US20070077256A1 - Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis and other infectious agents - Google Patents

Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis and other infectious agents Download PDF

Info

Publication number
US20070077256A1
US20070077256A1 US11/327,197 US32719706A US2007077256A1 US 20070077256 A1 US20070077256 A1 US 20070077256A1 US 32719706 A US32719706 A US 32719706A US 2007077256 A1 US2007077256 A1 US 2007077256A1
Authority
US
United States
Prior art keywords
als
candida
mice
rals1p
albicans
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/327,197
Other languages
English (en)
Inventor
John Edwards
Ashraf Ibrahim
Brad Spellberg
Yue Fu
Scott Filler
Michael Yeaman
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Harbor Ucla Medical Center
Original Assignee
Harbor Ucla Medical Center
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
Priority claimed from US09/715,876 external-priority patent/US7067138B1/en
Priority claimed from US10/245,802 external-priority patent/US8541008B2/en
Priority claimed from US11/123,873 external-priority patent/US20060083750A1/en
Application filed by Harbor Ucla Medical Center filed Critical Harbor Ucla Medical Center
Priority to US11/327,197 priority Critical patent/US20070077256A1/en
Assigned to LOS ANGELES BIOMEDICAL RESEARCH INSTITUTE AT HARBOR-UCLA MEDICAL CENTER reassignment LOS ANGELES BIOMEDICAL RESEARCH INSTITUTE AT HARBOR-UCLA MEDICAL CENTER ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: SPELLBERG, BRAD, EDWARDS, JOHN E., JR., FILLER, SCOTT G., FU, YUE, IBRAHIM, ASHRAF, YEAMAN, MICHAEL
Priority to JP2008549598A priority patent/JP5692963B2/ja
Priority to ES07709622T priority patent/ES2391580T3/es
Priority to PT07709622T priority patent/PT1982190E/pt
Priority to CA2636277A priority patent/CA2636277C/en
Priority to EP11008862.2A priority patent/EP2428800B1/de
Priority to US12/160,073 priority patent/US20090297562A1/en
Priority to ES12001586.2T priority patent/ES2608354T3/es
Priority to AU2007205065A priority patent/AU2007205065B2/en
Priority to CA2948424A priority patent/CA2948424A1/en
Priority to DK07709622.0T priority patent/DK1982190T3/da
Priority to ES11008862.2T priority patent/ES2608309T3/es
Priority to EP12001595A priority patent/EP2532361A1/de
Priority to PCT/US2007/000433 priority patent/WO2007081896A2/en
Priority to EP07709622A priority patent/EP1982190B1/de
Priority to EP12001586.2A priority patent/EP2532360B1/de
Publication of US20070077256A1 publication Critical patent/US20070077256A1/en
Priority to US12/987,949 priority patent/US20120014995A1/en
Priority to JP2012207831A priority patent/JP5775044B2/ja
Priority to CY20121100979T priority patent/CY1113693T1/el
Priority to US13/785,835 priority patent/US20140037689A1/en
Priority to JP2014105980A priority patent/JP2014148547A/ja
Priority to US15/014,440 priority patent/US10300120B2/en
Abandoned legal-status Critical Current

Links

Images

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
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/14Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from fungi, algea or lichens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5156Animal cells expressing foreign proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/58Medicinal preparations containing antigens or antibodies raising an immune response against a target which is not the antigen used for immunisation
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/02Bacterial antigens
    • A61K39/085Staphylococcus
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • This invention relates to Candida albicans surface adhesin proteins, to antibodies resulting from an immune response to vaccination with C. albicans surface adhesion proteins and to methods for the prevention and/or treatment of candidiasis and other bacterial infections with C. albicans surface adhesion proteins.
  • Candida albicans the major pathogen in this genus, can switch between two morphologies: the blastospore (budding yeast) and filamentous (hyphae and pseudohyphae) phases.
  • Candida mutants that are defective in genes regulating filamentation are reported to have reduced virulence in animal models. This reduced virulence suggests that the ability to change from a blastospore to a filament is a key virulence factor of C. albicans .
  • no essential effectors of these filamentation pathways have been identified in C. albicans . See Caesar-TonThat, T. C. and J. E. Cutler, “A monoclonal antibody to Candida albicans enhances mouse neutrophil candidacidal activity,” Infect. Immun. 65:5354-5357, 1997.
  • Staphylococcus aureus infections also are common and increasingly result in drug resistance to antibiotics.
  • S. aureus is a common cause of skin and skin structure infections, endocarditis and bacteremia in the U.S. and throughout the world.
  • CA- S. aureus formerly community acquired S. aureus infections were nearly uniformly susceptible to penicillinase-resistant beta lactams such as cefazolin, oxacillin, methicillin, penicillin and amoxicillin.
  • beta-lactam resistant S. aureus (MRSA) infection have been seen in multiple locales throughout the world, especially community acquired MRSA (CA-MRSA). In many places MRSA has become the predominant S.
  • the identification of effectors in the regulatory pathways of the organism that contribute to virulence offers the opportunity for therapeutic intervention with methods or compositions that are superior to existing antifungal agents.
  • the identification of cell surface proteins that affect a regulatory pathway involved in virulence is particularly promising because characterization of the protein enable immunotherapeutic techniques that are superior to existing antifungal agents when fighting a candidal infection.
  • Candida albicans The virulence of Candida albicans is regulated by several putative virulence factors of which adherence to host constituents and the ability to transform from yeast-to-hyphae are among the most critical in determining pathogenicity. While potent antifungal agents exist that are microbicidal for Candida , the attributable mortality of candidemia is approximately 38%, even with treatment with potent anti-fungal agents such as amphotericin B. Also, existing agents such as amphotericin B tend to exhibit undesirable toxicity. Although additional antifungals may be developed that are less toxic than amphotericin B, it is unlikely that agents will be developed that are more potent. Therefore, either passive or active immunotherapy to treat or prevent disseminated candidiasis is a promising alternative to standard antifungal therapy.
  • the invention provides a vaccine including an isolated Als protein family member having cell adhesion activity, or an immunogenic fragment thereof, with an adjuvant in a pharmaceutically acceptable medium.
  • the invention also provides a method of treating or preventing disseminated candidiasis.
  • the method includes administering an immunogenic amount of a vaccine an isolated Als protein family member having cell adhesion activity, or an immunogenic fragment thereof, in a pharmaceutically acceptable medium.
  • a method of treating or preventing disseminated candidiasis also is provided that includes administering an effective amount of an isolated Als protein family member having cell adhesion activity, or an functional fragment thereof, to inhibit the binding or invasion of Candida to a host cell or tissue.
  • the Als protein family member can be derived from a Candida strain selected from the group consisting of Candida albicans, Candida krusei, Candida tropicalis, Candida glabrata and Candida parapsilosis and the Als protein family member includes Als1p, Als3p, Als5p, Als6p, Als7p or Als9p. Also provided is a method of treating or preventing Staphylococcus aureus infections. The method includes administering an immunogenic amount of a vaccine an isolated Als protein family member having cell adhesion activity, or an immunogenic fragment thereof, in a pharmaceutically acceptable medium.
  • FIG. 1A, 1B show the mediation of Als1p adherence of C. albicans to human umbilical vein endothelial cells. Values represent the mean ⁇ SD of at least three independent experiments, each performed in triplicate.
  • Statistical treatment was obtained by Wilcoxon ran sum test and corrected for multiple comparisons with the Bonferroni correction. *P ⁇ 0.001 for all comparisons.
  • FIG. 2A -D shows the cell surface localization of Als1p on filaments of C. albicans indirect immunofluorescence.
  • Filamentation of C. albicans was induced by incubating yeast cells in RPMI 1640 medium with glutamine for 1.5 hours at 37° C.
  • Als1p was detected by incubating organisms first with anti-Als1p mouse mAb followed by FITC-labeled goat anti-mouse IgG.
  • C. albicans cell surface was also stained with anti- C. albicans polyclonal Ab conjugated with Alexa 594 (Molecular Probes, Eugene, Oreg.). Areas with yellow staining represent Als1p localization.
  • A C. albicans wild-type.
  • C als1/als1 complemented with wild type ALS1
  • D P ADH1 -ALS1 overexpression mutant.
  • FIG. 3A , 3B show the mediation of Als1p on C. albicans filamentation on solid medium.
  • C. albicans blastospores were spotted on Lee's agar plates and incubated at 37° C. for 4 days (A) or 3 days (B).
  • FIG. 4A, 4B show the control of ALS1 expression and the mediation of C. albicans filamentation by the EFG1 filamentation regulatory pathway.
  • A Northern blot analysis showing expression of ALS1 in (i) mutants deficient in different filamentation regulatory pathways. (ii) efg1/efg1 mutant complemented with either EFG1 or P ADH1 -ALS 1.
  • Total RNA was extracted from cells grown in RPM1 1640+ glutaine medium at 37° C. for 90 minutes to induce filamentation. Blots were probed with ALS1 and TEF1.
  • B Photomicrographs of the efg1/efg1 mutant and efg1/efg1 mutant complemented with P ADH1 -ALS1 grown on Lee's agar plates at 37° C. for 4 days.
  • FIG. 5A, 5B show the reduction of virulence in the mouse model of hematogenously disseminated candidiasis by
  • FIG. 6 shows the prophylactic effect of anti-ALS antibody against disseminated candidiasis as a function of surviving animals over a 30-day period for animals infused with anti-Als1p polyserum.
  • FIG. 7 is polypeptide sequence alignment of the N-terminal portion of select ALS polypeptides arranged by adherence phenotype.
  • the top three lines are the sequences from ALS1, 3 and 5 polypeptides (SEQ ID NOS: 1-3, respectively), which bind endothelial cells.
  • the bottom three are sequences from ALS6, 7 and 9 polypeptides (SEQ ID NOS; 4-6, respectively), which do not bind endothelial cells.
  • the last line represents the ALS polypeptide family consensus sequence (SEQ ID NO:7).
  • FIG. 8 shows Als proteins confer substrate-specific adherence properties when heterologously expressed in Saccharonzyces cerevisiae .
  • Each panel demonstrates the percentage adherence of one Alsp expression strain (filled bars) to a variety of substrates to which C. albicans is known to adhere.
  • Adherence of S. cerevisiae transformed with the empty vector (empty bars) is included in each panel as a negative control.
  • * p ⁇ 0.01 when compared with empty plasmid control by single factor analysis of variance. Results are the mean ⁇ S.D. of at least three experiments performed in triplicate.
  • FIG. 9 shows domain swapping demonstrates that substrate-specific adherence is determined by the composition of the N-terminal domain of Als proteins.
  • a representation of the ALS gene or construct being tested is depicted as a bar composed of sequences from ALS5 (black) or ALS6 (white).
  • Adherence properties of each mutant are displayed as a photomicrograph illustrating the adherence of transformed S. cerevisiae to fibronectin-coated beads and a graph demonstrating the adherence to gelatin (black bars) and endothelial cells (gray bars) as measured in the 6-well plate assay. Results are mean ⁇ S.D. of at least three experiments, each performed in triplicate.
  • FIG. 10 shows a subset of Als proteins mediate endothelial cell invasion when expressed in S. cerevisiae .
  • A endothelial cell adherence of S. cerevisiae strains expressing Als proteins or transformed with the empty plasmid (control). Data represent the total number of endothelial cell-associated organisms and are expressed as cells per high power field.
  • B degree of endothelial cell invasion of Alsp expressing S. cerevisiae strains presented as the number of intracellular organisms per high power field.
  • * p ⁇ 0.01 when compared with empty plasmid control by single factor analysis of variance. Results are the mean ⁇ S.D. of at least three experiments performed in triplicate.
  • FIG. 11 shows an alignment of the N-terminal amino acid sequence of Als proteins of known function demonstrates an alternating pattern of CRs and HVRs.
  • A percentage of consensus identity among the N-terminal regions of Als proteins of known function. Note that the signal peptide region (amino acids 1-20) is not shown. Open boxes indicate the regions designated as HVRs 1-7.
  • B schematic alignment of Als proteins (SEQ ID NOS:1-6, respectively) showing the composition of the individual HVRs. The sequences are arranged to compare proteins with an affinity to multiple substrates with those that bind few or no identified substrates. The number of amino acids in each conserved region is indicated in parentheses.
  • FIG. 12 shows CD and FTIR spectra of the Als1 protein N-terminal domain.
  • A circular dichroism spectrum of 10 ⁇ M Als1p in phosphate-buffered saline.
  • B FTIR spectrum of Als1p self-film hydrated with D 2 O vapor.
  • FIG. 13 shows a comparison of predicted physicochemical properties of N-terminal domains among the Als protein family. Hydrophobic, electrostatic, or hydrogen-bonding features are projected onto water-accessible surfaces of each domain. Hydrophobics are shown as follows: brown, most hydrophobic; blue, most hydrophilic. Electrostatics (spectral continuum) is shown as follows: red, most positive charge (+10 kcal/mol); blue, most negative charge ( ⁇ 10 kcal/mol). Hydrogen-bonding potential (H-binding) is shown as follows: red, donor; blue, acceptor. Als proteins are distinguishable into three groups based on the composite of these properties.
  • FIG. 14 Conceptual model of structural-functional relationships in Als family proteins.
  • Als proteins are composed of three general components: an N-terrninal domain, tandem repeats, and a serine/threonine-rich C-terminal domain containing a glycosylphosphatidylinositol anchor that interfaces with the C. albicans cell wall.
  • Als proteins contain multiple conserved anti-parallel ⁇ -sheet regions (CR1 ⁇ n) that are interposed by extended spans, characteristic of the immunoglobulin superfamily. Projecting from the ⁇ -sheet domains are loop/coil structures containing the HVRs.
  • the three-dimensional physicochemical properties of specific Als protein HVRs probably govern interactions with host substrates that confer adhesive and invasive functions to Candida .
  • For illustrative purposes only three N-terminal ⁇ -sheet/coil domains and their respective CR/HVR components are shown. Note that this projection is viewed at right angles to the structural images shown in FIG. 13 .
  • FIG. 18 Only the protective dose of rAls1p-N induces an increase in C. albicans -stimulated Th1 splenocytes.
  • FIG. 22 The rAls1p-N vaccine improves survival of immunocompetent mice with hematogenously disseminated candidiasis and reduces tissue fungal burden.
  • A) Survival of vaccinated or control BALB/c mice (n 7 or 10 per group for 2.5 or 5 ⁇ 10 5 inocula, respectively) mice subsequently infected via the tail-vein with C. albicans . Each experiment was terminated at 30 days post-infection with all remaining mice appearing well. *p ⁇ 0.05 vs. Control by Log Rank test.
  • FIG. 23 The rAls1p-N vaccine induces a DTH reaction in neutropenic mice and improves their survival during subsequent hematogenously disseminated candidiasis.
  • FIG. 26 shows an Als1p homology map versus S. aureus clumping factor A (cln67A). Consensus functional sites from C. albicans Als1p and S. aureus ClfA were mapped onto the Als1p homology model. Numerous residues from the N-termini of Als1p and ClfA map to a consensus cleft motif, which is where binding to substrate is predicted to occur for both adhesins.
  • FIG. 28 shows that antibody titers do not correlate with degree of protection in individual vaccinated mice, but they do distinguish unvaccinated from vaccinated mice.
  • FIG. 29 shows that the rAls1p-N vaccine protects outbred, CD1 mice from hematogenously disseminated candidiasis.
  • A) CD1 mice (n 8 per group) were vaccinated SQ with rAls1p-N (20 ⁇ g)+CFA, or CFA alone, and infected via the tail-vein with C. albicans SC5314 fourteen days after the boost.
  • B) CD1 mice (n 8 per group) were vaccinated SQ with rAls1p-N at various doses with alum, or with alum alone, and infected via the tail-vein with C. albicans SC5314 fourteen days after the boost.
  • FIG. 30 shows that the rAls1p-N vaccine improves the survival of Balb/c mice infected with one of several strains of C. albicans .
  • Survival of Balb/c mice immunized with rAls1p-N plus CFA versus CFA alone and infected via the tail-vein with C. albicans 15563 (7 ⁇ 10 5 blastospores), 16240 (4 ⁇ 10 5 blastospores), or 36082 (4 ⁇ 10 5 blastospores) (n 8 mice per group).
  • FIG. 31 shows that the rAls1p-N vaccine reduces tissue fungal burden in Balb/c mice infected with several non-albicans species of Candida .
  • Infectious inocula are shown in parentheses below the species names.
  • Kidney fungal burden was determined on day five post-infection.
  • the y axis reflects the lower limit of detection of the assay. *p ⁇ 0.05 vs. adjuvant control by non-parametric Steel test for multiple comparisons.
  • FIG. 32 shows that rAls3p-N-immunized mice generated antibodies that cross-reacted against rAls1p-N.
  • N 7 mice per group for CFA and CFA+rAls3p-N;
  • n 6 mice for CFA+rAls1p-N.
  • FIG. 33 shows that both rAls1p-N and rAls3p-N primed mice for in vivo delayed type hypersensitivity responses.
  • FIG. 34 shows that the rAls1p-N and rAls3p-N vaccines mediated similar efficacy against murine hematogenously disseminated candidiasis.
  • FIG. 35 shows that in vivo delayed-type hypersensitivity correlated with survival during disseminated candidiasis.
  • FIG. 37 shows that rAls3p-N reduced vaginal fungal burden compared to both CFA alone and CFA+rAls1p-N in murine candidal vaginitis.
  • the y axis reflects the lower limit of detection of the assay. *p ⁇ 0.02 vs CFA and CFA+rAls1p-N by Steel test for multiple comparisons.
  • Candida albicans and Staphylococcus aureus are common pathogen in humans.
  • C. albicans while normally a harmless commensal, this organism can cause a variety of conditions ranging from superficial mucocutaneous infection such as vaginal and/or oropharyngeal candidiasis, to deep organ involvement in disseminated candidiasis.
  • the fungus Prior to causing disease, the fungus colonizes the gastrointestinal tract, and in some cases skin and mucous membranes. Adherence to host mucosal surfaces is a key prerequisite for this initial step.
  • C. albicans enters the bloodstream via infected intravascular devices or by transmigration through gastrointestinal mucosa compromised by chemotherapy or stress ulcerations. Organisms then disseminate via the bloodstream, bind to and penetrate the vascular endothelium to egress from the vascular tree, and invade deep organs such as liver, spleen, and kidney.
  • the identification and functional characterizations of a variety of exemplary Als protein family members described herein allow this family of proteins to be effectively utilized in the treatment of candidiasis.
  • Specific binding activity to diverse substrates and other selective cell adhesion functions can be exploited in the production of vaccines for active or passive immunization, in the production of peptide, analogue of mimetic inhibitors of cell adhesion to reduce or prevent initial infection by inhibiting binding, adhesion or invasion of a host cell.
  • the differential binding and invasion profiles allow design and use of broad spectra or targeted inhibition of Als protein family member activities.
  • functional fragments that confer binding and/or invasive activity allow elimination of unwanted foreign protein sequences, thus, increasing the efficacy of the Als family protein member vaccine or therapeutic inhibitor.
  • Candida albicans Als1p an adhesin that is a downstream effector of the EFG1 filamentation pathway. Molecular Microbiology 44:61-72. Sheppard D C, Yeaman M R, Welch W H, Phan Q T, Fu Y, (2004) A S, Filler S G, Zhang M, Waring A J, Edwards, Jr., J E 2004. Functional and Structural Diversity in the Als Protein Family of Candida albicans. Journal Biological Chemistry. 279: 30480-30489. The ALS gene family of Candida albicans .
  • Candida albicans ALS1 domains related to a Saccharonzyces cerevisiae sexual agglutinin separated by a repeating motif. Mol. Microbiol. 15:39-54.
  • the human fungal pathogen Candida albicans colonizes and invades a wide range of host tissues. Adherence to host constituents plays an important role in this process.
  • Two members of the C. albicans Als protein family (Als1p and Als5p) have been found to mediate adherence and exemplify the binding, adhesion and cell invasion activities of Als protein family members.
  • members of the ALS gene family were cloned and expressed in S. cerevisiae to characterize their individual functions.
  • Distinct Als proteins conferred distinct adherence profiles to diverse host substrates. Using chimeric Als5p-Als6p constructs, the regions mediating substrate-specific adherence were localized to the N-terminal domains in Als proteins.
  • the invention provides a vaccine having an isolated Als protein family member having cell adhesion activity, or an immunogenic fragment thereof, and an adjuvant in a pharmaceutically acceptable medium.
  • the vaccine can be an Als protein family member derived from a Candida species such as Candida albicans, Candida krusei, Candida tropicalis, Candida glabrata or Candida, parapsilosis .
  • the Als protein family member can be, for example, Als1p, Als3p, Als5p, Als6p, Als7p and Als9p, or an immunogenic fragment thereof. All other Als protein family members within an Candida species can similarly be employed as a vaccine of the invention.
  • the present invention utilizes the gene product of C. albicans agglutinin like sequence protein family member as a vaccine to treat, prevent, or alleviate disseminated candidiasis.
  • the vaccine is effective against different strains of C. albicans as well as against different Candida species.
  • the Als protein family member can be, for example, Als1p, Als3p, Als5p, Als6p, Als7p and Als9p.
  • the invention exploits the role of the ALS gene products in the adherence of and invasion by C. albicans to endothelial and/or epithelial cells and the susceptibility of the Als protein family member-expressed surface protein for use as a vaccine to retard the pathogenesis of the organism.
  • an ALS family member gene encodes a surface adhesin that is selected as the target of an immunotherapeutic strategy against C. albicans .
  • the Als protein family members can be structurally characterized as having a signal peptide at the N-terminus, a glycosylphosphatidylinosine (GPI) anchorage sequence in the C-terminus, and a central region comprising repeats rich in threonine and serine.
  • GPI glycosylphosphatidylinosine
  • Als protein family members have N-, and 0-glycosylation sites, typical of proteins that are expressed on the cell surface.
  • This expression of ALs1p is increased during hyphal formation and is localized to the junction where the hyphal element extends from the blastospores as indicated by the diffused surface staining.
  • this monoclonal antibody blocked the enhanced adherence of C. albicans overexpression mutant to endothelial cells, thereby establishing the principle for immunotherapy applications using ALs1p. Functional characteristics as they relate to cell adhesion and invasion of other Als family members are described further below in Example VI.
  • the invention provides an Als family member surface adhesion protein, designated, for example, Als1p, Als3p, Als5p, Als6p, Als7p and Als9p, or a functional fragment, conjugate or analogue thereof, having useful properties when fonnulated in a pharmaceutical composition and administered as a vaccine with or without an adjuvant.
  • An Als protein family member, combination of two or more Als protein family members or one or more functional fragments, analogues, conjugates or derivatives thereof, can be obtained from, for example, Candida albicans .
  • Similar adhesin or invasin molecules or analogues or derivatives thereof can be of candidal origin and can be obtainable, for example, from species belonging to the genera Candida , for example Candida parapsilosis, Candida krusei, Candida glabrata and Candida tropicalis .
  • a surface adhesin or invasin protein according to the invention can be obtained in isolated or purified form, and thus, according to one embodiment of the invention a substantially pure Als protein family member Candida surface adhesin protein, or functional fragment, immunogenic fragment, analogue, conjugate or derivative thereof, is formulated as a vaccine to cause an immune response in a patient to elicit an immune response against Candida and/or to block adhesion of the organism to the endothelial cells.
  • Fragments of Als protein family members that exhibit similar binding, adhesion or invasion activity as an intact Als protein family member is referred to herein as a functional fragment.
  • Fragments of Als protein family members that are capable of eliciting an antibody or cellular immune response against a Candida species are referred immunogenic fragment.
  • Exemplary functional fragments include the N-terminal polypeptide region of the Als protein family member described further below in Example VI.
  • Exemplarily immogenic fragments include the N-terminal Als polypeptide region, the C-terminal Als polypeptide region as well as any other Als fragment that is sufficient to generate an antibody, cellular or both an antibody and cellular immune response.
  • Such immogenic fragments can be as small as about four amino acids and as large as the intact polypeptide as well as include all polypeptide lengths in between.
  • analogue or derivative of the surface adhesion protein according to the invention can be identified and further characterized by the criteria described herein for an ALS family member gene and/or gene product. For example, a null mutant of the analogue or derivative would share markedly reduced adhesion to endothelial cells compared to controls. Similarly, over-expression of the analogue or derivative in an appropriate model would show an increased adherence to endothelial cells compared to controls and would be confirmed as a cell surface adhesin in accord with the criteria described above. Also, antisera to an analogue or derivative can cross-react with anti-Als protein family member antibodies and can exhibit increased survival times when administered in a mouse model of disseminated candidiasis as disclosed herein.
  • the invention also provides a method of treating or preventing disseminated candidiasis.
  • the method includes administering an immunogenic amount of a vaccine an isolated Als protein family member having cell adhesion or invasion activity, or an immunogenic fragment thereof, in a pharmaceutically acceptable medium.
  • the vaccine can be administered with or without an adjuvant.
  • the Als protein family member can be derived from different Candida strains as well as from different Candida species such as Candida albicans, Candida krusei, Candida tropicalis, Candida glabrata and Candicla, parapsilosis .
  • An Als protein family member used in the method of treating or prevention disseminated candidias includes Als1p, Als3p, Als5p, Als6p, Als7p and Als9p.
  • Example V shows that anti-ALS antibodies are effective against mucosal and hematogenously disseminated candidal infections.
  • Example VII shows that vaccination with rAls1p-N improves survival during murine disseminated candidiasis by enhancing cell-mediated immunity.
  • Example VIII shows that the vaccines of the invention reduce fungal burden and improve survival in both immunocompetent and immunocompromised mice.
  • Example IX shows the effectiveness of the ALS vaccines of the invention against S. aureus infections.
  • Example X exemplifies that the vaccines of the invention are effective against different strains of C. albicans and against different species such as C. glabrata, C. krusei, C. parapsilosis and C. tropicalis as well as effectiveness in different animal models.
  • Example XI also exemplifies the effectiveness of the different vaccines of the invention in different animal models as well as provides a comparison of the different responses elicited and potency of two representative ALS vaccines.
  • the invention further provided is a method of treating or preventing disseminated candidiasis that includes administering an effective amount of an isolated Als protein family member having cell adhesion activity, or an functional fragment thereof, to inhibit the binding or invasion of Candida to a host cell or tissue.
  • the Als protein family member can be derived from Candida albicans, Candida krusei, Candida tropicalis, Candida glabrata, and Candida, parapsilosis .
  • An Als protein family member used in the method of treating or prevention disseminated candidias includes Als1p, Als3p, Als5p, Als6p, Als7p and Als9p.
  • the cell adhesion activity includes binding to gelatin, fibronectin, laminin, epithelial cells or endothelial cells and/or promoting cell invasion.
  • the invention also provides a method of treating or preventing Staphylococcus aureus infections using the Als protein family members described herein.
  • the method of treating or preventing Staphylococcus aureus infections includes administering an immunogenic amount of a vaccine an isolated Als protein family member having cell adhesion activity, or an immunogenic fragment thereof, in a pharmaceutically acceptable medium.
  • Als1p and Als3p are particularly efficacious because of significant homology to S. aureus cell surface proteins.
  • the sequence and structural homology of, for example, Als1p and Als3p, are described further below in Example IX.
  • the vaccines and inethods of the invention can be applied to the treatment of Candida and Staphylococcus infections alike.
  • the vaccines and methods of the invention also can be applied to other pathogens having cell surface polypeptides with similar immunogenicity, sequence and/or structural homology to the Als protein family members described herein, including fungus, bacteria and the like.
  • Immunotherapeutic and/or Als polypeptide inhibition of cell adhesion or invasion strategies against Candida or Staphylococcus infection can operate at the level of binding to the vascular endothelial cells as well as through a downstream effector of the filamentation regulatory pathway.
  • An immunotherapeutic strategy or inhibition of binding using a soluble Als protein family member or functional fragment is useful in this context because: (i) the morbidity and mortality associated with hematogenously disseminated candidiasis and other infectious pathogens remains unacceptably high, even with currently available antifungal therapy; (ii) a rising incidence of antifungal and antibiotic resistance is associated with the increasing use of antifungal and antibacterial agents, iii) the population of patients at risk for serious Candida and Staphylococcus infections is well-defined and very large, and includes post-operative patients, transplant patients, cancer patients and low birth weight infants; and iv) a high percentage of the patients who develop serious Candida infections are not neutropenic, and thus may respond to a vaccine or a competitive
  • Candida and Staphylococcus are attractive fungal and bacterial targets for passive immunotherapy, active immunotherapy or a combination of passive or active immunotherapy. Additionally, Candida also is attractive for competitive inhibition using an Als protein family member polypeptide, functional fragment thereof and/or a compound or mimetic thereof that binds to one or more Als family members and prevents binding of Candida to a host cell receptor.
  • immunotherapeutic methods well know in the art can be employed with the Als protein family members of the invention, immunogenic fragments, analogues, conjugates, and/or derivatives thereof, to use one or more of the molecule as an immunogen in a pharmaceutically acceptable composition administered as a vaccine with or without an adjuvant.
  • pharmaceutically acceptable composition refers to compositions formulated by known techniques to be non-toxic and, when desired, used with carriers or additives that can be safely administered to humans. Administration can be performed using well known routes including, for example, intravenous, intramuscular, intraperitoneal or sub-cutaneous injection.
  • Such vaccines of the inventions also can include buffers, salts or other solvents known to these skilled in the art to preserve the activity of the vaccine in solution.
  • any of a wide range of adjuvants well known in the art can be employed with the vaccine of the invention to elicit, promote or enhance a therapeutically effective immune response capable of reducing or blocking binding, invasion and/or infection of Candida or Staphylococcus to a susceptible host cell.
  • inhibitory formulations can similarly be administered using well known method in the art including, for example, intravenous intramuscular, intraperitoneal or sub-cutaneous injection.
  • inhibitory compositions that bind Als family member receptors and block an Als protein family member binding also can include buffers, salts or other solvents known to these skilled in the art to preserve the activity of the vaccine in solution.
  • any of a wide range of formulations well known in the art can be employed with the inhibitory compositions of the invention to target and/or enhance delivery to reduce or inhibit binding, invasion and/or infection of Candida or Staphylococcus to a susceptible host cell.
  • an Als protein family member can be truncated to yield an N-terminal fragment by truncation from the C-terminal end with preservation of the functional properties described above and further below in the Examples.
  • C-terminal fragments can be generated by truncation from the N-terminal end with preservation of their functional properties.
  • Other modifications in accord with the teachings and guidance provided herein can be made pursuant to this invention to create other Als protein family member functional fragments, immunogenic fragments, analogs or derivatives thereof, to achieve the therapeutically useful properties described herein with the native protein.
  • Als protein family members and methods of the invention achieves interference with regulation of filamentation, to block adherence of the organism to host constituents, and to enhance clearance of the organism by immunoeffector cells and other physiological mechanisms. Since endothelial cells cover the majority of the vasculature, strategies to block the adherence, invasion and/or both of the organism to endothelial cells using antibodies, Als family member proteins, polypeptide or peptides or any combination thereof include useful embodiment of the present invention. As described previously, such adherence and/or invasion blocking therapies include active or passive immunotherapy or inhibitory binding directed against the candidal adhesins, invasins, or cognate receptors disclosed herein.
  • any suitable host can be injected with protein and the serum collected to yield the desired anti-adhesin antibody after appropriate purification and/or concentration.
  • the adhesin or invasin protein or a combination thereof can be formulated in a suitable vehicle preferably a known immunostimulant such as a polysaccharide or delivery formulation such as liposomes or time-released compositions.
  • a suitable vehicle preferably a known immunostimulant such as a polysaccharide or delivery formulation such as liposomes or time-released compositions.
  • a pharmaceutical composition comprising a candidal adhesin or invasin protein together with one or more pharmaceutically acceptable excipients in a formulation for use as a vaccine or Als receptor inhibitor.
  • the method of the invention is ameliorating and/or preventing candidal or Staphylococcus infection by blocking the adherence of C. albicans to the endothelial or epithelial cells of a host constituent or by, for example, antibody binding to the Staphylococcus and allowing immune mechanisms remove the pathogen.
  • a pharmaceutical composition comprising an Als protein family member adhesin or invasin protein, functional or immunogenic fragment, derivative, analogue, or conjugate thereof is formulated as a vaccine or Als receptor inhibitor in a pharmaceutical composition containing a biocompatible carrier for injection or infusion and is administered to a patient.
  • antiserum raised against Als family member protein or isolated or recombinant Als family member protein can be used to block the adherence of C. albicans to a mammalian host constituent or effect the removal of a Staphylococcus pathogen.
  • Antiserum against adhesin protein can be obtained by known techniques, Kohler and Milstein, Nature 256: 495-499 (1975), and may be humanized to reduce antigenicity, see U.S. Pat. No. 5,693,762, or produced in transgenic mice leaving an unrearranged human immunoglobulin gene, see U.S. Pat. No. 5,877,397.
  • isolated or recombinant Als protein family member also can be produced using methods well known to those skilled in the art including, for example, the recombinant production described in the Examples below.
  • a still further use of the invention is using the Als protein family member adhesin or invasin protein to develop vaccine strategies for the prevention and/or amelioration of candidal or Staphylococcus infections.
  • standard immunology techniques can be employed to construct a multi-component vaccine strategy that can enhance and/or elicit immune response from a host constituent to bock adherence of C. albicans or to effect the elimination of Staphylococcus pathogens.
  • a still further use of the invention is developing DNA vaccine strategies.
  • the ALS family member polynucleotides encoding Als protein family member adhesin or invasin or a functional fragment thereof is administered according to a protocol designed to yield an immune response to the gene product. See e.g., Feigner U.S. Pat. No. 5,703,055.
  • anti-ALS protein family member antibodies may be used with antibodies in treating and/or preventing candidal or Staphylococcus infections. See U.S. Pat. No. 5,578,309.
  • Example 1 describes the preparation of an ALS1 null mutant and a strain of C. albicans characterized by overexpression of ALS1 to confirm the mediation of adherence to endothelial cells.
  • Example 2 describes the localization of Als1p and the implication of the efg filamentation regulatory pathway.
  • Example 3 describes the purification of ALS1 adhesin protein.
  • Example 4 describes the preparation of rabbit polyclonal antibodies raised against the ALS1 surface adhesin protein to be used to demonstrate the blocking of the surface adhesin protein.
  • Example 5 describes the blocking of adherence in vivo, using polyclonal antibodies raised against the ALS1 surface adhesion protein as described herein according to the invention to protect against disseminated candidiasis in a mouse model.
  • Example VI describes the structural and functional characteristics of Als protein family members.
  • the URA blaster technique was used to construct a null mutant of C albicans that lacks express of the Als1p.
  • the als1/als1 mutant was constructed in C. albicans strain CAI4 using a modification of the Ura-blaster methodology (Fonzi and Irwin, Genetics 134, 717 (1993)) as follows: Two separate als1-hisG-IRA3-hisG-als1 constructs were utilized to disrupt the two different alleles of the gene.
  • a 4.9 kb AsLS1 coding sequence was generated with high fidelity PCR (Boehringer Mannheim, Indianapolis, Ind.) using the primers: 5′-CCCTCGAGATGCTTCAACAATTTACATTGTTA-3′ (SEQ ID NO:8) and 5′-CCGCTCGAGTCACTAAATGAACAAGGACAATA-3′ (SEQ ID NO:9).
  • the PCR fragment was cloned into pGEM-T vector (Promega, Madison, Wis.), thus obtaining pGEM-T-ALS1.
  • the hisG-URA3-hisG construct was released from pMG-7 by digestion with Kpn1 and Hind3 and used to replace the portion of ALS1 released by Kpn1 and Hind3 digestion of pGEM-T-ALS1.
  • the final als1-hisG-URA3-hisG-als1 construct was released from the plasmid by digestion with Xhol and used to disrupt the first allele of ALS1 by transformation of strain CAI-4.
  • a second als1-hisG-URA3-hisG-als1 construct was generated in two steps. First, a Bgl2-Hind3 hisG-URA3-hisG fragment of pMB7 was cloned into the BamH1-Hind3 sites of pUC19, thereby generating pYC2. PYC2 was then digested with Hind3, partially filled in with dATP and dGTP using T4 DNA polymerase, and then digested with Sma1 to produce a new hisGURA3-hisG fragment. Second, to generate ALS1 complementary flanking regions, pGEM-T-ALS1 was digested with Xbal and then partially filled in with dCTP and dTTP.
  • This fragment was digested with Hpa1 to delete the central portion of ALS1 and then ligated to the hisG-URA3-hisG fragment generating pYC3.
  • This plasmid was then digested by Xhol to release a construct that was used to disrupt the second allele of the ALS 1. Growth curves were done throughout the experiment to ensure that the generated mutations had no effect on growth rates. All integrations were confirmed by Southern blot analysis using a 0.9 kb ALS1 specific probe generated by digestion of pYF5 with XbaI and HindIII.
  • the null mutant was compared to C. albicans CAI-12 (a URA+revertant strain) for its ability to adhere in vitro to human umbilical vein endothelial cells.
  • yeast cells from YPD 2% glucose, 2% peptone, and 1% yeast extract
  • RPMI RPMI with glutamine at 25° C. for 1 hour to induce Als1p expression.
  • 3 ⁇ 10 2 organisms in Hanks balanced salt solution (HBSS) Irvine Scientific, Irvine, Calif.
  • HBSS Hanks balanced salt solution
  • the nonadherent organisms were aspirated and the endothelial cell monolayers were rinsed twice with HBSS in a standardized manner.
  • the wells were over laid with YPD agar and the number of adherent organisms were determined by colony counting.
  • Statistical treatment was obtained by Wilcoxon rank sum test and corrected for multiple comparisons with the Bonferroni correction. P ⁇ 0.001.
  • a comparison of the ALS1/ALS1 and als1/als1 strain showed that the ALS1 null mutant was 35% less adherent to endothelial cells than C. albicans CAI-12.
  • the adherence of the wild-type strain grown under non-ALS1 expressing conditions was compared with a mutant autonomously expressing Als1p. This mutant was constructed by integrating a third copy of ALS1 under the control of the constitutive ADH1 promoter into the wild-type C. albicans . To achieve constitutive expression of the ALS1 in C.
  • a blunt-ended PCR generated URA3 gene is ligated into a blunt-edged Bg12 site of pOCUS-2 vector (Novagen, Madison, Wis.), yielding pOU-2.
  • a 2.4 kb Not1-Stu1 fragment which contained C. albicans alcohol dehydrogenase gene (ADH1) promoter and terminator (isolated from pLH-ADHpt, and kindly provided by A. Brown, Aberdeen, UK), was cloned into pOU-2 after digestion with Not1 and Stu1.
  • the new plasmid, named pOAU-3 had only one Bg12 site between the ADH1 promoter and terminator.
  • ALS1 coding sequence flanked by BamH1 restriction enzyme sites was generated by high fidelity PCR using pYF-5 as a template and the following primers: 5′-CGGGATCCAGATGCTTCA-ACAATTTACATTG-3′ (SEQ ID NO:10) and 5′-CGGGATCCTCACTAATGAACAAGGACAATA-3′ (SEQ ID NO:11).
  • This PCR fragment was digested with BamH1 and then cloned into the compatible Bg12 site of pOAU-3 to generate pAU-1.
  • pAU-1 was linearized by Xbal prior to transforming C. albicans CAI-4. The site-directed integration was confirmed by Southern Blot analysis. Referring to FIG.
  • overexpressing ALS1 in this P ADH1 -ALS1 strain resulted in a 76% increase in adherence to endothelial cells compared to the wild-type C. albicans .
  • yeast cells were grown overnight in YPD at 25° C. (non-inducing condition of Als1p).
  • Als1p expression was not induced to reduce the background adherence of the wile-type, thus magnifying the role of Als1p in adherence through P ADH1 -ALS1 hybrid gene.
  • the adherence assay was carried out as described above. Statistical treatment was obtained by Wilcoxon rank sum test and corrected for multiple comparisons with the Bonferroni correction. P ⁇ 0.001.
  • a monoclonal anti-Als1p murine IgG antibody was raised against a purified and truncated N-terminus of Als1p (amino acid #17 to #432) expressed using Clontech YEXpressTM Yeast Expression System (Palo Alto, Calif.).
  • the adherence blocking capability of these monoclonal anti-Als1p antibodies was assessed by incubating C. albicans cells with either anti-Als1 antibodies or mouse IgG (Sigma, St. Louis, Mo.) at a 1:50 dilution. After which the yeast cells were used in the adherence assay as described above.
  • Statistical treatment was obtained by Wilcoxon rank sum test and corrected for multiple comparisons with the Bonferroni correction. P ⁇ 0.001.
  • Als1p For Als1p to function as an adhesin, it must be located on the cell surface.
  • the cell surface localization of Als1p was verified using indirect immunofluorescence with the anti-Als1p monoclonal antibody. Diffuse staining was detected on the surface of blastospores during exponential growth. This staining was undetectable on blastospores in the stationary phase. Referring to FIG. 2A , when blastospores were induced to produce filaments, intense staining was observed that localized exclusively to the base of the emerging filament. No immunofluorescence was observed with the als1/als1 mutant, confirming the specificity of this antibody for Als1p. See FIG. 2B . These results establish that Als1p is a cell surface protein.
  • the specific localization of Als1p to the blastospore-filament junction implicates Als1p in the filamentation process.
  • the filamentation phenotype of the C. albicans ALS1 mutants was analyzed. Referring to FIG. 3A , the als1/als1 mutant failed to form filaments after a 4 day incubation on Lee's solid medium, while both the ALS1/ALS1 AND ALS1/als1 strains as well as the ALS1-complemented mutant produced abundant filaments at this time point.
  • the als1/als1 mutant was capable of forming filaments after longer periods of incubation.
  • overexpressing ALS1 augmented filamentation the P ADH1 -ALS1 strain formed profuse filaments after a 3 day incubation, whereas the wild-type strain produced scant filaments at this time point. See FIG. 3B .
  • a negative control was provided using mutant similar to the ALS1 overexpression mutant, except the coding sequence of the ALS1 was inserted in the opposite orientation.
  • the filamentation phenotype of the resulting strain was shown to be similar to that of the wild-type strain.
  • the filament-inducing properties of Als1p are specific to cells grown on solid media, because all of the strains described above filamented comparably in liquid media.
  • ALS1 in the efg1/efg1 strain should restore filamentation.
  • a functional allele of ALS1 under the control of the ADH1 promoter was integrated into the efg1/efg1 strain.
  • an Ura efg1 null mutant was transformed with linearized pAU-1.
  • Ura + clones were selected and integration of the third copy of ALS1 was confirmed with PCR using the primers: 5′-CCGTTTATACCATCCAATC-3′ (SEQ ID NO:13) and 5′-CTACATCCTCCAATGATATAAC-3′ (SEQ ID NO:14).
  • the resulting strain expressed ALS1 autonomously and regained the ability to filament on Lee's agar. See FIGS. 4B and C. Therefore, Efg1p induces filamentation through activation of ALS1 expression.
  • mice infected with the asl1/asl1 null mutant survived significantly longer than mice infected with the ALS1/ALS1 strain, the ALS1/asl1 mutant or the ALS1-complemented mutant.
  • the kidneys of mice infected with the asl1/asl1.mutant contained significantly fewer organisms (5.70 ⁇ 0.46 log 10 CFU/g) (P ⁇ 0.0006 for both comparisons).
  • C. albicans ALS1 encodes a cell surface protein that mediates both adherence to endothelial cells and filamentation.
  • Als1p is the only identified downstream effector of any known filamentation regulatory pathway in C. albicans . Additionally, Als1p contributes to virulence in hematogenous candidal infection. The cell surface location and dual functionality of Als1p make it an attractive target for both drug and immune-based therapies.
  • the ALS1 protein synthesized by E. coli is adequate as an immunogen. However eukaryotic proteins synthesized by E. coli may not be functional due to improper folding or lack of glycosylation. Therefore, to determine if the ALS1 protein can block the adherence of C. albicans to endothelial cells, the protein is, preferably, purified from genetically engineered C. albicans.
  • PCR was used to amplify a fragment of ALS1, from nucleotides 52 to 1296.
  • This 1246 bp fragment encompassed the N-terminus of the predicted ALS1 protein from the end of the signal peptide to the beginning of the tandem repeats.
  • This region of ALS1 was amplified because it likely encodes the binding site of the adhesin, based on its homology to the binding region of the S. cerevisiae Aga1 gene product.
  • this portion of the predicted ALS1 protein has few glycosylation sites and its size is appropriate for efficient expression in E. coli.
  • the fragment of ALS1 was ligated into pQE32 to produce pINS5.
  • the protein is expressed under control of the lac promoter and it has a 6-hits tag fused to its N-terminus so that it can be affinity purified.
  • the cell lysate was passed through a Ni 2+ -agarose column to affinity purify the ALS1-6His fusion protein. This procedure yielded substantial amounts of ALS1-6His.
  • the fusion protein was further purified by SDS-PAGE. The band containing the protein was excised from the gel so that polyclonal rabbit antiserum can be raised against it.
  • the sequence of Als1p is listed in FIG. 7 .
  • Sera from immunized rabbits were absorbed with whole cells of S. cerevisiae transformed with empty plasmid to remove antibodies that are reactive with components of the yeast other than ALS1 protein.
  • the titer of the antisera was determined by immunofluorescence using S. cerevisiae that express the ALS1 gene.
  • FITC-labeled anti-rabbit antibodies were purchased from commercial sources (Southern Biotechnology, Inc).
  • Affinity-purified secondary antibodies were essential because many commercially available sera contain antibodies reactive with yeast glucan and mannan.
  • the secondary antibodies were pretested using Candida albicans as well as S. cerevisiae transformed with the plasmid and were absorbed as needed to remove any anti- S. cerevisiae or anti- Candida antibodies.
  • Negative controls were 1) preimmune serum 2) S. cerevisiae transformed with the empty plasmid, and 3) S. cerevisiae transformed with the ALS gene but grown under conditions that suppress expression of the ALS gene
  • the antisera against the ALS proteins were first tested in the murine model of hematogenously disseminated candidiasis.
  • Affinity-purified anti-ALS antibodies are effective in preventing adhesion of yeast cells to various substrates (see EXAMPLE 3). Affinity-purification is useful in this system because antibody doses can be accurately determined.
  • the unfractionated antisera will undoubtedly contain large amounts of antibody directed toward antigens on the S. cerevisiae carrier cells. Many of these anti- Saccharomyces antibodies would likely bind to C. albicans and make interpretation of the results impossible. Additionally, it is quite possible that the procedure used to elute antibodies from S.
  • ALS protein may also elute small amounts of yeast mannan or glucan that could have adjuvant-like activity.
  • the immunoaffinity-purified antibodies are further purified before use. They may also be preabsorbed with mouse splenocytes.
  • Antibody doses may be administered to cover the range that brackets the levels of serum antibody that can be expected in most active immunization protocols and to cover the range of antibody doses that are typically used for passive immunization in murine models of candidiasis. See Dromer, F., J. Charreire, A. Contrepois, C. Carbon, and P. Yeni. 1987, Protection, of mice against experimental cryptococcosis by anti- Cryptococcus neofornwns monoclonal antibody, Infect. Immun. 55:749-752; Han, Y. and J. E. Cutler. 1995, Antibody response that protects against disseminated candidiasis, Infect. Immun.
  • mice (femal, 7 week old, the NCI) were given anti-ALS that had been absorbed with mouse splenic cells by an intraperitoneal (i.p.) injection.
  • Control mice received prebled serum that had been absorbed with mouse spenic cells, intact anti-ALS serum, or DPBS, respectively.
  • 2 ml of anti-ALS or prebled sera were mixed with 100 ⁇ l of mouse (BALB/c, 7 weeks old female, NCI) splenic cells (app. 9 ⁇ 10 6 cells per ml) at room temperature for 20 minutes. The mixture was washed with warm sterile DPBS by centrifugation (@300 ⁇ g) for 3 minutes. This procedure was repeated three times.
  • mice The volume of i.p. injection was 0.4 ml per mouse. Four hours later, the mice were challenged with C. albicans (strain CA-1; 5 ⁇ 10 5 hydrophilic yeast cells per mouse by i.v. injection. Then, their survival times were measured. See FIG. 6 .
  • C. albicans strain CA-1; 5 ⁇ 10 5 hydrophilic yeast cells per mouse by i.v. injection. Then, their survival times were measured. See FIG. 6 .
  • ALS1 encodes a cell surface protein that mediates adherence to endothelial and epithelial cells. Disruption of both copies of this gene in C. albicans is associated with a 35% reduction in adherence to endothelial cells, and overexpression of ALS1 increases adherence by 125% (Fu et al., Mol. Microbiol. 44:61-72 (2002)).
  • ALS1 is a member of a large C. albicans gene family consisting of at least eight members originally described by Hoyer et al. (Hoyer et al., Trends Microbiol. 9:176-180 (2001), Zhao et al., Microbiology 149:2947-2960. (2003)). These genes encode cell surface proteins that are characterized by three domains. The N-terminal region contains a putative signal peptide and is relatively conserved among Als proteins. This region is predicted to be poorly glycosylated (Zhao et al., Microbiology 149:2947-2960 (2003), Hoyer et al., Genetics 157:1555-1567 (2001)).
  • the central portion of these proteins consists of a variable number of tandem repeats ( ⁇ 36 amino. acids in length) and is followed by a.serine-threonine-rich C-terminal region that contains a glycosylphosphatidylinositol anchor sequence (supra).
  • the proteins encoded by this gene family are known to be expressed during infection (Hoyer et al., Infect. Immun. 67:4251-4255 (1999), Zhang et al., Genome Res. 13:2005-2017 (2003)), the function of the different Als proteins has not been investigated in detail.
  • S. cerevisiae strain S150-2B leu2 his3 trp1 ura3
  • C. albicans strain SC5314 was used for genomic cloning.
  • Plasmid pADH1 obtained from A. Brown (Aberdeen, UK) contains the C. albicans alcohol dehydrogenase gene (ADH1) promoter and terminator, which are functional in S. cerevisiae (Bailey et al., J. Bacteriol. 178:5353-5360 (1996)). This plasmid was used for constitutive expression of ALS genes in S. cerevisiae.
  • Human oral epithelial and vascular endothelial cells were obtained and cultured as follows.
  • the FaDu oral epithelial cell line isolated from a pharyngeal carcinoma, was purchased from the American Type Culture Collection (ATCC) and maintained as per their recommended protocol.
  • Endothelial cells were isolated from umbilical cord veins and maintained by our previously described modification of the method of Jaffe et al. (Fu et al., Mol. Microbiol. 44:61-72 (2002), Jaffe et al., J. Clin. Invest. 52:2745-2756 (1973)). All cell cultures were maintained at 37° C. in a humidified environment containing 5% CO2.
  • ALS genes genomic sequences of members of the ALS family were identified by BLAST searching of the Stanford data base (available on the World Wide Web at URL: sequence.stanford.edu/group/candida/search.html).
  • PCR primers were generated to specifically amplify each of the open reading frames that incorporated a 5′ BglII and a 3′ XhoI restriction enzyme site and are shown below in Table I (SEQ ID NOS:14-19 (ALS 1, 3, 5, 6, 7 and 9 sense primers, respectively); SEQ ID NOS:20-25 ((ALS 1, 3, 5, 6, 7 and 9 antisense primers, respectively)).
  • Each gene was cloned by PCR using the Expand® High Fidelity PCR system (Roche Applied Science).
  • ALS3, ALS6, and ALS7 were amplified from C. albicans SC5314 genomic DNA, whereas ALS1, ALS5, and ALS9 were amplified from plasmids that had been previously retrieved from C. albicans genomic libraries (Fu et al., Infect. Immune. 66:1783-1786 (1998), Gaur et al., Infect. Immune. 65:5289-5297 (1997), Lucinod et al., Proceedings of the 102 nd Annual Meeting of the American Society for Microbiology , pp. 204, American Society for Microbiology, Salt Lake City, Utah (2002)). PCR products were ligated into pGEM-T-Easy (Promega) for sequencing.
  • Sequence-verified ALS open reading frames were then released from pGEM-T-Easy by BglII-XhoI co-digestion and ligated into pADH1, such that the ALS gene of interest was under the control of the ADH1 promoter.
  • S. cerevisiae strain S150-2B was transformed with each of the ALS overexpression constructs as well as the empty pADH1 construct using the lithium acetate method. Expression of each ALS gene in S. cerevisiae was verified by Northern blot analysis before phenotypic analyses were performed. TABLE I PCR primers used to amplify the coding regions of ALS gene for heterologous expression in S.
  • ALS mRNA expression was detected by Northern blot analysis for each construct. Despite the use of three sets of primers, amplification of ALS2 and ALS4 from genomic DNA of C. albicans SC5314 was unsuccessful. Given the difficulty of sequencing and assembling across the tandem repeats of ALS genes, it is possible that this outcome reflects errors in the sequence assembly currently available on the published genome data base.
  • HBSS Hanks' balanced salt solution
  • Als5p expression in S. cerevisiae confered adherence to multiple substrates, including gelatin and endothelial cells
  • Als6p expression resulted in adherence to gelatin alone.
  • Als5p and Als6p are more than 80% identical at the amino-acid level.
  • the tandem repeat and C-terminal portions of these proteins are virtually identical, and the majority of the sequence differences are concentrated in the N termini of these two proteins.
  • chimeric Als5/Als6 proteins were constructed by exchanging the N termini of each protein.
  • Chimeric ALS5/6 genes were constructed as follows. A BglII-HpaI fragment of ALS5 encompassing the 5′ 2117 bp of the gene was isolated.
  • pGEM-T-ALS6 was then digested with BglII and HpaI to release the corresponding 5′ 2126 bp of ALS6, and the fragment consisting of pGEM-T-Easy plus the 3′ sequences of ALS6 was isolated and ligated to the 5′ ALS5 fragment to generate plasmid pGEM-T-5N6C.
  • An identical approach using the corresponding 5′ fragment of ALS6 and 3′ fragment of ALS5 was used to generate plasmid p-GEM-T-6N5C.
  • each chimeric ALS gene was released by BglII-XhoI digestion and subcloned into pADH1 as above.
  • S. cerevisiae S150-2B was then transformed with these constructs, and expression was verified by Northern blot analysis before characterization of their adherence properties.
  • the endothelial cells were then perneabilized in 0.2% Triton X-100 in phosphate-buffered saline for 10 min, after which the cell-associated organisms (the internalized plus adherent organisms) were again stained with the anti- C. albicans antiserum conjugated with Alexa 488, which fluoresces green.
  • the coverslips were then observed under epifluorescence.
  • the number of organisms that had been internalized by the endothelial cells was determined by subtracting the number of adherent organisms (fluorescing red) from the number of cell-associated organisms (fluorescing green). At least 100 organisms were counted on each coverslip, and all experiments were performed in triplicate on at least three separate occasions.
  • Fibronectin bead adherence assays also was performed to further characterize the binding characteristics of certain Als proteins.
  • Als5p was originally identified by virtue of the protein's ability to induce agglutination of fibronectin-coated beads when expressed on the surface of S. cerevisiae (Gaur et al., Infect. Immune. 65:5289-5297 (1997)). Therefore, S. cerevisiae strains transformied with ALS5, ALS6, 5N6C, and 6N5C for fibronectin were tested for bead adherence using this methodology (Gaur et al., Infect. Immune. 65:5289-5297 (1997), Gaur et al., Infect. Immun.
  • tosylated magnetic beads (Dynal Biotech) were coated with fibronectin following the manufacturer's instructions.
  • 10 ⁇ l of coated beads (10 6 beads) were mixed with 1 ⁇ 10 8 transformed S. cerevisiae in 1 ml of 1 ⁇ Tris-EDTA (TE) buffer, pH 7.0, and incubated with gentle mixing for 45 min.
  • the tubes were placed in a magnet to separate beads and adherent S. cerevisiae from nonadherent organisms.
  • the supernatant containing nonadherent organisms was removed by aspiration, and the remaining beads were washed three times by resuspending in 1 ml of TE buffer, followed by magnetic separation and aspiration of the supernatant. Finally, the washed beads and adherent organisms were resuspended in 100 ⁇ l of TE buffer and examined microscopically for co-agglutination.
  • Als proteins also were found to be homologous to adhesins and invasins of the immunoglobulin superfamily.
  • a knowledge-based search algorithm was used to identify molecules that share significant structural similarity with Als family members. Briefly, homology and energy-based modeling was conducted to compare overall physicochemical features of Als proteins.
  • SWISS-MODEL knowledge-based method
  • Resulting Als N-terminal domain models were prioritized based on three criteria: (i) most favorable strain energy (molecular mechanics); (ii) empirical positional energy functions; and (iii) preservation of the spatial arrangement of potential disulfide bridging (Godzik et al., J. Mol. Biol. 227:227-238 (1992), Bowie et al., Science 253:164-170 (1991), Eisenberg et al., Methods Enzymol. 277:396-404 (1997), Fischer et al., FASEB J. 10:126-136 (1996), Luthy et al., Nature 356:83-85 (1992)).
  • Als proteins were also determined to contain N-terminal hypervariable regions that map to predicted loop/coil structures. In this regard, despite the observed differences in substrate-specific adherence mediated by individual Als proteins, large regions of sequence in the N-terminal domains are conserved across this family. However, seven regions of significant divergence among Als proteins designated hypervariable regions (HVRs) 1-7, were found. These regions (composed of 8 or more amino acids) contained no apparent consensus identity across Als proteins and less than 50% consensus conservation. In contrast, the intervening conserved regions (CRs) 1-7, displayed more than 30% consensus identity and more than 50% consensus conservation across Als proteins.
  • HVRs hypervariable regions
  • FIGS. 11 , A and B An identity plot and schematic alignment of these amino acid sequences comprising the N-terminal domains (residues 1-420 ) of Als proteins with known function is presented in FIGS. 11 , A and B.
  • homology modeling revealed that the HVRs of different Als proteins, while distinguishable in sequence, are predicted to conform to similar loop/coil structures that project from the ⁇ -sheet components of the CRs.
  • the presence of these conserved HVRs indicate that they are available to interact with host constituents.
  • the instrument was routinely calibrated with (+)-10-camphorsulfonic acid (1 mg/ml in a 1-mm path length cell) (Johnson et al., Proteins 7:205-214 (1990)), and ellipticity was expressed as the mean residue ellipticity (1)MRE (degrees-cm2 dmol-1).
  • MRE mean residue ellipticity
  • the protein concentration was determined by absorbance at 280 nm based on aromatic amino acid composition of the expressed Als1p domain (Pace et al, Protein Sci 4:2411-2423 (1995)).
  • the CD spectra were deconvoluted into helix, ⁇ -sheet, turn, and disordered structures using Selcon (Sreerama et al, Protein Sci.
  • Infrared spectra of Als1p self-films were recorded at 25° C. on a Bruker Vector 22 FTIR spectrometer (Bruker Optics) fitted with a deuterated triglycine sulfate detector at a gain of 4, averaged over 256 scans, and at a resolution of 2 cm ⁇ 1.
  • Fifty micrograms of the protein in 50 ⁇ l of phosphate-buffered saline were spread onto the surface of a 50 ⁇ 20 ⁇ 2-mm germanium attenuated total reflectance sample crystal (Pike Technologies) and allowed to dry. The dry protein self-film was then hydrated with D2O for 1 h prior to recording the infrared spectra.
  • Amide I bands of the infrared spectra were analyzed for secondary conformations by area calculations of component peaks with curve-fitting software (GRAMS/32, Version 5; Galactic).
  • the frequency limits for the various conformations were as follows: ⁇ -helix (1662-1645 cm ⁇ 1), ⁇ -sheet (1637-1613 and 1710-1682 cm ⁇ 1), ⁇ -turn loops (1682-1662 cm ⁇ 1), and disordered structures (1645-1637 cm ⁇ 1) (50-52).
  • Circular dichroism results of the N-terminal domain of Als1 p are shown in FIG. 12A and reveal a dichroic minimun at 217 nm and strong positive dichroic maximum near 200 nm. These features are characteristic of a protein having a dominant anti-parallel ⁇ sheet component. Deconvolution of the CD spectra indicated that the protein assumed conformations of 50.1% ⁇ sheet, whereas other structure class contributions include disordered structures (26.9%), turn structures (19.3%), and ⁇ -helix (3.7%).
  • Curve fitting of the spectra indicated that the protein construct is ⁇ 57.2% antiparallel ⁇ -sheet.
  • Other secondary structural conformations from curve fitting of the IR spectra include disordered structures (20.5%), turn components (13.3%), and ⁇ -helix (9.0%).
  • Three-dimensional models further indicate Physicochemical distinctions among Als N-terminal domains.
  • molecular models indicated differences in predicted physicochemical attributes of the N-terminal domains of Als proteins that likely influence their interactions with host cells and several substrates.
  • Als proteins are separable into three distinct groups based on surface distributions of hydrophobicity, charge, and hydrogen bonding potential.
  • Als1p, Als3p, and Als5p each share similar patterns of these properties and thus are considered the Als group A.
  • the predicted physicochemical properties of Als6p and Als7p N-terminal domains (Als group B) have striking differences from those of the Als group A ( FIG. 13 ).
  • Als group A members are typically segregated from their neutral or anionic facets
  • positive charge is broadly distributed across the entire surface of the Als group B members Als6p and Als7p.
  • Als group C the Als group C proteins that differ structurally from either the Als group A or B proteins.
  • the Als group C proteins would appear to be more similar to the Als group A than Als group B proteins in terms of hydrophobic or electrostatic distribution.
  • Hwp1p has been shown to mediate adherence to buccal epithelial cells by acting as a substrate for mammalian transglutaminase (5).
  • EAP1 was recently identified by heterologous expression in S. cerevisiae and mediates adherence to polystyrene and renal epithelial cells in vitro (7).
  • Als1p and Als5p have been studied from a functional perspective.
  • Heterologous expression of Als1p has been shown to mediate binding to human vascular endothelial cells and epithelial cells, a finding that has been confirmed in C.
  • the physicochemical properties of protein domains as distributed in three-dimensional space are crucial structural features governing receptor-ligand interactions (Eisenberg et al, J. Mol. Biol. 179:125-142 (1984), Waring et al., Protein Peptidew Lett. 3:177-184 (1996), Hancock et al., Lancet 349:418-422 (1997)).
  • the Als proteins share conformational features characteristic of other adhesins and invasins of the immunoglobulin superfamily. However, individual Als proteins differed in their primary homolog, a finding consistent with the experimental data indicating that members of the Als family exhibit different substrate-binding profiles. Collectively, these patterns of Als homologies indicate that, whereas Als protein members share a global similarity in structure and predicted fold, there exists structural differences among distinct Als proteins that are responsible for their differences in function.
  • variable loop regions significantly alter substrate binding in these homologous proteins (Renz et al., J. Cell Biol. 125-1395-1406 (1994), Viney et al., J. Immunol. 157:2488-2497 (1996)).
  • the three-dimensional modeling results further indicate that N-terminal domains of individual Als proteins possess distinctive molecular signatures that relate to their adhesive profiles. These signatures incorporate parameters such as surface area, hydrophobicity, and electrostatic charge, yielding configurations that distinguish structural relationships among Als proteins.
  • Als proteins that bind to multiple substrates such as the Als group A members (Als1p, Als3p, and Als5p) have similar predicted N-terminal profiles in terms of steric bulk, hydrophobic distribution, and electrostatic potential. Yet, even within this group, specific physicochemical distinctions exist that can govern functional differences within the group ( FIG. 13 ).
  • Als proteins with reduced adhesive capacity have surface features predicted to be distinct from the Als group A proteins in multiple physicochemical properties, including hydrophobicity and electrostatic potential. It is highly likely that the aggregate effects of differences in these structural features confer the specific functional properties of distinct Als proteins.
  • the above results indicate an analogy between antibodies and Als proteins at both the structural and functional level.
  • the homology modeling underscores the similarities in structural configurations of these families, with hypervariability targeted to localized domains within an otherwise stable framework (e.g. HVRs of Als proteins and Fab regions in immunoglobulins).
  • the genetic variability of the ALS gene family may provide the opportunity for Candida to display a diverse array of proteins with a spectrum of specificity in adherence and invasion. The availability of such a group of related proteins is likely to improve the ability of the organism to colonize and invade different anatomical and physiological niches during infection.
  • Vaccination with rAls1p-N Improves Survival during Murine Disseminated Candidiasis by Enhancing Cell-Mediated, not Humoral, Immunity
  • the C. albicans used in the study was SC5314, a well-characterized clinical isolate that is highly virulent in animal models (Spellberg et al., Infect Immun. 71 :5756-5764 (2003)) was supplied by W. Fonzi (Georgetown Univeristy). The organism was serially passaged three times in yeast peptone dextrose broth (Difco) prior to infection.
  • mice strains used in the study were female BALB/c mice obtained from the National Cancer Institute (Bethesda, Md.). To explore the impact of age on vaccine efficacy, both juvenile mice (8-10 weeks) and retired breeders ( ⁇ 6 months) were utilized.
  • Female B cell-deficient mice bearing a homozygous deletion of the igh loci (C.129B6-IgH-Jhdtm1Dhu), T cell-deficient nude mice (C.Cg/AnBomTac-Foxn1nuN20), and congenic wild-type BALB/c littermates were obtained from Taconic Farms (Germantown, N.Y.).
  • mice were housed in filtered cages with irradiated food and autoclaved water ad libitum.
  • mice were immunized with varying doses of antigen (see below) and subsequently infected via the tail vein with the appropriate inoculum of C. albicans SC5314 blastospores, or PBS (Irvine Scientific, Irvine, Calif.) control. Results of replicate survival studies were combined if the individual datasets demonstrated no statistical heterogeneity (see below). All procedures involving mice were approved by the institutional animal use and care committee, following the National Institutes of Health guidelines for animal housing and care.
  • rAls1p-N immunization procedures described below were performed as follows. Briefly, rAls1p-N (amino acids 17 to 432 of Als1p) was produced in S. cerevisiae and purified by gel filtration and Ni-NTA matrix affinity purification (Fu et al., Molec. Microbiol. 44:61-72 (2002)). The amount of protein was quantified by modified Lowry assay. A high degree of purity ( ⁇ 90%) was confirmed by SDS-polyacrylamide gel electrophoresis as well as circular dichroism and FTIR, as described above.
  • mice were immunized by intraperitoneal (ip) injection of rAls1p-N mixed with complete Freund's adjuvant (CFA, Sigma-Aldrich) at day 0, boosted with another dose of the antigen with incomplete Freund's adjuvant (IFA, Sigma-Aldrich) at day 21, and infected two weeks following the boost.
  • CFA complete Freund's adjuvant
  • IFA incomplete Freund's adjuvant
  • Resultant Antibody titers were determined by ELISA in 96 well plates. Briefly, wells were coated with 100 ⁇ l per well of 5 ⁇ g/ml rAls1p-N in PBS. Mouse sera were incubated for 1 h at room temperature following a blocking step with tris buffer saline (TBS) (0.01 M TrisHCl, pH 7.4, 0.15 M NaCl) containing 3% bovine serum albumin. The wells were washed 3 times with TBS containing 0.05% Tween 20, followed by another 3 washes with TBS.
  • TBS tris buffer saline
  • Goat anti-mouse secondary antibody conjugated with horseradish peroxidase was added at a final dilution of 1:5000 and the plate was further incubated for 1 h at room temperature. Wells were washed with TBS and incubated with substrate containing 0.1 M citrate buffer (pH 5.0), 50 mg/ml of o-phenylenediamine (Sigma), and 10 ⁇ l of 30% H 2 O 2 . The color was allowed to develop for 30 min after which the reaction was terminated by adding 10% H 2 SO 4 and the optical density (OD) was determined at 490 nm in a microtiter plate reader.
  • Negative control wells received only diluent, and background absorbance was subtracted from the test wells to obtain final OD readings.
  • the ELISA titer was taken as the reciprocal of the last serum dilution that gave a positive OD reading (i.e.>mean OD of negative control samples+2standard deviation).
  • C. albicans -induced cytokine profiles were performed to determine the effect of the rAls1p-N vaccine on cell-mediated immunity and in vivo cytokine profiles. Mice were immunized as described above. Two weeks after the final boost, splenocytes were harvested and cultured in complete media at a density of4 ⁇ 10 6 cells/ml as previously described (Spellberg et al., Infect. Immun. 71:5756-5764 (2003)). To stimulate cytokine production, splenocytes were co-cultured with heat-killed C. albicans SC5314 germ tubes. We used heat-killed C.
  • the C. albicans cells were pre-germinated in RPMI-1640 with glutamine (Gibco BRL) for 90 minutes to induce expression of Als1p (Fu et al., Molec. Microbiol. 44:61-72 (2002)).
  • the resulting C. albicans germ tubes were heat-killed by incubation for 90 minutes at 60° C. (Ibrahim et al., Infect. Immun. 63:4368-74 (1995)).
  • the heat-killed fungi were added to the splenocyte cultures at a density of 2 ⁇ 10 7 pseudohyphae/ml (ratio of five fungi to one leukocyte). After 48 h, splenocytes were profiled for Th1 (CD4+IFN- ⁇ +IL-4 ⁇ ), Th2 (CD4+IFN- ⁇ -IL-4+), or CD4+IL-10+ frequencies by intracellular cytokine detection and flow cytometry, as previously described (Spellberg et al., Infect. Immun. 71:5756-5764 (2003)).
  • Three-color flow cytometry was performed on a Becton-Dickinson FACScan instrument calibrated with CaliBRITE beads (Becton Dickinson, San Jose, Calif.) using FACSComp software as per the manufacturer's recommendations.
  • CD4+ lymphocytes were gated by concatenation of forward and side scatter, and FITC-anti-CD4 antibody fluorescence properties. Data for each sample were acquired until 10,000 CD4+ lymphocytes were analyzed. Results are presented as the median ⁇ 25th and 75th quartiles of the percentage of all gated lymphocytes that were Th1 or Th2 cells.
  • Footpad swelling was determined by the method of Oomura et al (41). Briefly, mice were immunized with the appropriate dose of rAls1p-N or CFA alone as described above. Two weeks following the boost, baseline footpad sizes of immunized mice were measured using an electronic digital caliper. Fifty ⁇ g of rAls1p-N in 25 ⁇ l of PBS was injected into the right footpads, and PBS alone injected into the left footpads of the immunized mice. Twenty-four hours later the footpads were again measured. Antigen-specific footpad swelling was calculated as: (right footpad thickness at 24 h ⁇ right footpad thickness at baseline) ⁇ (left footpad thickness at 24 h ⁇ left footpad thickness at baseline).
  • the non-parametric Log Rank test was utilized to determine differences in survival times of the mice. Titers of antibody, frequency of Th1 or Th2 lymphocytes, and footpad swelling were compared by the Steel test for non-parametric multiple comparisons (Rhyne et al., Biometrics 23:539-49 (1967) or the Mann Whitney U test for unpaired comparisons, as appropriate. Correlations were calculated with the Spearman Rank sum test. To determine if heterogeneity existed in replicate survival studies, the Kolmogorov-Smimov test was utilized. P values ⁇ 0.05 were considered significant.
  • mice Female retired breeder BALB/c mice were immunized with rAls1p-N plus adjuvant (CFA/IFA) or adjuvant alone. Immunized mice were bled 2 weeks after boosting to determine anti-rAls1p-N antibody titers (see below). The mice were subsequently infected with a lethal inoculum of C. albicans (2 ⁇ 10 5 blastospores). The survival data from repeat experiments were combined since the individual experiments demonstrated no statistical heterogeneity (p>0.05 by Kolmogorov-Smirnov test).
  • To determine if the age of the mice influenced their response to the rAls1p-N vaccine, we tested it in juvenile mice. A similar survival benefit was found when juvenile mice were vaccinated and infected with the same high inoculum (p 0.02 by Log Rank test, FIG. 16B ).
  • mice immunized with the highest dose of antigen 200 ⁇ g had anti-rAls1p-N antibody titers in excess of 1:100,000, but had survival durations no different from mice immunized with lower doses of antigen whose titers were at the lower limit of detection ( ⁇ 1:100).
  • rAls1p-N Since humoral immunity did not correlate with rAls1p-N-induced protection, we examined the cell-mediated immune response induced by protective and non-protective doses of rAls1p-N. Mice were immunized with 0.2, 20, or 200 ⁇ g of rAls1p-N, or adjuvant alone, as above. Two weeks after the boost, splenocytes were harvested and cultured in the presence of heat-killed, pre-germinated C. albicans , which are known to express Als1p (Fu et al., Molec. Microbiol. 44:61-72 (2002)). Following 48 h of culture, splenocytes were harvested for intracellular cytokine detection by flow cytometry.
  • B cell-deficient, T-cell deficient nude, or congenic BALB/c wild-type control-mice were immunized with 20 ⁇ g of rAls1p-N plus adjuvant or adjuvant alone, and infected with a lethal inoculum (8 ⁇ 10 5 blastospores) of C. albicans .
  • the inverted U-shaped dose-response efficacy curve is reminiscent of the classical studies of Parish et al., who first described the inverse relationship between the induction of humoral and cell-mediated immunity by a given dose of antigen.
  • an inverted U-shaped dose-response efficacy curve could be explained if: 1) vaccine efficacy depended on cell-mediated immunity and, 2) intermediate doses of rAls1p-N stimulated superior cell-mediated immunity compared to the high, antibody-stimulating dose.
  • the inverted U-shaped dose response efficacy curve seen with the rAls1p-N vaccine was due to superior induction of cell-mediated immunity by the protective, intermediate doses of antigen.
  • Th1 cells were of functional significance in vivo
  • Th2 cells were activated in peripheral lymph nodes rather than the spleen.
  • other T cell populations e.g. NKT cells
  • NKT cells may have been responsible for inducing the high antibody titers seen in response to the 200 ⁇ g dose of rAls1p-N.
  • the Anti- Candida albicans rAls1p-N Vaccine Reduces Fungal Burden and Improves Survival in both Immunocompetent and Immunocompromised Mice
  • This example describes enhancement of the efficacy of the rAls1p-N vaccine described in example VII when administered by a subcutaneous (SQ) route in both immunocompetent and immunocompromised mice.
  • SQ subcutaneous
  • rAls1p-N encompassing amino acids 19-433 of the full length protein, was produced in S. cerevisiae and purified as described above. Control preparation was similarly purified from S. cerevisiae transformed with an empty plasmid.
  • mice 25-30 g were immunized by SQ injection of rAls1p-N (20 ⁇ g) or control preparation mixed with Complete Freund's Adjuvant (CFA) at day 0, followed by a booster dose in Incomplete Freund's Adjuvant (IFA) at day 21.
  • CFA Complete Freund's Adjuvant
  • IFA Incomplete Freund's Adjuvant
  • the efficacy of the rAls1p-N vaccine was evaluated by determining the impact of rAls1p-N vaccination on survival in infected BALB/c mice ( FIG. 22A ).
  • Vaccinated or control mice were infected via the tail-vein with rapidly lethal inocula (2.5-5 ⁇ 10 5 blastospores) of C. albicans .
  • Vaccination markedly prolonged time to death (p ⁇ 0.05 for both inocula by Log Rank test) and improved 30 day survival (50-57% vs. 0%, p ⁇ 0.05 for both inocula by Fisher's Exact test).
  • the efficacy of the rAls1p-N vaccine also was assessed in immunocompromised mice. Having demonstrated efficacy in immunocompetent mice, the potential for the rAls1p-N vaccine to induce immunity in and protect neutropenic mice from disseminated candidiasis also was evaluated.
  • Vaccinated BALB/c mice were made neutropenic by administration of cyclophosphamide (200 mg/kg ip on day ⁇ 2, and 100 mg/kg ip on day +9 relative to infection, resulting in approximately 12 days of neutropenia, as described (Sheppard et al., Antimicrob. Agents. Chemother. 48:1908-11 (2004)).
  • Footpad swelling reaction was performed 2 days after the first dose of cyclophosphamide.
  • Vaccinated neutropenic mice developed DTH reactions of similar magnitude to immunocompetent mice ( FIG. 23A vs. 1, experiments performed in parallel).
  • Vaccinated mice were treated with cortisone acetate (225 mg/kg SQ on days ⁇ 1, 1, and 3 relative to infection) and infected sublingually as described. Tongues were excised on day 5 post-infection.
  • OPC murine oropharyngeal candidiasis
  • mice developed marked fungal invasion of their tongues in numerous locations, while only two vaccinated mouse developed any tongue lesions.
  • mice were treated with estrogen (30 ⁇ g, given SQ) on day ⁇ 3 relative to infection and then challenged in the vagina with 10 ⁇ l phosphate buffered saline containing 10 6 blastospores of C. albicans .
  • kidney fungal burden indicative of a fatal infection is 5 log CFU/g; mice with kidney fungal burdens above this level typically die from infection, whereas mice with kidney fungal burdens below this burden survive the infection (Spellberg et al., J. Infect. Dis. In press (2005) and (Spellberg et al., Infect. Immun. 71:5756-5764 (2003)). Therefore, breakthrough deaths in the vaccinated group likely reflect high fungal burden in spite of vaccination.
  • the mouse to mouse variations in tissue fungal burden may reflect the complexities of host-pathogen interactions and/or variable vaccine responsiveness.
  • the rAls1p-N vaccine can be used for the treatment, reduction in severity and/or prevention of increasingly common and highly lethal disseminated candidiasis.
  • the vaccine is efficacious in immunocompetent mice, and efficacy is retained even in neutropenic and corticosteroid-treated hosts.
  • the vaccine can protect against mucocutaneous candidiasis including vaginal and oropharyngeal candidiasis
  • Als adhesins of C. albicans were identified to be significantly homologous to adhesins on S. aureus . This characteristic was used to design and implement an effective vaccine against S. aureus using Als adhesins.
  • the C. albicans ALS family is comprised of at least 9 genes (Hoyer et al., Genetics 157:1555-67 (2001); Hoyer L L., Trends Microbiol. 9:176-80 (2001)).
  • Als proteins function as adhesins to biologically relevant substrates (Fu et al., Molec. Microbiol. 44:61-72 (2002); Gaur and Klotz, Intfect. Immun.
  • homology calculation takes into account both features of sequence alignment and 3-dimensional surface structure.
  • Homology of Als1p was calculated to be greater than 95% 90% compared to collagen binding protein or clumping factor of S. aureus (r 2 ⁇ 90%; Sheppard et al., supra).
  • homology of Als3p was calculated to be greater than 95% 80% compared to collagen binding protein or clumping factor of S. aureus (r 2 ⁇ 90%).
  • the predicted functional cleft motif in Als1p maps to specific residues originating from hypervariable regions in the N-terminal region encompassing amino acid residues 30-300.
  • a monoclonal antibody against S. aureus also was identified that may reduce infections caused by C. albicans . As with the above structural findings, this characteristic also was used to design and implement an effective vaccine against S. aureus using Als adhesins.
  • exemplary Als adhesin vaccines were designed and shown to improve survival of mice infected with S. aureus .
  • the exemplary Als adhesins used to vaccinate were rAls1p-N or rAls3p-N, which were produced and used as described above. Briefly, to determine if these Als vaccines against Candida , rAls1p-N and rAls3p-N, can mediate cross-species protection against S.
  • mice Female Balb/c mice were vaccinated with the previously described regimen (Complete Freund's Adjuvant+20 ⁇ g of rAls1p-N or rAls3p-N on day 0, followed by a booster dose in Incomplete Freund's Adjuvant at 3 weeks, both administered subcutaneously). Two weeks following vaccination, mice were infected via the tail-vein with a lethal dose of S. aureus strain 67-0, which is methicillin-resistant and known to be virulent in animal models. The results showing mice survival are shown in FIG. 26 . As indicated, both the rAls1p-N and rAls3p-N vaccines mediated improved long-term survival in these infected mice ( FIG.
  • the Anti- Candida rAls1p-N Vaccine Mediates a Broad Range of Protection Against Disseminated Candidiasis
  • mice were obtained from the National Cancer Institute (Bethesda, Md.). All procedures involving mice were approved by the institutional animal use and care committee, following the National Institutes of Health guidelines for animal housing and care. The mice were vaccinated with rAls1p-N+Freund's adjuvant as previously described above and in, for example, Ibralim et al., Infect. Immun. 73:999-1005 (2005); Spellberg et al., Infect. Immun. 73:6191-93 (2005).
  • rAls1p-N (amino acids 17 to 432 of Als1p) was produced in S. cerevisiae and purified by gel filtration and Ni-NTA matrix affinity purification. A high degree of purity ( ⁇ 90%) was confirmed by SDS-polyacrylamide gel electrophoresis as well as circular dichroism and FTIR, as described above and in, for example, Sheppard et al., J Biol Chem 279:30480-89 (2004). Mice were immunized by SQ injection of rAls1p-N (20 ⁇ g) mixed with Complete Freund's Adjuvant (CFA; Sigma-Aldrich, St.
  • CFA Complete Freund's Adjuvant
  • mice were immunized with CFA/IFA alone.
  • immunized mice were infected via the tail-vein with C. albicans SC5314, as we have described previously Ul previously Ul et al., (2005) supra; and Spellberg et al. (2005), supra. Similar to our previous findings in Balb/c mice, the rAls1p-N vaccine markedly improved the survival of infected CD1 mice ( FIG. 29A ).
  • the rAls1p-N vaccine also was shown to improve the survival of Balb/c mice infected with several strains of C. albicans .
  • Particularly useful vaccines utilize an immunogen that can prime the immune system to recognize multiple strains of the target pathogen.
  • DNA sequence analysis we found that the predicted amino acid sequence of the N-terminal region of Als1p was 99.9% conserved amongst a diverse group of clinical C. albicans isolates from bloodstream (5 strains), urine (5 strains) and oropharyngeal (10 strains) infections (data not shown). These results indicated that the rAls1p-N vaccine can be effective against a broad array of C. albicans strains.
  • mice were vaccinated with rAls1p-N+Freund's adjuvant as above, and infected with one of several clinical isolates of C. albicans (Ibrahim et al., Infect Immun 63:1993-98 (1995)). As shown in FIG. 30 , the rAls1p-N vaccine significantly improved the survival of mice infected with each of these strains.
  • the rAls1p-N vaccine also was shown to reduce tissue fungal burden in mice infected with several non-albicans species of Candida .
  • the ALS gene family is present in other Candida species, including C. dubliniensis and C. tropicalis (Hoyer et al., Genetics 157:1555-67 (2001)).
  • an adhesin analogous to Als family members has been described in C. glabrata (Cormack et al., Science 285:578-82 (1999); Frieman et al., Mol Microbiol 46:479-92 (2002)).
  • mice were vaccinated with rAls1p-N+Freund's adjuvant as above, and infected via the tail-vein with C. glabrata 31028 (a clinical bloodstream isolate from the microbiology laboratory at Harbor-UCLA Medical Center), C. krusei 91-1159, (generously provided by Michael Rinaldi, San Antonio, Tex.), C. parapsilosis 22019 (clinical bloodstream isolate from Harbor-UCLA Medical Center), or C. tropicalis. 4243 (clinical bloodstream isolate from Harbor-UCLA Medical Center).
  • C. glabrata 31028 a clinical bloodstream isolate from the microbiology laboratory at Harbor-UCLA Medical Center
  • C. krusei 91-1159 (generously provided by Michael Rinaldi, San Antonio, Tex.)
  • C. parapsilosis 22019 clinical bloodstream isolate from Harbor-UCLA Medical Center
  • C. tropicalis. 4243 clinical bloodstream isolate from Harbor-UCLA Medical Center
  • the rAls1p-N vaccine is able to prevent and/or reduce the severity of an increasingly common and highly lethal disseminated candidiasis.
  • the vaccine is efficacious in both inbred and outbred mice, when mixed with alum as an adjuvant, against multiple strains of C. albicans , and against several non-albicans species of Candida .
  • This Example compares the efficacy of rAls3p-N to rAls1p-N vaccines in murine models of hematogenously disseminated, oropharyngeal, and vaginal candidiasis.
  • the ALS1 and ALS3 genes encode adhesins with the broadest array of substrate affinity.
  • Als1p mediated greater adherence to endothelial cells and gelatin, but inferior adherence to epithelial cells (Sheppard et al., J Biol Chem 279:30480-89 (2004)).
  • Their differences in adherence qualities suggested that rAls3p-N may have different efficacy as a vaccine immunogen compared to rAls1p-N.
  • rAls1p-N and rAls3p-N amino acids 17 to 432 Als1p or Als3p
  • the amount of protein was quantified by modified Lowry assay. A high degree of purity ( ⁇ 90%) was confirmed by SDS-polyacrylamide gel electrophoresis as well as circular dichroism and FTIR, as described above and in (2004) et al., (2005), supra; Spellberg et al., (2005), supra).
  • mice were immunized by subcutaneous (SQ) injection of 20 ⁇ g of rAls1p-N or rAls3p-N mixed with Complete Freund's adjuvant (CFA, Sigma-Aldrich, St. Louis, Mo.) at day 0, boosted with another dose of the antigen with Incomplete Freund's adjuvant (IFA, Sigma-Aldrich) at day 21, and infected two weeks following the boost.
  • CFA Complete Freund's adjuvant
  • IFA Incomplete Freund's adjuvant
  • mice vaccinated with CFA+rAls1p-N or rAls3p-N developed antibody titers significantly greater than mice receiving CFA alone.
  • mice vaccinated with rAls3p-N generated anti-rAls1p-N antibodies at equivalent titers to mice vaccinated with rAls1p-N ( FIG. 32 , top).
  • mice vaccinated with rAls1p-N generated smaller titers against rAls3p-N than did mice vaccinated with rAls3p-N ( FIG. 32 , bottom).
  • both rAls1p-N and rAls3p-N resulted in similar delayed type hypersensitivity responses in vivo as shown in FIG. 33 .
  • mice were vaccinated with CFA, CFA+rAls1p-N, or CFA+rAls3p-N, and subsequently infected via the tail-vein with C. albicans .
  • the results shown in FIG. 34 demonstrate that both the rAls1p-N and rAls3p-N vaccines resulted in significant improvement in survival.
  • mice sera were incubated for 1 h at room temperature following a blocking step with tris buffer saline (TBS) (0.01 M TrisHCl, pH 7.4, 0.15 M NaCl) containing 3% bovine serum albumin.
  • TBS tris buffer saline
  • the wells were washed 3 times with TBS containing 0.05% Tween 20, followed by another 3 washes with TBS without Tween.
  • Goat anti-mouse IgG secondary antibody conjugated with horseradish peroxidase (Sigma-Aldrich) was added at a final dilution of 1:5000 and the plate was further incubated for 1 h at room temperature.
  • mice were immunized with rAls1p-N, rAls3p-N, or CFA alone. Two weeks following the boost, baseline footpad sizes of immunized mice were measured using an electronic digital caliper. Fifty ⁇ g of rAls1p-N or rAls3p-N in 25 ⁇ l of PBS were injected into the right footpads, and PBS alone injected into the left footpads of the immunized mice. Twenty-four hours later the footpads were again measured. Antigen-specific footpad swelling was calculated as: (right footpad thickness at 24 h ⁇ right footpad thickness at baseline) ⁇ (left footpad thickness at 24 h ⁇ left footpad thickness at baseline).
  • the rAls3p-N vaccine also demonstrated more efficacy than rAls1p-N in two models of mucosal candidiasis. Because Als3p mediated superior adhesion to epithelial cells compared to Als1p, this observation indicates that rAls3p-N can exhibit unique efficacy in mucosal models of infection. The efficacy of rAls1p-N compared to rAls3p-N assessed in a steroid-treated, oropharyngeal model of infection and in a model of candidal vaginitis.
  • mice were immunocompromised by treatment with cortisone acetate (225 mg/kg SQ on days ⁇ 1, 1, and 3 relative to infection). On the day of infection, the mice were anesthetized by intraperitoneal injection with 8 mg xylazine and 110 mg ketamine per kg. Calcium alginate urethral swabs were saturated with C.
  • albicans by placing them in a suspension of 10 6 organisms per ml in HBSS at 30° C.
  • the saturated swabs were placed sublingually in the oral cavity of the mice for 75 min. After 5 days of infection, the tongue and hypoglossal tissue were excised, weighed, homogenized, and then quantitatively cultured to determine the oral fungal burden.
  • mice were treated with 30 ⁇ g of subcutaneous estradiol valerate dissolved in peanut oil (both from Sigma-Aldrich) on day ⁇ 3 relative to infection to induce pseudoestrus.
  • mice were sedated by ip administration of 100 mg/kg of ketamine. Sedated mice were infected intravaginally with 10 6 blastospores of C. albicans in 10 ⁇ l of HBSS.
  • vaginas and approximately one centimeter of each uterine horn were dissected en block, homogenized, and quantitatively cultured.
  • the overall magnitude of the benefit was ⁇ 0.3 log CFU/gram ( FIG. 36 ).
  • Antibody titers did not correlate with the protective effect of either vaccine during disseminated candidiasis, but induction of delayed type hypersensitivity in vivo did correlate with protection. These data also further corroborate the mechanism of vaccine-induced protection was induction of Type 1, cell-mediated immunity to the fungus. Both rAls1p-N and rAls3p-N induced equivalent titers of antibody against rAls1p-N, but that rAls3p-N induced significantly higher titers of anti-rAls3p-N antibodies than did rAls1p-N.
  • the anti-candidal rAls3p-N vaccine induced equivalent cell-mediated but broader antibody-based responses than did the rAls1p-N vaccine.
  • the immunogens resulted in an equivalent degree of protection against hematogenously disseminated candidiasis, but rAls3p-N mediated greater protection against both oropharyngeal and vaginal candidiasis.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Immunology (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Mycology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Oncology (AREA)
  • Diabetes (AREA)
  • Communicable Diseases (AREA)
  • Microbiology (AREA)
  • Epidemiology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Transplantation (AREA)
  • Emergency Medicine (AREA)
  • Endocrinology (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
US11/327,197 1999-11-19 2006-01-06 Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis and other infectious agents Abandoned US20070077256A1 (en)

Priority Applications (22)

Application Number Priority Date Filing Date Title
US11/327,197 US20070077256A1 (en) 1999-11-19 2006-01-06 Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis and other infectious agents
PCT/US2007/000433 WO2007081896A2 (en) 2006-01-06 2007-01-05 Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis and other infectious agents
EP07709622A EP1982190B1 (de) 2006-01-06 2007-01-05 Pharmazeutische zusammensetzungen und verfahren für die impfung gegen disseminierte candidiase und andere infektiöse mittel
EP12001586.2A EP2532360B1 (de) 2006-01-06 2007-01-05 Pharmazeutische Zusammensetzungen und Verfahren zum Impfen gegen vaginal Candidose
AU2007205065A AU2007205065B2 (en) 2006-01-06 2007-01-05 Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis and other infectious agents
CA2948424A CA2948424A1 (en) 2006-01-06 2007-01-05 Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis and other infectious agents
EP12001595A EP2532361A1 (de) 2006-01-06 2007-01-05 Pharmazeutische Zusammensetzungen und Verfahren zum Impfen gegen disseminierte Candidose und andere infektiöse Einheiten
PT07709622T PT1982190E (pt) 2006-01-06 2007-01-05 Composições farmacêuticas e métodos de vacinação contra a candidíase disseminada e outros agentes infecciosos
CA2636277A CA2636277C (en) 2006-01-06 2007-01-05 Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis and other infectious agents
EP11008862.2A EP2428800B1 (de) 2006-01-06 2007-01-05 Pharmazeutische Zusammensetzungen und Verfahren zum Impfen gegen mukosale Candidose
US12/160,073 US20090297562A1 (en) 2006-01-06 2007-01-05 Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis and other infectious agents
ES12001586.2T ES2608354T3 (es) 2006-01-06 2007-01-05 Composiciones farmacéuticas y métodos para vacunar contra candidiasis vaginales
ES07709622T ES2391580T3 (es) 2006-01-06 2007-01-05 Composiciones farmacéuticas y métodos para vacunar contra la candidiasis diseminada y otros agentes infecciosos
JP2008549598A JP5692963B2 (ja) 2006-01-06 2007-01-05 播種性カンジダ症および他の感染性因子に対するワクチン接種をするための医薬組成物および方法
DK07709622.0T DK1982190T3 (da) 2006-01-06 2007-01-05 Farmaceutiske sammensætningerbog fremgangsmåder til vaccination mod dissemineret candidiasis og andre smitstoffer
ES11008862.2T ES2608309T3 (es) 2006-01-06 2007-01-05 Composiciones farmacéuticas y métodos para vacunar contra candidiasis mucosales
US12/987,949 US20120014995A1 (en) 1999-11-19 2011-01-10 Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis and other infectious agents
JP2012207831A JP5775044B2 (ja) 2006-01-06 2012-09-21 播種性カンジダ症および他の感染性因子に対するワクチン接種をするための医薬組成物および方法
CY20121100979T CY1113693T1 (el) 2006-01-06 2012-10-18 Φαρμακευτικες συνθεσεις και μεθοδοι για τον εμβολιασμο κατα της διαχυτης καντιντιασης και αλλων μολυσματικων παραγοντων
US13/785,835 US20140037689A1 (en) 1999-11-19 2013-03-05 Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis and other infectious agents
JP2014105980A JP2014148547A (ja) 2006-01-06 2014-05-22 播種性カンジダ症および他の感染性因子に対するワクチン接種をするための医薬組成物および方法
US15/014,440 US10300120B2 (en) 2002-09-13 2016-02-03 Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis and other infectious agents

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US16666399P 1999-11-19 1999-11-19
US09/715,876 US7067138B1 (en) 1999-11-19 2000-11-18 Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis
US10/245,802 US8541008B2 (en) 1999-11-19 2002-09-13 Pharmaceutical compositions and methods to vaccinate against candidiasis
US11/123,873 US20060083750A1 (en) 1999-11-19 2005-05-05 Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis
US11/327,197 US20070077256A1 (en) 1999-11-19 2006-01-06 Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis and other infectious agents

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/123,873 Continuation-In-Part US20060083750A1 (en) 1999-11-19 2005-05-05 Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/987,949 Continuation US20120014995A1 (en) 1999-11-19 2011-01-10 Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis and other infectious agents

Publications (1)

Publication Number Publication Date
US20070077256A1 true US20070077256A1 (en) 2007-04-05

Family

ID=38256965

Family Applications (5)

Application Number Title Priority Date Filing Date
US11/327,197 Abandoned US20070077256A1 (en) 1999-11-19 2006-01-06 Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis and other infectious agents
US12/160,073 Abandoned US20090297562A1 (en) 2006-01-06 2007-01-05 Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis and other infectious agents
US12/987,949 Abandoned US20120014995A1 (en) 1999-11-19 2011-01-10 Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis and other infectious agents
US13/785,835 Abandoned US20140037689A1 (en) 1999-11-19 2013-03-05 Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis and other infectious agents
US15/014,440 Expired - Fee Related US10300120B2 (en) 2002-09-13 2016-02-03 Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis and other infectious agents

Family Applications After (4)

Application Number Title Priority Date Filing Date
US12/160,073 Abandoned US20090297562A1 (en) 2006-01-06 2007-01-05 Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis and other infectious agents
US12/987,949 Abandoned US20120014995A1 (en) 1999-11-19 2011-01-10 Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis and other infectious agents
US13/785,835 Abandoned US20140037689A1 (en) 1999-11-19 2013-03-05 Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis and other infectious agents
US15/014,440 Expired - Fee Related US10300120B2 (en) 2002-09-13 2016-02-03 Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis and other infectious agents

Country Status (10)

Country Link
US (5) US20070077256A1 (de)
EP (4) EP2532360B1 (de)
JP (3) JP5692963B2 (de)
AU (1) AU2007205065B2 (de)
CA (2) CA2636277C (de)
CY (1) CY1113693T1 (de)
DK (1) DK1982190T3 (de)
ES (3) ES2391580T3 (de)
PT (1) PT1982190E (de)
WO (1) WO2007081896A2 (de)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2007081896A3 (en) * 2006-01-06 2007-11-22 Los Angeles Biomed Res Inst 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
EP3552620A1 (de) * 2018-04-11 2019-10-16 Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center Verfahren zur behandlung von infektionen mit candida auris
US10653757B2 (en) 2011-07-22 2020-05-19 Novadigm Therapeutics, Inc. Methods and compositions for vaccinating against Staphylococcus aureus
US10857216B2 (en) 2016-03-09 2020-12-08 Novadigm Therapeutics, Inc. Methods and kits for use in preventing and treating vulvovaginal candidiasis
CN112203684A (zh) * 2018-04-10 2021-01-08 海港医学中心洛杉矶生物医学研究所 耳念珠菌感染的治疗方法

Families Citing this family (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060083750A1 (en) * 1999-11-19 2006-04-20 Los Angeles Biomedical Research Institute At Harbor-Ucla Medical Center Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis
US8541008B2 (en) * 1999-11-19 2013-09-24 Los Angeles Biomedical Research Institute At Harbor-Ucla Medical Center Pharmaceutical compositions and methods to vaccinate against candidiasis
MX2012000018A (es) * 2009-07-03 2012-03-29 Los Angeles Biomed Res Inst Hyr1 como objetivo para inmunizacion activa y pasiva contra candida.
WO2012163533A1 (en) 2011-06-01 2012-12-06 Eth Zurich T cell epitope of candida albicans
JP6566616B2 (ja) * 2014-08-19 2019-08-28 キヤノン株式会社 通信装置、その制御方法、およびプログラム
HRP20231453T1 (hr) 2015-11-16 2024-03-01 Medincell S.A. Postupak za morseliranje i/ili usmjeravanje farmaceutski aktivnih sastojaka na sinovijalno tkivo
US10308571B2 (en) * 2017-10-11 2019-06-04 Uop Llc Process for minimizing benzene, toluene, and a recycle loop in a zero benzene aromatics complex

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020102262A1 (en) * 1996-05-16 2002-08-01 Magnus Hook Collagen binding protein compositions and methods of use
US20030124134A1 (en) * 1999-11-19 2003-07-03 Harbor-Ucla Research And Education Institute Pharmaceutical compositions and methods to vaccinate against candidiasis
US20040175731A1 (en) * 2002-11-12 2004-09-09 The Brigham And Women's Hospital, Inc. Methods and products for treating staphylococcal infections
US20050287146A1 (en) * 2003-12-19 2005-12-29 Joseph Patti Method of inhibiting Candida-related infections using donor selected or donor stimulated immunoglobulin compositions
US20060083750A1 (en) * 1999-11-19 2006-04-20 Los Angeles Biomedical Research Institute At Harbor-Ucla Medical Center Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis
US7666438B1 (en) * 1998-08-31 2010-02-23 Inhibitex, Inc. Multicomponent vaccines

Family Cites Families (35)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4346074A (en) 1978-08-24 1982-08-24 National Research Development Corp. Pasteurellosis vaccines
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5703055A (en) 1989-03-21 1997-12-30 Wisconsin Alumni Research Foundation Generation of antibodies through lipid mediated DNA delivery
US5622939A (en) 1992-08-21 1997-04-22 Alpha-Beta Technology, Inc. Glucan preparation
US5877397A (en) 1990-08-29 1999-03-02 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5961970A (en) 1993-10-29 1999-10-05 Pharmos Corporation Submicron emulsions as vaccine adjuvants
AU2657495A (en) 1994-05-23 1995-12-18 Research And Development Institute, Inc. Candida albicans adhesin as a vaccine
US5668263A (en) 1994-12-16 1997-09-16 Smithkline Beecham Corporation Conserved yeast nucleic acid sequences
ES2155129T3 (es) 1995-06-07 2001-05-01 Pfizer Vacunacion in ovo contra la coccidiosis.
US6747137B1 (en) 1998-02-13 2004-06-08 Genome Therapeutics Corporation Nucleic acid sequences relating to Candida albicans for diagnostics and therapeutics
US6248329B1 (en) * 1998-06-01 2001-06-19 Ramaswamy Chandrashekar Parasitic helminth cuticlin nucleic acid molecules and uses thereof
US7067138B1 (en) 1999-11-19 2006-06-27 Los Angeles Biomedical Research Institute At Harbor-Ucla Medical Center Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis
US20070077256A1 (en) * 1999-11-19 2007-04-05 Los Angeles Biomedical Research Institute Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis and other infectious agents
US7435871B2 (en) 2001-11-30 2008-10-14 Amgen Fremont Inc. Transgenic animals bearing human Igλ light chain genes
CA2486618C (en) 2002-05-21 2013-04-09 Schering-Plough Ltd. Methods for the in vitro culture of sporozoea sp. and uses thereof
US7241613B1 (en) 2002-06-05 2007-07-10 Oscient Pharmaceuticals Corporation Identification of Candida cell surface proteins and their uses
MXPA06005639A (es) 2003-11-21 2006-08-17 Pfizer Prod Inc Adyuvante antimicrobiano.
GB0420466D0 (en) 2004-09-14 2004-10-20 Cassone Antonio Anti-glucan antibodies
WO2006036817A2 (en) 2004-09-24 2006-04-06 Microbia, Inc. Fungal variants and uses thereof
CA2489194A1 (en) 2004-12-03 2006-06-03 Guilhem Janbon Polypeptides involved in candida biofilm formation and uses thereof
WO2007126813A2 (en) 2006-03-29 2007-11-08 Inhibitex, Inc. Cross-reactive monoclonal antibodies recognizing als family proteins
US7732187B2 (en) 2006-04-17 2010-06-08 Intervet Inc. Recombinant attenuated clostridium organisms and vaccine
EP2164511A1 (de) 2007-05-22 2010-03-24 Baylor College Of Medicine Immunkomplex-impfung als strategie zur verstärkung der immunität von älteren menschen und anderen immumgeschwächten personen
JP2011521955A (ja) 2008-05-29 2011-07-28 インターベツト・インターナシヨナル・ベー・ベー コクシジウム症ワクチン
US20100150942A1 (en) 2008-12-03 2010-06-17 Cantor Thomas L Affinity purified human polyclonal antibodies and methods of making and using them
US8568735B2 (en) 2009-06-22 2013-10-29 Wyeth Llc Immunogenic compositions of Staphylococcus aureus antigens
MX2012000018A (es) 2009-07-03 2012-03-29 Los Angeles Biomed Res Inst Hyr1 como objetivo para inmunizacion activa y pasiva contra candida.
WO2012163533A1 (en) 2011-06-01 2012-12-06 Eth Zurich T cell epitope of candida albicans
JP2014521605A (ja) 2011-07-22 2014-08-28 ノバダイム セラピューティクス, インコーポレイテッド Staphylococcusaureusに対してワクチン接種するための方法および組成物
UA115305C2 (uk) 2011-09-14 2017-10-25 Лос-Анджелес Біомедікал Ресьорч Інститут Ет Харбор-Укла Медікал Центер Композиції hyr1-похідних і способи лікування ними
WO2014144222A2 (en) 2013-03-15 2014-09-18 Los Angeles Biomedical Research Institute At Harbor-Ucla Medical Center Compositions and methods of treating fungal and bacterial pathogens
AU2014227748B2 (en) 2013-03-15 2019-02-14 Los Angeles Biomedical Research Institute At Harbor-Ucla Medical Center Compositions and methods for treating fungal and bacterial pathogens
WO2014144024A1 (en) 2013-03-15 2014-09-18 Los Angeles Biomedical Research Institute At Harbor-Ucla Medical Center Hyr1 compositions and methods for treating fungal and bacterial pathogens
GB201503812D0 (en) 2015-03-06 2015-04-22 Univ Aberdeen Antibody molecules and uses thereof
WO2017155949A1 (en) 2016-03-09 2017-09-14 Los Angeles Biomedical Research Institute At Harbor-Ucla Medical Center Methods and kits for use in preventing and treating vulvovaginal candidiasis

Patent Citations (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20020102262A1 (en) * 1996-05-16 2002-08-01 Magnus Hook Collagen binding protein compositions and methods of use
US7666438B1 (en) * 1998-08-31 2010-02-23 Inhibitex, Inc. Multicomponent vaccines
US20100150956A1 (en) * 1998-08-31 2010-06-17 Patti Joseph M Multicomponent vaccines
US20030124134A1 (en) * 1999-11-19 2003-07-03 Harbor-Ucla Research And Education Institute Pharmaceutical compositions and methods to vaccinate against candidiasis
US20060083750A1 (en) * 1999-11-19 2006-04-20 Los Angeles Biomedical Research Institute At Harbor-Ucla Medical Center Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis
US8541008B2 (en) * 1999-11-19 2013-09-24 Los Angeles Biomedical Research Institute At Harbor-Ucla Medical Center Pharmaceutical compositions and methods to vaccinate against candidiasis
US20040175731A1 (en) * 2002-11-12 2004-09-09 The Brigham And Women's Hospital, Inc. Methods and products for treating staphylococcal infections
US20050287146A1 (en) * 2003-12-19 2005-12-29 Joseph Patti Method of inhibiting Candida-related infections using donor selected or donor stimulated immunoglobulin compositions

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
Jones et al. Cell Regulation 2: 1001-1009, 1991 *
Liu et al. 44th ICAAC Abstracts, Washington D.C., page 425, M-1144, October 30 - 02 November 2004. *
Rudinger et al. In: Peptide Hormones. (Ed) JA Parsons, University Park Press, pages 1-7, 1976 *
Schmidt et al. Vaccine 30: 7594-7600, 14 December 2012 *
Skolnick et al. Trends in Biotechnology 18: 34-39, 2000 *
von Eiff et al. Diagn. Microbiol. Infect. Dis. 58: 297-302, 2007 *
Wisplinghoff et al. Clin. Infect. Dis. 39: 309-317, 2004 *
Zhao et al. Microbiology 152: 2287-2299, 2006 *

Cited By (7)

* 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
WO2007081896A3 (en) * 2006-01-06 2007-11-22 Los Angeles Biomed Res Inst Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis and other infectious agents
US10653757B2 (en) 2011-07-22 2020-05-19 Novadigm Therapeutics, Inc. Methods and compositions for vaccinating against Staphylococcus aureus
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
CN112203684A (zh) * 2018-04-10 2021-01-08 海港医学中心洛杉矶生物医学研究所 耳念珠菌感染的治疗方法
EP3552620A1 (de) * 2018-04-11 2019-10-16 Los Angeles Biomedical Research Institute at Harbor-UCLA Medical Center Verfahren zur behandlung von infektionen mit candida auris

Also Published As

Publication number Publication date
JP5775044B2 (ja) 2015-09-09
EP2532361A1 (de) 2012-12-12
WO2007081896A2 (en) 2007-07-19
JP2012255031A (ja) 2012-12-27
EP2532360B1 (de) 2016-09-28
WO2007081896A3 (en) 2007-11-22
JP2014148547A (ja) 2014-08-21
US20120014995A1 (en) 2012-01-19
AU2007205065A1 (en) 2007-07-19
ES2608354T3 (es) 2017-04-10
EP2428800B1 (de) 2016-09-28
US10300120B2 (en) 2019-05-28
AU2007205065B2 (en) 2013-10-31
ES2391580T3 (es) 2012-11-28
US20090297562A1 (en) 2009-12-03
DK1982190T3 (da) 2012-10-22
EP1982190B1 (de) 2012-07-18
CA2948424A1 (en) 2007-07-19
CA2636277C (en) 2017-06-13
PT1982190E (pt) 2012-10-22
US20140037689A1 (en) 2014-02-06
CA2636277A1 (en) 2007-07-19
EP1982190A2 (de) 2008-10-22
CY1113693T1 (el) 2016-06-22
EP1982190A4 (de) 2009-12-23
EP2532360A1 (de) 2012-12-12
EP2428800A1 (de) 2012-03-14
US20160220648A1 (en) 2016-08-04
JP5692963B2 (ja) 2015-04-01
ES2608309T3 (es) 2017-04-07
JP2009524601A (ja) 2009-07-02

Similar Documents

Publication Publication Date Title
US10300120B2 (en) Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis and other infectious agents
US9364539B2 (en) Pharmaceutical compositions and methods to vaccinate against candidiasis
Wang et al. Vaccines in the treatment of invasive candidiasis
Sui et al. The vaccines and antibodies associated with Als3p for treatment of Candida albicans infections
AU2006244401B2 (en) Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis and other infectious agents
Mochon et al. Prospects of vaccines for medically important fungi Is a vaccine needed against Candida albicans?
Magliani et al. Protective antifungal yeast killer toxin-like antibodies
US7067138B1 (en) Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis
AU2012216520A1 (en) Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis and other infectious agents
AU2013203750A1 (en) Pharmaceutical compositions and methods to vaccinate against disseminated candidiasis and other infectious agents
Mochon et al. Prospects of vaccines for medically important fungi

Legal Events

Date Code Title Description
AS Assignment

Owner name: LOS ANGELES BIOMEDICAL RESEARCH INSTITUTE AT HARBO

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:EDWARDS, JOHN E., JR.;IBRAHIM, ASHRAF;SPELLBERG, BRAD;AND OTHERS;REEL/FRAME:017595/0227;SIGNING DATES FROM 20060217 TO 20060221

STCB Information on status: application discontinuation

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