identified as significantly surviving longer after receiving a single dose of the appropriate IgG than mice receiving the control IgG e.g., - 80% survival in the anti- polypeptide IgG vs. 0 % in the control arm, p ⁇ 0.0001 by Log Rank test) are taken as evidence of the effectiveness of the polypeptide antigen.
  • PBMCs are obtained from individuals vaccinated using an Hyrl at various time points following
  • PBMCs Collected PBMCs are stored at -80°C and thawed before use .
  • ELISpot plates coated with antibodies to specific human cytokines or chemokines, e.g. IFN- ⁇ , IL-17A, IL-4, or GRO are used.
  • PBMC samples are then activated in culture for 48 h and are distributed in 96-well ELISpot plates at ⁇ 200,000 cells per well.
  • polypeptides and/or combinations of polypeptides are added to triplicate wells and incubated for 48-96 h and then the supernatants from each well are removed for analysis.
  • the ELISpot plates are developed to reveal the spot forming units per well reflecting the number or cells in the well that produce the compound of interest.
  • Peptides or combination of peptides having an increase in cytokine or chemokine production relative to unstimulated PMBCs are taken as being useful in the invention.

Abstract

The invention features fragments of the Candida cell Hyr1 surface protein useful in immunizing a subject against fungal or bacterial infections or both.

Description

HYR1 COMPOSITIONS AND METHODS FOR TREATING
FUNGAL AND BACTERIAL PATHOGENS
FIELD OF THE INVENTION
The present invention relates generally to compositions and methods for detecting, treating and preventing infectious diseases in a subject.
BACKGROUND OF THE INVENTION
The fungus Candida, the third most common cause of healthcare- associated bloodstream infections, causes approximately 60,000 cases of hematogenously disseminated candidiasis per year in the United States, resulting in billions of dollars of healthcare expenditures. Despite current antifungal therapy, mortality remains unacceptably high. Because of the rising incidence of life-threatening candidiasis and high treatment failure rates, more effective prophylactic and therapeutic strategies are needed.
Lethal infections of antibiotic resistant pathogenic bacteria, like infections resulting from Candida, are becoming increasingly freguent. Moreover, the risk of contracting these lethal infections is extremely high for many at-risk patients in intensive care units (ICUs) every year as well as for soldiers deployed to front line combat zones. Acinetobacter species are a freguent source of
infection in hospitalized patients and soldiers, in particular the species Acinetobacter baumannii . Acinetobacter is a genus of gram negative bacteria belonging to the Gammaproteobacteria . Acinetobacter species contribute to the mineralization of aromatic compounds in the soil. Unfortunately, no technology presently exists that prevents
Acinetobacter infections, aside from standard hand washing and other infection control practices in hospital settings.
There accordingly exists a need for compounds and methods that reduce the risk of infectious diseases related to fungal and bacterial infections and provide effective therapies. The present invention satisfies this need and provides related advantages as well.
SUMMARY OF THE INVENTION
Fragments of the Candida cell surface proteins Hyrl useful in immunizing a subject against fungal and bacterial infections are described below.
Hyrl
The invention features fragments of Hyrl. The amino acid sequence of native C. albicans SC5314 Hyrl polypeptide is as follows :
1 MKWSNFIFTILLTLNLSAA LEWTSRIDRGGIQGFHGDV KVHSGATWAILGTTLCSFFG
61 GLEVEKGASLFIKSDNGPVL ALNVALSTLVRPVINNGVIS LNSKSSTSFSNFDIGGSSFT
121 NNGEIYLDSSGLVKSTAYLY AREWTNNGLIVAYQNQKAAG NIAFGTAYQTITNNGQICLR
181 HQDFVPATKIKGTGCVTADE DTWIKLGNTILSVEPTHNFY LKDSKS SLIVHAVS SNQTFT
241 VHGFGNGNKLGLTLPLTGNR DHFRFEYYPDTGILQLRADA LPQYFKIGKGYDSKLFRIVN
301 SRGLKNAVTYDGPVPNNEIP AVCLIPCTNGPSAPESESDL ΝΤΡΤΤΞΞΙΕΤΞΞΥΞΞΑΑΤΕΞ
361 SVVSESSSAVDSLTSSSLSS KSESSDVVSSTTNIESSSTA IETTMNSESSTDAGSSSISQ
421 ΞΕΞΞΞΤΑΙΤΞΞΞΕΤΞΞΞΕΞΜ SASSTTASNTSIETDSGIVS QSESSSNALSSTEQSITSSP
481 GQSTIYVNSTVTSTITSCDE NKCTEDVVTIFTTVPCSTDC VPTTGDIPMSTSYTQRTVTS
541 TITNCDEVSCSQDVVTYTTN VPHTTVDATTTTTTSTGGDN STGGNESGSNHGPGNGSTEG
601 SGNGSGAGSNEGSQSGPNNG SGSGSEGGSNNGSGSDSGSN NGSGSGSNNGSGSGSTEGSE
661 GGSGSNEGSQSGSGSQPGPN EGSEGGSGSNEGSNHGSNEG SGSGSGSGSNNGSGSGSQSG
721 SGSGSQSGSESGSNSGSNEG SNPGAGNGSNEGSGQGSGNG SEAGSGQGSGPNNGSGSGHN
781 DGSGSGSNQGSNPGAGSGSG SESGSKAGSHSGSNEGAKTD SIEGFHTESKPGFNTGAHTD
841 ATVTGNSVANPVTTSTESDT TISVTVSITSYMTGFDGKPK PFTTVDVIPVPHSMPSNTTD
901 SSSSVPTIDTNENGSSIVTG GKSILFGLIVSMWLFM
Select fragments of Hyrl are as follows .
Hyrl (154-350)
In another aspect, the invention features an isolated polypeptide optionally fused to a heterologous fusion partner, wherein the amino acid sequence of the polypeptide consists of an amino acid sequence havinq at least 95% identity to
QNQKAAG NIAFGTAYQTITNNGQICLR HQDFVPATKIKGTGCVTADE DTWIKLGNTILSVEPTHNFY LKDSKS SLIVHAVS SNQTFT
VHGFGNGNKLGLTLPLTGNR DHFRFEYYPDTGILQLRADA LPQYFKIGKGYDSKLFRIVN SRGLKNAVTYDGPVPNNEIP AVCLIPCTNGPSAPESESDL ΝΤΡΤΤΞΞΙΕΤ ( SEQ ID NO:
Hyrl (201-350)
In another aspect, the invention features an isolated polypeptide optionally fused to a heterologous fusion partner, wherein the amino acid sequence of the polypeptide consists of an amino acid sequence having at least 95% identity to
DTWIKLGNTILSVEPTHNFY LKDSKS SLIVHAVS SNQTFT
VHGFGNGNKLGLTLPLTGNR DHFRFEYYPDTGILQLRADA LPQYFKIGKGYDSKLFRIVN
SRGLKNAVTYDGPVPNNEIP AVCLIPCTNGPSAPESESDL ΝΤΡΤΤΞΞΙΕΤ (SEQ ID NO:
7) .
Hyrl (201-469)
In another aspect, the invention features an isolated polypeptide optionally fused to a heterologous fusion partner, wherein the amino acid sequence of the polypeptide consists of an amino acid sequence having at least 95% identity to
DTWIKLGNTILSVEPTHNFY LKDSKS SLIVHAVS SNQTFT
VHGFGNGNKLGLTLPLTGNR DHFRFEYYPDTGILQLRADA LPQYFKIGKGYDSKLFRIVN SRGLKNAVTYDGPVPNNEIP AVCLIPCTNGPSAPESESDL ΝΤΡΤΤΞΞΙΕΤΞΞΥΞΞΑΑΤΕΞ SVVSESSSAVDSLTSSSLSS KSESSDVVSSTTNIESSSTA IETTMNSESSTDAGSSSISQ ΞΕΞΞΞΤΑΙΤΞΞΞΕΤΞΞΞΕΞΜ SASSTTASNTSIETDSGIVS QSESSSNAL (SEQ ID NO: 9) .
Hyrl (154-469)
In another aspect, the invention features an isolated polypeptide optionally fused to a heterologous fusion partner, wherein the amino acid sequence of said polypeptide consists of an amino acid sequence having at least 95% identity to
QNQKAAG NIAFGTAYQTITNNGQICLR
HQDFVPATKIKGTGCVTADE DTWIKLGNTILSVEPTHNFY LKDSKS SLIVHAVS SNQTFT VHGFGNGNKLGLTLPLTGNR DHFRFEYYPDTGILQLRADA LPQYFKIGKGYDSKLFRIVN SRGLKNAVTYDGPVPNNEIP AVCLIPCTNGPSAPESESDL ΝΤΡΤΤΞΞΙΕΤΞΞΥΞΞΑΑΤΕΞ SVVSESSSAVDSLTSSSLSS KSESSDVVSSTTNIESSSTA IETTMNSESSTDAGSSSISQ ΞΕΞΞΞΤΑΙΤΞΞΞΕΤΞΞΞΕΞΜ SASSTTASNTSIETDSGIVS QSESSSNAL (SEQ ID NO In some embodiments, the polypeptide is SEQ ID NO: 6; SEQ ID NO: 7; SEQ ID NO: 9; or SEQ ID NO: 33.
In another aspect, the invention features an isolated nucleic acid molecule including a nucleic acid seguence which is substantially identical to any of the isolated nucleic acid molecule which encodes any of the polypeptides described herein.
The invention further features a vector including any of the nucleic acid molecules encoding the polypeptides described herein. The invention accordingly also provides vectors containing the nucleic acids of the invention. Suitable expression vectors are well-known in the art and include vectors capable of expressing a nucleic acid operatively linked to a regulatory seguence or element such as a promoter region or enhancer region that is capable of regulating expression of the nucleic acid. Appropriate expression vectors include vectors that are replicable in eukaryotic cells and/or prokaryotic cells and vectors that remain episomal or integrate into the host cell genome.
The invention also provides a method for expression of a
polypeptide as disclosed herein by culturing cells containing a nucleic acid that encodes the polypeptide under conditions suitable for expression of polypeptide. Thus, there is provided a method for the recombinant production of a polypeptide of the invention by expressing the nucleic acid seguences encoding the polypeptide in suitable host cells. Recombinant DNA expression systems that are suitable for production of polypeptides are described herein and are well-known in the art. For example, the above-described nucleotide seguences can be incorporated into vectors for further manipulation. Vectors can include a recombinant DNA or RNA plasmid or virus
containing discrete elements that are used to introduce heterologous DNA into cells for either expression or replication thereof.
Similarly, the invention features a cell including any of the nucleic acid molecules encoding polypeptides described herein.
In another aspect, the invention features a method of producing a recombinant polypeptide, the method including the steps of: (a) providing a cell transformed with the nucleic acid molecule of encoding an polypeptide described herein positioned for expression in the cell; (b) culturing the transformed cell under conditions for expressing the nucleic acid molecule, wherein the culturing results in expression of the recombinant polypeptide; and (c) isolating the recombinant polypeptide. In some embodiments, cell is a bacterium (e.g., E. coli) . In other embodiments, the cell is a yeast cell (e.g., Saccharomyces cerevisae) . In another aspect, the invention features a recombinant polypeptide produced according to this
aforementioned method.
In another aspect, the invention features a substantially pure antibody that specifically recognizes and binds to any one of the polypeptides described herein.
In yet another aspect, the invention features an antigenic composition including the aforementioned polypeptides and a
pharmaceutically acceptable carrier, diluent, and/or excipient . In some embodiments, the composition further includes an adjuvant.
In another aspect, the invention features a method of inducing an immune response in a mammal against an antigen including administering any of the aforementioned polypeptides, or the aforementioned
antigenic composition to the mammal (e.g., a human), wherein the polypeptide or the composition induces an immune response against the antigen in the mammal. Typically, the mammal is administered a single dose of the polypeptide or the composition. In some embodiments, the mammal is administered a plurality of doses of the polypeptide or the composition. In some embodiments, the plurality of doses are
administered at least one day apart (e.g., the plurality of doses are administered at least two weeks apart) . In yet other embodiments, the composition is administered twice.
In another aspect, the invention features a vaccine including an immunogenic amount of any of the aforementioned polypeptides, and a pharmaceutically acceptable excipient. In some embodiments, the vaccine includes a mixture of distinct polypeptides of any one of the aforementioned polypeptides. In some embodiments, the vaccine further includes an adjuvant (Alhydrogel) . The vaccine of the invention is useful for vaccination of a mammal (e.g., a human) against candidiasis or a bacterial infection such as one caused by Acinetobacter or both. Typically, the vaccine is to be administered by intramuscular, subcutaneous, or intradermal administration. The vaccine may also be administered by intramuscular administration. Vaccination may further includes administering a booster dose. Candidiasis may take many forms such as disseminated candidiasis (e.g., hematogenously
disseminated candidiasis) or mucosal candidiasis. Candidiasis is caused, for example, by Candida albicans, Candida glabrata, Candida krusei, Candida parapsilosis, or Candida tropicalis . In some
embodiments, vaccination is against Acinetobacter.
In other aspects, the invention features a method of producing a chimeric vaccine including the steps of: (a) providing a phage, yeast, or virus; (b) inserting into the phage, yeast, or virus a nucleic acid molecule that encodes any of the aforementioned polypeptides; (c) allowing expression of the polypeptide in the phage, yeast, or virus; (d) isolating the phage, yeast, or virus of step (c) including the expressed polypeptide; and (e) adding a pharmaceutically acceptable excipient to the isolated phage, yeast, or virus of step (d) . In some embodiments, the polypeptide is displayed on the surface of the phage, yeast, or virus following step (c).
In other aspects, the invention features an isolated monoclonal antibody that binds to any of the aforementioned polypeptides .
Typically, the antibody is human or humanized. The antibody may also be chimeric. The antibody may also be produced recombinantly . A diagnostic composition including these antibodies is within the invention .
Another aspect of the invention relates to a pharmaceutical composition including any of the aforementioned antibodies alone or in combination, and a pharmaceutically acceptable excipient. In some embodiments, the pharmaceutical composition includes a mixture of antibodies with a plurality of distinct specificities.
In still another aspect, the invention features a pharmaceutical composition including polyclonal antibodies that bind to the any of the polypeptides described herein, or that bind to a mixture of distinct polypeptides of such described polypeptides. In some embodiments, the pharmaceutical composition is for use in the passive immunization of a mammal (e.g., a mammal) against candidiasis or a bacterial infection. Typically, the pharmaceutical composition is administered by intramuscular, subcutaneous, or intradermal
administration. In some embodiments, the pharmaceutical composition is administered by intramuscular administration. In some embodiments, the candidiasis is disseminated candidiasis, for example,
hematogenously disseminated candidiasis. In other embodiments, the candidiasis is mucosal candidiasis. In some embodiments, candidiasis is caused by Candida albicans, Candida glabrata, Candida krusei,
Candida parapsilosis, or Candida tropicalis . In some embodiments, passive immunization is against Acinetobacter.
In another aspect, the invention features a method of passive immunization of a mammal (e.g., a human) against candidiasis or a bacterial infection such as one caused Acinetobacter including administering to the mammal an effective amount of any of the
pharmaceutical compositions disclosed herein, thereby passively immunizing the mammal against the candidiasis. In some embodiments, the pharmaceutical composition is administered by intramuscular, subcutaneous, or intradermal administration. In other embodiments, the pharmaceutical composition is administered by intramuscular administration. In some embodiments, the candidiasis is disseminated candidiasis, for example, hematogenously disseminated candidiasis. In some embodiments, the candidiasis is mucosal candidiasis. In some embodiments, the candidiasis is caused by Candida albicans, Candida glabrata, Candida krusei, Candida parapsilosis, or Candida tropicalis .
In still other aspects, the invention features compositions and methods as disclosed herein that are based, at least in part, on the proposition that an immune response, such as antibodies and other mechanisms, that target the a Candida Hyrl polypeptide and confer protection from Acinetobacter infection such as Acinetobacter
baumannii. Active or passive immunization approaches using the a Hyrl polypeptide fragment disclosed herein are useful to protect against infections caused by gram negative rod bacteria, including, but not limited to, Acinetobacter baumannii . Some uses of the compositions and methods disclosed herein include passive vaccination of acutely at-risk patients with a dose of anti-Hyrl antibody to prevent the acquisition of Acinetobacter baumannii infection. Additionally, patients with active Acinetobacter baumannii infection can be treated with the antibody alone or combined with other antibacterial agents . Alternatively, patients who are at risk of developing such infections, such as, for example, military personnel, can be actively vaccinated with a Hyrl polypeptides disclosed herein to prevent such infections.
In addition to vaccination of subjects susceptible to
Acinetobacter or Candida infections or both, the vaccine compositions of the present invention can also be used to treat,
immunotherapeutically, subjects suffering from a variety of gram negative bacterial infections. Accordingly, vaccines that contain one or more of the polypeptides and/or antibody compositions described herein in combination with adjuvants, can act for the purposes of prophylactic or therapeutic treatment of infections from gram negative bacteria. In one embodiment, vaccines of the present invention will induce the body's own immune system to seek out and inhibit gram negative bacteria or Candida or both.
Vaccines according to the invention refer to a composition that can be administered to an individual to protect the individual against an infectious disease. Vaccines protect against diseases by inducing or increasing an immune response in an animal against the infectious disease. An exemplary infectious disease amenable to treatment with the vaccines of the invention include severe pneumonia, infections of the urinary tract, infections of the bloodstream and infections of other parts of the body. The vaccine-mediated protection can be humoral and/or cell mediated immunity induced in host when a subject is challenged with, for example, or an immunogenic portion of a polypeptide or protein described herein.
Accordingly, in some embodiments, the invention provides a method of treating or preventing an infection from gram negative bacteria in a subject in need thereof by administering a therapeutically effective amount of a pharmaceutical composition as disclosed herein or a vaccine composition as disclosed herein. For example, the invention provides methods of treating or preventing infections caused by one or more gram negative bacteria including bacteria of the Acinetobacter genus, such as A. baumannii, A. iwoffii, A. haemolyticus, A.
calcoaceticus, A. johnsonii, A. radioresistens, and A. junii, bacteria of the Haemophilus genus, such as H. aegyptius, H.
aphrophilus, H. avium, H. ducreyi, H. felis, H. haemolyticus, H.
influenza, H. parainfluenzae, H. paracuniculus, H. parahaemolyticus, H. pittmaniae, and H. somnus, bacteria of the Bordetella genus, such as B. ansorpii, B avium, B. bronchiseptica, B. hinzii, B. holmesii, B. parapertussis, B. pertussis, B. petrii, and B. trematum, bacteria of the Salmonella genus, such as S. typhimurium, S. bongori , S. enterica subsp . enterica, S. enterica subsp. salamae, S. arizonae, S. enterica subsp . diarizonae, S. enterica subsp. houtenae, and S. enterica subsp. indica, bacteria of the Yersina genus, such as Yersina pseudotuber, Y. aldovae, Y. aleksiciae, Y. bercovieri, Y. enterocolitica, Y.
frederiksenii, Y. intermedia, Y. kristensenii, Y. mollaretii, Y.
pestis, Y. pseudotuberculosis, Y. rohdei, and Y. ruckeri, bacteria of the Escherichia genus, such as E. albertii, E. blattae, E. coli, E. fergusonii, E. hermannii and E. vulneris, bacteria of the Pedobacter genus, such as P. heparinus, P. roseus sp. nov. and P. aquatilis sp. nov, bacteria of the Pseudomonas genus, such as P. aeruginosa, P.
alcaligenes, P. mendocina, P. fluorescens, P. monteilii, P.
oryzihabitans, P. luteola, P. putida, P. cepacia, P. stutzeri, P.
maltophilia, P. putrefaciens, P. mallei and P. pseudomallei, or bacteria of the Klebsiella genus, such as K. pneumoniae, K. planticola K. oxytoca and K. rhinoscleromatis . In other embodiments, Candida species as disclosed herein may be treated or prevented.
By "adjuvant" is meant one or more substances that cause stimulation of the immune system. In this context, an adjuvant is used to enhance an immune response to one or more vaccine antigens or antibodies. An adjuvant may be administered to a subject before, in combination with, or after administration of the vaccine or antibody. Examples of chemical compounds used as adjuvants include, but are not limited to, aluminum compounds (e.g., alum, Alhydrogel), oils, block polymers, immune stimulating complexes, vitamins and minerals (e.g., vitamin E, vitamin A, selenium, and vitamin B12) , Quil A (saponins), bacterial and fungal cell wall components (e.g., lipopolysaccarides , lipoproteins, and glycoproteins), hormones, cytokines, and co- stimulatory factors.
By "antibody" is meant whole antibodies, immunoglobulins, or any antigen-binding fragment or single chains thereof. Antibodies, as used herein, can be mammalian (e.g., human or mouse), humanized, chimeric, recombinant, synthetically produced, or naturally isolated, and can be, e.g., monoclonal or polyclonal. In most mammals, including humans, whole antibodies have at least two heavy (H) chains and two light (L) chains connected by disulfide bonds. Each heavy chain consists of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region. The heavy chain constant region consists of three domains, CH1, CH2, and CH3 and a hinge region between CH1 and CH2. Each light chain consists of a light chain variable region (abbreviated herein as VL) and a light chain constant region. The light chain constant region consists of one domain, CL. The VH and VL regions can be further subdivided into regions of hypervariability, termed complementarity determining regions (CDR) , interspersed with regions that are more conserved, termed
framework regions (FR) . Each VH and VL is composed of three CDRs and four FRs, arranged from amino-terminus to carboxy-terminus in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4. The variable regions of the heavy and light chains contain a binding domain that interacts with an antigen. The constant regions of the antibodies may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component (Clg) of the classical complement system.
Antibodies of the present invention include all known forms of antibodies and other protein scaffolds with antibody-like properties. For example, the antibody can be a human antibody, a humanized antibody, a bispecific antibody, a chimeric antibody, or a protein scaffold with antibody-like properties, such as fibronectin or ankyrin repeats. The antibody also can be a Fab, Fab'2, scFv, SMIP, diabody, nanobody, aptamers, or a domain antibody. The antibody can have any of the following isotypes: IgG (e.g., IgGl, IgG2, IgG3, and IgG4), IgM, IgA (e.g., IgAl, IgA2, and IgAsec) , IgD, or IgE.
The term "antibody fragment, " as used herein, refers to one or more fragments of an antibody that retain the ability to specifically bind to an antigen. The antigen-binding function of an antibody can be performed by fragments of a full-length antibody, which include but are not limited to: (i) a Fab fragment, a monovalent fragment consisting of the VL, VH, CL, and CH1 domains; (ii) a F(ab')2 fragment, a bivalent fragment
comprising two Fab fragments linked by a disulfide bridge at the hinge region; (iii) a Fd fragment consisting of the VH and CH1 domains; (iv) a Fv fragment consisting of the VL and VH domains of a single arm of an antibody, (v) a dAb including VH and VL domains; (vi) a dAb fragment (Ward et al . , Nature 341:544-546 (1989)), which consists of a VH domain; (vii) a dAb which consists of a VH or a VL domain; (viii) an isolated
complementarity determining region (CDR) ; and (ix) a combination of two or more isolated CDRs which may optionally be joined by a synthetic linker. Furthermore, although the two domains of the Fv fragment, VL and VH, are coded for by separate genes, they can be joined, using recombinant methods, by a synthetic linker that enables them to be made as a single protein chain in which the VL and VH regions pair to form monovalent molecules (known as single chain Fv (scFv); see e.g., Bird et al.,
Science 242:423-426 (1988) and Huston et al . , Proc. Natl. Acad. Sci . USA 85:5879-5883 (1988)). These antibody fragments are obtained using conventional technigues known to those with skill in the art, and the fragments are screened for utility in the same manner as are intact antibodies. Antibody fragments can be produced by recombinant DNA technigues, or by enzymatic or chemical cleavage of intact
immunoglobulins .
By "antigen" is meant a molecule to which an antibody can
selectively bind. The target antigen may be a protein (e.g., an
antigenic peptide) , carbohydrate, nucleic acid, lipid, hapten, or other naturally occurring or synthetic compound. The target antigen may be a polypeptide or peptide mimic. An antigen may also be administered to an animal to generate an immune response in the animal .
By "carrier" in the context of a conjugate is meant a moiety or particle, e.g., KLH, CRM197, tetanus toxoid, a phage, a yeast, a virus, a virosome, or a recombinant virus-like particle, that is suitable for being linked to or displaying a polypeptide as described herein .
By "chimeric antibody" is meant an immunoglobulin or antibody whose variable regions derive from a first species and whose constant regions derive from a second species. Chimeric antibodies can be constructed, for example, by genetic engineering, from immunoglobulin gene segments belonging to different species (e.g., from a mouse and a human) .
By "chimeric vaccine" is meant a vaccine that includes at least two distinct antigens, e.g., joined covalently. An example of a chimeric vaccine is a composition that includes a polypeptide
displayed, e.g., on the surface of a particle such as a phage, virus, yeast, virosome, or recombinant virus-like particle.
By "conjugate" is meant a compound that includes a polypeptide of the invention linked to another moiety or particle, e.g., KLH, CRM197, tetanus toxoid, a phage, a yeast, a virus, a virosome, or a
recombinant virus-like particle.
By "conservative substitution" in an amino acid seguence is meant replacement of an amino acid for another within a family of amino acids that are related in the chemical nature of their side chains.
Genetically encoded amino acids can be divided into four
families: acidic (aspartate, glutamate) ; basic (lysine, arginine, histidine) ; nonpolar (alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan) ; and uncharged polar (glycine, asparagine, glutamine, cysteine, serine, threonine, tyrosine) .
Phenylalanine, tryptophan, and tyrosine are sometimes grouped as aromatic amino acids. In similar fashion, the amino acids can also be separated into the following groups: acidic (aspartate, glutamate); basic (lysine, arginine, histidine); alipathic (glycine, alanine, valine, leucine, isoleucine, serine, threonine), with serine and threonine optionally grouped separately as alipathic-hydroxyl ;
aromatic (phenylalanine, tyrosine, tryptophan) ; amide (asparagine, glutamine) ; and sulfur-containing (cysteine, methionine).
Whether a change in the amino acid seguence results in a functional variant can be determined by assessing the ability of the variant polypeptide to function in a fashion similar to the wild-type polypeptide using standard methods such as those described herein.
By "diagnostic composition" is meant a composition containing a polypeptide, conjugate, vaccine, or antibody of the invention, formulated for use in conjunction with a diagnostic method.
By "effective amount" in the context of passive immunization using a pharmaceutical composition, e.g., comprising an antibody, is meant the amount of the pharmaceutical composition reguired to passively immunize in a clinically relevant manner. An effective amount of pharmaceutical composition used to practice the methods of passive immunization described herein varies depending upon the manner of administration, the age, body weight, and general health of the subject. Ultimately, the prescribers will decide the appropriate amount and dosage regimen.
By "flanking amino acid" is meant an amino acid in a polypeptide seguence that is immediately adjacent to the N- or C-terminus of a particular defined seguence. Desirably, a flanking amino acid is present on the N- and/or C-terminus of the amino acid seguence of SC5314 Hyrl or rHyrl or a fragment thereof; and more desirably, 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 flanking amino acids are present at the N- and/or C-terminus of the amino acid seguence of SC5314 Hyrl or rHyrl, or fragment thereof .
By "fusion protein" is meant a protein that includes a
polypeptide of the invention, e.g., a peptide fragment or variant, and a fusion partner.
By "fusion partner" is meant a heterologous seguence that can be fused to a polypeptide or peptide of the invention , e.g., one or more of Peptide 3-11 or variants thereof. Examples of fusion partners are described herein and include detection markers, stabilizing domains, sequences which aid in production or purification of the protein, or domains which increase the antigenicity of the polypeptide.
By "immunogenic" is meant any substance that is capable of inducing an immune response in a subject.
By "immunogenic amount" in the context of a vaccine is meant an amount of the vaccine required to induce an immune response in a subject in a clinically relevant manner. An immunogenic amount of vaccine used to practice the methods of vaccination as described herein varies depending upon the manner of administration, the age, body weight, and general health of the subject. Ultimately, the prescribers will decide the appropriate amount and dosage regimen.
By "isolated" or "purified" is meant separated from other naturally accompanying components. Typically, a compound (e.g., nucleic acid, polypeptide, antibody, or small molecule) is
substantially isolated when it is at least 60%, by weight, free from the proteins and/or naturally occurring organic molecules with which it is naturally associated. The definition also extends, e.g., to a polypeptide or nucleic acid molecule separated from its flanking sequences (e.g., for an amino acid sequence, isolated refers to a sequence that is free from the flanking amino acids with which the sequence is naturally associated in a polypeptide) . In some
instances, the compound is at least 75%, more preferably at least 90%, and most preferably at least 99%, by weight, isolated. An isolated compound, e.g., polypeptide, may be obtained by standard techniques, for example, by extraction from a natural source (e.g., purification from a cell infected with Candida) ; by expression of a recombinant nucleic acid encoding an Hyrl or variant, or a fusion protein thereof; or by chemically synthesizing the polypeptide. Purity can be measured by any appropriate method, e.g., by column chromatography,
polyacrylamide gel electrophoresis, or HPLC analysis.
By "linked to" or "conjugated to" in the context of a conjugate is meant a covalent or non-covalent interaction between the
polypeptide and the carrier or fusion partner. Non-covalent
interactions include, but are not limited to, hydrogen bonding, ionic interactions among charged groups, electrostatic binding, van der Waals interactions, hydrophobic interactions among non-polar groups, lipophobic interactions, and LogP-based attractions.
By "monoclonal antibody" is meant an antibody obtained from a population of substantially homogeneous antibodies, i.e., the
individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single antigenic site. Furthermore, in contrast to conventional (polyclonal) antibody preparations which typically include different antibodies directed against different determinants (epitopes), each monoclonal antibody is directed against a single determinant on the antigen. Monoclonal antibodies can be prepared using any art recognized technigue and those described herein such as, for example, a hybridoma method, as described by Kohler et al . , Nature 256:495 (1975), a transgenic animal (e.g., Lonberg et al . , Nature 368 (6474) : 856-859 (1994)), recombinant DNA methods (e.g., U.S. Pat.
No. 4,816,567), or using phage, yeast, or synthetic scaffold antibody libraries using the technigues described in, for example, Clackson et al., Nature 352:624-628 (1991) and Marks et al . , J. Mol. Biol.
222 :581-597 (1991) .
By "nucleic acid molecule" is meant a molecule, e.g., RNA or DNA, having a seguence of two or more covalently bonded, naturally
occurring or modified nucleotides. The nucleic acid molecule may be, e.g., single or double stranded, and may include modified or
unmodified nucleotides, or mixtures or combinations thereof. Various salts, mixed salts, and free acid forms are also included.
By "patient" or "subject" is meant a mammal, including, but not limited to, a human or non-human mammal, such as a bovine, eguine, canine, ovine, or feline.
The terms "peptide," "polypeptide," and "protein" are used interchangeably and refer to any chain of two or more natural or unnatural amino acids, regardless of post-translational modification (e.g., glycosylation or phosphorylation), constituting all or part of a naturally-occurring or non-naturally occurring polypeptide or peptide, as is described herein. Such polypeptides typically are continuous and unbranched peptide. A peptide is a short polymer of amino acid monomers. "Proteins" are intended to include one or more polypeptides arranged in a biologically functional way. The amino acids comprising the polypeptides of the invention may be linked by peptide bonds or other bonds, for example, ester or ether bonds. The amino acids comprising the polypeptides of the invention can include non-genetically coded amino acids that either occur naturally or are chemically synthesized.
A polypeptide of the invention can also encompass one or more conservative substitutions . Conservative substitutions of encoded amino acids include, for example, amino acids that belong within the following groups: (1) non-polar amino acids (Gly, Ala, Val, Leu, and lie); (2) polar neutral amino acids (Cys, Met, Ser, Thr, Asn, and Gin); (3) polar acidic amino acids (Asp and Glu) ; (4) polar basic amino acids (Lys, Arg and His); and (5) aromatic amino acids (Phe, Trp, Tyr, and His) . Other minor modifications are also included within polypeptides of the invention so long as the polypeptide retains some or all of its function as described herein.
The invention polypeptides can also include derivatives, analogues and functional mimetics thereof, provided that such
polypeptide retains some or all of its function as disclosed herein. For example, derivatives can include chemical modifications of the polypeptide such as alkylation, acylation, carbamylation, iodination, or any modification that derivatizes the polypeptide. Such
derivatized molecules include, for example, those molecules in which free amino groups have been derivatized to form amine hydrochlorides, p-toluene sulfonyl groups, carbobenzoxy groups, t-butyloxycarbonyl groups, chloroacetyl groups or formyl groups. Free carboxyl groups can be derivatized to form salts, methyl and ethyl esters or other types of esters or hydrazides. Free hydroxyl groups can be
derivatized to form O-acyl or O-alkyl derivatives. The imidazole nitrogen of histidine can be derivatized to form N-im-benzylhistidine . Also included as derivatives or analogues are those peptides which contain one or more naturally occurring amino acid derivatives of the twenty standard amino acids, for example, 4-hydroxyproline, 5-hydroxylysine, 3-methylhistidine, homoserine, ornithine or carboxyglutamate, and can include amino acids that are not linked by peptide bonds. Polypeptides of the present invention also include any polypeptide having one or more additions and/or deletions of residues, relative to the seguence of a polypeptide whose seguence is shown herein, so long as immunogenic activity as disclosed herein is maintained .
The invention polypeptides can be isolated by a variety of methods well-known in the art, for example, recombinant expression systems, precipitation, gel filtration, ion-exchange, reverse-phase and affinity chromatography, and the like. Other well-known methods are described in Deutscher et al . , Guide to Protein Purification:
Methods in Enzymology Vol. 182, (Academic Press, (1990)).
Alternatively, the isolated polypeptides of the present invention can be obtained using well-known recombinant methods (see, for example, Ausubel et al . , "Immunology," Short Protocols in Molecular Biology, John Wiley & Sons, Inc. Chapter 11. Page 11.1-11.29 (1999); Sambrook and Russell, "Molecular Cloning: A Laboratory Manual," Cold Spring Harbor Laboratory (2001)). The methods and conditions for biochemical purification of a polypeptide of the invention can be chosen by those skilled in the art, and purification monitored, for example, by an immunological assay or a functional assay.
An example of the means for preparing an invention polypeptide is to express nucleic acids encoding a polypeptide of the invention in a suitable host cell, such as a bacterial cell, a yeast cell, an amphibian cell such as an oocyte, or a mammalian cell, using methods well known in the art, and recovering the expressed polypeptide, again using well-known purification methods, so described herein. Invention polypeptides can be isolated directly from cells that have been transformed with expression vectors as described herein. The
invention polypeptides can also be produced by chemical synthesis.
Methods for chemically synthesizing polypeptides are well known in the art and are commercially available.
Recombinantly expressed polypeptides of the invention can also be expressed as fusion proteins with appropriate fusion partners. An appropriate fusion partner can be an amino acid sequence that is not normally connected to the amino acid sequence such as an heterologous sequence, which serves a particular function or provides additional characteristic to the polypeptides of the invention. Non-limiting examples of suitable heterologous seguences include a detectable marker, a stabilizing domain, a carrier protein for the generation of antibodies, a linker seguence and a seguence that aids in the
purification of the polypeptide. Seguences that can aid in the purification of the invention polypeptides include affinity tags, such as glutathione S transferase (GST) or poly His. Thus, in some aspects, the invention provide a fusion protein having a polypeptide as disclosed herein fused to a heterologous seguence, a carrier protein, an affinity tag or a linker seguence or other polypeptides as disclosed herein.
As used herein, a natural amino acid is a natural -amino acid having the L-configuration, such as those normally occurring in natural polypeptides. Unnatural amino acid refers to an amino acid that normally does not occur in polypeptides, e.g., an epimer of a natural a-amino acid having the L configuration, that is to say an amino acid having the unnatural D-configuration; or a (D, L) -isomeric mixture thereof; or a homolog of such an amino acid, for example, a β- amino acid, an a, -disubstituted amino acid, or an a-amino acid wherein the amino acid side chain has been shortened by one or two methylene groups or lengthened to up to 10 carbon atoms, such as an a- amino alkanoic acid with 5 up to and including 10 carbon atoms in a linear chain, an unsubstituted or substituted aromatic (a-aryl or a- aryl lower alkyl), for example, a substituted phenylalanine or phenylglycine .
The terms "pharmaceutically acceptable carrier" and
"pharmaceutically acceptable excipient" are used interchangeably and mean a carrier or excipient that is physiologically acceptable to the treated patient while retaining the therapeutic properties of the compound with which it is administered. One exemplary
pharmaceutically acceptable carrier substance is physiological saline. Other physiologically acceptable carriers and their formulations are known to those skilled in the art and described, for example, in
Remington's Pharmaceutical Sciences, (20th edition), ed. A. Gennaro, 2000, Lippincott, Williams & Wilkins, Philadelphia, PA.
By "pharmaceutical composition" is meant a composition containing a polypeptide, conjugate, vaccine, or antibody of the invention, formulated with a pharmaceutically acceptable excipient, and
manufactured or sold with the approval of a governmental regulatory agency as part of a therapeutic regimen for the treatment or
prevention of a disease or event in a mammal. Pharmaceutical
compositions can be formulated, for example, for intravenous
administration (e.g., as a sterile solution free of particulate emboli and in a solvent system suitable for intravenous use) , for oral administration (e.g., a tablet, capsule, caplet, gelcap, or syrup), or any other formulation described herein, e.g., in unit dosage form.
By "specifically binds" is meant the preferential association of a binding moiety (e.g., an antibody, antibody fragment, receptor, ligand, or small molecule portion of an agent as described herein) to a target molecule (e.g., a polypeptide or conjugate including same) or to a cell or tissue bearing the target molecule (e.g., a cell surface antigen, such as a receptor or ligand) and not to non-target
molecules, cells, or tissues lacking the target molecule. It is recognized that a certain degree of non-specific interaction may occur between a binding moiety and a non-target molecule (present alone or in combination with a cell or tissue) . Nevertheless, specific binding may be distinguished as mediated through specific recognition of the target molecule. Specific binding results in a stronger association between the binding moiety (e.g., an antibody) and the target molecule (e.g., a polypeptide or conjugate including same) than between the binding moiety and, e.g., non-target molecules or other compositions lacking the target molecule. Specific binding typically results in greater than 2-fold, preferably greater than 5-fold, more preferably greater than 10-fold and most preferably greater than 100-fold increase in amount of bound binding moiety (per unit time) to e.g., a cell or tissue bearing the target molecule or marker as compared to a cell or tissue lacking that target molecule or marker. Binding moieties bind to the target molecule or marker with a dissociation constant of e.g., less than 1CT6M, less than 1CT7M, 1CT8M, 1CT9M, 1CT10M, 10_11 , or 1CT12M, or even less than 1CT13M, 1CT14M, or 10~15M. Specific binding to a protein under such conditions requires a binding moiety that is selected for its specificity for that particular protein. A variety of assay formats are appropriate for selecting binding moieties (e.g., antibodies) capable of specifically binding to a particular target molecule. For example, solid-phase ELISA
immunoassays are routinely used to select monoclonal antibodies specifically immunoreactive with a protein. See Harlow & Lane,
Antibodies, A Laboratory Manual, Cold Spring Harbor Publications, New York (1988), for a description of immunoassay formats and conditions that can be used to determine specific immunoreactivity .
By "substantially identical" is meant an amino acid sequence or nucleic acid sequence that exhibits at least 50% identity to a reference sequence. Such a sequence is generally at least, e.g., 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% identical at the amino acid level or nucleic acid level to a reference sequence. In general, for polypeptides, the length of comparison sequences can be at least five amino acids, e.g., 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 125, 150, 175, 200, 250, 300, or more amino acids, up to the entire length of the polypeptide. For nucleic acids, the length of comparison sequences can generally be at least 10, 20, 30, 40, 50, 60, 70, 80, 90, 100, 125, 150, 175, 200, 250, 300, 400, 500, 600, 700, 800, 900, or more nucleotides, up to the entire length of the nucleic acid molecule. It is understood that for the purposes of determining sequence identity when comparing a DNA sequence to an RNA sequence, a thymine nucleotide is equivalent to a uracil nucleotide.
As used herein, when a polypeptide or nucleic acid sequence is referred to as having "at least X% sequence identity" to a reference sequence, it is meant that at least X percent of the amino acids or nucleotides in the polypeptide or nucleic acid are identical to those of the reference sequence when the sequences are optimally aligned. An optimal alignment of sequences can be determined in various ways that are within the skill in the art, for instance, the Smith Waterman alignment algorithm (Smith et al . , J. Mol . Biol. 147:195-7, 1981) and BLAST (Basic Local Alignment Search Tool; Altschul et al . , J. Mol. Biol. 215: 403-10, 1990). These and other alignment algorithms are accessible using publicly available computer software such as "Best Fit" (Smith and Waterman, Advances in Applied Mathematics, 482-489, 1981) as incorporated into GeneMatcher PlusTM (Schwarz and Dayhof,
Atlas of Protein Seguence and Structure, Dayhoff, M.O., Ed pp 353-358, 1979), BLAST, BLAST-2, BLAST-P, BLAST-N, BLAST-X, WU-BLAST-2, ALIGN, ALIGN-2, CLUSTAL, or Megalign (DNASTAR) . In addition, those skilled in the art can determine appropriate parameters for measuring
alignment, including any algorithms needed to achieve optimal
alignment over the length of the seguences being compared.
A "target molecule" or "target cell" is meant a molecule (e.g., a polypeptide, epitope, antigen, receptor, or ligand) or cell to which a binding moiety (e.g., an antibody) can specifically bind. In some instances, target molecules are exposed on the exterior of a target cell (e.g., a cell surface or secreted protein), but target molecules may alternately or also be present in the interior of a target cell.
By "treating" or "treatment" is meant the medical management of a patient with the intent to cure, ameliorate, stabilize, reduce the likelihood of, or prevent a disease, pathological condition, disorder, or event, by administering a pharmaceutical composition. This term includes active treatment, that is, treatment directed specifically toward the improvement or associated with the cure of a disease, pathological condition, disorder, or event, and also includes causal treatment, that is, treatment directed toward removal of the cause of the associated disease, pathological condition, disorder, or event. In addition, this term includes palliative treatment, that is, treatment designed for the relief of symptoms rather than the curing of the disease, pathological condition, disorder, or event;
symptomatic treatment, that is, treatment directed toward
constitutional symptoms of the associated disease, pathological condition, disorder, or event; preventative treatment, that is, treatment directed to minimizing or partially or completely inhibiting the development of the associated disease, pathological condition, disorder, or event, e.g., in a patient who is not yet ill, but who is susceptible to, or otherwise at risk of, a particular disease, pathological condition, disorder, or event; and supportive treatment, that is, treatment employed to supplement another specific therapy directed toward the improvement of the associated disease,
pathological condition, disorder, or event.
By "vaccine," as used herein, is meant a composition that elicits an immune response in a subject to which it is administered.
By "vaccinate, " as used herein, is meant to treat a patient by administering a vaccine, e.g., to prevent or ameliorate a disease, pathological condition, disorder, or event.
By "variant" in the context of a polypeptide or portion thereof as described herein, or a nucleic acid molecule encoding same, is meant to include substitutions or alterations in the amino acid seguence or nucleic acid seguence, e.g., resulting in a substantially identical seguence. A polypeptide having a variant seguence may maintain at least one biological activity of the original polypeptide, e.g., immunogenic activity. The term "variant" includes, e.g., amino acid insertional derivatives such as amino and/or carboxylterminal fusions, as well as intraseguence insertions of single or multiple amino acids. Insertional amino acid variants are those in which one or more amino acid residues are introduced into a predetermined site in the protein. Random insertion is also possible with suitable screening of the resulting product. Deletional variants are
characterized by removal of one or more amino acids from the seguence. Substitutional amino acid variants are those in which at least one residue inserted in its place. Where the protein is derivatized by amino acid substitution, amino acids are generally replaced by conservative substitutions, e.g., other amino acids having similar physical chemical properties such as hydrophobicity, hydrophilicity, electronegativity, bulky sidechains and the like.
For purposes of the present invention, variants also include single or multiple substitutions, deletions and/or additions of any component (s) naturally or artificially associated with the portion of a naturally occurring protein from which the polypeptide may be derived, such as carbohydrate, lipid and/or other proteinaceous moieties. All such molecules are encompassed by the term "variant."
By "variant sequence" is meant the amino acid or nucleic acid sequence of a variant as defined herein.
Other features and advantages of the invention will be apparent from the following Detailed Description and the claims.
DETAILED DESCRIPTION OF THE INVENTION
The identification of the Hyrl polypeptide fragments and other compositions described herein allow, e.g., for the effective treatment of and vaccination against candidiasis or bacterial infections such as those caused by any pathogen disclosed herein.
The invention provides polypeptides, e.g., derived from Hyrl, conjugates, vaccines, antibodies, compositions, methods of vaccination using same, and methods of production of same, as described in further detail below.
Polypeptides
The invention features polypeptides derived from SC5413 Hyrl or rHyrl : Hyrl
The amino acid sequence of native C. albicans SC5314 Hyrl polypeptide is as follows:
1 MKWSNFIFTILLTLNLSAA LEWTSRIDRGGIQGFHGDV KVHSGATWAILGTTLCSFFG
61 GLEVEKGASLFIKSDNGPVL ALNVALSTLVRPVINNGVIS LNSKSSTSFSNFDIGGSSFT
121 NNGEIYLDSSGLVKSTAYLY AREWTNNGLIVAYQNQKAAG NIAFGTAYQTITNNGQICLR
181 HQDFVPATKIKGTGCVTADE DTWIKLGNTILSVEPTHNFY LKDSKS SLIVHAVS SNQTFT
241 VHGFGNGNKLGLTLPLTGNR DHFRFEYYPDTGILQLRADA LPQYFKIGKGYDSKLFRIVN
301 SRGLKNAVTYDGPVPNNEIP AVCLIPCTNGPSAPESESDL ΝΤΡΤΤΞΞΙΕΤΞΞΥΞΞΑΑΤΕΞ
361 SVVSESSSAVDSLTSSSLSS KSESSDVVSSTTNIESSSTA IETTMNSESSTDAGSSSISQ
421 ΞΕΞΞΞΤΑΙΤΞΞΞΕΤΞΞΞΕΞΜ SASSTTASNTSIETDSGIVS QSESSSNALSSTEQSITSSP
481 GQSTIYVNSTVTSTITSCDE NKCTEDVVTIFTTVPCSTDC VPTTGDIPMSTSYTQRTVTS
541 TITNCDEVSCSQDVVTYTTN VPHTTVDATTTTTTSTGGDN STGGNESGSNHGPGNGSTEG
601 SGNGSGAGSNEGSQSGPNNG SGSGSEGGSNNGSGSDSGSN NGSGSGSNNGSGSGSTEGSE
661 GGSGSNEGSQSGSGSQPGPN EGSEGGSGSNEGSNHGSNEG SGSGSGSGSNNGSGSGSQSG
721 SGSGSQSGSESGSNSGSNEG SNPGAGNGSNEGSGQGSGNG SEAGSGQGSGPNNGSGSGHN
781 DGSGSGSNQGSNPGAGSGSG SESGSKAGSHSGSNEGAKTD SIEGFHTESKPGFNTGAHTD 841 ATVTGNSVANPVTTSTESDT TISVTVSITSYMTGFDGKPK PFTTVDVIPVPHSMPSNTTD
901 SSSSVPTIDTNENGSSIVTGG KSILFGLIVSMVVLFM
The amino acid sequence of rHyrl polypeptide is:
The amino acid sequence of the rHyrl protein is as follows:
1 TSRI DRGGI QGFHGDVKVHS GATWAI LGTTLCSFFGGLEV EKGASLF IKSDNGPVLALNV
61 ALSTLVRPVINNGVI SLNSK SSTSFSNFDIGGSSFTNNGE IYLDSSGLVKSTAYLYAREW
121 TNNGLIVAYQNQKAAGNIAF GTAYQTITNNGQICLRHQDF VPATKIKGTGCVTADEDTWI
181 KLGNT I LSVEPTHNFYLKDS KSSLIVHAVSSNQTFTVHGF GNGNKLGLTLPLTGNRDHFR
241 FEYYPDTGI LQLRADALPQY FKIGKGYDSKLFRIVNSRGL KNAVTYDGPVPNNEIPAVCL
301 IPCTNGPSAPESESDLNTPT TSSIETSSYSSAATESSWS ESSSAVDSLTSSSLSSKSES
361 SDWSSTTNIESSSTAIETT MNSESSTDAGSSSISQSESS STAITSSSETSSSESMSASS
401 TTASNTSIETDSGIVSQSES SSNAL Select fraqments of Hyrl are as follows.
Hyrl (154-350)
The amino acid sequence of Hyrl (154-350) is :
QNQKAAG NIAFGTAYQTITNNGQICLR HQDFVPATKIKGTGCVTADE DTWIKLGNTILSVEPTHNFY LKDSKS SLIVHAVS SNQTFT
VHGFGNGNKLGLTLPLTGNR DHFRFEYYPDTGILQLRADA LPQYFKIGKGYDSKLFRIVN SRGLKNAVTYDGPVPNNEIP AVCLIPCTNGPSAPESESDL ΝΤΡΤΤΞΞΙΕΤ ( SEQ ID NO:
Hyrl (201-350)
The amino acid se ce of Hyrl (154-350) is :
DTWIKLGNTILSVEPTHNFY LKDSKS SLIVHAVS SNQTFT
VHGFGNGNKLGLTLPLTGNR DHFRFEYYPDTGILQLRADA LPQYFKIGKGYDSKLFRIVN SRGLKNAVTYDGPVPNNEIP AVCLIPCTNGPSAPESESDL ΝΤΡΤΤΞΞΙΕΤ (SEQ ID NO: 7) .
Hyrl (201-469)
The amino acid sequence of Hyrl (201-469) is:
DTWIKLGNTILSVEPTHNFY LKDSKS SLIVHAVS SNQTFT VHGFGNGNKLGLTLPLTGNR DHFRFEYYPDTGILQLRADA LPQYFKIGKGYDSKLFRIVN SRGLKNAVTYDGPVPNNEIP AVCLIPCTNGPSAPESESDL ΝΤΡΤΤΞΞΙΕΤΞΞΥΞΞΑΑΤΕΞ SVVSESSSAVDSLTSSSLSS KSESSDVVSSTTNIESSSTA IETTMNSESSTDAGSSSISO ΞΕΞΞΞΤΑΙΤΞΞΞΕΤΞΞΞΕΞΜ SASSTTASNTSIETDSGIVS QSESSSNAL (SEQ ID NO: 9) .
Hyrl (154-469)
The amino acid sequence of Hyrl (154-469) is
QNQKAAG NIAFGTAYQTITNNGQICLR
HQDFVPATKIKGTGCVTADE DTWIKLGNTILSVEPTHNFY LKDSKS SLIVHAVS SNQTFT VHGFGNGNKLGLTLPLTGNR DHFRFEYYPDTGILQLRADA LPQYFKIGKGYDSKLFRIVN SRGLKNAVTYDGPVPNNEIP AVCLIPCTNGPSAPESESDL ΝΤΡΤΤΞΞΙΕΤΞΞΥΞΞΑΑΤΕΞ SVVSESSSAVDSLTSSSLSS KSESSDVVSSTTNIESSSTA IETTMNSESSTDAGSSSISQ ΞΕΞΞΞΤΑΙΤΞΞΞΕΤΞΞΞΕΞΜ SASSTTASNTSIETDSGIVS QSESSSNAL (SEQ ID NO
In some instances, a modification to a polypeptide as described herein does not substantially reduce the biological activity, e.g., immunogenic activity, of the polypeptide. The modified polypeptide may have or may optimize a characteristic of a polypeptide, such as in vivo stability, bioavailability, toxicity, immunological activity, immunological identity, or conjugation properties.
Modifications include those by natural processes, such as posttranslational processing, or by chemical modification techniques known in the art. Modifications may occur anywhere in a polypeptide including the polypeptide backbone, the amino acid side chains, and the amino- or carboxy-terminus . The same type of modification may be present in the same or varying degrees at several sites in a given polypeptide, and a polypeptide may contain more than one type of modification .
A variant or otherwise modified polypeptide can also include one or more amino acid insertions, deletions, or substitutions, either conservative or non-conservative (e.g., D-amino acids, desamino acids) in the polypeptide sequence. For example, the addition of one or more cysteine residues to the amino or carboxy terminus of any of the polypeptides of the invention can facilitate conjugation of these polypeptides. Exemplary polypeptides having an N- or C-terminal cysteine .
Amino acid substitutions can be conservative (i.e., wherein a residue is replaced by another of the same general type or group) or non-conservative (i.e., wherein a residue is replaced by an ammo acid of another type) . In addition, a non-naturally occurring amino acid can be substituted for a naturally occurring amino acid (i.e., non- naturally occurring conservative amino acid substitution or a non- naturally occurring non-conservative amino acid substitution) .
Polypeptides made synthetically, e.g., using methods known in the art, can include substitutions of amino acids not naturally encoded by DNA (e.g., non-naturally occurring or unnatural amino acid). Examples of non-naturally occurring amino acids include D-amino acids, an amino acid having an acetylaminomethyl group attached to a sulfur atom of a cysteine, a pegylated amino acid, the omega amino acids of the formula NH2 (CH2) nCOOH wherein n is 2-6, neutral nonpolar amino acids, such as sarcosine, t-butyl alanine, t-butyl glycine, N-methyl isoleucine, and norleucine. Phenylglycine may substitute for Trp, Tyr, or Phe;
citrulline and methionine sulfoxide are neutral nonpolar, cysteic acid is acidic, and ornithine is basic. Proline may be substituted with hydroxyproline and retain the conformation conferring properties.
Variants may be generated by substitutional mutagenesis and retain or even increase the biological activity, e.g., immunogenic activity, of the original polypeptide.
The polypeptides described herein can be obtained, e.g., by chemical synthesis using a commercially available automated peptide synthesizer. The synthesized protein or polypeptide can be
precipitated and further purified, for example by high performance liguid chromatography (HPLC) . Alternatively, the proteins and polypeptides can be obtained by recombinant methods, e.g., that are well-known in the art.
Conjugates
Polypeptides of the invention may be conjugated to another moiety or particle.
Protein moieties
In some instances, it may be useful to conjugate the polypeptide to a protein that is immunogenic in the species to be immunized, e.g., keyhole limpet hemocyanin (KLH) , CRM197, tetanus toxoid, diptheria toxoid, serum albumin, bovine thyroglobulin, soybean trypsin inhibitor, or a polycation (poly-L-Lysine or poly-L-arginine) , e.g., using a bifunctional or derivatizing agent as known in the art, for example, maleimidobenzoyl sulfosuccinimide ester (conjugation through cysteine residues), N-hydroxysuccinimide (through lysine residues), glutaraldehyde, or succinic anhydride.
In some instances, the conjugate may be a recombinant fusion protein, e.g., to facilitate expression and purification of the polypeptide .
Particles for Conjugation or Display of Polypeptides
In some instances, polypeptides are conjugated to or displayed on a particle, e.g., a phage, a yeast, a virus, a virosome, or a
recombinant virus-like particle.
For example, one or more polypeptides may be conjugated to a phage, a yeast, or a virus particle, e.g., to the surface of the particle. In one embodiment, a nucleic acid molecule encoding the polypeptide is inserted into the phage, yeast, or virus particle, resulting in expression of the polypeptide in the phage, yeast, or virus, e.g., at the surface of the particle. The phage, yeast, or virus population containing the polypeptide may then be isolated and prepared, e.g., as a vaccine, by adding a pharmaceutically acceptable excipient .
In some embodiments, polypeptides as described herein are conjugated to a virosome or virus-like particle (VLP) . Virosomes and VLPs generally contain one or more proteins from a virus optionally combined or formulated with a phospholipid. They are generally nonpathogenic, non-replicating and generally do not contain any of the native viral genome. The viral proteins may be recombinantly produced or isolated from whole viruses . Viral proteins suitable for use in virosomes or VLPs include proteins derived from influenza virus (such as HA or NA) , Hepatitis B virus (such as core or capsid proteins), Hepatitis E virus, measles virus, Sindbis virus, Rotavirus, Foot-and- Mouth Disease virus, Retrovirus, Norwalk virus, human Papilloma virus, HIV, RNA-phages, Οβ-phage (such as coat proteins), GA-phage, fr-phage, AP205 phage, and Ty (such as retrotransposon Ty protein p 1) . Virosomes are discussed further in, e.g., Gluck et al . (2002), Vaccine 20:B10-B16, which is incorporated by reference in its entirety.
VLPs are discussed further, e.g., in Niikura et al . (2002), Virology 293:273-280; Lenz et al . (2001), J Immunol 166:5346-5355; Pinto et al . (2003), J Infect Dis 188:327-338; Gerber et al . (2001), Viral 75:4752-4760; WO03/024480; and WO03/024481, each of which is incorporated by reference in its entirety.
Antibodies
The invention features monoclonal and polyclonal antibodies that bind to the polypeptides or conjugates described herein.
Monoclonal Antibodies
Monoclonal antibodies may be made, e.g., using the hybridoma method first described by Kohler et al., Nature 256:495, 1975, or may be made by recombinant DNA methods (see, e.g., U.S. Patent No.
4,816,567). In the hybridoma method, a mouse or other appropriate host animal, such as a hamster or macague monkey, is immunized, e.g., using a polypeptide or conjugate described herein, to elicit
lymphocytes that produce or are capable of producing antibodies that will specifically bind to the polypeptide or conjugate used for immunization. Alternatively, lymphocytes may be immunized in vitro. Lymphocytes then are fused with myeloma cells using a suitable fusing agent, such as polyethylene glycol, to form a hybridoma cell (Goding, Monoclonal Antibodies: Principles and Practice, pp. 59-103, Academic Press, 1986) .
The hybridoma cells thus prepared are seeded and grown in a suitable culture medium that can contain one or more substances that inhibit the growth or survival of the unfused, parental myeloma cells . For example, if the parental myeloma cells lack the enzyme
hypoxanthine guanine phosphoribosyl transferase (HGPRT or HPRT) , the culture medium for the hybridomas typically will include hypoxanthine, aminopterin, and thymidine (HAT medium) , which substances prevent the growth of HGPRT-deficient cells . Exemplary myeloma cells are those that fuse efficiently, support stable high-level production of antibody by the selected antibody- producing cells, and are sensitive to a medium such as HAT medium. Among these, particular myeloma cell lines that may be considered for use are murine myeloma lines, such as those derived from MOPC-21 and MPC-11 mouse tumors available from the Salk Institute Cell
Distribution Center, San Diego, CA, USA, and SP-2 or X63-Ag8-653 cells available from the American Type Culture Collection, Manassas, VA, USA. Human myeloma and mouse-human heteromyeloma cell lines also have been described for the production of human monoclonal antibodies (Kozbor, J. Immunol. 133:3001, 1984; Brodeur et al . , Monoclonal
Antibody Production Technigues and Applications, pp. 51-63, Marcel Dekker, Inc., New York, 1987).
Culture medium in which hybridoma cells are growing is assayed for production of monoclonal antibodies directed against the antigen. The binding specificity of monoclonal antibodies produced by hybridoma cells can be determined by immunoprecipitation or by an in vitro binding assay, such as radioimmunoassay (RIA) or enzyme-linked immunoabsorbent assay (ELISA) .
After hybridoma cells are identified that produce antibodies of the desired specificity, affinity, and/or activity, clones may be subcloned by limiting dilution procedures and grown by standard methods (Goding, Monoclonal Antibodies: Principles and Practice, pp. 59-103, Academic Press, 1986). Suitable culture media for this purpose include, for example, D-MEM or RPMI-1640 medium. In addition, the hybridoma cells may be grown in vivo as ascites tumors in an animal .
The monoclonal antibodies secreted by the subclones are suitably separated from the culture medium, ascites fluid, or serum by
conventional immunoglobulin purification procedures such as, for example, protein A-Sepharose, hydroxylapatite chromatography, gel electrophoresis, dialysis, or affinity chromatography.
DNA encoding the monoclonal antibodies is readily isolated and seguenced using conventional procedures (e.g., by using
oligonucleotide probes that are capable of binding specifically to genes encoding the heavy and light chains of the monoclonal antibodies) . The hybridoma cells serve as a source of such DNA. Once isolated, the DNA may be placed into expression vectors, which are then transfected into host cells such as E. coli cells, simian COS cells, Chinese hamster ovary (CHO) cells, or myeloma cells that do not otherwise produce immunoglobulin protein, to obtain the synthesis of monoclonal antibodies in the recombinant host cells. Recombinant production of antibodies will be described in more detail below.
In a further embodiment, antibodies or antibody fragments can be isolated from antibody phage libraries generated using the technigues described, for example, in McCafferty et al . , Nature 348:552-554, 1990.
Clackson et al., Nature 352:624-628, 1991 and Marks et al., J. Mol . Biol. 222:581-597, 1991, describe the isolation of murine and human antibodies, respectively, using phage libraries. Subseguent publications describe the production of high affinity (nM range) human antibodies by chain shuffling (Marks et al . , Bio/Technology 10:779- 783, 1992), as well as combinatorial infection and in vivo
recombination as a strategy for constructing very large phage
libraries (Waterhouse et al . , Nucl . Acids. Res. 21:2265-2266, 1993). Thus, these technigues are viable alternatives to traditional
monoclonal antibody hybridoma technigues for isolation of monoclonal antibodies .
The DNA also may be modified, for example, by substituting the coding seguence for human heavy- and light-chain constant domains in place of the homologous murine seguences (U.S. Patent No. 4,816,567; Morrison et al . , Proc. Natl. Acad. Sci. U.S.A. 81:6851, 1984), or by covalently joining to the immunoglobulin coding seguence all or part of the coding seguence for a non-immunoglobulin polypeptide.
Typically, such non-immunoglobulin polypeptides are substituted for the constant domains of an antibody, or they are substituted for the variable domains of one antigen-combining site of an antibody to create a chimeric bivalent antibody comprising one antigen-combining site having specificity for an antigen and another antigen-combining site having specificity for a different antigen. Polyclonal Antibodies
Polyclonal antibodies are typically raised in animals by multiple injections, e.g., subcutaneous or intraperitoneal injections, of the relevant antigen and an adjuvant. In some instances, it may be useful to conjugate the polypeptide to a protein that is immunogenic in the species to be immunized, e.g., keyhole limpet hemocyanin (KLH) ,
CRM197, tetanus toxoid, diptheria toxoid, serum albumin, bovine thyroglobulin, soybean trypsin inhibitor, or a polycation (poly-L- Lysine or poly-L-arginine) , e.g., using a bifunctional or derivatizing agent as known in the art, for example, maleimidobenzoyl
sulfosuccinimide ester (conjugation through cysteine residues), N- hydroxysuccinimide (through lysine residues), glutaraldehyde, or succinic anhydride.
Vaccines and Antibody-Containing Pharmaceutical Compositions
Formulations for vaccines and antibody-containing pharmaceutical compositions (collectively "compositions") as described herein can be prepared using standard pharmaceutical formulation chemistries and methodologies that are readily available to the reasonably skilled artisan. For example, polypeptides, conjugates, or antibodies as described herein can be combined with one or more pharmaceutically acceptable excipients or vehicles. Auxiliary substances, such as wetting or emulsifying agents, pH buffering substances and the like, may be present in the excipient or vehicle. These excipients, vehicles and auxiliary substances are generally pharmaceutical agents that do not induce an immune response in the individual receiving the composition, and which may be administered without undue toxicity. Pharmaceutically acceptable excipients include, but are not limited to, liguids such as water, saline, polyethyleneglycol, hyaluronic acid, glycerol and ethanol . Pharmaceutically acceptable salts can also be included therein, for example, mineral acid salts such as hydrochlorides, hydrobromides, phosphates, sulfates, and the like; and the salts of organic acids such as acetates, propionates, malonates, benzoates, and the like. A thorough discussion of pharmaceutically acceptable excipients, vehicles and auxiliary substances is available in Remington's Pharmaceutical Sciences (Mack Pub. Co., N.J. 1991).
Such compositions may be prepared, packaged, or sold in a form suitable for bolus administration or for continuous administration. Injectable compositions may be prepared, packaged, or sold in unit dosage form, such as in ampoules or in multi-dose containers
containing a preservative. Compositions may include, but are not limited to, suspensions, solutions, emulsions in oily or agueous vehicles, pastes, and implantable sustained-release or biodegradable formulations. Such compositions may further comprise one or more additional ingredients including, but not limited to, suspending, stabilizing, or dispersing agents. In one embodiment of a composition for parenteral administration, the active ingredient is provided in dry (for e.g., a powder or granules) form for reconstitution with a suitable vehicle (e.g., sterile pyrogen-free water) prior to
parenteral administration of the reconstituted composition. The compositions may be prepared, packaged, or sold in the form of a sterile injectable agueous or oily suspension or solution. This suspension or solution may be formulated according to the known art, and may comprise, in addition to the active ingredient, additional ingredients such as the dispersing agents, wetting agents, or
suspending agents described herein. Such sterile injectable
formulations may be prepared using a non-toxic parenterally-acceptable diluent or solvent, such as water or 1,3-butane diol, for example. Other acceptable diluents and solvents include, but are not limited to, Ringer's solution, isotonic sodium chloride solution, and fixed oils such as synthetic mono- or di-glycerides .
Other parentally-administrable compositions that are useful include those which comprise the active ingredient in macrocrystalline form, in a liposomal preparation, or as a component of a biodegradable polymer systems. Compositions for sustained release or implantation may comprise pharmaceutically acceptable polymeric or hydrophobic materials such as an emulsion, an ion exchange resin, a sparingly soluble polymer, or a sparingly soluble salt. Alternatively, the polypeptides, conjugates, and antibodies described herein may be encapsulated, adsorbed to, or associated with particulate carriers. Suitable particulate carriers include those derived from polymethyl methacrylate polymers, as well as PLG
microparticles derived from poly ( lactides ) and poly ( lactide-co- glycolides) . See, e.g., Jeffery et al . (1993) Pharm. Res. 10:362-368. Other particulate systems and polymers can also be used, for example, polymers such as polylysine, polyarginine, polyornithine, spermine, spermidine, as well as conjugates of these molecules.
The formulated compositions will include an amount of one or more polypeptides or conjugates described herein that is sufficient to mount an immunological response. An immunogenic amount can be readily determined by one of skill in the art. Such an amount will fall in a relatively broad range that can be determined through routine trials. The compositions may contain from about 0.1% to about 99.9% of the polypeptides, conjugates, or antibodies, and can be administered directly to the subject or, alternatively, delivered ex vivo, to cells derived from the subject, using methods known to those skilled in the art .
Compositions can include a mixture of distinct polypeptides, conjugates, or antibodies as described herein. For example, vaccines may include, e.g., 2, 3, or 4, or even 5, or more distinct
polypeptides or conjugates as described herein, e.g., containing or consisting of the amino acid seguences disclosed herein, or a variant seguence thereof having up to three substitutions, deletions, or additions to the amino acid seguence of any one of amino acid
seguences disclosed herein. In one embodiment, a vaccine includes eight distinct polypeptides, wherein the amino acid seguence of the eight polypeptides consist of the seguence of the amino acid seguences disclosed herein. In another embodiment, antibody-containing
pharmaceutical compositions may include a mixture of monoclonal or polyclonal antibodies, e.g., having distinct specificities to
polypeptides or conjugates as described herein.
Substances that stimulate the immune response, e.g., adjuvants, may be included in the compositions, e.g., in vaccines. Examples of chemical compounds used as adjuvants include, but are not limited to, aluminum compounds (e.g., alum, Alhydrogel) , oils, block polymers, immune stimulating complexes, vitamins and minerals (e.g., vitamin E, vitamin A, selenium, and vitamin B12), Quil A (saponins), bacterial and fungal cell wall components (e.g., lipopolysaccarides ,
lipoproteins, and glycoproteins), hormones, cytokines, and co- stimulatory factors.
Methods of Treatment
The invention features methods of vaccinating a mammal against candidiasis including administering to the animal a vaccine as described herein, thereby vaccinating the mammal against candidiasis. Additionally, the invention features methods of passive immunization of a mammal against candidiasis including administering to the mammal an effective amount of a pharmaceutical composition as described herein, thereby passively immunizing the mammal against candidiasis. Candidiasis may include, e.g., disseminated candidiasis, e.g., hematogenously disseminated candidiasis, or mucosal candidiasis. In some instances, the candidiasis is caused, e.g., by Candida albicans, Candida glabrata, Candida krusei, Candida parapsilosis, or Candida tropicalis . Other Candida species include Candida lusitaniae and Candida stellatoidea .
Vaccines and antibody-containing pharmaceutical compositions (collectively "compositions") as described herein can be administered prophylactically or therapeutically on their own or in combination with other art-known compositions that induce protective responses against pathogens (e.g., viral, bacterial, fungal, or parasitic pathogens), tumors or cancers, allergens, autoimmune disorders, or graft rejection. For example, the compositions can be administered simultaneously, separately, or seguentially, e.g., with another immunization vaccine, such as a vaccine for, e.g., influenza, malaria, tuberculosis, smallpox, measles, rubella, mumps, or any other vaccines known in the art .
Compositions as described herein can be delivered to a mammalian subject (e.g., a human or other mammal described herein) using a variety of known routes and techniques. For example, a composition can be provided as an injectable solution, suspension, or emulsion, and administered via intramuscular, subcutaneous, intradermal, intracavity, parenteral, epidermal, intraarterial, intraperitoneal, or intravenous injection using a conventional needle and syringe, or using a liquid jet injection system. Compositions can also be administered topically to skin or mucosal tissue, such as nasally, intratracheally, intestinal, rectally or vaginally, or provided as a finely divided spray suitable for respiratory or pulmonary
administration. Other modes of administration include oral
administration, suppositories, and active or passive transdermal delivery techniques.
The compositions described herein can be administered to a mammalian subject (e.g., a human or other mammal described herein) in an amount that is compatible with the dosage formulation and that will be prophylactically and/or therapeutically effective. An appropriate effective amount will fall in a relatively broad range but can be readily determined by one of skill in the art by routine trials. The "Physicians Desk Reference" and "Goodman and Gilman's The
Pharmacological Basis of Therapeutics" are useful for the purpose of determining the amount needed.
Prophylaxis or therapy can be accomplished by a single direct administration at a single time point or by multiple administrations, optionally at multiple time points. Administration can also be delivered to a single or to multiple sites. Those skilled in the art can adjust the dosage and concentration to suit the particular route of delivery. In one embodiment, a single dose is administered on a single occasion. In an alternative embodiment, a number of doses are administered to a subject on the same occasion but, for example, at different sites. In a further embodiment, multiple doses are
administered on multiple occasions. Such multiple doses may be administered in batches, i.e. with multiple administrations at different sites on the same occasion, or may be administered
individually, with one administration on each of multiple occasions (optionally at multiple sites) . Any combination of such
administration regimes may be used.
In one embodiment, different compositions of the invention may be administered at different sites or on different occasions as part of the same treatment regime.
Different administrations may be performed on the same occasion, on the same day, one, two, three, four, five or six days apart, or one, two, three, four or more weeks apart. In some instances, administrations are 1 to 5 weeks apart, e.g., 2 to 4 weeks apart, such as 2 weeks, 3 weeks or 4 weeks apart. The schedule and timing of such multiple administrations can be optimised for a particular vaccine or pharmaceutical composition by one of skill in the art by routine trials .
Dosages
An adeguate dose of the vaccines or antibody-containing
pharmaceutical compositions described herein may vary depending on such factors as preparation method, administration method, age, body weight and sex of the patient, severity of symptoms, administration time, administration route, rate of excretion, and responsivity . A physician of ordinary skill in the art will easily determine and diagnose the administration dose effective for treatment.
Compositions may be prepared into unit-dose or multiple-dose preparations by those skilled in the art using a pharmaceutically acceptable carrier and/or excipient according to a method known in the art .
Vectors
The invention also provides vectors containing the nucleic acids encoding the polypeptides disclosed herein. Suitable expression vectors are well-known in the art and include vectors capable of expressing a nucleic acid operatively linked to a regulatory seguence or element such as a promoter region or enhancer region that is capable of regulating expression of the nucleic acid. Appropriate expression vectors include vectors that are replicable in eukaryotic cells and/or prokaryotic cells and vectors that remain episomal or integrate into the host cell genome.
The terms "vector", "cloning vector" and "expression vector" mean the vehicle by which a nucleic acid can be introduced into a host cell. The vector can be used for propagation or harboring a nucleic acid or for polypeptide expression of an encoded sequence. A wide variety of vectors are known in the art and include, for example, plasmids, phages and viruses. Exemplary vectors can be found
described in, for example, Sambrook et al., "Molecular Cloning: A Laboratory Manual," 3rd Edition. Cold Spring Harbor Laboratories, Cold Spring Harbor, N.Y., 2001; and Ausubel et al . , "Current Protocols in Molecular Biology," John Wiley and Sons, Baltimore, MD (1999)).
Promoters or enhancers, depending upon the nature of the
regulation, can be constitutive or regulated. The regulatory
sequences or regulatory elements are operatively linked to a nucleic acid of the invention such that the physical and functional
relationship between the nucleic acid and the regulatory sequence allows transcription of the nucleic acid.
Vectors useful for expression in eukaryotic cells can include, for example, regulatory elements including the SV40 early promoter, the cytomegalovirus (CMV) promoter, the mouse mammary tumor virus (MMTV) steroid-inducible promoter, Moloney murine leukemia virus (MMLV) promoter, and the like. The vectors of the invention are useful for subcloning and amplifying a nucleic acid molecule and for recombinantly expressing a polypeptide as disclosed herein. A vector of the invention can include, for example, viral vectors such as a bacteriophage, a baculovirus or a retrovirus; cosmids or plasmids; and, particularly for cloning large nucleic acid molecules, bacterial artificial chromosome vectors (BACs) and yeast artificial chromosome vectors (YACs). Such vectors are commercially available, and their uses are well known in the art. One skilled in the art will know or can readily determine an appropriate promoter for expression in a particular host cell.
The invention additionally provides recombinant cells containing nucleic acids of the invention. The recombinant cells are generated by introducing into a host cell a vector containing a nucleic acid molecule. The recombinant cells are transducted, transfected or otherwise genetically modified. Exemplary host cells that can be used to express recombinant molecules include mammalian primary cells; established mammalian cell lines, such as COS, CHO, HeLa, NIH3T3, HEK 293 and PC12 cells; amphibian cells, such as Xenopus embryos and oocytes; and other vertebrate cells. Exemplary host cells also include insect cells such as Drosophila, yeast cells such as
Saccharomyces cerevisiae, Saccharomyces pombe, or Pichia pastoris, and prokaryotic cells such as Escherichia coli.
Embodiments of the present invention also provide specific Hyrl polypeptides that can act as antigens for generating an immune response to Candida, gram negative rod bacteria including bacteria of the Acinetobacter genus, for example, Acinetobacter baumannii .
In some aspects of the invention, the polypeptides of the invention include substantially the same amino acid seguence set forth in any one of the amino acid seguences described herein. For example, the amino acid seguence can have at least 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98% or 99% seguence identity to any one of polypeptides described herein. In other aspects, such polypeptides are immunogenic and capable of eliciting production of an anti-Hyrl antibody or immunogenic response in a subject.
As described herein, the polypeptides of the invention can encompass substantially similar amino acid seguences having at least about 65% identity with respect to the reference amino acid seguence, and retaining comparable functional and biological activity
characteristic of the reference amino acid seguence. In one aspect, polypeptides having substantially the same amino acid seguence will have at least 50% or 60% identity, at least 65% identity, at least 70% identity, at least 75% identity, at least 80% identity, at least 85% identity, at least 90% identity, at least 95% identity, at least 98% identity, or at least 99% identity. It is recognized, however, that polypeptides, or encoding nucleic acids, containing less than the described levels of seguence identity arising as splice variants or that are modified by conservative amino acid substitutions, or by substitution of degenerate codons are also encompassed within the scope of the present invention.
ASSESSMENT
The following examples are intended to illustrate the invention. These are not meant to limit the invention in any way.
Methods and materials for evaluating treatment of candidiasis
Candida strains and growth conditions
C. albicans 15663, C. glabrata 31028, C. parapsilosis 22019 and C. tropicalis 4243 are clinical bloodstream isolates collected from
Harbor-UCLA Medical Center. C. krusei 91-1159 was generously provided by Michael Rinaldi, San Antonio, TX. C. albicans strains CAAH-31 and THE31 are as described in the literature. All tested strains were routinely grown in YPD (2% Bacto Peptone, 1% yeast extract, 2% dextrose) . Cell densities were determined by counting in a
hemacytometer .
Recombinant polypeptides and rabbit polyclonal antibodies
Recombinant polypeptides are generated according to standard methods. To generate antibodies, the peptides may be purified and conjugated to keyhole limpet hemocyanin (KLH) before raising rabbit antiserum individually using a standard immunization protocol. Total IgG from pooled serum is affinity purified using Pierce Protein A plus Agarose (Thermo Scientific, Rockford, IL) prior to administering in passive immunization studies.
Immunization protocol and animal studies
All active vaccinations are conducted according to standard methods. In brief, juvenile (10-12 week) Balb/C mice are vaccinated subcutaneously with 30 μg of a polypeptide mixed with alum (2%
Alhydrogel; Brenntag Biosector, Frederikssund, Denmark) as an adjuvant in phosphate buffered saline (PBS) on day 0, boosted with the same dose on day 21, then infected via the tail vein on day 35. Control mice are vaccinated with alum alone. To test the efficacy of the vaccine in immunocompromised mice, mice are vaccinated as above prior to inducing neutropenia by
intraperitoneal injection of 200 mg/kg of cyclophosphamide on day -2 followed by another dose of 100 mg/kg on day +7 relative to infection. This regimen results in approximately 10 days of leucopenia with reduction in neutrophil, lymphocyte and monocyte counts according to standard methods . For both immunocompetent and neutropenic mice differences in survival between vaccinated and adjuvant vaccinated mice are compared by the Log Rank test.
For passive immunization, immune IgG is administered
intraperitoneally to naive mice 2 h before infecting i.v. with C.
albicans . Control mice are given isotype matching IgG (Innovative Research, USA) . IgG doses are repeated 3 days after infection, and survival of mice was monitored twice daily.
Quantitative culturing of kidneys from vaccinated or control mice to be infected with different species of Candida is performed
according to standard methods. In brief, mice are infected through tail veins. Kidneys are harvested 3 day post infection, homogenized, serially diluted in 0.85% saline, and guantitatively cultured on YPD that contained 50 g/ L chloramphenicol. Colonies are counted after incubation of the plates at 37°C for 24 to 48 h, and results are expressed as log CFU per gram of infected organ.
Concomitant with the fungal burden experiment, kidneys are removed aseptically from two mice per group for histopathological examination. Kidneys are immersed in zinc formalin fixative until examination. Fixed organs are dehydrated in graded alcohol solutions, embedded in paraffin, and cut into 6- m-thick sections. Mounted sections are stained with Gomori methenamine silver and examined by light microscopy (Davis et al . (2000) Infect Immun 68: 5953-5959).
Enzyme-linked immunosorbent assay (ELISA)
To test if a polypeptide induces an immune response, antibody titers of serum samples are collected from vaccinated and control mice are determined by ELISA in 96-well plates as previously described (Ibrahim et al . (2005) Infect Immun 73: 999-1005). Wells are coated at 100 μΐ per well with a peptide (e.g., one of more of peptide 2-11) at 5 g/ml in PBS. Mouse sera are incubated for 1 h at room temperature following a blocking step with Tris-buffered saline (TBS; 0.01 M Tris HC1 [pH 7.4], 0.15 M NaCl) containing 3% bovine serum albumin. The wells are then washed three times with TBS containing 0.05% Tween 20, followed by another three washes with TBS. Goat anti-mouse secondary antibody conjugated with horseradish peroxidase (Sigma) is added at a final dilution of 1:5000, and the plate is further incubated for 1 h at room temperature. Wells are then washed with TBS and incubated with substrate containing 0.1 M citrate buffer (pH 5.0), 50 mg of o- phenylenediamine (Sigma), and 10 μΐ of 30% H202. The color is allowed to develop for 30 min, after which the reaction is terminated by addition of 10% H2S04 and the optical density (OD) at 490 nm is determined in a microtiter plate reader. Negative control wells received only diluent, and background absorbance is subtracted from the test wells to obtain final OD readings. The ELISA titer is taken as the reciprocal of the last serum dilution that gave a positive OD reading (i.e., more than the mean OD of negative control samples plus 2 standard deviations).
F(ab') 2 blocking assay
To study the mechanism of protection mediated by anti-polypetides (e.g., one described herein) antibodies in phagocyte-mediated killing of C. albicans, HL-60 cells that have been differentiated to
neutrophil-like phenotype are employed (Luo et al . , (2010) J Infect Dis 201: 1718-1728) . A killing assay is conducted in the presence of anti-peptide IgG or F(ab') 2 fragments as described before (Luo (2010) J Infect Dis 201: 1718-1728). In brief, HL-60 cells are induced with 2.5 μΜ of retinoic acid and 1.3% DMSO for three days at 37°C with 5%C02. Immune anti-Hyrl polypeptide sera are, if desired, pooled and total IgG is isolated using protein A agarose (Thermo Scientific) . Serum collected from the same rabbits prior to immunization with the polypeptides serves as control serum. The F(ab') 2 fragments from immune or control IgG is purified with Pierce F(ab')2 Preparation Kit according to the manufacturer's instruction. SDS-PAGE analysis is utilized to indicate >95% of Fc fragment is digested. Next, C.
albicans cells overexpressing or suppressing Hyrl is incubated with 50 μg/ml of vaccinated or control F(ab') 2 fragments on ice for 45 min. C. albicans cocultured with the F(ab') 2 fragments is incubated with HL-60 derived neutrophils for 1 h at 37°C with 5% C02 prior to sonication and guantitative culturing on YPD plates. % killing is calculated by dividing the number of CFU after coculturing with HL-60 derived neutrophils by the number of CFU from C. albicans incubated with media without neutrophil-like cells.
Statistical analysis
The nonparametric log rank test is used to determine differences in the survival times of the mice. Neutrophil killing assay, titers of antibody, and tissue fungal burden is compared by the Mann-Whitney U test for unpaired comparisons. Correlations are calculated with the Spearman rank sum test. P values of <0.05 are considered significant.
Expected Results
Polypeptides that significantly improved survival and decreased fungal burden in immunocompetent mice challenged i.v. with C. albicans are taken as being useful in the invention. Similarly, polypeptides that statistically protect immunocompromised mice against candidiasis are useful in the invention. Mice protected from fungal infection after receiving purified IgG targeting a polypeptide disclosed herein in a dose specific manner are not only taken as an indication of the usefulness of passive immunization strategies for treating candidiasis but also for the usefulness of the polypeptide antigen used to raise an immune response. Polypeptide vaccines that substantially reduce tissue fungal burden in BALB/c mice challenged with several non- albicans species of Candida are likewise taken as being useful in the invention .
Methods and materials for evaluating treatment of an Acinteobacter infection
Recombinant polypeptides disclosed herein are produces according to standard methods, for example, using E. coli expression system. The recombinant polypeptide is then used to actively vaccinate mice. Mice, for example, are immunized with aluminum hydroxide alone or the recombinant polypeptide mixed with aluminum hydroxide (n=9) on day 0, and boosted on day 21. The vaccinated mice are subsequently infected with A. baumannii on day 35. Polypeptide vaccines providing
statistically significant survival compared to control mice are taken as useful in the invention. Additionally, measurement of bacterial burden in the tissue of mice vaccinated and infected similarly are examined. The bacterial burden as measured by the number of colony forming units per gram of tissue showing that tissue isolates from kidney, lung and spleen have a lower bacterial burden as compared to control tissue samples are also taken as indicative of useful
polypeptide vaccines.
In another working example, overall passive immunization against Acinetobacter baumannii infection may also be assayed in diabetic mice. Purified IgG from the eight different polyclonal antibodies are given to diabetic mice 2 hours prior to infection. Commercially available unrelated rabbit IgG is given to diabetic control mice. The mice are then infected with a lethal dose of Acinetobacter baumannii via tail vein injection. Mice identified as significantly surviving longer after receiving a single dose of the appropriate IgG than mice receiving the control IgG (e.g., - 80% survival in the anti- polypeptide IgG vs. 0 % in the control arm, p<0.0001 by Log Rank test) are taken as evidence of the effectiveness of the polypeptide antigen.
Additional assessment utilizing human PBMCs
Useful polypeptide antigens described herein are also identified using standard human PBMCs. PBMCs are obtained from individuals vaccinated using an Hyrl at various time points following
vaccination. Collected PBMCs are stored at -80°C and thawed before use .
For an assay, ELISpot plates coated with antibodies to specific human cytokines or chemokines, e.g. IFN-γ, IL-17A, IL-4, or GRO are used. PBMC samples are then activated in culture for 48 h and are distributed in 96-well ELISpot plates at ~200,000 cells per well.
Specific polypeptides and/or combinations of polypeptides are added to triplicate wells and incubated for 48-96 h and then the supernatants from each well are removed for analysis. The ELISpot plates are developed to reveal the spot forming units per well reflecting the number or cells in the well that produce the compound of interest. Peptides or combination of peptides having an increase in cytokine or chemokine production relative to unstimulated PMBCs are taken as being useful in the invention.
Other Embodiments
All publications, patents, and patent applications mentioned in the above specification are hereby incorporated by reference. Various modifications and variations of the described methods of the invention will be apparent to those skilled in the art without departing from the scope and spirit of the invention. Although the invention has been described in connection with specific embodiments, it should be understood that the invention as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the described modes for carrying out the invention that are obvious to those skilled in the art are intended to be within the scope of the invention .
Other embodiments are in the claims.
What is claimed is:

Claims

Claims
1. An isolated polypeptide optionally fused to a heterologous fusion partner, wherein the amino acid seguence of said polypeptide consists of an amino acid seguence having at least 95% identity to
QNQKAAG NIAFGTAYQTITNNGQICLR
HQDFVPATKIKGTGCVTADE DTWIKLGNTILSVEPTHNFY LKDSKS SLIVHAVS SNQTFT
VHGFGNGNKLGLTLPLTGNR DHFRFEYYPDTGILQLRADA LPQYFKIGKGYDSKLFRIVN
SRGLKNAVTYDGPVPNNEIP AVCLIPCTNGPSAPESESDL ΝΤΡΤΤΞΞΙΕΤ ( SEQ ID NO: 6)
2. An isolated polypeptide optionally fused to a heterologous fusion partner, wherein the amino acid seguence of said polypeptide consists of an amino acid seguence having at least 95% identity to
DTWIKLGNTILSVEPTHNFY LKDSKS SLIVHAVS SNQTFT
VHGFGNGNKLGLTLPLTGNR DHFRFEYYPDTGILQLRADA LPQYFKIGKGYDSKLFRIVN
SRGLKNAVTYDGPVPNNEIP AVCLIPCTNGPSAPESESDL ΝΤΡΤΤΞΞΙΕΤ (SEQ ID NO: 7)
3. An isolated polypeptide optionally fused to a heterologous fusion partner, wherein the amino acid seguence of said polypeptide consists of an amino acid seguence having at least 95% identity to
DTWIKLGNTILSVEPTHNFY LKDSKS SLIVHAVS SNQTFT
VHGFGNGNKLGLTLPLTGNR DHFRFEYYPDTGILQLRADA LPQYFKIGKGYDSKLFRIVN
SRGLKNAVTYDGPVPNNEIP AVCLIPCTNGPSAPESESDL ΝΤΡΤΤΞΞΙΕΤΞΞΥΞΞΑΑΤΕΞ
SVVSESSSAVDSLTSSSLSS KSESSDVVSSTTNIESSSTA IETTMNSESSTDAGSSSISQ
ΞΕΞΞΞΤΑΙΤΞΞΞΕΤΞΞΞΕΞΜ SASSTTASNTSIETDSGIVS QSESSSNAL (SEQ ID NO: 9) .
4. An isolated polypeptide optionally fused to a heterologous fusion partner, wherein the amino acid seguence of said polypeptide consists of an amino acid seguence having at least 95% identity to
QNQKAAG NIAFGTAYQTITNNGQICLR
HQDFVPATKIKGTGCVTADE DTWIKLGNTILSVEPTHNFY LKDSKS SLIVHAVS SNQTFT VHGFGNGNKLGLTLPLTGNR DHFRFEYYPDTGILQLRADA LPQYFKIGKGYDSKLFRIVN SRGLKNAVTYDGPVPNNEIP AVCLIPCTNGPSAPESESDL ΝΤΡΤΤΞΞΙΕΤΞΞΥΞΞΑΑΤΕΞ SVVSESSSAVDSLTSSSLSS KSESSDVVSSTTNIESSSTA IETTMNSESSTDAGSSSISQ ΞΕΞΞΞΤΑΙΤΞΞΞΕΤΞΞΞΕΞΜ SASSTTASNTSIETDSGIVS QSESSSNAL (SEQ ID NO
5. An isolated nucleic acid molecule which encodes a polypeptide of any one of claims 1-4.
6. An isolated nucleic acid molecule comprising a nucleic acid seguence which is substantially identical to the isolated nucleic acid molecule of claim 5.
7. A vector comprising the nucleic acid molecule of claim 5 or 6.
8. A cell comprising the nucleic acid molecule of claim 5 or 6.
9. A method of producing a recombinant polypeptide, said method comprising the steps of:
(a) providing a cell transformed with the nucleic acid molecule of claim 5 or 6 encoding an polypeptide positioned for expression in the cell;
(b) culturing the transformed cell under conditions for
expressing the nucleic acid molecule, wherein said culturing results in expression of said recombinant polypeptide; and
(c) isolating the recombinant polypeptide.
10. The method of claim 9, wherein said cell is a bacterium.
11. The method of claim 10, wherein said cell is a yeast.
12. A recombinant polypeptide produced according to the method of claim 9.
13. A substantially pure antibody that specifically recognizes and binds to any one of the polypeptides of claims 1-4.
14. An antigenic composition comprising the polypeptide of any one of claims 1-4 a pharmaceutically acceptable carrier, diluent, and/or excipient .
15. The composition of claim 14, further comprising an adjuvant.
16. A method of inducing an immune response in a mammal against an antigen comprising administering a polypeptide of any one of claims 1 4, or the antigenic composition of claim 14 to said mammal, wherein said polypeptide or said composition induces an immune response against said antigen in said mammal.
17. The method of claim 16, wherein the mammal is administered a single dose of said polypeptide or said composition.
18. The method of claim 16, wherein the mammal is administered a plurality of doses of said polypeptide or said composition.
19. The method of claim 16, wherein said plurality of doses are administered at least one day apart.
20. The method of claim 18, wherein said composition is administered twice .
21. The method of claim 18, wherein said plurality of doses are administered at least two weeks apart.
22. The method of any one of claims 16-21, wherein said mammal is a human .
23. A vaccine comprising an immunogenic amount of the polypeptide of any one of claims 1-4, and a pharmaceutically acceptable excipient .
24. The vaccine of claim 23, comprising a mixture of distinct polypeptides of any one of claims 1-4.
25. The vaccine of claim 23 or 24, further comprising an adjuvant.
26. The vaccine of claim 25, wherein said adjuvant is Alhydrogel .
27. The vaccine of any one of claims 23-26 for use in the vaccination of a mammal against candidiasis or a gram negative rod bacterium.
28. The vaccine of claim 27, wherein said mammal is a human.
29. The vaccine of claim 27 or 28, wherein said vaccine is to be administered by intramuscular, subcutaneous, or intradermal
administration .
30. The vaccine of claim 29, wherein said vaccine is to be
administered by intramuscular administration.
31. The vaccine of any one of claims 23-30, wherein said vaccination further comprises administering a booster dose.
32. The vaccine of any one of claims 27-31, wherein said candidiasis is disseminated candidiasis.
33. The vaccine of claim 32, wherein said disseminated candidiasis is hematogenously disseminated candidiasis.
34. The vaccine of any one of claims 27-31, wherein said candidiasis is mucosal candidiasis.
35. The vaccine of any one of claims 27-34, wherein said candidiasis is caused by Candida albicans, Candida glabrata, Candida krusei, Candida parapsilosis, or Candida tropicalis .
36. A method of vaccinating a mammal against candidiasis comprising administering to said mammal the vaccine of claim 23, thereby
vaccinating said mammal against candidiasis or a gram negative rod bacterium.
37. The method of claim 36, wherein said mammal is a human.
38. The method of claim 36 or 37, wherein said vaccine is administered by intramuscular, subcutaneous, or intradermal
administration .
39. The method of claim 36 or 37, wherein said vaccine is
administered by intramuscular administration.
40. The method of any one of claims 36-39, wherein said administering further comprises administering a booster dose.
41. The method of any one of claims 36-40, wherein said candidiasis is disseminated candidiasis.
42. The method of claim 41, wherein said disseminated candidiasis is hematogenously disseminated candidiasis.
43. The method of any one of claims 36-40, wherein said candidiasis is mucosal candidiasis.
44. The method of any one of claims 36-43, wherein said candidiasis is caused by Candida albicans, Candida glabrata, Candida krusei, Candida parapsilosis, or Candida tropicalis .
45. A method of producing a chimeric vaccine comprising the steps of:
(a) providing a phage, yeast, or virus;
(b) inserting into said phage, yeast, or virus a nucleic acid molecule that encodes the polypeptide of any one of claims 1-4;
(c) allowing expression of said polypeptide in said phage, yeast, or virus;
(d) isolating said phage, yeast, or virus of step (c) comprising said expressed polypeptide; and
(e) adding a pharmaceutically acceptable excipient to said isolated phage, yeast, or virus of step (d) .
46. The method of claim 45, wherein said polypeptide is displayed on the surface of said phage, yeast, or virus following step (c) .
47. An isolated monoclonal antibody that binds to the polypeptide of any one of claims 1-4.
48. The antibody of claim 47, wherein said antibody is human or humanized .
49. The antibody of claim 47, wherein said antibody is chimeric.
50. The antibody of any one of claims 47-49, wherein said antibody is produced recombinantly .
51. A diagnostic composition comprising the antibody of any one of claims 47-50.
52. A pharmaceutical composition comprising the antibody of any one of claims 47-50, and a pharmaceutically acceptable excipient .
53. The pharmaceutical composition of claim 52, comprising a mixture of antibodies of any one of claims 47-50 with a plurality of distinct specificities .
54. A pharmaceutical composition comprising polyclonal antibodies that bind to the polypeptide of any one of claims 1-4, or that bind to a mixture of distinct polypeptides of any one of claims 1-4.
55. The pharmaceutical composition of any one of claims 51-53 for use in the passive immunization of a mammal against candidiasis or a gram negative rod bacterium.
56. The composition of claim 55, wherein said mammal is a human.
57. The composition of claim 55 or 56, wherein said pharmaceutical composition is administered by intramuscular, subcutaneous, or intradermal administration.
58. The composition of claim 57, wherein said pharmaceutical composition is administered by intramuscular administration.
59. The composition of any one of claims 55-58, wherein said candidiasis is disseminated candidiasis.
60. The composition of claim 59, wherein said disseminated
candidiasis is hematogenously disseminated candidiasis.
61. The composition of any one of claims 55-58, wherein said candidiasis is mucosal candidiasis.
62. The composition of any one of claims 55-61, wherein said candidiasis is caused by Candida albicans, Candida glabrata, Candida krusei, Candida parapsilosis, or Candida tropicalis .
63. A method of passive immunization of a mammal against candidiasis or a gram negative rod bacterium comprising administering to said mammal an effective amount of the pharmaceutical composition of claim 55, thereby passively immunizing said mammal against said candidiasis or said gram negative rod bacterium.
64. The method of claim 63, wherein said mammal is a human.
65. The method of claim 63 or 64, wherein said pharmaceutical composition is administered by intramuscular, subcutaneous, or intradermal administration.
66. The method of claim 65, wherein said pharmaceutical composition is administered by intramuscular administration.
67. The method of any one of claims 63-66, wherein said candidiasis is disseminated candidiasis.
68. The method of claim 167, wherein said disseminated candidiasis is hematogenously disseminated candidiasis.
69. The method of any one of claims 63-66, wherein said candidiasis is mucosal candidiasis.
70. The method of any one of claims 63-69, wherein said candidiasis is caused by Candida albicans, Candida glabrata, Candida krusei,
Candida parapsilosis, or Candida tropicalis .
71. In any aforementioned claim, wherein said gram negative rod bacterium is Acinetobacter.
PCT/US2014/028256 2013-03-15 2014-03-14 Hyr1 compositions and methods for treating fungal and bacterial pathogens WO2014144024A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201361791181P 2013-03-15 2013-03-15
US61/791,181 2013-03-15

Publications (1)

Publication Number Publication Date
WO2014144024A1 true WO2014144024A1 (en) 2014-09-18

Family

ID=51537592

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2014/028256 WO2014144024A1 (en) 2013-03-15 2014-03-14 Hyr1 compositions and methods for treating fungal and bacterial pathogens

Country Status (1)

Country Link
WO (1) WO2014144024A1 (en)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10130691B2 (en) 2013-03-15 2018-11-20 Los Angeles Biomedical Research Institute At Harbor-Ucla Medical Center Compositions and methods for treating fungal and bacterial pathogens
US10300120B2 (en) 2002-09-13 2019-05-28 Los Angeles Biomedical Research Institute At Harbor-Ucla Medical Center Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis and other infectious agents
WO2020174366A1 (en) 2019-02-28 2020-09-03 Universidade Do Minho Antisense oligomers for controlling candida albicans infections
US10857216B2 (en) 2016-03-09 2020-12-08 Novadigm Therapeutics, Inc. Methods and kits for use in preventing and treating vulvovaginal candidiasis

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120107316A1 (en) * 2004-09-14 2012-05-03 Novartis Vaccines And Diagnostics, Inc. Protective anti-glucan antibodies with preference for beta-1,3-glucans
US20120237534A1 (en) * 2009-07-03 2012-09-20 Los Angeles Biomedical Research Institute At Harbor-Ucla Medical Center Hyr1 as a target for active and passive immunization against candida

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20120107316A1 (en) * 2004-09-14 2012-05-03 Novartis Vaccines And Diagnostics, Inc. Protective anti-glucan antibodies with preference for beta-1,3-glucans
US20120237534A1 (en) * 2009-07-03 2012-09-20 Los Angeles Biomedical Research Institute At Harbor-Ucla Medical Center Hyr1 as a target for active and passive immunization against candida

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10300120B2 (en) 2002-09-13 2019-05-28 Los Angeles Biomedical Research Institute At Harbor-Ucla Medical Center Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis and other infectious agents
US10130691B2 (en) 2013-03-15 2018-11-20 Los Angeles Biomedical Research Institute At Harbor-Ucla Medical Center Compositions and methods for treating fungal and bacterial pathogens
US10857216B2 (en) 2016-03-09 2020-12-08 Novadigm Therapeutics, Inc. Methods and kits for use in preventing and treating vulvovaginal candidiasis
WO2020174366A1 (en) 2019-02-28 2020-09-03 Universidade Do Minho Antisense oligomers for controlling candida albicans infections

Similar Documents

Publication Publication Date Title
US20190030141A1 (en) Compositions and methods for treating fungal and bacterial pathogens
US10160790B2 (en) HYR1-derived compositions and methods of treatment using same
JP2669594B2 (en) Polypeptide for vaccine against herpes simplex virus infection
US9809620B2 (en) Prion disease-specific epitopes and methods of use thereof
WO2014144024A1 (en) Hyr1 compositions and methods for treating fungal and bacterial pathogens
US5656451A (en) OspE, OspF, and S1 polypeptides in borrelia burgdorferi
PT1446422E (en) Methylated heparin-binding hemagglutinin recombinant mycobacterial antigen, preparation method and immunogenic compositions comprising same
AU2010300380B2 (en) Methods of improving vaccine immunogenicity
US20160030533A1 (en) Compositions and methods of treating fungal and bacterial pathogens
CN109369809B (en) Multi-epitope antigen, preparation method thereof and application of multi-epitope antigen in preparation of medicine for preventing and treating chlamydia psittaci infection
CN111948387B (en) Application of mycobacterium tuberculosis antigen protein Rv1485 in preparation of tuberculosis vaccine
US10590178B2 (en) Chimeric vaccine against fungal infections
US9376476B2 (en) Prion epitopes and methods of use thereof
US20030017162A1 (en) Outer membrane protein A, peptidoglycan-associated lipoprotein, and murein lipoprotein as therapeutic targets for treatment of sepsis
US20190269771A1 (en) Peptide mimotope that induces an immune response against mycobacterium tuberculosis lipoarabinomannan (lam)
WO2023227563A1 (en) Protective staphylococcal exotoxin vaccine
WO2023247747A1 (en) Protective staphylococcal exotoxin vaccine
Good et al. Evidence implicating MHC genes in the immunological nonresponsiveness to the Plasmodium falciparum CS protein.
BR102015017724B1 (en) MIMETIC PEPTIDES FROM LEISHMANIA SPP., PROCESS FOR OBTAINING THEM AND APPLICATIONS

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 14762901

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 14762901

Country of ref document: EP

Kind code of ref document: A1