US20070172503A1 - Compositions and Methods to Elicit Immune Responses Against Pathogenic Organisms Using Yeast Based Vaccines - Google Patents

Compositions and Methods to Elicit Immune Responses Against Pathogenic Organisms Using Yeast Based Vaccines Download PDF

Info

Publication number
US20070172503A1
US20070172503A1 US11/610,457 US61045706A US2007172503A1 US 20070172503 A1 US20070172503 A1 US 20070172503A1 US 61045706 A US61045706 A US 61045706A US 2007172503 A1 US2007172503 A1 US 2007172503A1
Authority
US
United States
Prior art keywords
spp
fungal
cells
composition
subject
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
US11/610,457
Inventor
Claude Selitrennikoff
Tamara Miller
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.)
GlobeImmune Inc
Original Assignee
Mycologics Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Mycologics Inc filed Critical Mycologics Inc
Priority to US11/610,457 priority Critical patent/US20070172503A1/en
Assigned to MYCOLOGICS, INC. reassignment MYCOLOGICS, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MILLER, TAMARA K., SELITRENNIKOFF, CLAUDE P.
Publication of US20070172503A1 publication Critical patent/US20070172503A1/en
Assigned to GLOBEIMMUNE, INC. reassignment GLOBEIMMUNE, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MYCOLOGICS, INC.
Priority to US12/570,947 priority patent/US8460919B2/en
Priority to US13/800,094 priority patent/US9333246B2/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
    • 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
    • A61K2039/52Bacterial cells; Fungal cells; Protozoal cells
    • A61K2039/521Bacterial cells; Fungal cells; Protozoal cells inactivated (killed)
    • 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
    • A61K2039/52Bacterial cells; Fungal cells; Protozoal cells
    • A61K2039/523Bacterial cells; Fungal cells; Protozoal cells expressing foreign proteins

Definitions

  • the present invention provides for methods, compositions and kits for reducing a fungal infection and/or inducing a protective or therapeutic response against a pathogenic or non-pathogenic organism.
  • Coccidioidomycosis also known as San Joaquin Valley Fever
  • Coccidioides immitis that is endemic in portions of Southern Arizona, central California, southern New Mexico and west Texas. At least 100,000 new cases are reported each year.
  • the migration of not only permanent residents, but also agricultural workers to these areas increases exposure to C. immitis spores that lie dormant in the soil, and as the soil is disturbed, the spores become airborne and are inhaled. Once in the lungs, the arthroconidia transform into spherules.
  • Cryptococcus neoformans is an encapsulated pathogenic yeast that causes pulmonary infections and meningoencephalitis in humans and other animals.
  • Cryptococcus neoformans is an encapsulated pathogenic yeast that causes pulmonary infections and meningoencephalitis in humans and other animals.
  • Loss of CD4 + T cells predisposes patients to progressive infection with C. neoformans —this emphasizes the role of cell-mediated immunity in host resistance.
  • C. neoformans is a basidiomycetous fungus that is generally isolated as a haploid yeast, although diploids have been identified in nature. There are two mating types that can undergo recognition and fusion, and form a mycelium. A structure called a basidium is made and spores are produced from the surface of the basidium. Under specific conditions, haploid cells can undergo sporulation. Recently, a stable diploid was shown to grow as a yeast at 37° C. and formed hyphae and produced spores at 24° C. C. neoformans has been the subject of many studies, the var. grubii strain H99, as the source of the candidate immunogens. Var.
  • grubii are serotype A strains, have a worldwide distribution, and are the most common variety to cause disease in the United States.
  • Strain H99 has several advantages. First, molecular genetic analysis, including gene deletion and allelic replacement experiments, are well established in H99. Second, the genome sequence of H99 has been determined. The random shotgun and assembly phases of the genome are complete and the assembled genome has been released. Third, a congenic mating partner for H99 has recently been developed. Lastly, several reproducible animal models have been well developed to assess virulence for this strain.
  • Cryptococcosis is a disease that is acquired by inhalation of the organism from the soil or avian droppings into the lungs. Both immunocompetent and immunocompromised patients can be infected with the organism. In immunocompetent patients, the disease is usually contained in a lung granuloma and induces an antibody response. In contrast, individuals whose cellular immunity has been compromised by viral infection, suppression due to tissue transplantation, or anti-neoplastic chemotherapy are particularly susceptible to disseminated disease, followed by often-fatal meningoencephalitis. For example, an estimated 7-10% of AIDS patients acquire cryptococcosis during the course of their HIV disease.
  • Cryptococcal pneumonia is the next most frequent manifestation of cryptococcosis in immunocompromised patients. It occurs as a primary infection in approximately 4% of cases and is associated with general symptoms of pneumonia such as fever, cough, pleuritic chest pain, and/or dyspnea.
  • Aspergillus fumigatus is a ubiquitous spore-bearing fungus that causes multiple diseases in humans. These include allergic asthma, aspergillomas, and invasive pulmonary disease of hosts with predisposing underlying conditions.
  • 10190 aspergillosis-related hospitalizations In the United States in 1996, there were an estimated 10,190 aspergillosis-related hospitalizations; these resulted in 1,970 deaths, 176,300 hospital days, and $633 million in costs. The average hospitalization lasted 17.3 days and cost ⁇ $62,000.
  • aspergillosis-related hospitalizations account for a small percentage of hospitalizations in the United States, patients hospitalized with the condition have lengthy hospital stays and high mortality rates. The high mortality rates (in some instances over 90%) are due, in part, to the lack of rapid and sensitive diagnostic tests (all too often the diagnosis is made post mortem), as well as the lack of effective anti-fungal therapeutics.
  • A. fumigatus is the most frequently found fungus in airborne spore surveys. The organism grows in a variety of environments including air ducts, houseplant soil, compost piles, and at a wide range of temperatures, from 12° C. to 55° C. Of the genus Aspergillus, A. fumigatus is the most common pathogen of man.
  • Embodiments of the present invention concern methods and compositions for reducing the onset of, preventing or as treatment for a fungal infection.
  • a composition derived at least in part from non-viable fungal cells can be used to vaccinate a subject against or treat for a fungal infection.
  • fungal cells were heated at a temperature of about 56° C. for about one hour. In other embodiments, fungal cells were heated at temperatures ranging from about 50° C. to about 70° C. for from about 30 minutes to about 2 hours. Time and temperature may vary depending on the fungal organism, as well as, other factors such as the type of vaccine or therapeutic desired.
  • non-viable fungal cell compositions including fungal cells that were heated at a temperature of about 56° C. for about one hour, or a derivative thereof.
  • Other particular embodiments concern vaccines, wherein the vaccines include a composition derived from non-viable fungal cells and wherein fungal cells were heated at a temperature of about 56° C. for about one hour.
  • the fungal cells can be transgenic, non-transgenic or a combination thereof.
  • the vaccine compositions contemplated herein are capable of inducing an immune response against viable and non-viable fungal cells when administered to a subject.
  • Fungal cells can include, but are not limited to, Saccharomyces spp., Aspergillus spp.
  • compositions can include fungal cells transformed with an oligonucleotide, a peptide, a protein, a chimeric molecule or combination thereof.
  • transgenic fungal cells can contain one or more virulence factor.
  • a derivative of a non-viable fungal composition can be an active portion of non-viable fungal cells or a fraction thereof that is capable of inducing an immune response when introduced to a subject.
  • the derivative can be used a prophylactic against, prevention of and/or treatment for a fungal infection.
  • the fungal cells comprise transgenic, non-transgenic or a combination thereof.
  • the fungal cells can include, but are not limited to, Saccharomyces spp., Aspergillus spp., Cryptococcus spp., Coccidioides spp., Neurospora spp., Histoplasma spp., Blastomyces spp., and a combination thereof.
  • Saccharomyces spp. cells can be spores, vegetative cells, germlings, or a combination thereof.
  • the Neurospora spp. cells can be vegetative hyphae, aerial hyphae, macroconidia, germinating macroconidia, microconidia, germinating microconidia, ascospores, germinating ascospores, or a combination thereof.
  • the Aspergllus spp. cells can be vegetative hyphae, conidia, or germinating conidia, or combinations thereof.
  • Coccidioides spp. cells can be arthoconidia, germinating arthroconidia, hyphae, spherules, germinating spherules, endospores, germinating endospores, or a combination thereof.
  • Cryptococcus spp. cells can be yeast cells.
  • Histoplasma spp. cells can be yeast cells, vegetative hyphae, microcondia, germinating microconidia, macroconidia, germinating macroconidia, or combinations thereof.
  • Blastomyces spp. cells can be yeasts, hyphae, blastoconidia, germinating blastoconidia or combinations thereof.
  • the fungal cells may further include a tag.
  • transgenic fungal cells can contain at least one oligonucleotide.
  • the oligonucleotide can encode a peptide or a protein.
  • the transgenic fungal cells contain at least one virulence factor.
  • Embodiments of the present invention concern administering to a subject a vaccine that includes, at least in part, non-viable fungal cells.
  • vaccines of the present invention induce an immune response in the subject directed against one or more fungal organisms.
  • the subject is a mammal, bird or reptile.
  • the subject is a human.
  • the vaccine can be administered in a single dose, a few doses or multiple doses to reduce the onset of a fungal infection, preventing and/or treating a fungal infection.
  • the vaccination can occur yearly, monthly, or weekly or even more frequent depending on need.
  • a subject having a fungal infection can be treated prophylactically to reduce progression of the fungal infection and or eliminate the infection.
  • the subject is a human predisposed to a fungal infection or a fungal organism (e.g., predisposed can mean at risk of exposure or having been exposed to a fungal infection or fungal organism).
  • the subject is an immunocompromised subject.
  • the immunocompromised subject is a subject having a viral infection, tissue transplantation, anti-neoplastic chemotherapy or combination thereof.
  • kits for treating and/or preventing a fungal infection can include non-viable fungal cells.
  • FIG. 1 represents exemplary growth curves of yeast containing a gene encoding an antigen grown in the presence of various concentrations of CuSO 4 .
  • FIG. 2 illustrates an exemplary growth curve of yeast cells containing and expressing an antigen protein.
  • FIG. 3 illustrates an exemplary schematic of a method to test a vaccine disclosed herein.
  • FIG. 4 illustrates exemplary survival curves for mice infected in vitro with A. fumigatus.
  • FIG. 5 represents an exemplary electrophoresis gel of separated nucleic acid sequences from PCR reactions.
  • FIG. 6 illustrates an exemplary schematic of a method to test a vaccine disclosed herein.
  • FIG. 7 illustrates exemplary survival curves for mice infected in vitro with C immitis.
  • FIG. 8 represents an exemplary Western blot of separated lysed yeast cells containing and expressing genes for hemolysin, aspf2 or antigen-2/PRA.
  • FIG. 9 illustrates an exemplary schematic of a method to test a vaccine disclosed herein.
  • FIG. 10 represents an exemplary electrophoresis gel of separated molecules of yeast lysates containing and expressing the gene encoding Chitin deacetylase and Laccase.
  • FIG. 11 illustrates an exemplary survival curves for mice infected in vitro. with Cryptococcosis neoformans
  • FIG. 12 illustrates an exemplary scattergram of protective efficacy of Aspergillus hemolysin antigen expressed in Saccharomyces spp. against systemic aspergillosis in mice.
  • Embodiments of the present invention provide for compositions and methods for reducing the onset of, preventing and/or treating a fungal infection.
  • methods and compositions when administered to a subject are capable of inducing a protective immune response against a target.
  • these embodiments include a fungal pathogen.
  • a fungal pathogenic organism includes, but is not limited to, Aspergillus spp., Cryptococcus spp., and Coccidioides spp.
  • pathogenic fungus may include, but are not limited to, Aspergillus fumigatus, A. flavus, A. niger, A. terreus, A.
  • the immune response may be induced by injection of a fungal-based vaccine.
  • a fungal-based vaccine may comprise, but are not limited to, suspensions, solutions, extracts, homogenates, precipitates and/or other processed or unprocessed forms of Saccharomyces cerevisiae .
  • the vaccines may comprise yeast transformed with genetic markers, such as a c-myc tag sequence.
  • the vaccines may include a non-transgenic fungus or fungi.
  • the vaccines may include non-transgenic or transgenic yeast.
  • Embodiments of the present invention provide for combination treatments of a subject in need of a vaccine against an organism such as a pathogenic fungal organism.
  • any vaccine treatment detailed herein may be combined with, but is not limited to, treatments for cryptococcosis, aspergillosis, or coccidoidomycosis such as amphotericin B or the azoles, fluconazole or itraconazole, 5-flucytosine, voriconazole, and fluconazole.
  • other treatments may include other antifungal treatments, such as any of the echinocandin class antifungals or any other antifungal agent.
  • any of the vaccines disclosed may be administered to a subject having or suspected of developing a condition due to exposure or potential exposure to a fungal pathogen.
  • a subject may include a human.
  • a subject may include a human, other mammals, birds and reptiles.
  • the subject can include a domesticated animal. Any of the methods disclosed herein may be used in combination with other antifungal agents or therapies to treat a subject in order to achieve the desired results.
  • a subject can be vaccinated and/or treated using a composition against a fungal pathogen disclosed herein before infection, during infection, after infection or combination thereof.
  • a composition disclosed herein may be used to vaccinate a subject against a particular fungal organism to reduce the risk of a fungal infection in the subject.
  • a composition disclosed herein may be used to vaccinate a subject against one or more fungal organisms to reduce the risk of a fungal infection in the subject.
  • a vaccine containing non-viable fungal cells of one fungal type when administered to a subject may reduce the onset or progression of fungal infection of one or more additional fungal types in the subject.
  • a vaccine or treatment of use in a subject can contain non-viable fungal cells where the fungal cells are transgenic, non-transgenic or a combination thereof.
  • Transgenic cells of the present invention may include an oligonucleotide.
  • the oligonucleotide can encode one or more peptides or proteins.
  • the fungal cells expressing each fungal protein may be engineered so that it is not secreted but rather is retained intracellularly. This likely obviates the need for large amounts of recombinant fungal antigen protein to be produced and purified, ultimately under cGMP conditions.
  • fungal cells are easily transformed and manipulated.
  • putative antigens can be tested rather inexpensively and ineffective antigens can be eliminated quickly.
  • whole, heat-killed yeast as a vaccine delivery vehicle is novel and presents antigens directly to dendritic cells to induce the immune system.
  • several individual antigen-expressing yeast strains can be mixed to form a vaccine, a possibility not readily available to current vaccine projects.
  • the yeast-based vaccine technology has each of the characteristics of a successful vaccine: generates a productive immune response; not neutralized; capable of generating a response to multiple epitopes; and stable and easy to manufacture.
  • a beneficial therapeutic or a vaccine might consist of a vector with no pathogenic potential that can deliver several antigens to antigen-presenting cells (APCs).
  • APCs antigen-presenting cells
  • the use of recombinant yeast proves to be an ideal vector for vaccine development.
  • a vaccine contemplated herein could consist of a vector with no pathogenic potential that can deliver several antigens to APCs.
  • the use of recombinant yeast proves to be an ideal vector for vaccine development.
  • the yeast Saccharomyces cerevisiae is avidly taken up by professional APCs, such as neutrophils, macrophages and dendritic cells.
  • yeast-based vaccine formulations do not require extensive purification to remove potentially toxic contaminants.
  • the organism S. cerevisiae is designated by the FDA as GRAS (Generally Regarded As Safe, FDA Proposed Rule 62FR18938, Apr. 17, 1997).
  • the heterologous proteins expressed in recombinant yeast serve as antigens that elicit CD8+ CTL-mediated immune responses in vitro and in vivo.
  • the yeast formulation was successful at protecting vaccinated animals from tumor growth (refer to FIGS. 4, 7 , 11 and 12 as examples of animal model testing).
  • the immunogens may be of a single formulation or a combination formulation.
  • immunocompetent animals or immuncompromised animals may be used in order to assess what formulation may be used to treat a subject.
  • the test can include vaccination and then a challenge of the vaccination.
  • the appropriate dose of any disclosed therapeutic or vaccine may be determined by the methods disclosed herein including, but not limited to optimum formulation, the appropriate number of treatments or vaccinations and the optimum time for treatment or vaccinations in order to instill maximum treatment or protection against a given organism.
  • methods and compositions disclosed herein are directed toward making and using an anti-fungal vaccine or anti-fungal treatment.
  • Fungal organisms targeted in the present invention include, but are not limited to, Saccharomyces spp., Aspergillus spp. Cryptococcus spp., Coccidioides spp., Neurospora spp., Histoplasma spp., Blastomyces spp., and a combination thereof.
  • methods, compositions and treatments for a fungal organism infection can concern treatment for or prevention of an Aspergillus associated-condition.
  • the most common species of Aspergillus causing invasive disease include A. fumigatus, A. flavus, A. niger, A. terreus and A. nidulans.
  • A. fumigatus is the most frequently found fungus in airborne spore surveys.
  • A. fumigatus pneumonia and systemic aspergillosis occur primarily in patients who have immunosuppression or T-cell or phagocytic impairment. The immunodeficiency detected in these patients may be congenital, acquired or iatrogenic.
  • a subject having or suspected of developing a disease derived from an Aspergillus species can be administered a composition disclosed herein.
  • a subject having or suspected of developing a disease derived from an Aspergillus species can be administered a composition including heated, transformed or non-transformed fungi for example, heated Aspergillus fungi.
  • A. fumigatus Three main types of diseases are attributed to A. fumigatus ; these include asthma, aspergillomas and invasive aspergillosis.
  • the most serious disease involves invasion of hosts with predisposing underlying conditions. Patients undergoing organ transplants, bone marrow transplants, leukemics, or cancer chemotherapy are particularly at risk for invasive aspergillosis.
  • Aspergillosis is often diagnosed when there is an unexplained pulmonary infiltrate, a patient is unresponsive to antibacterials and/or there is a fever of unknown origin.
  • the prognosis for patients with invasive disease is high (mortality rates >50%) due to the lack of a rapid diagnostic test confirming A. fumigatus infection and the lack of safe and effective antifungal drugs.
  • Coccidioidomycosis also known as San Joaquin Valley Fever, is a fungal disease caused by Coccidioides immitis that is endemic in portions of Southern Arizona, central California, southern New Mexico and west Texas. At least 100,000 new cases are reported each year.
  • the migration of not only permanent residents, but also agricultural workers to these areas increases exposure to Coccidioides spp. spores that lie dormant in the soil, and as the soil is disturbed, the spores become airborne and are inhaled. Once in the lungs, the arthroconidia transform into spherules. An acute respiratory infection occurs between seven days to three weeks after exposure and often resolves rapidly.
  • PMNs Polymorphonuclear leukocytes
  • Polymorphonuclear leukocytes are only able to partially inhibit growth of the pathogen, and are unable to kill the organism. PMNs are slightly more effective in inhibiting growth of arthroconidia than mature spherules. Since mature fungal spherules are typically 40-120 ⁇ m in diameter, a single PMN is unable to phagocytose the fungal cell. Endospores, on the other hand, are more sensitive to growth inhibition by these host cells. Most investigators of cellular immune response to Coccidioides spp. believe that macrophages are the pivotal effector cells in coccidioidomycosis.
  • methods, compositions and treatments for a fungal organism infection can concern treatment for or prevention of a Cryptococcosis -associated condition.
  • C. neoformans can be an intracellular as well as an extracellular pathogen. It is easily found extracellularly in cerebral spinal fluid and lung tissue. It can also be localized inside macrophages and neutrophils where it has been shown to replicate.
  • Antibody and cell-mediated responses can provide important protection even against an intracellular pathogen.
  • the mammalian host has several defenses against C. neoformans that include components from innate, humoral, and cell-mediated immunity. Subjects are continually exposed to C. neoformans from the environment, a powerful innate immunity represents part of the protection afforded the normal host. This appears to be inherent in normal macrophage, monocyte, and neutrophil function.
  • Vaccination against cryptococcosis presents the innovative idea of not only aiming to protect an immunocompromised population, but also possibly expecting an immune response in that population.
  • a vaccine against cryptococcosis is contemplated for example, to treat a non-immunosuppressed or immunosuppressed subject.
  • immunosuppressed patients such as HIV patients are contemplated.
  • an HIV patient may be vaccinated immediately after diagnosis, but before their CD4 + T cell population decreases.
  • any of the vaccines detailed herein may be directed towards other fungal organisms, for example, Coccidioides and Aspergillus . It is contemplated herein that any fungal pathogen disclosed may have one or more virulence factors which may include surface proteins, cell-wall carbohydrates, secreted factors, anchored surface molecules, modes of action to invade a host, etc. Any virulence factor associated with a fungal pathogen may be used herein as a potential target to transform yeast or other fungi of the present invention to generate a vaccine against that fungal pathogen.
  • the fungal cells comprise transgenic cells, non-transgenic cells or a combination thereof.
  • the fungal cells can include, but are not limited to, Saccharomyces spp., Aspergillus spp., Cryptococcus spp., Coccidioides spp., Neurospora spp., Histoplasma spp., Blastomyces spp., and a combination thereof.
  • Saccharomyces spp. cells can be spores, vegetative cells, germlings, or a combination thereof.
  • the Neurospora spp. cells can be vegetative hyphae, aerial hyphae, macroconidia, germinating macroconidia, microconidia, germinating microconidia, ascospores, germinating ascospores, or a combination thereof.
  • a yeast vaccine formulation directly accesses DCs, the critical immune responsive cells of the body.
  • DCs the critical immune responsive cells of the body.
  • immature DCs must be activated to mature, as evidenced by the up-regulation of MHC and co-stimulatory molecules.
  • Mature DCs are then capable of prolonged antigen presentation and the production of cytokines, such as IL-12, that are critical for the induction of cellular immune responses.
  • Uptake of yeast by DCs increased surface expression of the co-stimulatory molecules CD40, CD80 and CD86, MHC class II, and the adhesion molecule ICAM, to levels comparable to that induced by exposure to bacterial lipopolysaccharide (LPS), a potent DC maturation factor.
  • LPS bacterial lipopolysaccharide
  • yeast vaccine technology can be formulated with a variety of antigens, including 5 from HIV, 3 from Hepatitis B, 1 from Hepatitis C, and 2 from lung cancer cells and, in each case, the yeast-expressed antigen effectively stimulated protective cell-mediated immunity.
  • yeast as a vaccine vehicle provides an adjuvant effect, such that dendritic cells are triggered to directly take up yeast, causing the DCs to mature and to present the yeast-associated antigens to the host immune system.
  • heat-killed yeast cells expressing putative antigens are as immunogenic and protective as live yeast cells expressing putative antigens.
  • yeast cells expressing antigens elicited Th1 responses.
  • recombinant-yeast vaccine formulations may elicit systemic, antigen-specific, CD4 and CD8-based protective immunity.
  • DCs have been shown to internalize yeast and present recombinant antigens to naive class I and class II MHC-restricted T cells. It has been shown that yeast exhibit adjuvant activity for DC maturation and immune signalling. Yeast might be providing an adjuvant effect to promote immune responses to the yeast-associated antigens.
  • a CD8-dependent protective immunity has been elicited in vaccinated mice.
  • EL-4 CD4 + T lymphoma cells (H-2 b ) transfected with cDNA encoding chicken ovalbumin (E.G7-OVA) and ovalbumin-expressing melanoma (B16-OVA) mouse tumor models have been employed to test vaccine candidates that induce protective CTL response.
  • Protective immunity was CD8-dependent, since OVAX-vaccinated animals were not protected from tumor challenge in CD8 ⁇ / ⁇ knockout mice.
  • kits for the methods described herein in one embodiment, a fungal organism treatment or (such as a pathogenic or non-pathogenic fungus) prevention kit is contemplated.
  • a kit for prophylactic treatment of a subject having or suspected of developing a fungal infection is contemplated.
  • a kit for prevention or treatment of a subject having or suspected of developing a fungal-induced infection is contemplated.
  • the kit may be used to treat or vaccinate a subject for or against a fungal infection.
  • kits may include a composition including at least a portion of a non-viable fungus within a tube, a vial or other suitable vessel.
  • the kit may include one or more dose(s) of the composition for administration to a subject having or exposed to a fungal organism infection by a healthcare provider.
  • the kit may be a portable kit for use at a specified location outside of a healthcare facility.
  • the container means of any of the kits will generally include at least one vial, test tube, flask, bottle, syringe or other container means, into which the testing agent, may be preferably and/or suitably aliquoted.
  • isolated nucleic acids may be used to modify or transform a fungal organism contemplated in the present invention.
  • the isolated nucleic acid may be derived from genomic DNA, RNA or complementary DNA (cDNA).
  • isolated nucleic acids such as chemically or enzymatically synthesized DNA, may be of use for generation of oligonucleotides for transformation of a fungal organism.
  • nucleic acid includes single-stranded and double-stranded molecules, as well as DNA, RNA, chemically modified nucleic acids and nucleic acid analogs. It is contemplated that a nucleic acid may be of 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98,
  • Isolated nucleic acids may be made by any method known in the art, for example using standard recombinant methods, synthetic techniques, or combinations thereof.
  • the nucleic acids may be cloned, amplified, or otherwise constructed.
  • Nucleic acids of use in the present invention may include oligonucleotides that include at least a portion of sequences from a virulence factor (e.g., including but not limited to, antigen-2/proline rich antigen, hemolysin, aspf2, dpp5, dpp4, chitin deacetylase, catalase), a disease-associated factor (e.g., tumor associated antigens, cytokines, viral-associated antigens, bacterial-associated antigens) or other peptides or proteins.
  • a multi-cloning site comprising one or more endonuclease restriction sites may be added.
  • a nucleic acid may be attached to a vector, adapter, or linker for cloning of a nucleic acid. Additional sequences may be added to such cloning and sequences to optimize their function, to aid in isolation of the nucleic acid, or to improve the introduction of the nucleic acid into a cell. Use of cloning vectors, expression vectors, adapters, and linkers is well known in the art.
  • Isolated nucleic acids may be obtained from fungal, bacterial, viral or other sources using any number of cloning methodologies known in the art.
  • oligonucleotide probes which selectively hybridize, under stringent conditions, to the nucleic acids are used to identify a sequence of interest.
  • Methods for construction of nucleic acid libraries are known and any such known methods may be used. (See, e.g., Current Protocols in Molecular Biology, Ausubel, et al., Eds., Greene Publishing and Wiley-Interscience, New York (1995); Sambrook, et al., Molecular Cloning: A Laboratory Manual, 2nd Ed., Cold Spring Harbor Laboratory Vols. 1-3 (1989); Methods in Enzymology, Vol. 152, Guide to Molecular Cloning Techniques, Berger and Kimmel, Eds., San Diego: Academic Press, Inc. (1987)).
  • Genomic DNA, RNA or cDNA may be screened for the presence of an identified genetic element of interest using a probe based upon one or more sequences.
  • Various degrees of stringency of hybridization may be employed in the assay. As the conditions for hybridization become more stringent, there must be a greater degree of complementarity between the probe and the target for duplex formation to occur.
  • the degree of stringency may be controlled by temperature, ionic strength, pH and/or the presence of a partially denaturing solvent such as formamide.
  • the stringency of hybridization is conveniently varied by changing the concentration of formamide within the range of 0% to 50%.
  • the degree of complementarity (sequence identity) required for detectable binding will vary in accordance with the stringency of the hybridization medium and/or wash medium.
  • High stringency conditions for nucleic acid hybridization are well known in the art.
  • conditions may comprise low salt and/or high temperature conditions, such as provided by about 0.02 M to about 0.15 M NaCl at temperatures of about 50° C. to about 70° C.
  • Other exemplary conditions are disclosed in the following Examples. It is understood that the temperature and ionic strength of a desired stringency are determined in part by the length of the particular nucleic acid(s), the length and nucleotide content of the target sequence(s), the charge composition of the nucleic acid(s), and to the presence or concentration of formamide, tetramethylammonium chloride or other solvent(s) in a hybridization mixture.
  • Nucleic acids may be completely complementary to a target sequence or may exhibit one or more mismatches.
  • Nucleic acids of interest may also be amplified using a variety of known amplification techniques. For instance, polymerase chain reaction (PCR) technology may be used to amplify target sequences directly from RNA or cDNA. PCR and other in vitro amplification methods may also be useful, for example, to clone nucleic acid sequences, to make nucleic acids to use as probes for detecting the presence of a target nucleic acid in samples, for nucleic acid sequencing, or for other purposes. Examples of techniques of use for nucleic acid amplification are found in Berger, Sambrook, and Ausubel, as well as Mullis et al., U.S. Pat. No.
  • Isolated nucleic acids may be prepared by direct chemical synthesis by methods such as the phosphotriester method of Narang et al., Meth. Enzymol. 68:90-99 (1979); the phosphodiester method of Brown et al., Meth. Enzymol. 68:109-151 (1979); the diethylphosphoramidite method of Beaucage et al., Tetra. Lett. 22:859-1862 (1981); the solid phase phosphoramidite triester method of Beaucage and Caruthers, Tetra. Letts.
  • Chemical synthesis generally produces a single stranded oligonucleotide. This may be converted into double stranded DNA by hybridization with a complementary sequence, or by polymerization with a DNA polymerase using the single strand as a template. While chemical synthesis of DNA is best employed for sequences of about 100 bases or less, longer sequences may be obtained by the ligation of shorter sequences.
  • cross-linking agents may be used to bind, label, detect, and/or cleave nucleic acids.
  • Vlassov, V. V., et al., Nucleic Acids Res (1986) 14:4065-4076 disclose covalent bonding of a single-stranded DNA fragment with alkylating derivatives of nucleotides complementary to target sequences.
  • a report of similar work by the same group is that by Knorre, D. G., et al., Biochimie (1985) 67:785-789.
  • tag nucleic acids may be used to trace or identify a particular nucleic acid sequence contemplated herein.
  • labels such as fluorophores, chromophores, radio-isotopes, enzymatic tags, antibodies, chemiluminescent, electroluminescent, affinity labels, etc.
  • enzymatic tags include urease, alkaline phosphatase or peroxidase.
  • Colorimetric indicator substrates can be employed with such enzymes to provide a detection means visible to the human eye or spectrophotometrically.
  • a well-known example of a chemiluminescent label is the luciferin/luciferase combination.
  • the label may be a fluorescent, phosphorescent or chemiluminescent label.
  • exemplary photodetectable labels may be selected from the group consisting of Alexa 350, Alexa 430, AMCA, aminoacridine, BODIPY 630/650, BODIPY 650/665, BODIPY-FL, BODIPY-R6G, BODIPY-TMR, BODIPY-TRX, 5-carboxy-4′,5′-dichloro-2′,7′-dimethoxy fluorescein, 5-carboxy-2′,4′,5′,7′-tetrachlorofluorescein, 5-carboxyfluorescein, 5-carboxyrhodamine, 6-carboxyrhodamine, 6-carboxytetramethyl amino, Cascade Blue, Cy2, Cy3, Cy5,6-FAM, dansyl chloride, Fluorescein, HEX, 6-JOE, NBD (7-nitrobenz-2-oxa-1,3-
  • a vaccine will be administered to a subject for vaccination against and/or treatment of a fungal infection. It is also contemplated herein that any of the vaccines disclosed may be administered to a subject having or suspected of developing a condition due to exposure or potential exposure to a fungal pathogen.
  • a subject may include a human. In other embodiments a subject may include other mammals, birds, and reptiles. Any of the methods disclosed herein may be used in combination with other anti-fungal agents or therapies to treat a subject in order to achieve the desired results.
  • a subject can be vaccinated and/or treated using a composition against a fungal pathogen disclosed herein before infection, during infection, after infection or combination therefore of.
  • a composition disclosed herein may be used to vaccinate a subject against a particular fungal organism to reduce the risk of a fungal infection in the subject.
  • a composition disclosed herein may be used to vaccinate a subject against one or more fungal organism to reduce the risk of a fungal infection in the subject.
  • a vaccine containing non-viable fungal cells of one fungal type when administered to a subject may reduce the onset or progression of fungal infection of another fungal type in the subject.
  • a dose may consist of around 1 ⁇ 10 7 yeast cells to 20 ⁇ 10 7 yeast cells per dose. And may be given before infection, during infection, after infection or a combination therefore of The dose may be given daily, every other day, biweekly, weekly, seasonally or yearly until a desired effect is achieved.
  • a composition of the present invention may be administered to a subject in an appropriate carrier or diluent or pharmaceutically acceptable carrier, co-administered with enzyme inhibitors or in an appropriate carrier such as liposomes.
  • pharmaceutically acceptable carrier as used herein is intended to include diluents such as saline and aqueous buffer solutions.
  • Enzyme inhibitors include pancreatic trypsin inhibitor, diisopropylfluorophosphate (DEP) and trasylol.
  • Liposomes include water-in-oil-in-water emulsions as well as conventional liposomes.
  • the active agent may also be administered parenterally or intraperitoneally.
  • Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations may contain a preservative to prevent the growth of microorganisms.
  • compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion.
  • the composition must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria.
  • the pharmaceutically acceptable carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyetheylene glycol, and the like), and suitable mixtures thereof.
  • the proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • Prevention of microorganisms can be achieved by various antibacterial and antifungal agents (i.e., parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like).
  • isotonic agents for example, sugars, polyalcohols such as manitol, sorbitol, sodium chloride in the composition.
  • a compound such as aluminum monostearate and gelatin can be included to prolong absorption of the injectable compositions.
  • Sterile injectable solutions can be prepared by incorporating active compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization.
  • dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a dispersion medium and other required ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum drying and freeze-drying which yields a powder of the active ingredient (i.e., a chemical agent, antibody, etc.) plus any additional desired ingredient from a previously sterile-filtered solution thereof
  • the composition may be orally administered (or otherwise indicated), for example, with an inert diluent or an assimilable edible carrier. It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined quantity of compound such as non-viable fungal cells calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • the biological material can be extensively dialyzed to remove undesired small molecular weight molecules and/or lyophilized for more ready formulation into a desired vehicle, where appropriate.
  • the active compounds will then generally be formulated for parenteral administration (i.e., formulated for injection via the intravenous, intramuscular, sub-cutaneous, intralesional, or even intraperitoneal routes).
  • parenteral administration i.e., formulated for injection via the intravenous, intramuscular, sub-cutaneous, intralesional, or even intraperitoneal routes.
  • the preparation of an aqueous composition that contains an active component or ingredient will be known.
  • such compositions can be prepared as injectables, either as liquid solutions or suspensions; solid forms suitable for use in preparing solutions or suspensions upon the addition of a liquid prior to injection can also be prepared; and the preparations can also be emulsified.
  • the pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions; formulations including sesame oil, peanut oil or aqueous propylene glycol; and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions.
  • the form must be sterile and must be fluid. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
  • Solutions of the active compounds as free-base or pharmacologically acceptable salts can be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required.
  • dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above.
  • the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof.
  • the preparation of more, or highly, concentrated solutions for direct injection is also contemplated, where the use of DMSO as solvent is envisioned to result in extremely rapid penetration, delivering high concentrations of the active agents to a small area.
  • solutions Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective.
  • the formulations are easily administered in a variety of dosage forms, such as the type of injectable solutions described above, but drug release capsules and the like can also be employed.
  • aqueous solutions For parenteral administration in an aqueous solution, for example, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose.
  • aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration.
  • sterile aqueous media that can be employed will be known to those of skill in the art in light of the present disclosure.
  • other pharmaceutically acceptable forms include, e.g., liposomal formulations; time-release capsules; and any other form currently used.
  • nasal solutions or sprays, aerosols or inhalants may be used to deliver the compound of interest.
  • Nasal solutions are usually aqueous solutions designed to be administered to the nasal passages in drops or sprays.
  • Nasal solutions are prepared so that they are similar in many respects to nasal secretions.
  • the aqueous nasal solutions usually are isotonic and slightly buffered to maintain a pH of 5.5 to 6.5.
  • antimicrobial preservatives similar to those used in ophthalmic preparations, and appropriate drug stabilizers, if required, may be included in the formulation.
  • Various commercial nasal preparations are known and include, for example, antibiotics and antihistamines and are used for asthma prophylaxis.
  • Inhalation preparations may include solutions or dry powder formulations that are commonly used along with a propellant in the formulation of therapeutics used for the treatment of asthmatics.
  • Oral formulations include such normally employed excipients as, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate and the like. These compositions take the form of solutions, suspensions, tablets, pills, capsules, sustained release formulations or powders.
  • oral pharmaceutical compositions will comprise an inert diluent or assimilable edible carrier, or they may be enclosed in hard or soft shell gelatin capsule, or they may be compressed into tablets, or they may be incorporated directly with the food of the diet.
  • the active compounds may be incorporated with excipients and used in the form of ingestible tablets, buccal tables, troches, capsules, elixirs, suspensions, syrups, wafers, and the like.
  • Such compositions and preparations should contain at least 0.1% of active compound.
  • the percentage of the compositions and preparations may, of course, be varied and may conveniently be between about 2 to about 75% of the weight of the unit, or preferably between 25-60%.
  • the amount of active compounds in such therapeutically useful compositions is such that a suitable dosage will be obtained.
  • compositions may contain any of a variety of known aerosol propellants useful for endopulmonary and/or intranasal inhalation administration.
  • water may be present, with or without any of a variety of cosolvents, surfactants, stabilizers (e.g., antioxidants, chelating agents, inert gases and buffers).
  • Proteins to be tested Each protein from a fungal pathogen was chosen because of its identification as a virulence factor (e.g., strains lacking the protein were less virulent that the wild type), as an immunogen (infected animals generated antibodies to the candidate protein), and/or was found on the cell surface of conidia or hyphae. In addition, none of the proteins had cognates with significant ( ⁇ 35%) homology to mammals and to yeast.
  • the Open Reading Frame (ORF) of each gene was obtained by PCR using gene specific primers and in certain embodiments, a high quality cDNA library as template.
  • the ORF of candidate genes was obtained using plasmid DNA containing the gene of interest as a template for amplification by PCR.
  • Each PCR product was gel purified and ligated into the TA cloning vector, pCR2.1, using the pCR2.1 TOPO kit from InVitrogen (Carlsbad, Calif.). Resulting plasmid DNAs were used to transform competent E. coli cells and plasmid DNA isolated from midi-preps using methods standard to those of skill in the art.
  • the gene specific PCR primers encoded a c-myc epitope such that the c-myc epitope would be generated in the protein at the carboxy terminus.
  • each ORF was excised from each plasmid DNA using the restriction endonuclease EcoR1 and ligated into the EcoR1 site of pYEX-BX. Each resulting ligated plasmid was used to transform competent E. coli cells and plasmid DNA isolated from midi-preps. Each ORF DNA was sequenced to ensure that it is in frame with the copper-inducible promoter and and no PCR errors were present.
  • an empty vector control was generated.
  • an empty vector control was generated a pYEX-BX (Clonetech) vector containing the DNA encoding a c-myc epitope but lacking a fungal gene of interest.
  • a 51 base pair template for PCR was created that contained a methionine, the sequence that encodes the c-myc protein and two stop codons.
  • the PCR primers also included a 5′ BamH I site and a 3′ EcoR I restriction site that could be used to excise the tag from the pCR 2.1 vector and inserted into the pYEX-BX vector (Clonetech).
  • the DNA and amino acid sequences are shown in SEQ ID NOs:1 and 2: c-myc control sequence atg gaa cag aag ttg att tcc gaa gaa gac ctc gag Met Glu Gln Lys Leu Ile Ser Glu Glu Asp Leu Glu
  • This template was amplified by PCR under the following conditions: 25 ⁇ L reactions were performed in PCR Ready Bead tubes containing 500 ng of cDNA library, 2 ⁇ M primers, and 13 ⁇ L sterile water. Reactions were performed using a Perkin Elmer 2400 Thermocycler under the following conditions: an initial cycle at 94° C. for 5 min, 30 cycles at 94° C. for 1 min, 50° C. for 1 min, 72° C. for 1 min, and a final extension at 72° C. for 7 min.
  • the PCR reactions were separated by agarose gel electrophoresis using a 3% (w/v) agarose gel.
  • the 51 bp band was excised from the gel, purified and ligated into pCR 2.1 (Invitrogen).
  • the ligated vector was used to transform competent E. coli cells and vector DNA was subsequently isolated using standard techniques. DNA isolated from several isolates was used as template for PCR reactions to confirm the presence of the c-myc insert.
  • exemplary fungal cells that can be transformed to express vaccine proteins can include, but are not limited to, Saccharomyces spp., Aspergillus spp., Cryptococcus spp., Coccidioides spp., Neurospora spp., Histoplasma spp., Blastomyces spp., and a combination thereof.
  • Exemplary method of transformation of S. cerevisiae After isolating each of the genes of interest and forming each yeast expression vector (as described), DNA vectors to individually transform yeast cells were used.
  • the transformation protocol was as follows: 50 ⁇ L of YPD broth was inoculated with W303-1B Saccharomyces cerevisiae and the culture grown overnight at 30° C. with shaking. The desired OD600 for transformation was between 0.1-0.6 ( ⁇ 2 ⁇ 107 c/mL). Cells were harvested by centrifugation at 2000 ⁇ g for 10 minutes, the pellet was washed in 25 mL sterile water and cells were again centrifuged at 2000 ⁇ g for 10 minutes.
  • the pellet was resuspended in 1 mL 01M lithium acetate (LiAC), centrifuged at 1000 ⁇ g for 30 seconds and the supernatant removed. This pellet was again resuspended in 400 ⁇ L 0.1M LiAC and the cell suspension mixed until the cells were completely suspended. Fifty milliliters of this cell suspension was used for each transformation. The cells were centrifuged at 3000 ⁇ g for 30 seconds and the supernatant discarded. To the cells was added 240 ⁇ L 50% (w/v) PEG, 36 ⁇ L 1M LiAC, 25 ⁇ L denatured carrier DNA, and 50 ⁇ L of previously diluted water containing plasmid DNA (0.1-10 ⁇ g).
  • LiAC lithium acetate
  • Yeast Nitrogen Base medium e.g., Difco
  • yeast colony was obtained from YNB minus uracil plates and grown for overnight at 30° C. with shaking in 250 mL flasks containing 50 mL YNB minus uracil medium. Each culture was diluted to a final concentration of 0.2 (OD600) in 50 mL YNB minus uracil interim cultures. The cultures were grown at 30° C. until the OD600 had doubled (approximately 5-6 hours). The OD600 of the interim cultures was determined a final time before the cultures were diluted to a final concentration of 0.02 OD600 units in 50 mL of YNB minus uracil. These cultures were grown for 1 hour with shaking at 30° C.
  • Induced and uninduced cultures were harvested after overnight incubation by centrifugation at 550 ⁇ g for 10 minutes. Each cell pellet was washed with 25 mL ice cold water to remove any residual medium and the cells were harvested again by centrifugation. Each pellet was then resuspended in 1 mL of ice cold water and divided into two 1.5 mL eppendorf tubes for centrifugation. After the removal of the supernatant, one tube was frozen at ⁇ 80° C. for storage and the other was lysed for protein determination.
  • Cells were lysed in the following manner: 100 ⁇ L of glass beads were added followed by the addition of 200 ⁇ L 2 ⁇ sample lysis buffer (0.1M Tris, pH 6.8, 20% (v/v) glycerol, 4% (w/v) SDS, 0.5% (v/v) ⁇ -mercaptoethanol). Tubes were mixed by vortexing for 1 min to resuspend and lyse the cells and mixtures were boiled for 3 min to denature the proteins for SDS-PAGE gel electrophoresis. Debris was removed by centrifugation at 550 ⁇ g for 5 min. and the cleared cell lysates were transferred to new tubes.
  • sample lysis buffer 0.1M Tris, pH 6.8, 20% (v/v) glycerol, 4% (w/v) SDS, 0.5% (v/v) ⁇ -mercaptoethanol. Tubes were mixed by vortexing for 1 min to resuspend and lyse the cells and mixtures were boiled for 3 min to denature the
  • PVDF membranes were blocked in TBST (100 mM Tris, 1.5 M NaCl and 0.1% [v/v] Tween 20) containing 5% (w/v) powdered milk (Carnation) for 1 hour at room temperature. Blocked membranes were then incubated with primary anti-c-myc epitope antibody (1:1000 dilution) in TBST containing 1% milk overnight at 4o C. The next morning the membranes were washed five times for 15 minutes each in TBST before secondary antibody was added. Secondary antibody conjugated to horse radish peroxidase (HRP) was added in TBST containing 1% (w/v) milk and incubated for 1 hour at room temperature.
  • HRP horse radish peroxidase
  • the membrane was washed five times for 5 minutes each in TBST before exposure to ECL reagents.
  • the membrane was exposed to ECL (Pierce Chemical Company, Rockford, Ill.) for 1 minute then developed using an Alpha-Innotech (San Leandro, Calif.) digital system.
  • overnight cultures were started by adding 1 mL of a glycerol stock of yeast cells to 50 mL of YNB minus uracil medium in a 250 mL flask. Cultures were grown overnight at 30° C. with shaking. The next morning the OD600 was determined and a 50 mL interim culture was inoculated at a final OD600 of 0.2 and grown at 30o C with shaking until it had doubled (4-5 hours). The optical density was subsequently determined for the interim culture which was used to inoculate a final 250 mL culture at 0.02 (final OD600) and grown for 1 hour at 30° C. with shaking.
  • cells from cultures were harvested by centrifugation at 1900 g for 5 min at 4o C. Cultures were washed 4 times with 100 mL room temperature PBS (137 mM NaCl, 2.7 mM KCl, 10 mM Na2HPO 4 , 2 mM KH2PO 4 ) to remove any residual medium and copper from the cells. Cells were resuspended in 3 mL room temperature PBS and then added to 200 mL PBS that had been pre-warmed to 56° C. The cells were subsequently heat killed by incubation at 56° C. in a water bath for 1 hour, with inversion of tubes every 10 min. to ensure that all cells were heat killed.
  • PBS room temperature PBS
  • Cells were harvested by centrifugation (1900 ⁇ g for 5 min at 4o C) and then washed 4 times with 100 mL with room temperature PBS. Cells were counted and resuspended at a final concentration of 4 ⁇ 108 c/mL in PBS, aliquoted in 200 mL aliquots and stored at 4° C.
  • Yeast cells can be heat-killed by incubation in sterile, pyrogen-free saline at 56° C. for 1 hr, washed in saline, resuspended at ⁇ 2 ⁇ 10 8 cells/mL in sterile saline, and stored at 4° C. Each batch is expected to yield ⁇ 2 ⁇ 10 10 yeast cells, of which a vaccine dose for the animal studies will consist of ⁇ 1 ⁇ 10 7 to 20 ⁇ 10 7 cells.
  • the fungal organism Aspergillus was examined.
  • Efficacy of four A. fumigatus antigens protecting mice from systemic aspergillosis the protective efficacy of 4 A. fumigatus antigens expressed in Saccharomyces sp. in an experimental model of systemic murine aspergillosis was tested. Efficacy was evaluated in terms of survival and tissue burden. See FIG. 3 as one exemplary strategy for vaccine generation and animal testing.
  • vaccines preparations containing 6 ⁇ 10 7 heat-killed yeast cells/150 microliters were administered s.c. using two injection sites (0.075 ml each) on days 28, 21 and 14 before infection. Groups pretreated with Myc (yeast containing and expressing only the c-myc tag) or PBS were used as controls.
  • Inoculum A total of 1.3 ⁇ 10 7 conidia of A. fumigatus , strain AF-10, were inoculated via lateral tail vein into the CD-1 mice.
  • HEMO A. fumigatus antigen expressed in Saccharomyces sp. in a systemic murine model of aspergillosis
  • the animal model showed low mortality in the control groups i.e., PBS and MYC, and HEMO pretreatment did not significantly prolonged the survival of the animals.
  • the tissue burden study demonstrated that animals receiving three doses of the HEMO antigen before the infection had significantly lower CFU of the fungus in the kidneys and brain in comparison with the PBS-treated control animals ( FIG. 12 ).
  • a 16 amino acid signal sequence was predicted by PSORTII (http://psort.nibb.ac.jp/cgi-bin). The signal sequence was subsequently removed as PSORTII predicted that the protein would be extracellular, which suggested that the protein would be secreted by the S. cerevisiae if the signal sequence was left intact. A methionine was added before the 17th amino acid to ensure that the protein would be translated.
  • PCR reactions were separated using agarose gel electrophoresis and a photograph of a representative gel data not shown.
  • a band of approximately 936 bases corresponding to the amplified AspF 2 ORF contains a 3′ c-myc tag.
  • the band was excised from the gel, purified and ligated into pCR 2.1.
  • the ligated vector was used to transform competent E. coli cells and vector DNA isolated from 5 mL mini cultures using standard alkaline lysis techniques. DNA isolated from several miniprep isolates was digested with EcoRI and separated by agarose gel electrophoresis to confirm the presence of the 936 base AspF 2/c-myc insert.
  • the DNA of an isolate was excised from the pCR 2.1 vector (Invitrogen) with a BamHI/EcoRI double digest and the reactions were separated by electrophoresis so that the DNA band encoding the AspF 2 ORF could be isolated.
  • This DNA fragment was then ligated into the pYEX-BX vector using the same restriction enzymes.
  • the ligated vector was then used to transform competent E. coli and the plasmid DNA isolated by standard alkaline lysis mini-preps.
  • the DNA sequence of salient portions of the vector was determined to ensure the correct alignment of the ORF with the TATA box. Then portions of the vector were sequenced data not shown.
  • the TATA box, the aspf-2 coding sequence, the c-myc tag sequence, and the stop codons were identified (SEQ ID NOs: 1 and 2).
  • Exemplary cloning method for Hemolysin Published DNA and predicted amino acid sequences of Hemolysin to design PCR primers to amply the ORF from the A. fumigatus cDNA library were used. The DNA and amino acid sequences are presented in diagram form in SEQ ID NOs:7 and 8, along with the positions of the 5′ and 3′ primers used to obtain the predicted PCR product are shown. No signal sequence was predicted by PSORTII (http://psort.nibb.ac.jp/cgi-bin). SEQ ID NOs: 7 and 8 - nucleic acid and corresponding amino acid sequence of Hemolysin
  • the following conditions were used to amplify hemolysin from the cDNA library: 25 ⁇ L reactions were performed in PCR Ready Bead tubes containing 500 ng of cDNA library, 2 ⁇ M primers, and 13 ⁇ L sterile water. Reactions were performed using a Perkin Elmer 2400 Thermocycler under the following conditions: an initial cycle at 94° C. for 5 min, 30 cycles at 94° C. for 1 min, 50° C. for 1 min, 72° C. for 1 min, and a final extension at 72° C. for 7 min.
  • the PCR reactions were separated using agarose gel electrophoresis and a band of approximately 444 bp corresponding to the amplified hemolysin ORF was excised from the gel, purified and ligated into pCR 2.1.
  • the ligated vector was used to transform competent E. coli cells and vector DNA was subsequently isolated using standard techniques. DNA isolated from several isolates was used as template for PCR reactions to confirm the presence of the hemolysin insert. Not all isolates contained the hemolysin insert due to self ligation of the pCR 2.1 vector. See FIG. 5 .
  • the DNA of one isolate was treated with BamH I/EcoRI and the DNA band encoding the hemolysin ORF/c-myc tag was isolated. This DNA fragment was then ligated into vector pYEX-BX that had also been digested with BamH I/EcoRI. The ligated vector was then used to transform competent E. coli and the plasmid DNA was subsequently isolated. The DNA sequence of the relevant portions of the vector was determined to ensure the correct alignment of the ORF with the TATA box and the c-myc tag was in frame with the rest of the ORF. The sequenced portion of the vector is presented in SEQ ID NO:11. In addition, this vector was used for Examples 2 and 3. SEQ IN NO: 11 - Portions of pYEX-BX that were sequenced
  • an empty vector control was generated as previously described in Methods (This template was amplified by PCR as discussed previously. DNA isolated from several isolates was used as template for PCR reactions to confirm the presence of the c-myc insert. (SEQ ID NOs:1 and 2) The DNA of one isolate was treated with BamH I/EcoRI and the DNA band encoding the c-myc tag was isolated. This DNA fragment was then ligated into pYEX-BX which had also been digested with BamHI and EcoRI. The ligated vector was used to transform competent E. coli cells and vector DNA isolated using standard techniques. DNA isolated from several miniprep isolates was used as template for PCR reactions to confirm the presence of the c-myc control insert. (SEQ ID NOs:1 and 2)
  • transformants expressing each A. fumigatus protein were identified and the cleared cell lysates were generated and stored as previously discussed.
  • transformants were determined by western blot by methods known in the art. In one exemplary method growth of transformants were determined. Transformants that were determined to produce a protein of the correct molecular weight above were grown in YNB minus uracil for 2-3 days at 30° C. with shaking. Once the cultures were in stationary phase, the cells were counted and frozen in 50% glycerol at a final concentration of 1 ⁇ 108 c/mL (10 yeast units) and stored at ⁇ 80° C. until use.
  • each of four A. fumigatus genes from a cDNA library using gene-specific PCR primers were cloned.
  • the DNA encoding each ORF was sequenced to confirm that the correct gene had been cloned followed by an inframe c-myc tag and that no PCR errors were introduced.
  • the ORF of each gene was inserted into the copper inducible vector pYEX-BX. Portions of each ligated vector were sequenced to ensure the correct position of the gene relative to the TATA box.
  • Each pYEX-BX vector containing a gene of interest was used to transform yeast and yeast transformants expressing each of the four proteins identified by Western blot analysis.
  • Each S. cerevisiae strain that produced the correct protein was subsequently produced and used to vaccinate an animal as discussed above.
  • HEMO antigen was administered s.c. using two injection sites dorsally (0.075 ml each) on days 28, 21 and 14 before infection, receiving 6 ⁇ 10 7 yeast cells per animal. Control animals received PBS (diluent control) or MYC (yeast not expressing immunogen control) at 6 ⁇ 10 7 yeast cells per animal.
  • mice Thirty six weeks-old male CD-1 mice were used. Animals were housed in standard conditions in cages of 5 animals. Groups of 10 mice per group were established.
  • Tissue burden 16 days after infection surviving animals were euthanatized by CO2 anoxia. The kidney and brain were removed aseptically, homogenized in 0.9% saline, diluted and plated on SDA plates for CFU determination.
  • FIG. 12 represents an exemplary Log 10 CFU per studied organ.
  • No differences were found between MYC and PBS groups.
  • the protective effect of the A. fumigatus antigen (HEMO) expressed in Saccharomyces was tested in a systemic murine model of aspergillosis.
  • the MYC only yeast control group did show a reduction in the brain organ burden.
  • the tissue burden study demonstrated that animals receiving three doses of the HEMO antigen before the infection had statistically significantly lower CFU of the fungus in the kidneys and brain in comparison with the PBS-treated control animals.
  • the fungal organism Coccidioides was examined.
  • FIG. 6 See one exemplary schematic for production of and testing for an anti-fungal vaccination in a Coccidioides animal model.
  • the feasibility of using heat-killed yeast cells expressing a C. immitis antigen as a vaccine against C. immitis infections was examined.
  • cDNA was obtained for each the C. immitis gene whose protein has been shown in other work to elicit an immune response in infected animals.
  • yeast were transformed with each cDNA and strains selected that expressed a high levels of the antigen.
  • a schematic of the strategy is depicted in FIG. 6 .
  • the protective effect of Coccidioides immitis antigen expressed by Saccharomyces cerevisiae was analyzed and used as a killed whole vaccine preparation in a systemic murine model of coccidioidomycosis.
  • DNA encoding for Ag2 also called PRA
  • S. cerevisiae was cloned into S. cerevisiae and gene expression induced using for example, copper. These yeast cells were heat killed and used to vaccinate mice prior to infection with Coccidioides immitis.
  • mice Six-week-old male CD-1 mice were purchased from Charles River Laboratories. Groups consisted on 20 mice, housed in standard conditions. After infection, mice were housed in micro-isolator cages. Animals were provided food and acidified water ad libitum.
  • Antigen in this exemplary example, a C. immitis antigen expressed in Saccharomyces was used. This was a protein present in the alkali-soluble and water-soluble preparation of the spherule wall and called antigen 2 (Ag2/PRA).
  • the vaccine was administered subcutaneously (s.c.) in a volume of 50 ⁇ l three times before infection (i.e., 21, 14 and 7 days prior to the infection) at a concentration of 4 ⁇ 10 8 cells/ml.
  • PBS was used as the control.
  • Coccidioides immitis (Silveira strain) was used. The strain was grown on glucose yeast-extract (GYE) plates in a Class 3 biosafety cabinet at room temperature for 2 weeks. Arthroconidia were harvested as a suspension in saline. The number of arthroconidia was determined by hemacytometer and verified for viability by cfu (colony forming units) determination done by plating inoculum dilutions on GYE medium. Animals were infected intravenously (i.v.) with a suspension containing 376 viable arthroconidia per mouse. Statistical analysis of survival was done using a log rank test.
  • FIG. 8 illustrates an exemplary a Western Blot of extracts of yeasts containing and expressing Hemolysin (hemo), aspf 2, or antigen 2 (the Coccidioides protein described above).
  • the numbers indicate the amount of lysate separated by SDS-GEL electrophoresis.
  • the blots were treated with a c-myc epitope specific antibody and the antibody detected using a secondary horse radish peroxidase antibody.
  • Various amounts in micrograms indicated by the numbers at the bottom of the figure) of c-myc peptide were spotted.
  • Antigen 2/proline-rich antigen has been previously shown to be protectivein murine models of coccidiomycosis. Thus antigen 2 was chosen. Published DNA and predicted amino acid sequences were used to design PCR primers to amplify the ORF from the Cox cDNA library (SEQ ID NOs:12 and 13). The DNA and amino acid sequences of antigen 2 are presented in diagram form, data not shown. An 18 amino acid signal sequence was predicted by PSORTII (http://psort.nibb.ac.jp/cgi-bin), but was not removed as PSORTII also predicted that the protein would be cell wall bound, most likely GPI anchored. This prediction suggests that the protein would not be secreted by the S. cerevisiae . SEQ ID NOs: 12 and 13
  • Exemplary conditions used to amplify antigen 2 from the cDNA library were as follows: 25 ⁇ L reactions were performed in PCR Ready Bead tubes (Amersham Pharmacia) containing 500 ng of cDNA library, 2 ⁇ M primers, and 13 ⁇ L sterile water. Reactions were performed using the Perkin Elmer 2400 Thermocycler (PE Applied Biosystems) under the following conditions: an initial cycle at 94° C. for 5 min, 30 cycles at 94° C. for 1 min, 50° C. for 1 min and 72° C. for 1 min, and a final extension at 72° C. for 7 min.
  • PCR reactions were separated and a band of approximately 582 bases corresponding to the amplified antigen 2 ORF was excised.
  • the band was excised from the gel, purified and ligated into pCR 2.1.
  • the ligated vector was used to transform competent E. coli cells and vector DNA isolated from 5 mL mini cultures using standard alkaline lysis techniques. DNA isolated from several miniprep isolates was digested with EcoRI to confirm the presence of the 582 base antigen 2 insert. Only one isolate contained the antigen 2 insert.
  • the DNA of the isolate was excised from the pCR 2.1 vector and the salient portions of the vector was determined to ensure the correct alignment of the ORF with the TATA box and the c-myc tag, as described previously. Published DNA and predicted amino acid sequences were used to design PCR primers to amply the ORF from the cDNA library for antigen 2.
  • an empty vector control for animal models was designed as previously indicated; that is, the pYEX-BX vector containing the c-myc tag but lacking a C. immitis gene of interest.
  • Several isolates were grown, cells harvested, plasmid DNA isolated and the DNA sequence of the c-myc control insert were determined. The DNA sequence of pertinent portions of the vector was determined to ensure the correct alignment of the c-myc with the TATA box.
  • Transformation of S. cerevisiae was performed as previously described. After obtaining at least 10 transformants for each C. immitis gene of interest, cells are grown and induced the expression of each gene by adding copper sulfate. Uninduced and induced cells were harvested by centrifugation, lysed in SDS-gel lysis buffer and the protein concentration of each extract determined. Equal protein amounts of each lysate were separated by SDS-PAGE and the separated proteins transferred to membranes. The presence of the c-myc tagged proteins was detected by Western blot analysis using an anti-c-myc antibody. Then the transformants were grown as previously described. Once transformants of each construct were isolated, the proteins were expressed via copper induction as previously discussed. Then cultures were grown overnight at 30° C. with shaking. These cultures were used for the identification of transformants that were expressing each of the C. immitis proteins.
  • Cells were lysed in the following manner: 100 ⁇ L of glass beads were added to the tube containing cells followed by the addition of 200 ⁇ L 2 ⁇ sample lysis buffer (0.1M Tris, pH 6.8, 20% (v/v) glycerol, 4% (w/v) SDS, 6.2 mg DTT). Tubes were mixed by vortexing for 1 min and boiled for 3 min to resuspend and lyse the cells. Debris was removed by centrifugation and the cleared supernatant was transferred to a new tube. The cleared lysate was used for western blots.
  • sample lysis buffer 0.1M Tris, pH 6.8, 20% (v/v) glycerol, 4% (w/v) SDS, 6.2 mg DTT. Tubes were mixed by vortexing for 1 min and boiled for 3 min to resuspend and lyse the cells. Debris was removed by centrifugation and the cleared supernatant was transferred to a new tube. The cleared lysate
  • C. immits proteins were determined by exemplary western blot procedures known in the art.
  • Each of three C. immitis genes from a cDNA library using gene-specific PCR primers was obtained.
  • the DNA encoding each ORF was sequenced to confirm that the correct gene had been cloned and that no PCR errors were introduced.
  • the ORF of each gene was inserted into the copper inducible vector pYEX-BX. Each ligated vector was sequenced to ensure the correct position of the gene relative to the TATA box and that the c-myc tag was in frame with each protein.
  • the fungal organism Cryptococcosis was analyzed (see exemplary schematic FIG. 9 )
  • Cryptococcosis model In one exemplary method, an inhalation model of cryptococcosis was chosen because 1) it most closely mimics the natural course of C. neoformans infection, 2) it is highly reproducible and 3) it is commonly used to assess virulence.
  • the inhalation cryptococcosis model performed was essentially as previously described (see: Cox, G. M., Mukherjee, J., Cole, G. T., et al Urease as a virulence factor in experimental cryptococcosis. Infect Immun. (2000) 68:443-448. incorporated herein by reference).
  • mice are vaccinated with various vaccine formulations, and challenged with C. neoformans , and monitored for protection.
  • C. neoformans H 99 was used as the challenge organism for all infections.
  • C. neoformans H99 yeast for the inoculum was prepared as follows. Briefly, C. neoformans strain H99 was grown at 30° C. with shaking for two days in liquid YPD medium. The cells were harvested by centrifugation, washed in endotoxin-free PBS, and resuspended in endotoxin-free PBS. The cells were counted on a hemocytometer and diluted to 1 ⁇ 10 6 cells per ml. The inoculum was serially diluted and plated onto YPD to confirm the number of cells that are inoculated.
  • mice Four-week-old female A/J Cr mice were obtained from Charles River Laboratories for the inhalation model of cryptococcosis. Mice are commonly used for virulence assays with the serotype A strain, H99, and have been used in previous assessments of antibody protection against C. neoformans.
  • mice from a specific pathogen-free colony were vaccinated with 150 ⁇ L of each yeast vaccine preparation on day 21, 14 and 7 before being challenged with 50 uL containing 5 ⁇ 10 4 H 99 C. neoformans cells.
  • experiments were performed to determine whether yeast strains expressing C. neoformans proteins could be used as a vaccine against C. neoformans infections.
  • cDNA of certain genes whose proteins are likely to be important in infection were generated.
  • yeast cells were transformed with each cDNA and select strains that express high levels of each putative antigen.
  • each vaccine formulation was tested for its ability to protect vaccinated mice against a challenge of C. neoformans .
  • FIG. 9 represents a schematic of these experiments.
  • the PCR reactions were separated using agarose gel electrophoresis and a band of approximately 744 bases corresponding to the amplified CDA ORF containing a 3′ c-myc tag was excised. The band was purified and ligated into pCR 2.1. The ligated vector was used to transform competent E. coli cells and vector DNA isolated from 5 mL mini cultures using standard alkaline lysis techniques. DNA isolated from several miniprep isolates was digested with EcoRI to confirm the presence of the 744 base CDA/c-myc insert.
  • the DNA of the isolate was excised from the pCR 2.1 vector and ligated into the pYEX-BX vector as previously described and the salient portions of the vector was determined to ensure the correct alignment of the ORF with the TATA box
  • Laccase As indicated above, the same strategy to clone Laccase (lac) from a plasmid containing the gene as used to clone CDA. DNA sequence and plasmid DNA was used to predicted the amino acid sequences to design PCR primers to amplify the ORF plus the c-myc tag from the plasmid. The DNA and amino acid sequences are presented in diagram form in SEQ ID NOs:18 and 19. The positions of the 5′ and 3′ primers used to obtain the predicted PCR product are shown. A 20 amino acid signal sequence was predicted by PSORTII (http://psort.nibb.ac.jp/cgi-bin).
  • the signal sequence was subsequently removed as PSORTII predicted that the protein would be extracellular, which suggested that the protein would be secreted by the S. cerevisiae if the signal sequence was left intact.
  • An endogenous methionine at amino acid 21 (Glutamic acid) was used as the first amino acid in the new protein to ensure that the protein would be translated.
  • the PCR reactions were separated, isolated and digested with EcoRI as previously described to confirm the presence of the 1857 base Lac/c-myc insert.
  • Transformation of S. cerevisiae was made and used as previously described in methods.
  • C. neoformans proteins were determined by an exemplary western blot technique known in the art. Ten CDA transformants were screened to obtain a transformant that was positive for induction via copper. Transformant 10 was chosen for vaccine production as it had the most protein as determined by counting pixels. Five Lac transformants were screened to obtain a transformant that was positive for induction via copper. Please note that the top band is of the correct size. Transformant number 2 had the least amount of protein in the un-induced sample and the most (by counting pixels) in the copper induced sample
  • Transformants that were determined to produce a protein of the correct molecular weight were grown in YNB minus uracil for 2-3 days at 30° C. with shaking. Once the cultures were in stationary phase, the cells were counted and frozen in 50% glycerol at a final concentration of 1 ⁇ 108 c/mL (10 yeast units) and stored at ⁇ 80° C. until use.
  • Yeast cells containing and expressing chitin deacetylase or laccase were harvested and lysed as described. The indicated number of microliters of each extract were separated by SDS-PAGE, transfer to membranes and the position of each protein detected by Western blot analysis as described. The results of the western blot are shown in FIG. 10 .
  • Lane 1-4 are various amounts in microliters of lysed yeast cells expressing hitin deacetylase (cda; MW.26.6 kDa), while lanes 6-9 are various amounts in microliters of lysed yeast cells expressing laccase (lac; MW. 67.4 kDa).
  • FIG. 11 represents exemplary linear survival plots in an animal model after exposure to a challenge fungal organism.
  • Mice were vaccinated with yeast cells containing and expressing the DNA encoding laccase, (lac), chitin deacetylase (cda), the MYC yeast control, PBS, trr and plb (two additional C. neoformans proteins) as described above on days 21, 14 and 7 prior to challenge by C. neoformans H99 cells.
  • Mice were weighed daily and mice that had lost >15% of their body weight euthanized.
  • mice vaccinated with lac-containing yeast were 100% protected (p ⁇ 0.05) while those vaccinated with cda containing yeast were 80% protected (p ⁇ 0.05) as were those vaccinated with MYC (empty vector control). These results demonstrate that mice vaccinated with yeast expressing either laccase or chitin deacetylase were highly protected against a challenge of C. neoformans .
  • PCR primers 5′ AspF2/Bam 5′GGA TCC ATG CTT GTG GCC ACC CTC CCT 3′ (SEQ ID NO:20) 3′ AspF2/myc 5′GAA TTC TTA TCA CTC GAG GTC TTC TTC GGA AAT CAA CTT AGT GCA ATG AAG CTG TCC ACC TTC ATG GGT ATG GC 3′ (SEQ ID NO:21) 5′DppV/Bam 5′GGA TTC ATG CTT ACA CCT GAG CAG CTA ATC ACT GCT CCA CGG 3′ (SEQ ID NO:22) 3′DppV/myc 5′GGA TTC TTA TCA CTC GAG GTC TTC TTC GGA AAT CAA CTT CTG TTC CGG GAC AAC GGT GTC CTC CAG GCT GAC GGC GTT GGG G 3′ (SEQ ID NO:23) 5′Fos-1/SalI 5′G

Abstract

Embodiments of the present invention illustrate methods of treating and preventing infection due to a pathogen such as a fungal pathogen. In particular, the present invention relates to compositions and methods for vaccinations against or treatment for a fungal organism in a non-immunocompromised or immunocompromised subject.

Description

    RELATED APPLICATIONS
  • This application claims the benefit of priority under 35 U.S.C. §119(e) of U.S. provisional application No. 60/750,029, filed on Dec. 13, 2005, which is incorporated herein by reference in its entirety, and of U.S. provisional application No. 60/817,300, filed on Jun. 28, 2006, which is incorporated herein by reference in its entirety.
  • FEDERALLY FUNDED RESEARCH
  • The studies disclosed herein were supported in part by grants 1 R43 AI 052632-01A1, 1 R43 AI054020-01A1 and 1 R41 AI062482-01 from the Small Business Innovation Research Program (SBIR) and STTR Program of the National Institutes of Health. The U.S. government may have certain rights to practice the subject invention.
  • FIELD
  • The present invention provides for methods, compositions and kits for reducing a fungal infection and/or inducing a protective or therapeutic response against a pathogenic or non-pathogenic organism.
  • BACKGROUND
  • Many conditions in humans and animals are caused by fungal infections and current therapeutics for the prevention of and treatments for these infections are largely ineffective. Coccidioidomycosis, also known as San Joaquin Valley Fever, is a fungal disease caused by Coccidioides immitis that is endemic in portions of Southern Arizona, central California, southern New Mexico and west Texas. At least 100,000 new cases are reported each year. The migration of not only permanent residents, but also agricultural workers to these areas increases exposure to C. immitis spores that lie dormant in the soil, and as the soil is disturbed, the spores become airborne and are inhaled. Once in the lungs, the arthroconidia transform into spherules. An acute respiratory infection occurs between seven days to three weeks after exposure and often resolves rapidly. However, in a significant number of cases, chronic pulmonary conditions or dissemination to the meninges, bones, and joints can result, leading to acute, life-threatening disease. One population, migrant laborers, is exposed to C. immitis and are a highly susceptible to get infected. A variety of approaches have been used to fight coccidioidomycosis, including soil treatments, but only a vaccine can completely eliminate this “emerging disease.”
  • Another pathogen, Cryptococcus neoformans is an encapsulated pathogenic yeast that causes pulmonary infections and meningoencephalitis in humans and other animals. During the last 20 years, there has been a dramatic increase in the incidence of cryptococcosis throughout the world that mirrors the increase in not only HIV infections, but also in the growing number of immunocompromised patients. Loss of CD4+ T cells predisposes patients to progressive infection with C. neoformans—this emphasizes the role of cell-mediated immunity in host resistance.
  • C. neoformans is a basidiomycetous fungus that is generally isolated as a haploid yeast, although diploids have been identified in nature. There are two mating types that can undergo recognition and fusion, and form a mycelium. A structure called a basidium is made and spores are produced from the surface of the basidium. Under specific conditions, haploid cells can undergo sporulation. Recently, a stable diploid was shown to grow as a yeast at 37° C. and formed hyphae and produced spores at 24° C. C. neoformans has been the subject of many studies, the var. grubii strain H99, as the source of the candidate immunogens. Var. grubii are serotype A strains, have a worldwide distribution, and are the most common variety to cause disease in the United States. Strain H99 has several advantages. First, molecular genetic analysis, including gene deletion and allelic replacement experiments, are well established in H99. Second, the genome sequence of H99 has been determined. The random shotgun and assembly phases of the genome are complete and the assembled genome has been released. Third, a congenic mating partner for H99 has recently been developed. Lastly, several reproducible animal models have been well developed to assess virulence for this strain.
  • Cryptococcosis is a disease that is acquired by inhalation of the organism from the soil or avian droppings into the lungs. Both immunocompetent and immunocompromised patients can be infected with the organism. In immunocompetent patients, the disease is usually contained in a lung granuloma and induces an antibody response. In contrast, individuals whose cellular immunity has been compromised by viral infection, suppression due to tissue transplantation, or anti-neoplastic chemotherapy are particularly susceptible to disseminated disease, followed by often-fatal meningoencephalitis. For example, an estimated 7-10% of AIDS patients acquire cryptococcosis during the course of their HIV disease. In compromised hosts, patients with cryptococcosis generally present with symptoms of meningitis such as fever, headache, and malaise. Cryptococcal pneumonia is the next most frequent manifestation of cryptococcosis in immunocompromised patients. It occurs as a primary infection in approximately 4% of cases and is associated with general symptoms of pneumonia such as fever, cough, pleuritic chest pain, and/or dyspnea.
  • Aspergillus fumigatus is a ubiquitous spore-bearing fungus that causes multiple diseases in humans. These include allergic asthma, aspergillomas, and invasive pulmonary disease of hosts with predisposing underlying conditions. In the United States in 1996, there were an estimated 10,190 aspergillosis-related hospitalizations; these resulted in 1,970 deaths, 176,300 hospital days, and $633 million in costs. The average hospitalization lasted 17.3 days and cost ˜$62,000. Although aspergillosis-related hospitalizations account for a small percentage of hospitalizations in the United States, patients hospitalized with the condition have lengthy hospital stays and high mortality rates. The high mortality rates (in some instances over 90%) are due, in part, to the lack of rapid and sensitive diagnostic tests (all too often the diagnosis is made post mortem), as well as the lack of effective anti-fungal therapeutics.
  • The most common species of Aspergillus causing invasive disease include A. fumigatus, A. flavus, A. niger, A. terreus and A. nidulans. A. fumigatus is the most frequently found fungus in airborne spore surveys. The organism grows in a variety of environments including air ducts, houseplant soil, compost piles, and at a wide range of temperatures, from 12° C. to 55° C. Of the genus Aspergillus, A. fumigatus is the most common pathogen of man.
  • Currently, there are a number of anti-fungal vaccine and anti-fungal treatment effort that use a variety of approaches including selected recombinantly-expressed antigens. Safe and effective vaccines against fungal organisms as well as against other similar pathogens are needed.
  • SUMMARY
  • Embodiments of the present invention concern methods and compositions for reducing the onset of, preventing or as treatment for a fungal infection. In accordance with these embodiments, a composition derived at least in part from non-viable fungal cells can be used to vaccinate a subject against or treat for a fungal infection. In certain embodiments, fungal cells were heated at a temperature of about 56° C. for about one hour. In other embodiments, fungal cells were heated at temperatures ranging from about 50° C. to about 70° C. for from about 30 minutes to about 2 hours. Time and temperature may vary depending on the fungal organism, as well as, other factors such as the type of vaccine or therapeutic desired.
  • Certain embodiments of the present invention concern non-viable fungal cell compositions including fungal cells that were heated at a temperature of about 56° C. for about one hour, or a derivative thereof. Other particular embodiments concern vaccines, wherein the vaccines include a composition derived from non-viable fungal cells and wherein fungal cells were heated at a temperature of about 56° C. for about one hour. In accordance with these embodiments, the fungal cells can be transgenic, non-transgenic or a combination thereof. The vaccine compositions contemplated herein are capable of inducing an immune response against viable and non-viable fungal cells when administered to a subject. Fungal cells can include, but are not limited to, Saccharomyces spp., Aspergillus spp. Cryptococcus spp., Coccidioides spp., Neurospora spp., Histoplasma spp., Blastomyces spp., and a combination thereof. In other embodiments, the fungal cells may further comprise a tag. In certain exemplary transgenic vaccines, compositions can include fungal cells transformed with an oligonucleotide, a peptide, a protein, a chimeric molecule or combination thereof. In addition, transgenic fungal cells can contain one or more virulence factor.
  • In some embodiments, a derivative of a non-viable fungal composition can be an active portion of non-viable fungal cells or a fraction thereof that is capable of inducing an immune response when introduced to a subject. In accordance with these embodiments, the derivative can be used a prophylactic against, prevention of and/or treatment for a fungal infection.
  • In certain embodiments of the present invention, the fungal cells comprise transgenic, non-transgenic or a combination thereof. In other embodiments, the fungal cells can include, but are not limited to, Saccharomyces spp., Aspergillus spp., Cryptococcus spp., Coccidioides spp., Neurospora spp., Histoplasma spp., Blastomyces spp., and a combination thereof.
  • In certain particular embodiments, Saccharomyces spp. cells can be spores, vegetative cells, germlings, or a combination thereof. In other embodiments, the Neurospora spp. cells can be vegetative hyphae, aerial hyphae, macroconidia, germinating macroconidia, microconidia, germinating microconidia, ascospores, germinating ascospores, or a combination thereof. In other embodiments, the Aspergllus spp. cells can be vegetative hyphae, conidia, or germinating conidia, or combinations thereof.
  • In certain embodiments, Coccidioides spp. cells can be arthoconidia, germinating arthroconidia, hyphae, spherules, germinating spherules, endospores, germinating endospores, or a combination thereof. In other particular embodiments, Cryptococcus spp. cells can be yeast cells.
  • In other embodiments, Histoplasma spp. cells can be yeast cells, vegetative hyphae, microcondia, germinating microconidia, macroconidia, germinating macroconidia, or combinations thereof.
  • In other embodiments, Blastomyces spp. cells can be yeasts, hyphae, blastoconidia, germinating blastoconidia or combinations thereof. In some embodiments of the present invention, the fungal cells may further include a tag. In other particular embodiments, transgenic fungal cells can contain at least one oligonucleotide. In certain examples, the oligonucleotide can encode a peptide or a protein. Alternatively, the transgenic fungal cells contain at least one virulence factor.
  • Embodiments of the present invention concern administering to a subject a vaccine that includes, at least in part, non-viable fungal cells. In certain embodiments, vaccines of the present invention induce an immune response in the subject directed against one or more fungal organisms. In accordance with these embodiments, the subject is a mammal, bird or reptile. In one particular embodiment, the subject is a human. In certain examples, the vaccine can be administered in a single dose, a few doses or multiple doses to reduce the onset of a fungal infection, preventing and/or treating a fungal infection. In other embodiments, the vaccination can occur yearly, monthly, or weekly or even more frequent depending on need.
  • In other embodiments, a subject having a fungal infection can be treated prophylactically to reduce progression of the fungal infection and or eliminate the infection.
  • In certain embodiments of the present invention, the subject is a human predisposed to a fungal infection or a fungal organism (e.g., predisposed can mean at risk of exposure or having been exposed to a fungal infection or fungal organism). In other embodiments, the subject is an immunocompromised subject. In accordance with this embodiment, the immunocompromised subject is a subject having a viral infection, tissue transplantation, anti-neoplastic chemotherapy or combination thereof.
  • Other embodiments of the present invention concern kits for treating and/or preventing a fungal infection. In accordance with these embodiments, the kits can include non-viable fungal cells.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • The following drawings form part of the present specification and are included to further demonstrate certain embodiments of the present invention. The embodiments may be better understood by reference to one or more of these drawings in combination with the detailed description of specific embodiments presented herein.
  • FIG. 1 represents exemplary growth curves of yeast containing a gene encoding an antigen grown in the presence of various concentrations of CuSO4.
  • FIG. 2 illustrates an exemplary growth curve of yeast cells containing and expressing an antigen protein.
  • FIG. 3 illustrates an exemplary schematic of a method to test a vaccine disclosed herein.
  • FIG. 4 illustrates exemplary survival curves for mice infected in vitro with A. fumigatus.
  • FIG. 5. represents an exemplary electrophoresis gel of separated nucleic acid sequences from PCR reactions.
  • FIG. 6. illustrates an exemplary schematic of a method to test a vaccine disclosed herein.
  • FIG. 7 illustrates exemplary survival curves for mice infected in vitro with C immitis.
  • FIG. 8 represents an exemplary Western blot of separated lysed yeast cells containing and expressing genes for hemolysin, aspf2 or antigen-2/PRA.
  • FIG. 9 illustrates an exemplary schematic of a method to test a vaccine disclosed herein.
  • FIG. 10. represents an exemplary electrophoresis gel of separated molecules of yeast lysates containing and expressing the gene encoding Chitin deacetylase and Laccase.
  • FIG. 11 illustrates an exemplary survival curves for mice infected in vitro. with Cryptococcosis neoformans
  • FIG. 12 illustrates an exemplary scattergram of protective efficacy of Aspergillus hemolysin antigen expressed in Saccharomyces spp. against systemic aspergillosis in mice.
  • DESCRIPTION OF ILLUSTRATIVE EMBODIMENTS
  • Definitions
  • As used herein, “a” or “an” may mean one or more than one of an item.
  • As used herein, “about” can mean plus or minus ten percent.
  • DETAILED DESCRIPTION
  • In the following sections, various exemplary compositions and methods are described in order to detail various embodiments of the invention. It will be obvious to one skilled in the art that practicing the various embodiments does not require the employment of all or even some of the specific details outlined herein, but rather that sequences chosen, proteins selected, samples, concentrations, times and other specific details may be modified through routine experimentation. In some cases, well-known methods or components have not been included in the description.
  • Embodiments of the present invention provide for compositions and methods for reducing the onset of, preventing and/or treating a fungal infection. In particular embodiments, methods and compositions when administered to a subject are capable of inducing a protective immune response against a target. For example, these embodiments include a fungal pathogen. In accordance with these embodiments a fungal pathogenic organism includes, but is not limited to, Aspergillus spp., Cryptococcus spp., and Coccidioides spp. In exemplary embodiments, such pathogenic fungus may include, but are not limited to, Aspergillus fumigatus, A. flavus, A. niger, A. terreus, A. nidulans, Coccidioides immitis, Coccidioides posadasii and Cryptococcus neoformans. The immune response may be induced by injection of a fungal-based vaccine. Such vaccines may comprise, but are not limited to, suspensions, solutions, extracts, homogenates, precipitates and/or other processed or unprocessed forms of Saccharomyces cerevisiae. Although in exemplary embodiments the vaccines may comprise yeast transformed with genetic markers, such as a c-myc tag sequence. In one embodiment, the vaccines may include a non-transgenic fungus or fungi. In one particular embodiment, the vaccines may include non-transgenic or transgenic yeast.
  • Embodiments of the present invention provide for combination treatments of a subject in need of a vaccine against an organism such as a pathogenic fungal organism. In accordance with these embodiments, any vaccine treatment detailed herein may be combined with, but is not limited to, treatments for cryptococcosis, aspergillosis, or coccidoidomycosis such as amphotericin B or the azoles, fluconazole or itraconazole, 5-flucytosine, voriconazole, and fluconazole. In another embodiment, other treatments may include other antifungal treatments, such as any of the echinocandin class antifungals or any other antifungal agent.
  • It is contemplated herein that any of the vaccines disclosed may be administered to a subject having or suspected of developing a condition due to exposure or potential exposure to a fungal pathogen. In one embodiment, a subject may include a human. In other embodiments a subject may include a human, other mammals, birds and reptiles. In yet another embodiment, the subject can include a domesticated animal. Any of the methods disclosed herein may be used in combination with other antifungal agents or therapies to treat a subject in order to achieve the desired results.
  • In another embodiment, it is contemplated that a subject can be vaccinated and/or treated using a composition against a fungal pathogen disclosed herein before infection, during infection, after infection or combination thereof. In one particular embodiment, a composition disclosed herein may be used to vaccinate a subject against a particular fungal organism to reduce the risk of a fungal infection in the subject. In another embodiment, a composition disclosed herein may be used to vaccinate a subject against one or more fungal organisms to reduce the risk of a fungal infection in the subject. In certain embodiments, a vaccine containing non-viable fungal cells of one fungal type when administered to a subject may reduce the onset or progression of fungal infection of one or more additional fungal types in the subject.
  • In certain embodiments of the present invention, a vaccine or treatment of use in a subject can contain non-viable fungal cells where the fungal cells are transgenic, non-transgenic or a combination thereof. Transgenic cells of the present invention may include an oligonucleotide. In certain examples, the oligonucleotide can encode one or more peptides or proteins.
  • Advantages
  • Approaches disclosed herein have several advantages over current fungal vaccines. First, the fungal cells expressing each fungal protein may be engineered so that it is not secreted but rather is retained intracellularly. This likely obviates the need for large amounts of recombinant fungal antigen protein to be produced and purified, ultimately under cGMP conditions. Second, fungal cells are easily transformed and manipulated. Third, putative antigens can be tested rather inexpensively and ineffective antigens can be eliminated quickly. Fourth, whole, heat-killed yeast as a vaccine delivery vehicle is novel and presents antigens directly to dendritic cells to induce the immune system. Fifth, several individual antigen-expressing yeast strains can be mixed to form a vaccine, a possibility not readily available to current vaccine projects. Sixth, the yeast-based vaccine technology has each of the characteristics of a successful vaccine: generates a productive immune response; not neutralized; capable of generating a response to multiple epitopes; and stable and easy to manufacture.
  • One example of a beneficial therapeutic or a vaccine might consist of a vector with no pathogenic potential that can deliver several antigens to antigen-presenting cells (APCs). The use of recombinant yeast proves to be an ideal vector for vaccine development. In one embodiment, a vaccine contemplated herein could consist of a vector with no pathogenic potential that can deliver several antigens to APCs. In this regard, the use of recombinant yeast proves to be an ideal vector for vaccine development. The yeast Saccharomyces cerevisiae is avidly taken up by professional APCs, such as neutrophils, macrophages and dendritic cells. Multiple antigens can be engineered for expression within a single yeast formulation, and these formulations share many advantages with DNA vaccines, including ease of construction, low expense of mass production and biological stability. Unlike DNA vaccines, yeast-based vaccine formulations do not require extensive purification to remove potentially toxic contaminants. Furthermore, while the FDA has not evaluated yeast as a vaccine vector, the organism S. cerevisiae is designated by the FDA as GRAS (Generally Regarded As Safe, FDA Proposed Rule 62FR18938, Apr. 17, 1997). As described below, the heterologous proteins expressed in recombinant yeast serve as antigens that elicit CD8+ CTL-mediated immune responses in vitro and in vivo. In animal trials as a tumor vaccine, the yeast formulation was successful at protecting vaccinated animals from tumor growth (refer to FIGS. 4, 7, 11 and 12 as examples of animal model testing).
  • In another embodiment, it is contemplated that additional immunogens for in vivo efficacy may be tested using methods disclosed herein.
  • In addition, it is contemplated that the immunogens may be of a single formulation or a combination formulation. To test a formulation, immunocompetent animals or immuncompromised animals may be used in order to assess what formulation may be used to treat a subject. The test can include vaccination and then a challenge of the vaccination. In another embodiment, the appropriate dose of any disclosed therapeutic or vaccine may be determined by the methods disclosed herein including, but not limited to optimum formulation, the appropriate number of treatments or vaccinations and the optimum time for treatment or vaccinations in order to instill maximum treatment or protection against a given organism.
  • In certain embodiments, methods and compositions disclosed herein are directed toward making and using an anti-fungal vaccine or anti-fungal treatment. Fungal organisms targeted in the present invention include, but are not limited to, Saccharomyces spp., Aspergillus spp. Cryptococcus spp., Coccidioides spp., Neurospora spp., Histoplasma spp., Blastomyces spp., and a combination thereof.
  • Aspergillus
  • In certain embodiments of the present invention, methods, compositions and treatments for a fungal organism infection can concern treatment for or prevention of an Aspergillus associated-condition. The most common species of Aspergillus causing invasive disease include A. fumigatus, A. flavus, A. niger, A. terreus and A. nidulans. A. fumigatus is the most frequently found fungus in airborne spore surveys. A. fumigatus pneumonia and systemic aspergillosis occur primarily in patients who have immunosuppression or T-cell or phagocytic impairment. The immunodeficiency detected in these patients may be congenital, acquired or iatrogenic. Patients with chronic granulomatous diseases, neutrophil dysfunction, and with severe immunodeficiency are at risk for the development of this predominantly fatal infection. Although no important protective antibody response was detected, CD4+ Th1 cytokines appear to be important in rendering protection in these patients. In one embodiment of the present invention, a subject having or suspected of developing a disease derived from an Aspergillus species can be administered a composition disclosed herein. In one particular embodiment, a subject having or suspected of developing a disease derived from an Aspergillus species can be administered a composition including heated, transformed or non-transformed fungi for example, heated Aspergillus fungi.
  • Three main types of diseases are attributed to A. fumigatus; these include asthma, aspergillomas and invasive aspergillosis. The most serious disease involves invasion of hosts with predisposing underlying conditions. Patients undergoing organ transplants, bone marrow transplants, leukemics, or cancer chemotherapy are particularly at risk for invasive aspergillosis. Aspergillosis is often diagnosed when there is an unexplained pulmonary infiltrate, a patient is unresponsive to antibacterials and/or there is a fever of unknown origin. The prognosis for patients with invasive disease is high (mortality rates >50%) due to the lack of a rapid diagnostic test confirming A. fumigatus infection and the lack of safe and effective antifungal drugs.
  • Coccidioides
  • Coccidioidomycosis, also known as San Joaquin Valley Fever, is a fungal disease caused by Coccidioides immitis that is endemic in portions of Southern Arizona, central California, southern New Mexico and west Texas. At least 100,000 new cases are reported each year. The migration of not only permanent residents, but also agricultural workers to these areas increases exposure to Coccidioides spp. spores that lie dormant in the soil, and as the soil is disturbed, the spores become airborne and are inhaled. Once in the lungs, the arthroconidia transform into spherules. An acute respiratory infection occurs between seven days to three weeks after exposure and often resolves rapidly. However, in a significant number of cases, chronic pulmonary conditions or dissemination to the meninges, bones, and joints can result, leading to acute, life-threatening disease. Migrant laborers who are exposed to Coccidioides spp. are a highly mobile and underrepresented population, and unfortunately, this disease is under-reported and receives insufficient attention from the general medical community. A variety of approaches have been used to fight coccidioidomycosis, including soil treatments, but only a vaccine can completely eliminate this “emerging disease.”
  • Cellular Defenses Against Coccidioides spp.
  • Polymorphonuclear leukocytes (PMNs) are only able to partially inhibit growth of the pathogen, and are unable to kill the organism. PMNs are slightly more effective in inhibiting growth of arthroconidia than mature spherules. Since mature fungal spherules are typically 40-120 μm in diameter, a single PMN is unable to phagocytose the fungal cell. Endospores, on the other hand, are more sensitive to growth inhibition by these host cells. Most investigators of cellular immune response to Coccidioides spp. believe that macrophages are the pivotal effector cells in coccidioidomycosis. This concept has arisen from the general paradigm for granulomatous diseases: activated T-lymphocytes secrete cytokines, which activate macrophages, inducing the formation of a granuloma, which kills or contains the organism. As the spherule develops and matures, it becomes more resistant to macrophages, so that less than 10% of mature spherules are killed. It has been suggested that specific immunologic suppression elicited by Coccidioides spp. antigens prevents an effective T-cell response.
  • Cryptococcosis
  • In certain embodiments of the present invention, methods, compositions and treatments for a fungal organism infection can concern treatment for or prevention of a Cryptococcosis-associated condition. C. neoformans can be an intracellular as well as an extracellular pathogen. It is easily found extracellularly in cerebral spinal fluid and lung tissue. It can also be localized inside macrophages and neutrophils where it has been shown to replicate.
  • Antibody and cell-mediated responses can provide important protection even against an intracellular pathogen. The mammalian host has several defenses against C. neoformans that include components from innate, humoral, and cell-mediated immunity. Subjects are continually exposed to C. neoformans from the environment, a powerful innate immunity represents part of the protection afforded the normal host. This appears to be inherent in normal macrophage, monocyte, and neutrophil function.
  • Vaccination against cryptococcosis presents the innovative idea of not only aiming to protect an immunocompromised population, but also possibly expecting an immune response in that population. In one embodiment, a vaccine against cryptococcosis is contemplated for example, to treat a non-immunosuppressed or immunosuppressed subject. In one particular example, immunosuppressed patients such as HIV patients are contemplated. In accordance with this embodiment, an HIV patient may be vaccinated immediately after diagnosis, but before their CD4+ T cell population decreases.
  • In other embodiments, any of the vaccines detailed herein may be directed towards other fungal organisms, for example, Coccidioides and Aspergillus. It is contemplated herein that any fungal pathogen disclosed may have one or more virulence factors which may include surface proteins, cell-wall carbohydrates, secreted factors, anchored surface molecules, modes of action to invade a host, etc. Any virulence factor associated with a fungal pathogen may be used herein as a potential target to transform yeast or other fungi of the present invention to generate a vaccine against that fungal pathogen.
  • In certain embodiments of the present invention, the fungal cells comprise transgenic cells, non-transgenic cells or a combination thereof. In other embodiments, the fungal cells can include, but are not limited to, Saccharomyces spp., Aspergillus spp., Cryptococcus spp., Coccidioides spp., Neurospora spp., Histoplasma spp., Blastomyces spp., and a combination thereof.
  • In certain particular embodiments, Saccharomyces spp. cells can be spores, vegetative cells, germlings, or a combination thereof. In other embodiments, the Neurospora spp. cells can be vegetative hyphae, aerial hyphae, macroconidia, germinating macroconidia, microconidia, germinating microconidia, ascospores, germinating ascospores, or a combination thereof.
  • Recombinant Yeast as an Antigen Delivery System
  • In one example, a yeast vaccine formulation directly accesses DCs, the critical immune responsive cells of the body. In order for DCs to present antigens efficiently to naive T cells, immature DCs must be activated to mature, as evidenced by the up-regulation of MHC and co-stimulatory molecules. Mature DCs are then capable of prolonged antigen presentation and the production of cytokines, such as IL-12, that are critical for the induction of cellular immune responses. Uptake of yeast by DCs increased surface expression of the co-stimulatory molecules CD40, CD80 and CD86, MHC class II, and the adhesion molecule ICAM, to levels comparable to that induced by exposure to bacterial lipopolysaccharide (LPS), a potent DC maturation factor. As further evidence of yeast-induced activation, DCs incubated with yeast produced significant amounts of IL-12 that rivalled levels induced by exposure to LPS. These results show that yeast trigger DC maturation, responses that would be essential for vaccine-induced immunity.
  • It has been shown that certain yeast vaccine technology can be formulated with a variety of antigens, including 5 from HIV, 3 from Hepatitis B, 1 from Hepatitis C, and 2 from lung cancer cells and, in each case, the yeast-expressed antigen effectively stimulated protective cell-mediated immunity. In addition, yeast as a vaccine vehicle provides an adjuvant effect, such that dendritic cells are triggered to directly take up yeast, causing the DCs to mature and to present the yeast-associated antigens to the host immune system. Further, recent results have shown that heat-killed yeast cells expressing putative antigens are as immunogenic and protective as live yeast cells expressing putative antigens. In addition, yeast cells expressing antigens elicited Th1 responses. Therefore, recombinant-yeast vaccine formulations may elicit systemic, antigen-specific, CD4 and CD8-based protective immunity. Also, DCs have been shown to internalize yeast and present recombinant antigens to naive class I and class II MHC-restricted T cells. It has been shown that yeast exhibit adjuvant activity for DC maturation and immune signalling. Yeast might be providing an adjuvant effect to promote immune responses to the yeast-associated antigens. These results show that i) yeast uptake triggers DC maturation; ii) yeast-expressed recombinant antigens are efficiently presented by MHC class I and MHC class II pathways of DCs, and, iii) yeast provides an adjuvant effect to enhance DC-stimulation of naive T cells.
  • In another example, a CD8-dependent protective immunity has been elicited in vaccinated mice. For example, EL-4 CD4+ T lymphoma cells (H-2b) transfected with cDNA encoding chicken ovalbumin (E.G7-OVA) and ovalbumin-expressing melanoma (B16-OVA) mouse tumor models have been employed to test vaccine candidates that induce protective CTL response. Mice vaccinated with OVAX, but not mock-vaccinated animals, were protected from E.G7-OVA or B16-OVA tumor formation. Protective immunity was CD8-dependent, since OVAX-vaccinated animals were not protected from tumor challenge in CD8−/− knockout mice.
  • Kits
  • In still further embodiments, the present invention concerns kits for the methods described herein. In one embodiment, a fungal organism treatment or (such as a pathogenic or non-pathogenic fungus) prevention kit is contemplated. In another embodiment, a kit for prophylactic treatment of a subject having or suspected of developing a fungal infection is contemplated. In a more particular embodiment, a kit for prevention or treatment of a subject having or suspected of developing a fungal-induced infection is contemplated. In accordance with this embodiment, the kit may be used to treat or vaccinate a subject for or against a fungal infection.
  • The kits may include a composition including at least a portion of a non-viable fungus within a tube, a vial or other suitable vessel. In addition, the kit may include one or more dose(s) of the composition for administration to a subject having or exposed to a fungal organism infection by a healthcare provider. In another embodiment, the kit may be a portable kit for use at a specified location outside of a healthcare facility.
  • The container means of any of the kits will generally include at least one vial, test tube, flask, bottle, syringe or other container means, into which the testing agent, may be preferably and/or suitably aliquoted.
  • Nucleic Acids
  • In various embodiments, isolated nucleic acids may be used to modify or transform a fungal organism contemplated in the present invention. The isolated nucleic acid may be derived from genomic DNA, RNA or complementary DNA (cDNA). In other embodiments, isolated nucleic acids, such as chemically or enzymatically synthesized DNA, may be of use for generation of oligonucleotides for transformation of a fungal organism.
  • A “nucleic acid” includes single-stranded and double-stranded molecules, as well as DNA, RNA, chemically modified nucleic acids and nucleic acid analogs. It is contemplated that a nucleic acid may be of 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, about 110, about 120, about 130, about 140, about 150, about 160, about 170, about 180, about 190, about 200, about 210, about 220, about 230, about 240, about 250, about 275, about 300, about 325, about 350, about 375, about 400, about 425, about 450, about 475, about 500, about 525, about 550, about 575, about 600, about 625, about 650, about 675, about 700, about 725, about 750, about 775, about 800, about 825, about 850, about 875, about 900, about 925, about 950, about 975, about 1000, about 1100, about 1200, about 1300, about 1400, about 1500, about 1750, about 2000 or greater nucleotide residues in length, up to a full length protein encoding or regulatory genetic element.
  • Construction of Nucleic Acids
  • Isolated nucleic acids may be made by any method known in the art, for example using standard recombinant methods, synthetic techniques, or combinations thereof. In some embodiments, the nucleic acids may be cloned, amplified, or otherwise constructed.
  • Nucleic acids of use in the present invention may include oligonucleotides that include at least a portion of sequences from a virulence factor (e.g., including but not limited to, antigen-2/proline rich antigen, hemolysin, aspf2, dpp5, dpp4, chitin deacetylase, catalase), a disease-associated factor (e.g., tumor associated antigens, cytokines, viral-associated antigens, bacterial-associated antigens) or other peptides or proteins. In another example, a multi-cloning site comprising one or more endonuclease restriction sites may be added. A nucleic acid may be attached to a vector, adapter, or linker for cloning of a nucleic acid. Additional sequences may be added to such cloning and sequences to optimize their function, to aid in isolation of the nucleic acid, or to improve the introduction of the nucleic acid into a cell. Use of cloning vectors, expression vectors, adapters, and linkers is well known in the art.
  • Recombinant Methods for Constructing Nucleic Acids
  • Isolated nucleic acids may be obtained from fungal, bacterial, viral or other sources using any number of cloning methodologies known in the art. In some embodiments, oligonucleotide probes which selectively hybridize, under stringent conditions, to the nucleic acids are used to identify a sequence of interest. Methods for construction of nucleic acid libraries are known and any such known methods may be used. (See, e.g., Current Protocols in Molecular Biology, Ausubel, et al., Eds., Greene Publishing and Wiley-Interscience, New York (1995); Sambrook, et al., Molecular Cloning: A Laboratory Manual, 2nd Ed., Cold Spring Harbor Laboratory Vols. 1-3 (1989); Methods in Enzymology, Vol. 152, Guide to Molecular Cloning Techniques, Berger and Kimmel, Eds., San Diego: Academic Press, Inc. (1987)).
  • Nucleic Acid Screening and Isolation
  • Genomic DNA, RNA or cDNA may be screened for the presence of an identified genetic element of interest using a probe based upon one or more sequences. Various degrees of stringency of hybridization may be employed in the assay. As the conditions for hybridization become more stringent, there must be a greater degree of complementarity between the probe and the target for duplex formation to occur. The degree of stringency may be controlled by temperature, ionic strength, pH and/or the presence of a partially denaturing solvent such as formamide. For example, the stringency of hybridization is conveniently varied by changing the concentration of formamide within the range of 0% to 50%. The degree of complementarity (sequence identity) required for detectable binding will vary in accordance with the stringency of the hybridization medium and/or wash medium.
  • High stringency conditions for nucleic acid hybridization are well known in the art. For example, conditions may comprise low salt and/or high temperature conditions, such as provided by about 0.02 M to about 0.15 M NaCl at temperatures of about 50° C. to about 70° C. Other exemplary conditions are disclosed in the following Examples. It is understood that the temperature and ionic strength of a desired stringency are determined in part by the length of the particular nucleic acid(s), the length and nucleotide content of the target sequence(s), the charge composition of the nucleic acid(s), and to the presence or concentration of formamide, tetramethylammonium chloride or other solvent(s) in a hybridization mixture. Nucleic acids may be completely complementary to a target sequence or may exhibit one or more mismatches.
  • Nucleic Acid Amplification
  • Nucleic acids of interest may also be amplified using a variety of known amplification techniques. For instance, polymerase chain reaction (PCR) technology may be used to amplify target sequences directly from RNA or cDNA. PCR and other in vitro amplification methods may also be useful, for example, to clone nucleic acid sequences, to make nucleic acids to use as probes for detecting the presence of a target nucleic acid in samples, for nucleic acid sequencing, or for other purposes. Examples of techniques of use for nucleic acid amplification are found in Berger, Sambrook, and Ausubel, as well as Mullis et al., U.S. Pat. No. 4,683,202 (1987); and, PCR Protocols A Guide to Methods and Applications, Innis et al., Eds., Academic Press Inc., San Diego, Calif. (1990). PCR-based screening methods have been disclosed. (See, e.g., Wilfinger et al. BioTechniques, 22(3): 481-486 (1997))
  • Synthetic Methods for Constructing Nucleic Acids
  • Isolated nucleic acids may be prepared by direct chemical synthesis by methods such as the phosphotriester method of Narang et al., Meth. Enzymol. 68:90-99 (1979); the phosphodiester method of Brown et al., Meth. Enzymol. 68:109-151 (1979); the diethylphosphoramidite method of Beaucage et al., Tetra. Lett. 22:859-1862 (1981); the solid phase phosphoramidite triester method of Beaucage and Caruthers, Tetra. Letts. 22(20):1859-1862 (1981), using an automated synthesizer as in Needham-VanDevanter et al., Nucleic Acids Res., 12:6159-6168 (1984); or by the solid support method of U.S. Pat. No. 4,458,066. Chemical synthesis generally produces a single stranded oligonucleotide. This may be converted into double stranded DNA by hybridization with a complementary sequence, or by polymerization with a DNA polymerase using the single strand as a template. While chemical synthesis of DNA is best employed for sequences of about 100 bases or less, longer sequences may be obtained by the ligation of shorter sequences.
  • Covalent Modification of Nucleic Acids
  • A variety of cross-linking agents, alkylating agents and radical generating species may be used to bind, label, detect, and/or cleave nucleic acids. For example, Vlassov, V. V., et al., Nucleic Acids Res (1986) 14:4065-4076, disclose covalent bonding of a single-stranded DNA fragment with alkylating derivatives of nucleotides complementary to target sequences. A report of similar work by the same group is that by Knorre, D. G., et al., Biochimie (1985) 67:785-789. Iverson and Dervan also showed sequence-specific cleavage of single-stranded DNA mediated by incorporation of a modified nucleotide which was capable of activating cleavage (J Am Chem Soc (1987) 109:1241-1243). Meyer, R. B., et al., J Am Chem Soc (1989) 111:8517-8519 disclose covalent crosslinking to a target nucleotide using an alkylating agent complementary to the single-stranded target nucleotide sequence. A photoactivated crosslinking to single-stranded oligonucleotides mediated by psoralen was disclosed by Lee, B. L., et al., Biochemistry (1988) 27:3197-3203. Use of crosslinking in triple-helix forming probes was also disclosed by Home, et al., J Am Chem Soc (1990) 112:2435-2437. Use of N4, N4-ethanocytosine as an alkylating agent to crosslink to single-stranded oligonucleotides has also been disclosed by Webb and Matteucci, J Am Chem Soc (1986) 108:2764-2765; Nucleic Acids Res (1986) 14:7661-7674; Feteritz et al., J. Am. Chem. Soc. 113:4000 (1991). Various compounds to bind, detect, label, and/or cleave nucleic acids are known in the art. See, for example, U.S. Pat. Nos. 5,543,507; 5,672,593; 5,484,908; 5,256,648; and, 5,681,941.
  • Nucleic Acid Labeling
  • In various embodiments, tag nucleic acids may be used to trace or identify a particular nucleic acid sequence contemplated herein. A number of different labels may be used, such as fluorophores, chromophores, radio-isotopes, enzymatic tags, antibodies, chemiluminescent, electroluminescent, affinity labels, etc. One of skill in the art will recognize that these and other label moieties not mentioned herein can be used. Examples of enzymatic tags include urease, alkaline phosphatase or peroxidase. Colorimetric indicator substrates can be employed with such enzymes to provide a detection means visible to the human eye or spectrophotometrically. A well-known example of a chemiluminescent label is the luciferin/luciferase combination.
  • In certain embodiments, the label may be a fluorescent, phosphorescent or chemiluminescent label. Exemplary photodetectable labels may be selected from the group consisting of Alexa 350, Alexa 430, AMCA, aminoacridine, BODIPY 630/650, BODIPY 650/665, BODIPY-FL, BODIPY-R6G, BODIPY-TMR, BODIPY-TRX, 5-carboxy-4′,5′-dichloro-2′,7′-dimethoxy fluorescein, 5-carboxy-2′,4′,5′,7′-tetrachlorofluorescein, 5-carboxyfluorescein, 5-carboxyrhodamine, 6-carboxyrhodamine, 6-carboxytetramethyl amino, Cascade Blue, Cy2, Cy3, Cy5,6-FAM, dansyl chloride, Fluorescein, HEX, 6-JOE, NBD (7-nitrobenz-2-oxa-1,3-diazole), Oregon Green 488, Oregon Green 500, Oregon Green 514, Pacific Blue, phthalic acid, terephthalic acid, isophthalic acid, cresyl fast violet, cresyl blue violet, brilliant cresyl blue, para-aminobenzoic acid, erythrosine, phthalocyanines, azomethines, cyanines, xanthines, succinylfluoresceins, rare earth metal cryptates, europium trisbipyridine diamine, a europium cryptate or chelate, diamine, dicyanins, La Jolla blue dye, allopycocyanin, allococyanin B, phycocyanin C, phycocyanin R, thiamine, phycoerythrocyanin, phycoerythrin R, REG, Rhodamine Green, rhodamine isothiocyanate, Rhodamine Red, ROX, TAMRA, TET, TRIT (tetramethyl rhodamine isothiol), Tetramethylrhodamine, and Texas Red. These and other labels are available from commercial sources, such as Molecular Probes (Eugene, Oreg.).
  • In certain embodiments, it is contemplated that a vaccine will be administered to a subject for vaccination against and/or treatment of a fungal infection. It is also contemplated herein that any of the vaccines disclosed may be administered to a subject having or suspected of developing a condition due to exposure or potential exposure to a fungal pathogen. In one embodiment, a subject may include a human. In other embodiments a subject may include other mammals, birds, and reptiles. Any of the methods disclosed herein may be used in combination with other anti-fungal agents or therapies to treat a subject in order to achieve the desired results.
  • In another embodiment, it is contemplated that a subject can be vaccinated and/or treated using a composition against a fungal pathogen disclosed herein before infection, during infection, after infection or combination therefore of. In one particular embodiment, a composition disclosed herein may be used to vaccinate a subject against a particular fungal organism to reduce the risk of a fungal infection in the subject. In another embodiment, a composition disclosed herein may be used to vaccinate a subject against one or more fungal organism to reduce the risk of a fungal infection in the subject. In certain embodiments, a vaccine containing non-viable fungal cells of one fungal type when administered to a subject may reduce the onset or progression of fungal infection of another fungal type in the subject.
  • Exemplary Dosing: In one embodiment, a dose may consist of around 1×107 yeast cells to 20×107 yeast cells per dose. And may be given before infection, during infection, after infection or a combination therefore of The dose may be given daily, every other day, biweekly, weekly, seasonally or yearly until a desired effect is achieved.
  • A composition of the present invention may be administered to a subject in an appropriate carrier or diluent or pharmaceutically acceptable carrier, co-administered with enzyme inhibitors or in an appropriate carrier such as liposomes. The term “pharmaceutically acceptable carrier” as used herein is intended to include diluents such as saline and aqueous buffer solutions. To administer a compound that stimulates an immune response by other than parenteral administration, it may be necessary to coat the compound with, or co-administer the compound with, a material to prevent its inactivation. Enzyme inhibitors include pancreatic trypsin inhibitor, diisopropylfluorophosphate (DEP) and trasylol. Liposomes include water-in-oil-in-water emulsions as well as conventional liposomes. The active agent may also be administered parenterally or intraperitoneally. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations may contain a preservative to prevent the growth of microorganisms.
  • Pharmaceutical compositions suitable for injectable use include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersion. In all cases, the composition must be sterile and must be fluid to the extent that easy syringability exists. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria. The pharmaceutically acceptable carrier can be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyetheylene glycol, and the like), and suitable mixtures thereof. The proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prevention of microorganisms can be achieved by various antibacterial and antifungal agents (i.e., parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, and the like). In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as manitol, sorbitol, sodium chloride in the composition. A compound such as aluminum monostearate and gelatin can be included to prolong absorption of the injectable compositions.
  • Sterile injectable solutions can be prepared by incorporating active compound in the required amount in an appropriate solvent with one or a combination of ingredients enumerated above, as required, followed by filtered sterilization. Generally, dispersions are prepared by incorporating the active compound into a sterile vehicle that contains a dispersion medium and other required ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum drying and freeze-drying which yields a powder of the active ingredient (i.e., a chemical agent, antibody, etc.) plus any additional desired ingredient from a previously sterile-filtered solution thereof
  • When the active agent is suitably protected, as described above, the composition may be orally administered (or otherwise indicated), for example, with an inert diluent or an assimilable edible carrier. It is especially advantageous to formulate parenteral compositions in dosage unit form for ease of administration and uniformity of dosage. Dosage unit form as used herein refers to physically discrete units suited as unitary dosages for the mammalian subjects to be treated; each unit containing a predetermined quantity of compound such as non-viable fungal cells calculated to produce the desired therapeutic effect in association with the required pharmaceutical carrier.
  • If needed for a particular use, the biological material can be extensively dialyzed to remove undesired small molecular weight molecules and/or lyophilized for more ready formulation into a desired vehicle, where appropriate. The active compounds will then generally be formulated for parenteral administration (i.e., formulated for injection via the intravenous, intramuscular, sub-cutaneous, intralesional, or even intraperitoneal routes). The preparation of an aqueous composition that contains an active component or ingredient will be known. Typically, such compositions can be prepared as injectables, either as liquid solutions or suspensions; solid forms suitable for use in preparing solutions or suspensions upon the addition of a liquid prior to injection can also be prepared; and the preparations can also be emulsified.
  • The pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions; formulations including sesame oil, peanut oil or aqueous propylene glycol; and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases the form must be sterile and must be fluid. It must be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms, such as bacteria and fungi.
  • Solutions of the active compounds as free-base or pharmacologically acceptable salts can be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.
  • Sterile injectable solutions are prepared by incorporating the active compounds in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the required other ingredients from those enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation are vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient plus any additional desired ingredient from a previously sterile-filtered solution thereof. The preparation of more, or highly, concentrated solutions for direct injection is also contemplated, where the use of DMSO as solvent is envisioned to result in extremely rapid penetration, delivering high concentrations of the active agents to a small area.
  • Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective. The formulations are easily administered in a variety of dosage forms, such as the type of injectable solutions described above, but drug release capsules and the like can also be employed.
  • For parenteral administration in an aqueous solution, for example, the solution should be suitably buffered if necessary and the liquid diluent first rendered isotonic with sufficient saline or glucose. These particular aqueous solutions are especially suitable for intravenous, intramuscular, subcutaneous and intraperitoneal administration. In this connection, sterile aqueous media that can be employed will be known to those of skill in the art in light of the present disclosure.
  • In addition to the compounds formulated for parenteral administration, such as intravenous or intramuscular injection, other pharmaceutically acceptable forms include, e.g., liposomal formulations; time-release capsules; and any other form currently used.
  • In another embodiment, nasal solutions or sprays, aerosols or inhalants may be used to deliver the compound of interest. Nasal solutions are usually aqueous solutions designed to be administered to the nasal passages in drops or sprays. Nasal solutions are prepared so that they are similar in many respects to nasal secretions. Thus, the aqueous nasal solutions usually are isotonic and slightly buffered to maintain a pH of 5.5 to 6.5. In addition, antimicrobial preservatives, similar to those used in ophthalmic preparations, and appropriate drug stabilizers, if required, may be included in the formulation. Various commercial nasal preparations are known and include, for example, antibiotics and antihistamines and are used for asthma prophylaxis. Inhalation preparations may include solutions or dry powder formulations that are commonly used along with a propellant in the formulation of therapeutics used for the treatment of asthmatics.
  • Oral formulations include such normally employed excipients as, for example, pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate and the like. These compositions take the form of solutions, suspensions, tablets, pills, capsules, sustained release formulations or powders. In certain defined embodiments, oral pharmaceutical compositions will comprise an inert diluent or assimilable edible carrier, or they may be enclosed in hard or soft shell gelatin capsule, or they may be compressed into tablets, or they may be incorporated directly with the food of the diet. For oral therapeutic administration, the active compounds may be incorporated with excipients and used in the form of ingestible tablets, buccal tables, troches, capsules, elixirs, suspensions, syrups, wafers, and the like. Such compositions and preparations should contain at least 0.1% of active compound. The percentage of the compositions and preparations may, of course, be varied and may conveniently be between about 2 to about 75% of the weight of the unit, or preferably between 25-60%. The amount of active compounds in such therapeutically useful compositions is such that a suitable dosage will be obtained.
  • Particularly preferred are methods in which the therapeutic compound(s) are directly administered as a pressurized aerosol or nebulized formulation to the patient's lungs via inhalation. Such formulations may contain any of a variety of known aerosol propellants useful for endopulmonary and/or intranasal inhalation administration. In addition, water may be present, with or without any of a variety of cosolvents, surfactants, stabilizers (e.g., antioxidants, chelating agents, inert gases and buffers).
  • EXAMPLES
  • The following examples are included to demonstrate preferred embodiments of the invention. It should be appreciated by those of skill in the art that the techniques disclosed in the examples which follow represent techniques discovered by the inventors to function well in the practice of the invention, and thus can be considered to constitute preferred modes for its practice. However, those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made in the specific embodiments which are disclosed and still obtain a like or similar result without departing from the spirit and scope of the invention.
  • Methods
  • Proteins to be tested Each protein from a fungal pathogen was chosen because of its identification as a virulence factor (e.g., strains lacking the protein were less virulent that the wild type), as an immunogen (infected animals generated antibodies to the candidate protein), and/or was found on the cell surface of conidia or hyphae. In addition, none of the proteins had cognates with significant (<35%) homology to mammals and to yeast.
  • Exemplary isolation of DNA containing the open reading frame encoding each protein: The Open Reading Frame (ORF) of each gene was obtained by PCR using gene specific primers and in certain embodiments, a high quality cDNA library as template. In other embodiments, the ORF of candidate genes was obtained using plasmid DNA containing the gene of interest as a template for amplification by PCR. Each PCR product was gel purified and ligated into the TA cloning vector, pCR2.1, using the pCR2.1 TOPO kit from InVitrogen (Carlsbad, Calif.). Resulting plasmid DNAs were used to transform competent E. coli cells and plasmid DNA isolated from midi-preps using methods standard to those of skill in the art.
  • In certain embodiments the gene specific PCR primers encoded a c-myc epitope such that the c-myc epitope would be generated in the protein at the carboxy terminus.
  • In certain methods, each ORF was excised from each plasmid DNA using the restriction endonuclease EcoR1 and ligated into the EcoR1 site of pYEX-BX. Each resulting ligated plasmid was used to transform competent E. coli cells and plasmid DNA isolated from midi-preps. Each ORF DNA was sequenced to ensure that it is in frame with the copper-inducible promoter and and no PCR errors were present.
  • In one exemplary method an empty vector control was generated. As one exemplary control, an empty vector control was generated a pYEX-BX (Clonetech) vector containing the DNA encoding a c-myc epitope but lacking a fungal gene of interest. A 51 base pair template for PCR was created that contained a methionine, the sequence that encodes the c-myc protein and two stop codons. The PCR primers also included a 5′ BamH I site and a 3′ EcoR I restriction site that could be used to excise the tag from the pCR 2.1 vector and inserted into the pYEX-BX vector (Clonetech). The DNA and amino acid sequences are shown in SEQ ID NOs:1 and 2:
    c-myc control sequence
    atg gaa cag aag ttg att tcc gaa gaa gac ctc gag
    Met Glu Gln Lys Leu Ile Ser Glu Glu Asp Leu Glu
  • This template was amplified by PCR under the following conditions: 25 μL reactions were performed in PCR Ready Bead tubes containing 500 ng of cDNA library, 2 μM primers, and 13 μL sterile water. Reactions were performed using a Perkin Elmer 2400 Thermocycler under the following conditions: an initial cycle at 94° C. for 5 min, 30 cycles at 94° C. for 1 min, 50° C. for 1 min, 72° C. for 1 min, and a final extension at 72° C. for 7 min.
  • The PCR reactions were separated by agarose gel electrophoresis using a 3% (w/v) agarose gel. The 51 bp band was excised from the gel, purified and ligated into pCR 2.1 (Invitrogen). The ligated vector was used to transform competent E. coli cells and vector DNA was subsequently isolated using standard techniques. DNA isolated from several isolates was used as template for PCR reactions to confirm the presence of the c-myc insert.
  • Several isolates were grown, cells harvested, plasmid DNA isolated and the DNA sequence of the c-myc were determined. A comparison the sequence obtained empirically with the published sequence revealed that no PCR errors were apparent. An ATG was added by the PCR primers to ensure the translation of the c-myc peptide.
  • Transformation of Fungal Cells Several exemplary fungal cells that can be transformed to express vaccine proteins can include, but are not limited to, Saccharomyces spp., Aspergillus spp., Cryptococcus spp., Coccidioides spp., Neurospora spp., Histoplasma spp., Blastomyces spp., and a combination thereof.
  • Exemplary method of transformation of S. cerevisiae. After isolating each of the genes of interest and forming each yeast expression vector (as described), DNA vectors to individually transform yeast cells were used. The transformation protocol was as follows: 50 μL of YPD broth was inoculated with W303-1B Saccharomyces cerevisiae and the culture grown overnight at 30° C. with shaking. The desired OD600 for transformation was between 0.1-0.6 (˜2×107 c/mL). Cells were harvested by centrifugation at 2000×g for 10 minutes, the pellet was washed in 25 mL sterile water and cells were again centrifuged at 2000×g for 10 minutes. The pellet was resuspended in 1 mL 01M lithium acetate (LiAC), centrifuged at 1000×g for 30 seconds and the supernatant removed. This pellet was again resuspended in 400 μL 0.1M LiAC and the cell suspension mixed until the cells were completely suspended. Fifty milliliters of this cell suspension was used for each transformation. The cells were centrifuged at 3000×g for 30 seconds and the supernatant discarded. To the cells was added 240 μL 50% (w/v) PEG, 36 μL 1M LiAC, 25 μL denatured carrier DNA, and 50 μL of previously diluted water containing plasmid DNA (0.1-10 μg). Cells were mixed until the pellet completely resuspended, mixtures were incubated at 30° C. for 30 minutes, and heat shocked at 42° C. for 20-25 min. The cells were harvested by centrifugation at 3000×g for 30 seconds, gently resuspended in imL of water and spread onto Yeast Nitrogen Base medium (YNB) (e.g., Difco) minus uracil plates. The plates were incubated at 30° C. for 3-4 days.
  • Identification of yeast cells expressing candidate proteins In one example, a single yeast colony was obtained from YNB minus uracil plates and grown for overnight at 30° C. with shaking in 250 mL flasks containing 50 mL YNB minus uracil medium. Each culture was diluted to a final concentration of 0.2 (OD600) in 50 mL YNB minus uracil interim cultures. The cultures were grown at 30° C. until the OD600 had doubled (approximately 5-6 hours). The OD600 of the interim cultures was determined a final time before the cultures were diluted to a final concentration of 0.02 OD600 units in 50 mL of YNB minus uracil. These cultures were grown for 1 hour with shaking at 30° C. before 0.4 mM CuSO4 (final concentration) was added. As a control, to a duplicate culture, 0.4 mM NaSO4 was added to show that the protein is not produced in the absence of copper. Once the CuSO4 and NaSO4 were added, the cultures were grown overnight (18 hours) at 30° C. with shaking (FIG. 1).
  • Induced and uninduced cultures were harvested after overnight incubation by centrifugation at 550×g for 10 minutes. Each cell pellet was washed with 25 mL ice cold water to remove any residual medium and the cells were harvested again by centrifugation. Each pellet was then resuspended in 1 mL of ice cold water and divided into two 1.5 mL eppendorf tubes for centrifugation. After the removal of the supernatant, one tube was frozen at −80° C. for storage and the other was lysed for protein determination. Cells were lysed in the following manner: 100 μL of glass beads were added followed by the addition of 200 μL 2× sample lysis buffer (0.1M Tris, pH 6.8, 20% (v/v) glycerol, 4% (w/v) SDS, 0.5% (v/v) β-mercaptoethanol). Tubes were mixed by vortexing for 1 min to resuspend and lyse the cells and mixtures were boiled for 3 min to denature the proteins for SDS-PAGE gel electrophoresis. Debris was removed by centrifugation at 550×g for 5 min. and the cleared cell lysates were transferred to new tubes.
  • Cell lysates were loaded into the wells of 15% Tris-HCl SDS-PAGE gels (example: Bio Rad, Hercules, Calif.) in 15 μL aliquots and separated by electrophoresis at 60-80 V for 2-3 hours. Proteins were then transferred from the gel to a PVDF [poly(vinylidine fluoride)] membrane using a mini-genie transfer apparatus (Idea Scientific, Minneapolis, Minn.) for 1 hour at 12 volts.
  • PVDF membranes were blocked in TBST (100 mM Tris, 1.5 M NaCl and 0.1% [v/v] Tween 20) containing 5% (w/v) powdered milk (Carnation) for 1 hour at room temperature. Blocked membranes were then incubated with primary anti-c-myc epitope antibody (1:1000 dilution) in TBST containing 1% milk overnight at 4o C. The next morning the membranes were washed five times for 15 minutes each in TBST before secondary antibody was added. Secondary antibody conjugated to horse radish peroxidase (HRP) was added in TBST containing 1% (w/v) milk and incubated for 1 hour at room temperature. The membrane was washed five times for 5 minutes each in TBST before exposure to ECL reagents. The membrane was exposed to ECL (Pierce Chemical Company, Rockford, Ill.) for 1 minute then developed using an Alpha-Innotech (San Leandro, Calif.) digital system.
  • Determination of Optimal Growth Conditions of Yeast Expressing Candidate Proteins
  • Initial 50 mL cultures were started with 800 μL of each glycerol stock that was frozen at a concentration of 1×108 cells/mL. Cultures were grown overnight at 30° C. with shaking. The next morning the cultures were counted using a hemocytometer as well as for the OD600 determination using a SpectraMax 340 plate reader (Molecular Devices, Sunnyvale, Calif.). Each culture was then diluted to OD600 of 0.1, 0.2, or 0.3. Duplicate cultures were grown in 250 mL Erlenmeyer flasks (50 mL cultures) as well as in 300 mL Klett flasks (containing 60 mL of medium). Samples were taken every hour from the 250 mL Erlenmeyer flasks for cell counting and OD600 readings, while Klett readings were taken using a Klett-Summerson Photoelectric colorimeter (Klett-Summerson, NY). By using these methods, not only could the growth curve be characterized, but the OD600 could be correlated, cell counts, and Klett readings thereby allowing use of any of the three methods in future experiments. The following correlation was determined; 50 Klett units=0.25 OD600 units=2×107 c/mL. From these data, the doubling time of the culture was determined to be approximately 5 hours. An exemplary growth curve for one such transformant is shown in FIG. 2.
  • Once the growth curves at each of the three initial inoculation concentrations were determined, the new variable of various CuSO4 concentrations was added. Copper sulfate was tested at the following concentrations: 0, 0.2, 0.3, and 0.4 mM. The protocol to induce pYEX-BX suggested a CuSO4 range of 0.2-0.5 mM and it had previously been determined that 0.5 mM was potentially toxic to the yeast. As in the experiment above, duplicate cultures were inoculated in 250 mL Erlenmeyer flasks and 300 mL Klett flasks. Samples were taken every hour from the 250 mL Erlenmyer flasks for cell counting and OD600 readings, while Klett readings were taken from the Klett flasks. The lower inoculum grew the best upon the addition of CuSO4. The correlation between Klett units, OD600 units, and cell counts was the same as determined previously. The doubling time of the each culture was again 5 hours and was independent of copper concentration. The growth curve with copper for one such transformant at each innoculumis shown in FIG. 1.
  • Preparation of Vaccine
  • In one exemplary method, overnight cultures were started by adding 1 mL of a glycerol stock of yeast cells to 50 mL of YNB minus uracil medium in a 250 mL flask. Cultures were grown overnight at 30° C. with shaking. The next morning the OD600 was determined and a 50 mL interim culture was inoculated at a final OD600 of 0.2 and grown at 30o C with shaking until it had doubled (4-5 hours). The optical density was subsequently determined for the interim culture which was used to inoculate a final 250 mL culture at 0.02 (final OD600) and grown for 1 hour at 30° C. with shaking. After the hour of growth, 0.4 mM CuSO4 (final concentration) was added to induce the production of each candidate protein. This induced culture was grown for 16 to 18 hours at 30° C. with shaking. Because the doubling time for copper-induced cells was approximately 5 hours, these induced cultures were in mid to late log after about 18 hours of growth.
  • Heat-Killed Preparations
  • In one example, cells from cultures were harvested by centrifugation at 1900 g for 5 min at 4o C. Cultures were washed 4 times with 100 mL room temperature PBS (137 mM NaCl, 2.7 mM KCl, 10 mM Na2HPO4, 2 mM KH2PO4) to remove any residual medium and copper from the cells. Cells were resuspended in 3 mL room temperature PBS and then added to 200 mL PBS that had been pre-warmed to 56° C. The cells were subsequently heat killed by incubation at 56° C. in a water bath for 1 hour, with inversion of tubes every 10 min. to ensure that all cells were heat killed. Cells were harvested by centrifugation (1900×g for 5 min at 4o C) and then washed 4 times with 100 mL with room temperature PBS. Cells were counted and resuspended at a final concentration of 4×108 c/mL in PBS, aliquoted in 200 mL aliquots and stored at 4° C.
  • Yeast cells can be heat-killed by incubation in sterile, pyrogen-free saline at 56° C. for 1 hr, washed in saline, resuspended at ˜2×108 cells/mL in sterile saline, and stored at 4° C. Each batch is expected to yield ˜2×1010 yeast cells, of which a vaccine dose for the animal studies will consist of ˜1×107 to 20×107 cells.
  • Test for Live Cells in Heat-Killed Preparations
  • In one method, to ensure that the heat treatment has inactivated >99.99% of the yeast cells, 1×107 heat-killed cells were incubated on agar-solidified rich medium for three days at 30° C. and the number of resulting yeast colonies (if any) determined. Only batches of cells with survival rates of <0.01% are to be used. To confirm that no live microorganisms, e.g., bacteria, were present, fluid thioglycolate medium (as described in the USP 24 NF19) may be inoculated with each batch of heat-killed yeast cells. The test mixtures were incubated for 14 days at 32° C. and examined visually for growth. Only batches of heat-killed cells in which minimal to no microbial growth was observed were used.
  • Endotoxin Levels of Each Batch of Vaccine
  • Bacterial cell-wall fragments and other pyrogens can often obscure immune responses. Pyrogen-free containers, liquids, and media were used to minimize spurious endotoxin contamination. Recently, the FDA has allowed use of a test for bacterial endotoxin levels to substitute for rabbit pyrogenicity tests. The endotoxin levels of each vaccine formulation were quantitated using a G test (Fungitec, Seikagaku Corp., Tokyo, Japan) under the U.S. Pharmacopeia (USP) guidelines. This test relies on the factor G component of horseshoe crab amoebocyte lysate. Unfortunately, this component is sensitive to (1,3)β-glucan while factors B and C are sensitive to endotoxin. Since it is expected that a small amount of fungal cell-wall glucan is present in heat-killed yeast preparations and these may lead to false positives, all samples prior to test were treated with (1,3)β-glucanase, which will destroy any (1,3)β-glucan. This procedure thus will provide an accurate determination of the endotoxin levels present. Only batches of heat-killed cells that contain <0.5 EU/mL were used.
  • Example 1
  • In certain exemplary methods, the fungal organism Aspergillus was examined.
  • Efficacy of four A. fumigatus antigens protecting mice from systemic aspergillosis In one exemplary method, the protective efficacy of 4 A. fumigatus antigens expressed in Saccharomyces sp. in an experimental model of systemic murine aspergillosis was tested. Efficacy was evaluated in terms of survival and tissue burden. See FIG. 3 as one exemplary strategy for vaccine generation and animal testing.
  • Antigens In this example, vaccines preparations containing 6×107 heat-killed yeast cells/150 microliters were administered s.c. using two injection sites (0.075 ml each) on days 28, 21 and 14 before infection. Groups pretreated with Myc (yeast containing and expressing only the c-myc tag) or PBS were used as controls.
  • Animals 60 CD-1 male mice weighting 30 g approx. were used.
  • Inoculum A total of 1.3×107 conidia of A. fumigatus, strain AF-10, were inoculated via lateral tail vein into the CD-1 mice.
  • In this exemplary method, 16 days after infection, animals were euthanatized by CO2 anoxia and kidney and brain were removed. Each organ was homogenized, diluted and plated on SDA plates for CFU determination.
  • Survival
  • Comparisons showed that animals immunized with Hemolysin (Hemo or HEMO) antigen had an increased survival time in comparison with the PBS control group (p=0.012). In addition, the MYC control treated group did have an increased survival time in comparison with the PBS control group although this did not reach statistical significance (p=0.089) (FIG. 4).
  • In another example, the protective effect of the A. fumigatus antigen (HEMO) expressed in Saccharomyces sp. in a systemic murine model of aspergillosis was evaluated. The animal model showed low mortality in the control groups i.e., PBS and MYC, and HEMO pretreatment did not significantly prolonged the survival of the animals. However, the tissue burden study demonstrated that animals receiving three doses of the HEMO antigen before the infection had significantly lower CFU of the fungus in the kidneys and brain in comparison with the PBS-treated control animals (FIG. 12). These results demonstrate that HEMO pretreatment provides protective resistance against systemic aspergillosis.
  • Exemplary Cloning of AspF 2. Published DNA and predicted amino acid sequences to design PCR primers to amplify the ORF from the Aspergillus fumigatus cDNA library were used. The DNA and amino acid sequences of AspF 2 are presented in diagram form in SEQ ID NOs:3 and 4. The positions of the 5′ and 3′ primers use to obtain the predicted PCR product are also shown in SEQ ID NOs:3 and 4.
    atg ctt gtg gcc acc ctc cct acc tcc ccc gtc ccc atc gcg gcg cga  48
    Met Leu Val Ala Thr Leu Pro Thr Ser Pro Val Pro Ile Ala Ala Arg
    1               5                   10                  15
    gca acc ccc cac gaa ccc gtc ttc ttc tcc tgg gac gct ggc gcg gtg  96
    Ala Thr Pro His Glu Pro Val Phe Phe Ser Trp Asp Ala Gly Ala Val
                20                  25                  30
    acc tcg ttc ccc atc cac tcc agc tgc aat gcg acc cag cgc cgg cag 144
    Thr Ser Phe Pro Ile His Ser Ser Cys Asn Ala Thr Gln Arg Arg Gln
            35                  40                  45
    atc gag gcc ggc ctg aac gag gcg gtc gag ctc gcc cgg cac gcc aag 192
    Ile Glu Ala Gly Leu Asn Glu Ala Val Glu Leu Ala Arg His Ala Lys
        50                  55                  60
    gcc cac atc ctc cgc tgg ggc aac gag agc gag atc tac cgg aag tac 240
    Ala His Ile Leu Arg Trp Gly Asn Glu Ser Glu Ile Tyr Arg Lys Tyr
    65                  70                  75                  80
    ttt ggc aac cgg ccc acc atg gag gcc gtc ggt gcc tac gat gtc atc 288
    Phe Gly Asn Arg Pro Thr Met Glu Ala Val Gly Ala Tyr Asp Val Ile
                    85                  90                  95
    gtg aac ggg gac aag gcc aac gtg ctc ttc cgg tgt gac aac ccc gac 336
    Val Asn Gly Asp Lys Ala Asn Val Leu Phe Arg Cys Asp Asn Pro Asp
                100                 105                 110
    ggc aac tgt gct ttg gaa ggc tgg ggc ggc cac tgg cgc ggc gcg aac 384
    Gly Asn Cys Ala Leu Glu Gly Trp Gly Gly His Trp Arg Gly Ala Asn
            115                 120                 125
    gcc acc tcc gaa acc gtc atc tgt gat cgc agc tac acc acc cgc cgc 432
    Ala Thr Ser Glu Thr Val Ile Cys Asp Arg Ser Tyr Thr Thr Arg Arg
        130                 135                 140
    tgg ctt gtc tcc atg tgc tcc cag ggc tac acc gtc gcc ggc tcc gag 480
    Trp Leu Val Ser Met Cys Ser Gln Gly Tyr Thr Val Ala Gly Ser Glu
    145                 150                 155                 160
    acc aac acc ttc tgg gct tcg gac ctg atg cac cgt ctg tac cat gtg 528
    Thr Asn Thr Phe Trp Ala Ser Asp Leu Met His Arg Leu Tyr His Val
                    165                 170                 175
    cct gct gtg ggt caa ggc cgg gtc gac cac ttc gcc gac ggc tac gac 576
    Pro Ala Val Gly Gln Gly Arg Val Asp His Phe Ala Asp Gly Tyr Asp
                180                 185                 190
    gag gtg att gcc ctg gcc aag agc aac ggc acc gag tcc acg cat gac 624
    Glu Val Ile Ala Leu Ala Lys Ser Asn Gly Thr Glu Ser Thr His Asp
            195                 200                 205
    tcg gag gcg ttg cag tat ttc gcc ctt gag gcg tat gcg ttt gat att 672
    Ser Glu Ala Leu Gln Tyr Phe Ala Leu Glu Ala Tyr Ala Phe Asp Ile
        210                 215                 220
    gcc gct ccc ggt gtc gga tgt gct ggc gag agt cac ggc cct gac cag 720
    Ala Ala Pro Gly Val Gly Cys Ala Gly Glu Ser His Gly Pro Asp Gln
    225                 230                 235                 240
    gga cat gac acc ggg tct gcc tcg gcg cct gcg tct acc tcc acc tct 768
    Gly His Asp Thr Gly Ser Ala Ser Ala Pro Ala Ser Thr Ser Thr Ser
                    245                 250                 255
    agc tcc agc tcg ggc tcg ggc tcg ggc gcc acg act acc ccg acg gat 816
    Ser Ser Ser Ser Gly Ser Gly Ser Gly Ala Thr Thr Thr Pro Thr Asp
                260                 265                 270
    tct ccc agt gcc act att gat gtg ccg tcg aac tgc cat acc cat gaa 864
    Ser Pro Ser Ala Thr Ile Asp Val Pro Ser Asn Cys His Thr His Glu
            275                 280                 285
    ggt gga cag ctt cat tgc act 885
    Gly Gly Gln Leu His Cys Thr
        290                 295
  • A 16 amino acid signal sequence was predicted by PSORTII (http://psort.nibb.ac.jp/cgi-bin). The signal sequence was subsequently removed as PSORTII predicted that the protein would be extracellular, which suggested that the protein would be secreted by the S. cerevisiae if the signal sequence was left intact. A methionine was added before the 17th amino acid to ensure that the protein would be translated.
  • The following conditions were used to amplify AspF 2 from an Aspergillus fumigatus cDNA library: 25 μL reactions were performed in PCR Ready Bead tubes (Amersham Pharmacia) containing 500 ng of cDNA library, 2 μM primers, and 13 μL sterile water. Reactions were performed using the Perkin Elmer 2400 Thermocycler (PE Applied Biosystems) under the following conditions: an initial cycle at 94° C. for 5 min, 30 cycles at 94° C. for 1 min, 50° C. for 1 min and 72° C. for 1 min, and a final extension at 72° C. for 7 min.
  • The PCR reactions were separated using agarose gel electrophoresis and a photograph of a representative gel data not shown. A band of approximately 936 bases corresponding to the amplified AspF 2 ORF contains a 3′ c-myc tag.
  • The band was excised from the gel, purified and ligated into pCR 2.1. The ligated vector was used to transform competent E. coli cells and vector DNA isolated from 5 mL mini cultures using standard alkaline lysis techniques. DNA isolated from several miniprep isolates was digested with EcoRI and separated by agarose gel electrophoresis to confirm the presence of the 936 base AspF 2/c-myc insert.
  • Several isolates were grown, cells harvested, plasmid DNA isolated and the DNA sequence of the AspF 2 insert determined. A comparison of the sequence obtained empirically with the published sequence was determined (SEQ ID NOs: 5 and 6). There were no apparent PCR errors.
    SEQ ID NO: 5 - AspF2
    accaacacct tctgggcttc ggacctgatg caccgtctgt accatgtgcc tgctgtgggt  60
    caaggccggg tcgaccactt cgccgacggc tacgacgagg tgattgccct ggccaagagc 120
    aacggcaccg agtccacgca tgactcggag gcgttgcagt atttcgccct tgaggcgtat 180
    gcgtttgata ttgccgctcc cggtgtcgga tgtgctggcg agagtcacgg ccctgaccag 240
    ggacatgaca ccgggtctgc ctcggcgcct gcgtctacct ccacctctag ctccagctcg 300
    ggctcgggct cgggcgccac gactaccccg acggattctc ccagtgccac tattgatgtg 360
    ccgtcgaact gccataccca tgaaggtgga cagcttcatt gcactgaaca gaagttgatt 420
    tccgaagaag acctcgag 438
    SEQ ID NO: 6 - AspF2 PCR
    accaacacct tctgggcttc ggacctgatg caccgtctgt accatgtgcc tgctgtgggt  60
    caaggccggg tcgaccactt cgccgacggc tacgacgagg tgattgccct ggccaagagc 120
    aacggcaccg agtccacgca tgactcggag gcgttgcagt atttcgccct tgaggcgtat 180
    gcgtttgata ttgccgctcc cggtgtcgga tgtgctggcg agagtcacgg ccctgaccag 240
    ggacatgaca ccgggtctgc ctcggcgcct gcgtctacct ccacctctag ctccagctcg 300
    ggctcgggct cgggcgccac gactaccccg acggattctc ccagtgccac tattgatgtg 360
    ccgtcgaact gccataccca tgaaggtgga cagcttcatt gcactgaaca gaagttgatt 420
    tccgaagaag acctcgag 438
  • The DNA of an isolate was excised from the pCR 2.1 vector (Invitrogen) with a BamHI/EcoRI double digest and the reactions were separated by electrophoresis so that the DNA band encoding the AspF 2 ORF could be isolated. This DNA fragment was then ligated into the pYEX-BX vector using the same restriction enzymes. The ligated vector was then used to transform competent E. coli and the plasmid DNA isolated by standard alkaline lysis mini-preps. The DNA sequence of salient portions of the vector was determined to ensure the correct alignment of the ORF with the TATA box. Then portions of the vector were sequenced data not shown. The TATA box, the aspf-2 coding sequence, the c-myc tag sequence, and the stop codons were identified (SEQ ID NOs: 1 and 2).
  • Exemplary cloning method for Hemolysin Published DNA and predicted amino acid sequences of Hemolysin to design PCR primers to amply the ORF from the A. fumigatus cDNA library were used. The DNA and amino acid sequences are presented in diagram form in SEQ ID NOs:7 and 8, along with the positions of the 5′ and 3′ primers used to obtain the predicted PCR product are shown. No signal sequence was predicted by PSORTII (http://psort.nibb.ac.jp/cgi-bin).
    SEQ ID NOs: 7 and 8 - nucleic acid and corresponding amino acid sequence of Hemolysin
    Figure US20070172503A1-20070726-C00001
  • The following conditions were used to amplify hemolysin from the cDNA library: 25 μL reactions were performed in PCR Ready Bead tubes containing 500 ng of cDNA library, 2 μM primers, and 13 μL sterile water. Reactions were performed using a Perkin Elmer 2400 Thermocycler under the following conditions: an initial cycle at 94° C. for 5 min, 30 cycles at 94° C. for 1 min, 50° C. for 1 min, 72° C. for 1 min, and a final extension at 72° C. for 7 min.
  • The PCR reactions were separated using agarose gel electrophoresis and a band of approximately 444 bp corresponding to the amplified hemolysin ORF was excised from the gel, purified and ligated into pCR 2.1. The ligated vector was used to transform competent E. coli cells and vector DNA was subsequently isolated using standard techniques. DNA isolated from several isolates was used as template for PCR reactions to confirm the presence of the hemolysin insert. Not all isolates contained the hemolysin insert due to self ligation of the pCR 2.1 vector. See FIG. 5.
  • Several isolates were grown, cells harvested, plasmid DNA isolated and the DNA sequence of the hemolysin were determined. A comparison the sequence obtained empirically with the published sequence revealed that no PCR errors were apparent, the sequence as shown in SEQ ID NOs:9 and 10.
    SEQ ID NO: 9 - Hemo
    Figure US20070172503A1-20070726-C00002
    SEQ ID NO: 10 - Hemo PCR
    Figure US20070172503A1-20070726-C00003
  • The DNA of one isolate was treated with BamH I/EcoRI and the DNA band encoding the hemolysin ORF/c-myc tag was isolated. This DNA fragment was then ligated into vector pYEX-BX that had also been digested with BamH I/EcoRI. The ligated vector was then used to transform competent E. coli and the plasmid DNA was subsequently isolated. The DNA sequence of the relevant portions of the vector was determined to ensure the correct alignment of the ORF with the TATA box and the c-myc tag was in frame with the rest of the ORF. The sequenced portion of the vector is presented in SEQ ID NO:11. In addition, this vector was used for Examples 2 and 3.
    SEQ IN NO: 11 - Portions of pYEX-BX that were sequenced
    Figure US20070172503A1-20070726-C00004
  • In one exemplary method an empty vector control was generated as previously described in Methods (This template was amplified by PCR as discussed previously. DNA isolated from several isolates was used as template for PCR reactions to confirm the presence of the c-myc insert. (SEQ ID NOs:1 and 2) The DNA of one isolate was treated with BamH I/EcoRI and the DNA band encoding the c-myc tag was isolated. This DNA fragment was then ligated into pYEX-BX which had also been digested with BamHI and EcoRI. The ligated vector was used to transform competent E. coli cells and vector DNA isolated using standard techniques. DNA isolated from several miniprep isolates was used as template for PCR reactions to confirm the presence of the c-myc control insert. (SEQ ID NOs:1 and 2)
  • Several isolates were grown, cells harvested, plasmid DNA isolated and the DNA sequence of the c-myc control insert were determined. The DNA sequence of pertinent portions of the vector was determined to ensure the correct alignment of the c-myc control with the TATA box.
  • In one exemplary method, transformants expressing each A. fumigatus protein were identified and the cleared cell lysates were generated and stored as previously discussed.
  • After gel separation, expression of A. fumigatus proteins was determined by western blot by methods known in the art. In one exemplary method growth of transformants were determined. Transformants that were determined to produce a protein of the correct molecular weight above were grown in YNB minus uracil for 2-3 days at 30° C. with shaking. Once the cultures were in stationary phase, the cells were counted and frozen in 50% glycerol at a final concentration of 1×108 c/mL (10 yeast units) and stored at −80° C. until use.
  • The production of vaccines for animal models was used as previously disclosed. Each of four A. fumigatus genes from a cDNA library using gene-specific PCR primers were cloned. The DNA encoding each ORF was sequenced to confirm that the correct gene had been cloned followed by an inframe c-myc tag and that no PCR errors were introduced. The ORF of each gene was inserted into the copper inducible vector pYEX-BX. Portions of each ligated vector were sequenced to ensure the correct position of the gene relative to the TATA box.
  • Each pYEX-BX vector containing a gene of interest was used to transform yeast and yeast transformants expressing each of the four proteins identified by Western blot analysis. Each S. cerevisiae strain that produced the correct protein was subsequently produced and used to vaccinate an animal as discussed above.
  • Aspergillus. In another exemplary method, the protective efficacy of HEMO antigen expressed in Saccharomyces spp. against systemic aspergillosis was tested in mice.
  • Antigen. HEMO antigen was administered s.c. using two injection sites dorsally (0.075 ml each) on days 28, 21 and 14 before infection, receiving 6×107 yeast cells per animal. Control animals received PBS (diluent control) or MYC (yeast not expressing immunogen control) at 6×107 yeast cells per animal.
  • Animals. Thirty six weeks-old male CD-1 mice were used. Animals were housed in standard conditions in cages of 5 animals. Groups of 10 mice per group were established.
  • Infection. A total of 5.8×106 viable Aspergillus fumigatus conidia per mouse were inoculated intravenously via lateral tail vein.
  • Tissue burden. 16 days after infection surviving animals were euthanatized by CO2 anoxia. The kidney and brain were removed aseptically, homogenized in 0.9% saline, diluted and plated on SDA plates for CFU determination.
  • Statistics. Comparisons of survival were done by a log rank test and tissue burden by Mann Whitney test using GraphPad 3.03 for Windows.
  • FIG. 12 represents an exemplary Log 10 CFU per studied organ. Pretreatment with HEMO antigen significantly reduced tissue burdens from brain and kidney in comparison to the diluent control (p=0.003 and 0.023, respectively). In addition the yeast control (MYC) reduced organs burdens as well but did not reach statistical significance (p=0.089 in both organs). No differences were found between MYC and PBS groups. Importantly, 50 and 60% of the animals receiving MYC or HEMO, respectively, did not show detectable amount of A. fumigatus in brain but all PBS-pretreated animals showed fungal grown in this organ.
  • The protective effect of the A. fumigatus antigen (HEMO) expressed in Saccharomyces was tested in a systemic murine model of aspergillosis. The MYC only yeast control group did show a reduction in the brain organ burden. Importantly, the tissue burden study demonstrated that animals receiving three doses of the HEMO antigen before the infection had statistically significantly lower CFU of the fungus in the kidneys and brain in comparison with the PBS-treated control animals. These results show that the vaccine containing either yeast alone (MYC) or Hemo protected mice against an infection of Aspergillus fumigatus.
  • Example 2
  • In certain exemplary methods, the fungal organism Coccidioides was examined.
  • See one exemplary schematic for production of and testing for an anti-fungal vaccination in a Coccidioides animal model (FIG. 6). In one exemplary method, the feasibility of using heat-killed yeast cells expressing a C. immitis antigen as a vaccine against C. immitis infections was examined. cDNA was obtained for each the C. immitis gene whose protein has been shown in other work to elicit an immune response in infected animals. Then yeast were transformed with each cDNA and strains selected that expressed a high levels of the antigen. A schematic of the strategy is depicted in FIG. 6.
  • In another exemplary method, the protective effect of Coccidioides immitis antigen expressed by Saccharomyces cerevisiae was analyzed and used as a killed whole vaccine preparation in a systemic murine model of coccidioidomycosis. DNA encoding for Ag2 (also called PRA) was cloned into S. cerevisiae and gene expression induced using for example, copper. These yeast cells were heat killed and used to vaccinate mice prior to infection with Coccidioides immitis.
  • Animals. Six-week-old male CD-1 mice were purchased from Charles River Laboratories. Groups consisted on 20 mice, housed in standard conditions. After infection, mice were housed in micro-isolator cages. Animals were provided food and acidified water ad libitum.
  • Antigen. In this exemplary example, a C. immitis antigen expressed in Saccharomyces was used. This was a protein present in the alkali-soluble and water-soluble preparation of the spherule wall and called antigen 2 (Ag2/PRA). The vaccine was administered subcutaneously (s.c.) in a volume of 50 μl three times before infection (i.e., 21, 14 and 7 days prior to the infection) at a concentration of 4×108 cells/ml. In this example, PBS was used as the control.
  • Inoculum. Coccidioides immitis (Silveira strain) was used. The strain was grown on glucose yeast-extract (GYE) plates in a Class 3 biosafety cabinet at room temperature for 2 weeks. Arthroconidia were harvested as a suspension in saline. The number of arthroconidia was determined by hemacytometer and verified for viability by cfu (colony forming units) determination done by plating inoculum dilutions on GYE medium. Animals were infected intravenously (i.v.) with a suspension containing 376 viable arthroconidia per mouse. Statistical analysis of survival was done using a log rank test.
  • FIG. 7 illustrates an exemplary the survival curve of the groups of mice included in the study that was vaccinated with yeast expressing the antigen-2/PRA protein. Deaths of control animals and pretreated animals began on day 13 after infection and by day 21 only 10% of the PBS treated mice were still alive. This was in contrast to the mice treated with the Ag-2 containing yeast (25% still alive) Statistical comparisons between groups showed a significant difference between the control group and Ag2 immunized animals (p=0.04). This example shows that mice vaccinated with yeast cells expressing the antigen-2/PRA protein survived to a greater extent than untreated mice.
  • FIG. 8 illustrates an exemplary a Western Blot of extracts of yeasts containing and expressing Hemolysin (hemo), aspf 2, or antigen 2 (the Coccidioides protein described above). The numbers indicate the amount of lysate separated by SDS-GEL electrophoresis. The blots were treated with a c-myc epitope specific antibody and the antibody detected using a secondary horse radish peroxidase antibody. Various amounts in micrograms (indicated by the numbers at the bottom of the figure) of c-myc peptide were spotted.
  • Antigen 2/proline-rich antigen has been previously shown to be protectivein murine models of coccidiomycosis. Thus antigen 2 was chosen. Published DNA and predicted amino acid sequences were used to design PCR primers to amplify the ORF from the Cox cDNA library (SEQ ID NOs:12 and 13). The DNA and amino acid sequences of antigen 2 are presented in diagram form, data not shown. An 18 amino acid signal sequence was predicted by PSORTII (http://psort.nibb.ac.jp/cgi-bin), but was not removed as PSORTII also predicted that the protein would be cell wall bound, most likely GPI anchored. This prediction suggests that the protein would not be secreted by the S. cerevisiae.
    SEQ ID NOs: 12 and 13
    Figure US20070172503A1-20070726-C00005
    Figure US20070172503A1-20070726-C00006
    Figure US20070172503A1-20070726-C00007
    Figure US20070172503A1-20070726-C00008
    Figure US20070172503A1-20070726-C00009
    Figure US20070172503A1-20070726-C00010
    Figure US20070172503A1-20070726-C00011
    Figure US20070172503A1-20070726-C00012
    Figure US20070172503A1-20070726-C00013
    Figure US20070172503A1-20070726-C00014
    Figure US20070172503A1-20070726-C00015
    Figure US20070172503A1-20070726-C00016
    Figure US20070172503A1-20070726-C00017
  • Exemplary conditions used to amplify antigen 2 from the cDNA library were as follows: 25 μL reactions were performed in PCR Ready Bead tubes (Amersham Pharmacia) containing 500 ng of cDNA library, 2 μM primers, and 13 μL sterile water. Reactions were performed using the Perkin Elmer 2400 Thermocycler (PE Applied Biosystems) under the following conditions: an initial cycle at 94° C. for 5 min, 30 cycles at 94° C. for 1 min, 50° C. for 1 min and 72° C. for 1 min, and a final extension at 72° C. for 7 min. As previously discussed, PCR reactions were separated and a band of approximately 582 bases corresponding to the amplified antigen 2 ORF was excised. The band was excised from the gel, purified and ligated into pCR 2.1. The ligated vector was used to transform competent E. coli cells and vector DNA isolated from 5 mL mini cultures using standard alkaline lysis techniques. DNA isolated from several miniprep isolates was digested with EcoRI to confirm the presence of the 582 base antigen 2 insert. Only one isolate contained the antigen 2 insert.
  • Several isolates were grown, cells harvested, plasmid DNA isolated and the DNA sequence of the antigen 2 insert determined. A comparison of the sequence obtained empirically with the published sequence is shown in SEQ ID NOs:14 and 15. There were no apparent PCR errors.
    SEQ ID NO:14 Antigen-2
    ATGCAGTTCTCTCACGCTCTCATCGCTCTCGTCGCTGCCGGCCTCGCCAG
    TGCCCAGCTCCCAGACATCCCACCTTGCGCTCTCAACTGCTTCGTTGAGG
    CTCTCGGCAACGATGGCTGCACTCGCTTGACCGACTTCAAGTGCCACTGC
    TCCAAGCCTGAGCTCCCAGGACAGATCACTCCTTGCGTTGAGGAGGCCTG
    CCCTCTCGACGCCCGTATCTCCGTCTCCAACATCGTCGTTGACCAGTGCT
    CCAAGGCCGGTGTCCCAATTGACATCCCACCAGTTGACACCACCGCCGCT
    CCCGAGCCATCCGAGACCGCTGAGCCCACCGCTGAGCCAACCGAGGAGCC
    CACTGCCGAGCCTACCGCTGAGCCCACCGCTGAGCCGACTCATGAGCCCA
    CCGAGGAGCCCACTGCCGTCCCAACCGGCACTGGCGGTGGTGTCCCCACT
    GGCACCGGTTCCTTCACCGTCACTGGCAGACCAACTGCCTCCACCCCAGC
    TGAGTTCCCAGGTGCTGGCTCCAACGTCCGTGCCAGCGTTGGCGGCATTG
    CTGCTGCTCTCCTCGGTCTCGCTGCCTACCTG
    SEQ ID NO:15: Antigen-2 PCR
    ATGCAGTTCTCTCACGCTCTCATCGCTCTCGTCGCTGCCGGCCTCGCCAG
    TGCCCAGCTCCCAGACATCCCACCTTGCGCTCTCAACTGCTTCGTTGAGG
    CTCTCGGCAACGATGGCTGCACTCGCTTGACCGACTTCAAGTGCCACTGC
    TCCAAGCCTGAGCTCCCAGGACAGATCACTCCTTGCGTTGAGGAGGCCTG
    CCCTCTCGACGCCCGTATCTCCGTCTCCAACATCGTCGTTGACCAGTGCT
    CCAAGGCCGGTGTCCCAATTGACATCCCACCAGTTGACACCACCGCCGCT
    CCCGAGCCATCCGAGACCGCTGAGCCCACCGCTGAGCCAACCGAGGAGCC
    CACTGCCGAGCCTACCGCTGAGCCCACCGCTGAGCCGACTCATGAGCCCA
    CCGAGGAGCCCACTGCCGTCCCAACCGGCACTGGCGGTGGTGTCCCCACT
    GGCACCGGTTCCTTCACCGTCACTGGCAGACCAACTGCCTCCACCCCAGC
    TGAGTTCCCAGGTGCTGGCTCCAACGTCCGTGCCAGCGTTGGCGGCATTG
    CTGCTGCTCTCCTCGGTCTCGCTGCCTACCTG
  • The DNA of the isolate was excised from the pCR 2.1 vector and the salient portions of the vector was determined to ensure the correct alignment of the ORF with the TATA box and the c-myc tag, as described previously. Published DNA and predicted amino acid sequences were used to design PCR primers to amply the ORF from the cDNA library for antigen 2.
  • In one exemplary method an empty vector control for animal models was designed as previously indicated; that is, the pYEX-BX vector containing the c-myc tag but lacking a C. immitis gene of interest. Several isolates were grown, cells harvested, plasmid DNA isolated and the DNA sequence of the c-myc control insert were determined. The DNA sequence of pertinent portions of the vector was determined to ensure the correct alignment of the c-myc with the TATA box.
  • Transformation of S. cerevisiae was performed as previously described. After obtaining at least 10 transformants for each C. immitis gene of interest, cells are grown and induced the expression of each gene by adding copper sulfate. Uninduced and induced cells were harvested by centrifugation, lysed in SDS-gel lysis buffer and the protein concentration of each extract determined. Equal protein amounts of each lysate were separated by SDS-PAGE and the separated proteins transferred to membranes. The presence of the c-myc tagged proteins was detected by Western blot analysis using an anti-c-myc antibody. Then the transformants were grown as previously described. Once transformants of each construct were isolated, the proteins were expressed via copper induction as previously discussed. Then cultures were grown overnight at 30° C. with shaking. These cultures were used for the identification of transformants that were expressing each of the C. immitis proteins.
  • Identification of transformants expressing each C. immitis protein. Induced and uninduced cultures were harvested after overnight incubation by centrifugation at 550×g for 10 minutes. Each cell pellet was washed with 25 mL ice cold water to remove any residual medium and the cells were harvested again by centrifugation. Each pellet was resuspended in 1 mL of ice cold water and divided into two 1.5 mL eppendorf tubes for centrifugation. After the removal of the supernatant, one tube was frozen at −80° C. for storage and the other was lysed for protein determination. Cells were lysed in the following manner: 100 μL of glass beads were added to the tube containing cells followed by the addition of 200 μL 2× sample lysis buffer (0.1M Tris, pH 6.8, 20% (v/v) glycerol, 4% (w/v) SDS, 6.2 mg DTT). Tubes were mixed by vortexing for 1 min and boiled for 3 min to resuspend and lyse the cells. Debris was removed by centrifugation and the cleared supernatant was transferred to a new tube. The cleared lysate was used for western blots.
  • Cell lysates were loaded into the wells of SDS page gels and expression of the C. immits proteins were determined by exemplary western blot procedures known in the art. Each of three C. immitis genes from a cDNA library using gene-specific PCR primers was obtained. The DNA encoding each ORF was sequenced to confirm that the correct gene had been cloned and that no PCR errors were introduced. The ORF of each gene was inserted into the copper inducible vector pYEX-BX. Each ligated vector was sequenced to ensure the correct position of the gene relative to the TATA box and that the c-myc tag was in frame with each protein.
  • Example 3
  • In certain exemplary methods, the fungal organism Cryptococcosis was analyzed (see exemplary schematic FIG. 9)
  • Cryptococcosis model. In one exemplary method, an inhalation model of cryptococcosis was chosen because 1) it most closely mimics the natural course of C. neoformans infection, 2) it is highly reproducible and 3) it is commonly used to assess virulence. The inhalation cryptococcosis model performed was essentially as previously described (see: Cox, G. M., Mukherjee, J., Cole, G. T., et al Urease as a virulence factor in experimental cryptococcosis. Infect Immun. (2000) 68:443-448. incorporated herein by reference).
  • To test the in vivo efficacy of each vaccine formulation to protect vaccinated animals against a challenge of C. neoformans, mice are vaccinated with various vaccine formulations, and challenged with C. neoformans, and monitored for protection.
  • Organism. C. neoformans H 99 was used as the challenge organism for all infections. C. neoformans H99 yeast for the inoculum was prepared as follows. Briefly, C. neoformans strain H99 was grown at 30° C. with shaking for two days in liquid YPD medium. The cells were harvested by centrifugation, washed in endotoxin-free PBS, and resuspended in endotoxin-free PBS. The cells were counted on a hemocytometer and diluted to 1×106 cells per ml. The inoculum was serially diluted and plated onto YPD to confirm the number of cells that are inoculated.
  • Mice. Four-week-old female A/J Cr mice were obtained from Charles River Laboratories for the inhalation model of cryptococcosis. Mice are commonly used for virulence assays with the serotype A strain, H99, and have been used in previous assessments of antibody protection against C. neoformans.
  • Briefly, mice from a specific pathogen-free colony were vaccinated with 150 μL of each yeast vaccine preparation on day 21, 14 and 7 before being challenged with 50 uL containing 5×104H99 C. neoformans cells.
  • In one exemplary method, experiments were performed to determine whether yeast strains expressing C. neoformans proteins could be used as a vaccine against C. neoformans infections. cDNA of certain genes whose proteins are likely to be important in infection were generated. Then yeast cells were transformed with each cDNA and select strains that express high levels of each putative antigen. Finally, each vaccine formulation was tested for its ability to protect vaccinated mice against a challenge of C. neoformans. FIG. 9 represents a schematic of these experiments.
  • DNA vector construction and Yeast Transformation were performed in this example as previously described. Primers are listed in Table 4 (SEQ ID NOs: 30-41). Then the cloned genes were used as template and amplified each gene using the gene-specific PCR primers. The DNA sequence of each ligated pYEX-BX vector containing a gene of interest was determined to ensure that the correct gene was cloned, the start codon was on the same DNA strand as the TATA box (initiation of transcription), and that the c-myc tag was in frame with each gene followed by two stop codons (TGA TAA).
  • Cloning of CDA: Published DNA was used and predicted amino acid sequences to design PCR primers to amplify the ORF from the clone, as described above. The DNA and amino acid sequences of CDA are presented in diagram form in SEQ ID NOs: 16-17. The positions of the 5′ and 3′ primers use to obtain the predicted PCR product are shown. A 19 amino acid signal sequence was predicted by PSORTII (http://psort.nibb.ac.jp/cgi-bin). The signal sequence was subsequently removed as PSORTII predicted that the protein would be extra-cellular, which suggested that the protein would be secreted by the S. cerevisiae if the signal sequence was left intact. A methionine was added before the 20th amino acid (Serine) to ensure that the protein would be translated.
    SEQ ID NOs: 16 and 17 - DNA and corresponding amino acid
    sequence CDA
    atg aag ttc atc aca agc ctc ttt gcc gtt ctt gcc att ctc tca agt  48
    Met Lys Phe Ile Thr Ser Leu Phe Ala Val Leu Ala Ile Leu Ser Ser
    1```````````````5```````````````````10``````````````````15
    gtc tct gct tct cct acc atg aag aaa cgt gcg acc gtc gaa act atc  96
    Val Ser Ala Ser Pro Thr Met Lys Lys Arg Ala Thr Val Glu Thr Ile
    ````````````20``````````````````25``````````````````30
    aac aac tgt aat cag cag ggc act gtt gct ctg acc ttt gac gat ggc 144
    Asn Asn Cys Asn Gln Gln Gly Thr Val Ala Leu Thr Phe Asp Asp Gly
    ````````35``````````````````40``````````````````45
    cct tac aat tac gaa gcc caa gtt gct tct gcc ctt gac ggg ggt aag 192
    Pro Tyr Asn Tyr Glu Ala Gln Val Ala Ser Ala Leu Asp Gly Gly Lys
    ````50``````````````````55``````````````````60
    ggt act ttt ttc ctc aac ggc gcg aat tat gtc tgc atc tac gac aag 240
    Gly Thr Phe Phe Leu Asn Gly Ala Asn Tyr Val Cys Ile Tyr Asp Lys
    65``````````````````70``````````````````75``````````````````80
    gcc gat gaa atc aga gct ttg tat gat gcc ggc cac act ctt ggt tct 288
    Ala Asp Glu Ile Arg Ala Leu Tyr Asp Ala Gly His Thr Leu Gly Ser
    ````````````````85``````````````````90``````````````````95
    cac act tgg tct cac gcc gac ctt acc cag tta gat gaa tcc ggg atc 336
    His Thr Trp Ser His Ala Asp Leu Thr Gln Leu Asp Glu Ser Gly Ile
    ````````````100`````````````````105`````````````````110
    aac gag gaa ctc tcc aag gtc gaa gat gcc ttt gtc aag atc ctt ggt 384
    Asn Glu Glu Leu Ser Lys Val Glu Asp Ala Phe Val Lys Ile Leu Gly
    ````````115`````````````````120`````````````````125
    gtc aag cct cga tac ttc cga ccc cct tac ggt aac atc aac gac aac 432
    Val Lys Pro Arg Tyr Phe Arg Pro Pro Tyr Gly Asn Ile Asn Asp Asn
    ````130`````````````````135`````````````````140
    gtc ttg aac gtc ctc agt gaa agg ggt tac acg aag gtg ttt ttg tgg 480
    Val Leu Asn Val Leu Ser Glu Arg Gly Tyr Thr Lys Val Phe Leu Trp
    145`````````````````150`````````````````155`````````````````160
    tct gat gac act ggg gat gcc aac ggc gag tcg gtc agt tac tcc gag 528
    Ser Asp Asp Thr Gly Asp Ala Asn Gly Glu Ser Val Ser Tyr Ser Glu
    ````````````````165`````````````````170`````````````````175
    ggg gta ttg gac aac gtt atc cag gat tat cct aac cct cat ctt gtc 576
    Gly Val Leu Asp Asn Val Ile Gln Asp Tyr Pro Asn Pro His Leu Val
    ````````````180`````````````````185`````````````````190
    ctt gat cac tct acc atc gag acg acc tcc tcc gag gtt ctc cct tac 624
    Leu Asp His Ser Thr Ile Glu Thr Thr Ser Ser Glu Val Leu Pro Tyr
    ````````195`````````````````200`````````````````205
    gct gta ccc aag ctc cag agt gct ggc tac caa ctg gtc act gtc ggt 672
    Ala Val Pro Lys Leu Gln Ser Ala Gly Tyr Gln Leu Val Thr Val Gly
    ````210`````````````````215`````````````````220
    gaa tgt ctc ggc acc gac gaa tct cct tac gaa tgg gtt gat tgc cct 720
    Glu Cys Leu Gly Thr Asp Glu Ser Pro Tyr Glu Trp Val Asp Cys Pro
    225`````````````````230`````````````````235`````````````````240
    gga gag agg gat agc tct tgg caa tgc 747
    Gly Glu Arg Asp Ser Ser Trp Gln Cys
    ````````````````245
  • The following conditions were used to amplify CDA from the plasmid: 25 μL reactions were performed in PCR Ready Bead tubes (Amersham Pharmacia) containing 500 ng of the plasmid containing CDA, 2 μM primers, and 13 μL sterile water. Reactions were performed using the Perkin Elmer 2400 Thermocycler (PE Applied Biosystems) under the following conditions: an initial cycle at 94° C. for 5 min, 30 cycles at 94° C. for 1 min, 50° C. for 1 min and 72° C. for 1 min, and a final extension at 72° C. for 7 min.
  • The PCR reactions were separated using agarose gel electrophoresis and a band of approximately 744 bases corresponding to the amplified CDA ORF containing a 3′ c-myc tag was excised. The band was purified and ligated into pCR 2.1. The ligated vector was used to transform competent E. coli cells and vector DNA isolated from 5 mL mini cultures using standard alkaline lysis techniques. DNA isolated from several miniprep isolates was digested with EcoRI to confirm the presence of the 744 base CDA/c-myc insert.
  • In one exemplary method, the DNA of the isolate was excised from the pCR 2.1 vector and ligated into the pYEX-BX vector as previously described and the salient portions of the vector was determined to ensure the correct alignment of the ORF with the TATA box
  • One exemplary method concerns cloning of Laccase: As indicated above, the same strategy to clone Laccase (lac) from a plasmid containing the gene as used to clone CDA. DNA sequence and plasmid DNA was used to predicted the amino acid sequences to design PCR primers to amplify the ORF plus the c-myc tag from the plasmid. The DNA and amino acid sequences are presented in diagram form in SEQ ID NOs:18 and 19. The positions of the 5′ and 3′ primers used to obtain the predicted PCR product are shown. A 20 amino acid signal sequence was predicted by PSORTII (http://psort.nibb.ac.jp/cgi-bin). The signal sequence was subsequently removed as PSORTII predicted that the protein would be extracellular, which suggested that the protein would be secreted by the S. cerevisiae if the signal sequence was left intact. An endogenous methionine at amino acid 21 (Glutamic acid) was used as the first amino acid in the new protein to ensure that the protein would be translated.
    atg cgg gga gta gtc aag ctc ttc ttt cta tct tgt tcc ctc gtt tcg 48
    Met Arg Gly Val Val Lys Leu Phe Phe Leu Ser Cys Ser Leu Val Ser
    1```````````````5```````````````````10``````````````````15
    ctg gtc agc agc gag gag act ggc aag tcg cca acc gcg aac tat gac 96
    Leu Val Ser Ser Glu Glu Thr Gly Lys Ser Pro Thr Ala Asn Tyr Asp
    ````````````20``````````````````25``````````````````30
    cat tat atg ccg aag gcg aca gca acc att gat cct agt gta ttc gct 144
    His Tyr Met Pro Lys Ala Thr Ala Thr Ile Asp Pro Ser Val Phe Ala
    ````````35``````````````````40``````````````````45
    ctt tca aat gac ttt gaa ata aca gat gtt ccg acg acg agg gag tat 192
    Leu Ser Asn Asp Phe Glu Ile Thr Asp Val Pro Thr Thr Arg Glu Tyr
    ````50``````````````````55``````````````````60
    acc ttc gat atc acc aaa gcg ttg gcc agc cct gat ggt tat gaa cga 240
    Thr Phe Asp Ile Thr Lys Ala Leu Ala Ser Pro Asp Gly Tyr Glu Arg
    65``````````````````70``````````````````75``````````````````80
    gag gtt tac gtt gtc aac aac atg ttc cct gga cct gtg ata gag gct 288
    Glu Val Tyr Val Val Asn Asn Met Phe Pro Gly Pro Val Ile Glu Ala
    ````````````````85``````````````````90``````````````````95
    aac acc ggg gat act att atc gta cat gtc aac aat cat ttg gag gaa 336
    Asn Thr Gly Asp Thr Ile Ile Val His Val Asn Asn His Leu Glu Glu
    ````````````100`````````````````105`````````````````110
    gga caa agt atc cac tgg cat ggt ttg cgg cag ctt ggc acg gct ttc 384
    Gly Gln Ser Ile His Trp His Gly Leu Arg Gln Leu Gly Thr Ala Phe
    ````````115`````````````````120`````````````````125
    atg gac ggt gtc cct ggt ata aca cag tgt cct att ccc cct gga agc 432
    Met Asp Gly Val Pro Gly Ile Thr Gln Cys Pro Ile Pro Pro Gly Ser
    ````130`````````````````135`````````````````140
    tca ttt acc tac caa ttc acc gta agc cat cag tca ggc acg ttt tgg 480
    Ser Phe Thr Tyr Gln Phe Thr Val Ser His Gln Ser Gly Thr Phe Trp
    145`````````````````150`````````````````155`````````````````160
    tgg cat tcc cat tat tcc aat tcc atg gcc gac ggc att tgg ggc ccc 528
    Trp His Ser His Tyr Ser Asn Ser Met Ala Asp Gly Ile Trp Gly Pro
    ````````````````165`````````````````170`````````````````175
    tta att atc cat tcg ccc aat gaa ccc ctc caa agg gga cga gac tat 576
    Leu Ile Ile His Ser Pro Asn Glu Pro Leu Gln Arg Gly Arg Asp Tyr
    ````````````180`````````````````185`````````````````190
    gac gag gat cga atc gtt ttt ata act gac tgg gtg cat gac aac tca 624
    Asp Glu Asp Arg Ile Val Phe Ile Thr Asp Trp Val His Asp Asn Ser
    ````````195`````````````````200`````````````````205
    gaa gtc gtt att gca gct cta gct act cca gaa ggg tac aaa gga agc 672
    Glu Val Val Ile Ala Ala Leu Ala Thr Pro Glu Gly Tyr Lys Gly Ser
    ````210`````````````````215`````````````````220
    cct gct ccg cca caa ggt gat gcg att ctc atc aat gga cgt ggc caa 720
    Pro Ala Pro Pro Gln Gly Asp Ala Ile Leu Ile Asn Gly Arg Gly Gln
    225`````````````````230`````````````````235`````````````````240
    acc aac tgc aca gcc act ggt tcc tcc tca tgc acc tat ccg cct cct 768
    Thr Asn Cys Thr Ala Thr Gly Ser Ser Ser Cys Thr Tyr Pro Pro Pro
    ````````````````245`````````````````250`````````````````255
    ccc gag att cac gtg cca gtc aat tgc agg gtt cgt ctg cgc ttt atc 816
    Pro Glu Ile His Val Pro Val Asn Cys Arg Val Arg Leu Arg Phe Ile
    ````````````260`````````````````265`````````````````270
    agt gcg acc gcc cat ccc atg tac cgc ata act atc gac aac cac cct 864
    Ser Ala Thr Ala His Pro Met Tyr Arg Ile Thr Ile Asp Asn His Pro
    ````````275`````````````````280`````````````````285
    ttg gaa gtt gtg gaa acc gac ggt aca gcc gtc tat ggg ccc aca gtc 912
    Leu Glu Val Val Glu Thr Asp Gly Thr Ala Val Tyr Gly Pro Thr Val
    ````290`````````````````295`````````````````300
    cat gaa atc tcc att gca cct ggg gaa cgg tac tct gca att atc aac 960
    His Glu Ile Ser Ile Ala Pro Gly Glu Arg Tyr Ser Ala Ile Ile Asn
    305`````````````````310`````````````````315`````````````````320
    acc tca gaa ggg aag gaa ggt gat gcg ttc tgg ctg agg aca agt gtt 1008
    Thr Ser Glu Gly Lys Glu Gly Asp Ala Phe Trp Leu Arg Thr Ser Val
    ````````````````325`````````````````330`````````````````335
    gct ctg ggc tgt atg ttt ggt gga ata gat cag gtg gga ttg gcg gtt 1056
    Ala Leu Gly Cys Met Phe Gly Gly Ile Asp Gln Val Gly Leu Ala Val
    ````````````340`````````````````345`````````````````350
    gtg agg tat acg ggt aat gga atg gtt agt act gaa gag cct caa act 1104
    Val Arg Tyr Thr Gly Asn Gly Met Val Ser Thr Glu Glu Pro Gln Thr
    ````````355`````````````````360`````````````````365
    act gct tgg agt gat cta gcg gga gct aca act cct tgt gct gga ctg 1152
    Thr Ala Trp Ser Asp Leu Ala Gly Ala Thr Thr Pro Cys Ala Gly Leu
    ````370`````````````````375`````````````````380
    gac caa aca tat act ctt tca cca cga gag agt ttt agt gca cct cgt 1200
    Asp Gln Thr Tyr Thr Leu Ser Pro Arg Glu Ser Phe Ser Ala Pro Arg
    385`````````````````390`````````````````395`````````````````400
    gaa ttt tca caa agc cat gtc ttc aat agc cag cga gga gcc ttt gtg 1248
    Glu Phe Ser Gln Ser His Val Phe Asn Ser Gln Arg Gly Ala Phe Val
    ````````````````405`````````````````410`````````````````415
    aat gtt tat ggc aac acc ttc caa ggt tat ggg ttt aac aat atc tca 1296
    Asn Val Tyr Gly Asn Thr Phe Gln Gly Tyr Gly Phe Asn Asn Ile Ser
    ````````````420`````````````````425`````````````````430
    tat cag aac caa atc ttc aac cct cta ctt tca atc gtc caa cgc ggt 1344
    Tyr Gln Asn Gln Ile Phe Asn Pro Leu Leu Ser Ile Val Gln Arg Gly
    ````````435`````````````````440`````````````````445
    ggc tct tgc gag agc aca cta gta gcc agt aca act ttc ccc gac ctc 1392
    Gly Ser Cys Glu Ser Thr Leu Val Ala Ser Thr Thr Phe Pro Asp Leu
    ````450`````````````````455`````````````````460
    gga tca ggg aac att atc atc aac aat ctt gat ggc gtt atc gac cat 1440
    Gly Ser Gly Asn Ile Ile Ile Asn Asn Leu Asp Gly Val Ile Asp His
    465`````````````````470`````````````````475`````````````````480
    cct tac cac ctg cac ggc aac gag ttc cag gtg ata gga cga gga act 1488
    Pro Tyr His Leu His Gly Asn Glu Phe Gln Val Ile Gly Arg Gly Thr
    ````````````````485`````````````````490`````````````````495
    gga gct ctc agc ctt gat aac ctg aca aat att gac ttc aat ttg gac 1536
    Gly Ala Leu Ser Leu Asp Asn Leu Thr Asn Ile Asp Phe Asn Leu Asp
    ````````````500`````````````````505`````````````````510
    aac cct gtg aga aag gat acc ctc tgg ata cag ggc gga agt tgg gtg 1584
    Asn Pro Val Arg Lys Asp Thr Leu Trp Ile Gln Gly Gly Ser Trp Val
    ````````515`````````````````520`````````````````525
    gta ctg agg atc acg acg gat aac cct gga gtt tgg gcc ttg cac tgt 1632
    Val Leu Arg Ile Thr Thr Asp Asn Pro Gly Val Trp Ala Leu His Cys
    ````530`````````````````535`````````````````540
    cat att ggg tgg cat ctt act gag gga aag ttg gct gtg gtt gtc att 1680
    His Ile Gly Trp His Leu Thr Glu Gly Lys Leu Ala Val Val Val Ile
    545`````````````````550`````````````````555`````````````````560
    caa cca ggt gcg att gga cat atg gag ggc ccc gag tct tgg acg aat 1728
    Gln Pro Gly Ala Ile Gly His Met Glu Gly Pro Glu Ser Trp Thr Asn
    ````````````````565`````````````````570`````````````````575
    ctc tgt gct aac act gat ccc aat gca ttt gga ccc gca cga cgc tca 1776
    Leu Cys Ala Asn Thr Asp Pro Asn Ala Phe Gly Pro Ala Arg Arg Ser
    ````````````580`````````````````585`````````````````590
    cct tct cca tct att caa tcc tct aag aca tcc act ttc cag tat ctg 1824
    Pro Ser Pro Ser Ile Gln Ser Ser Lys Thr Ser Thr Phe Gln Tyr Leu
    ````````595`````````````````600`````````````````605
    cgc gaa gtg aaa ggg aag gtc gtt aaa cgt aga ggt gct cga gag gcg 1872
    Arg Glu Val Lys Gly Lys Val Val Lys Arg Arg Gly Ala Arg Glu Ala
    ````610`````````````````615`````````````````620
  • The following conditions were used to amplify laccase from the plasmid: 25 μL reactions were performed in PCR Ready Bead tubes containing 500 ng of plasmid containing Laccase, 2 μM primers, and 13 μL sterile water. Reactions were performed using a Perkin Elmer 2400 Thermocycler under the following conditions: an initial cycle at 94o C for 5 min, 30 cycles at 94o C for 1 min, 50o C for 1 min, 72o C for 1 min, and a final extension at 72o C for 7 min.
  • The PCR reactions were separated, isolated and digested with EcoRI as previously described to confirm the presence of the 1857 base Lac/c-myc insert.
  • Several isolates were grown, cells harvested, plasmid DNA isolated and the DNA sequence of the Lac insert determined. The DNA of the isolate was excised from the pCR 2.1 vector and the DNA band encoding the Laccase ORF could be isolated, data not shown. This DNA fragment was then ligated into the pYEX-BX vector as described and then used to transform competent E. coli and the plasmid DNA isolated by standard alkaline lysis mini-preps. The DNA sequence of salient portions of the vector was determined to ensure the correct alignment of the ORF with the TATA box.
  • An empty vector control was made and used as previously described in methods.
  • Transformation of S. cerevisiae was made and used as previously described in methods.
  • Identification of yeast transformants expression each C. neoformans protein. After obtaining transformed S. cerevisiae cells that contained each gene of interest 5 transformants were screened for each gene of interest were screened to determine the expression of each gene and the transformants were isolated as previously described. Once transformants containing each construct were isolated, each of the proteins was expressed as described previously. Then transformants expressing each C. neoformans protein were identified by one exemplary method previously disclosed.
  • Expression of C. neoformans proteins were determined by an exemplary western blot technique known in the art. Ten CDA transformants were screened to obtain a transformant that was positive for induction via copper. Transformant 10 was chosen for vaccine production as it had the most protein as determined by counting pixels. Five Lac transformants were screened to obtain a transformant that was positive for induction via copper. Please note that the top band is of the correct size. Transformant number 2 had the least amount of protein in the un-induced sample and the most (by counting pixels) in the copper induced sample
  • Growth of transformants: Transformants that were determined to produce a protein of the correct molecular weight were grown in YNB minus uracil for 2-3 days at 30° C. with shaking. Once the cultures were in stationary phase, the cells were counted and frozen in 50% glycerol at a final concentration of 1×108 c/mL (10 yeast units) and stored at −80° C. until use.
  • Yeast cells containing and expressing chitin deacetylase or laccase were harvested and lysed as described. The indicated number of microliters of each extract were separated by SDS-PAGE, transfer to membranes and the position of each protein detected by Western blot analysis as described. The results of the western blot are shown in FIG. 10. Lane 1-4 are various amounts in microliters of lysed yeast cells expressing hitin deacetylase (cda; MW.26.6 kDa), while lanes 6-9 are various amounts in microliters of lysed yeast cells expressing laccase (lac; MW. 67.4 kDa).
  • FIG. 11 represents exemplary linear survival plots in an animal model after exposure to a challenge fungal organism. Mice were vaccinated with yeast cells containing and expressing the DNA encoding laccase, (lac), chitin deacetylase (cda), the MYC yeast control, PBS, trr and plb (two additional C. neoformans proteins) as described above on days 21, 14 and 7 prior to challenge by C. neoformans H99 cells. Mice were weighed daily and mice that had lost >15% of their body weight euthanized. Note that mice vaccinated with lac-containing yeast were 100% protected (p<<0.05) while those vaccinated with cda containing yeast were 80% protected (p<0.05) as were those vaccinated with MYC (empty vector control). These results demonstrate that mice vaccinated with yeast expressing either laccase or chitin deacetylase were highly protected against a challenge of C. neoformans.
    TABLE 1
    Candidate antigens
    Number of Base pairs
    Protein amino acids of the ORF Accession numbers
    fos-1 708 2,124 Protein: AAK27436
    DNA: AF257496
    hemolysin 131 721 Protein: BAA03951.1
    DNA: D16501
    catalase 728 2,184 Protein: AAB71223
    DNA: U97574
    abr-1 664 1,992 Protein: AAF03353
    DNA: AF116901
    rod A 159 477 Protein: AAB60172.1
    DNA: U06121
    dpp-5 721 2,163 Protein: AAB67282
    DNA: L48074
    dpp-4 765 2,295 Protein: AAC34310
    DNA: U87950
  • TABLE 2
    Cryptococcus antigens
    Number of Base pairs
    Protein amino acids of the ORF Accession numbers
    CDA—Chitan 250 750 Protein: CAD10036
    deacetylase DNA: AJ414580
    CnFOS—histidine 1367 4101 Protein: AAW42353
    kinase DNA: NA*
    Lac—Laccase 625 1875 Protein: NA*
    DNA: L22866
    Plb—phospholipse 638 1914 Protein: AAF65220.1
    B DNA: AF223383
    Ure—Urease 833 2499 Protein: AAC62257.1
    DNA: AF006062

    *NA—not available
  • TABLE 3
    PCR primers
    PCR primers
    5′ AspF2/Bam 5′GGA TCC ATG CTT GTG GCC ACC CTC CCT
    3′
    (SEQ ID NO:20)
    3′ AspF2/myc 5′GAA TTC TTA TCA CTC GAG GTC TTC TTC
    GGA AAT CAA CTT AGT GCA ATG AAG CTG
    TCC ACC TTC ATG GGT ATG GC 3′
    (SEQ ID NO:21)
    5′DppV/Bam 5′GGA TTC ATG CTT ACA CCT GAG CAG CTA
    ATC ACT GCT CCA CGG 3′
    (SEQ ID NO:22)
    3′DppV/myc 5′GGA TTC TTA TCA CTC GAG GTC TTC TTC
    GGA AAT CAA CTT CTG TTC CGG GAC AAC
    GGT GTC CTC CAG GCT GAC GGC GTT GGG
    G 3′
    (SEQ ID NO:23)
    5′Fos-1/SalI 5′GTC GAC ATG GCC CTC GAC AAG GAG
    C 3′
    (SEQ ID NO:24)
    3′Fos-1/myc 5′CTG CAG TTA TCA CTC GAG GTC TTC TTC
    GGA AAT CAA CTT CTG TTC CAT CTT GGA
    TGA TTC CTC AAA AGC TGC CAC CAT CCG
    CGC TTC AGC CGC 3′
    (SEQ ID NO:25)
    5′Hemo/Bam 5′GGA TTC ATG GCA TCG GTC CAA GCT TAC
    GCA CAG TGG 3′
    (SEQ ID NO:26)
    3′Hemo/myc 5′GAA TTC TTA TCA CTC GAG GTC TTC TTC
    GGA AAT CAA CTT CTG TTC ACA GTT GCC
    AAT GGC ACC ACC ATA CTT GTT CCA CGT
    TCC AAT TTC GAC CCA G 3′
    (SEQ ID NO:27)
    5′RodA/SalI 5′GTC GAC ATG AAG TTC TCT TTG AGC GCT
    GCT GTC CTC GC 3′
    (SEQ ID NO:28)
    3′RodA/myc 5′GAA TTC TTA TCA CTC GAG GTC TTC TTC
    GGA AAT CAA CTT CTG TTC CAG GAT AGA
    ACC AAG GGC AAT GCA AGG AAG ACC CAG
    TCC AAT GAG GG 3′
    (SEQ ID NO:29)
  • TABLE 4
    PCR primers
    PCR primers
    5′ CDA/Bam GGA TCC ATG TCT CCT ACC ATG AAG AAA
    CGT GCG
    (SEQ ID NO:30)
    3′ CDA/myc GAA TTC TTA TCA CTC GAG GTC TTC TTC
    GGA AAT CAA CTT CTG TTC GCA TTG CCA
    AGA GCT ATG CCT CTC TCC AGG GCA ATC
    AAC CCA TTC G
    (SEQ ID NO:31)
    5′ CnFos/Sal GTC GAC ATG TCC CTC CCC GAT GCC TAC
    CCT CCG GTC ATA GCC ACC
    (SEQ ID NO:32)
    3′ CnFos/myc GAA TTC TTA TCA CTC GAG GTC TTC TTC
    GGA AAT CAA CTT CTG TTC ACT TCT GTT
    GGC CAT TTC AGC TTC CTG TCT CGC C
    (SEQ ID NO:33)
    5′ Lac/Bam GGA TCC ATG GAG GAG ACT GGC AAG TCG
    CCA ACC GCG
    (SEQ ID NO:34)
    3′ Lac/myc GAA TTC TTA TCA CTC GAG GTC TTC TTC
    GGA AAT CAA CTT CTG TTC CGC CTC TCG
    AGC ACC TCT ACG TTT AAC GAC CTT CCC
    TTT CAC TTC GCG CAG
    (SEQ ID NO:35)
    5′ Plb/Bam GGA TCC ATG GCT GTT CCT CCC GAG ACT
    CCG CGG
    (SEQ ID NO:36)
    3′ Plb/myc GAA TTC TTA TCA CTC GAG GTC TTC TTC
    GGA AAT CAA CTT CTG TTC AGT GAG AGA
    GCG CAT ACC ATT GAG CAA CAT TTC GTT
    GGC
    (SEQ ID NO:37)
    5′ TRR/Bam GGA TCC ATG CAC TCC AAG GTT GTT ATC
    ATC GGC TCT GG
    (SEQ ID NO:38)
    3′ TRR/myc GAA TTC TTA TCA CTC GAG GTC TTC TTC
    GGA AAT CAA CTT CTG TTC CTC CTT GTC
    AGT GCC GAA GTA GTG CTC GGC AGG GAC
    ATG CAC ATC TTC GGT CTG G
    (SEQ ID NO:39)
    5′ URE/Bam GGA TCC ATG CAT CTC CTC CCG AGA GAA
    ACG
    (SEQ ID NO:40)
    3′ URE/myc GTC GAC TTA TCA CTC GAG GTC TTC TTC
    GGA AAT CAA CTT CTG TTC GTA AAC GAA
    GTA TCT CCT GGT CAA TGG GAG TTT GTC
    CGC GGG TGG GAC
    (SEQ ID NO:41)
  • All of the COMPOSITIONS and METHODS disclosed and claimed herein may be made and executed without undue experimentation in light of the present disclosure. While the COMPOSITIONS and METHODS have been described in terms of preferred embodiments, it will be apparent to those of skill in the art that variation may be applied to the COMPOSITIONS and METHODS and in the steps or in the sequence of steps of the METHODS described herein without departing from the concept, spirit and scope of the invention. More specifically, it will be apparent that certain agents which are both chemically and physiologically related may be substituted for the agents described herein while the same or similar results would be achieved. All such similar substitutes and modifications apparent to those skilled in the art are deemed to be within the spirit, scope and concept of the invention as defined by the appended claims.

Claims (22)

1. A non-viable fungal cell composition comprising, fungal cells that were heated at a temperature of about 56° C. for about one hour, or a derivative thereof.
2. The composition of claim 1, wherein the fungal cells comprise transgenic, non-transgenic or a combination thereof.
3. The composition of claim 1, wherein the fungal cells are selected from the group consisting of Saccharomyces spp., Aspergillus spp. Cryptococcus spp., Coccidioides spp., Neurospora spp., Histoplasma spp., Blastomyces spp., and a combination thereof.
4. The composition of claim 1, wherein the fungal cells comprise Saccharomyces spp.
5. The composition of claim 4, wherein fungal cells are spores, vegetative cells, germlings, or a combination thereof.
6. The composition of claim 1, wherein the fungal cells comprise Neurospora spp.
7. The composition of claim 6, wherein the fungal cells are vegetative hyphae, aerial hyphae, macroconidia, microconidia, ascospores, or a combination thereof.
8. The composition of claim 1, wherein the fungal cells further comprise a tag.
9. The composition of claim 2, wherein the transgenic fungal cells comprise an oligonucleotide, a peptide, a protein, a chimeric molecule or combination thereof.
10. The composition of claim 2, wherein the transgenic fungal cells further comprise a virulence factor.
11. A method of inducing an immune response against a fungus comprising:
administering a non-viable fungal composition to a subject, wherein administering the composition induces an immune response against a fungus in the subject and wherein the non-viable fungal composition comprises fungal cells that were heated at a temperature of about 56° C. for about one hour, or a derivative thereof.
12. The method of claim 11, wherein the non-viable fungal composition is administered prophylactically to reduce or prevent a fungal infection.
13. The method of claim 11, wherein the non-viable fungal composition is administered therapeutically to a subject with a fungal infection.
14. The method of claim 11, wherein the subject is a mammal, bird or reptile.
15. The method of claim 11, wherein the subject is a human.
16. The method of claim 11, wherein the non-viable fungal composition induces an immune response against a fungus selected from the group consisting of Saccharomyces spp., Aspergillus spp. Cryptococcus spp., Coccidioides spp., Neurospora spp., Histoplasma spp., Blastomyces spp., and a combination thereof.
17. The method of claim 11, wherein the subject is an immunocompromised subject.
18. A method of treating a subject in need of an antifungal therapy comprising:
administering to a subject in need of such treatment a non-viable fungal composition comprising fungal cells that were heated at a temperature of about 56° C. for about one hour, or a derivative thereof.
19. The method of claim 18, wherein the fungal cells comprise transgenic, non-transgenic or a combination thereof.
20. A method of preparing an immune response stimulating antifungal composition, said method comprising, heating fungal cells to about 56° C. for about one hour such that fungal cells are non-viable but are capable of inducing an immune response against a fungus when administered to a subject.
21. The method of claim 20, wherein the fungal cells comprise transgenic, non-transgenic or a combination thereof.
22. The method of claim 20, wherein the fungal cells are selected from the group consisting of Saccharomyces spp., Aspergillus spp. Cryptococcus spp., Coccidioides spp., Neurospora spp., Histoplasma spp., Blastomyces spp., and a combination thereof
US11/610,457 2005-12-13 2006-12-13 Compositions and Methods to Elicit Immune Responses Against Pathogenic Organisms Using Yeast Based Vaccines Abandoned US20070172503A1 (en)

Priority Applications (3)

Application Number Priority Date Filing Date Title
US11/610,457 US20070172503A1 (en) 2005-12-13 2006-12-13 Compositions and Methods to Elicit Immune Responses Against Pathogenic Organisms Using Yeast Based Vaccines
US12/570,947 US8460919B2 (en) 2005-12-13 2009-09-30 Compositions and methods to elicit immune responses against pathogenic organisms using yeast based vaccines
US13/800,094 US9333246B2 (en) 2005-12-13 2013-03-13 Compositions and methods to elicit immune responses against pathogenic organisms using yeast-based vaccines

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US75002905P 2005-12-13 2005-12-13
US81730006P 2006-06-28 2006-06-28
US11/610,457 US20070172503A1 (en) 2005-12-13 2006-12-13 Compositions and Methods to Elicit Immune Responses Against Pathogenic Organisms Using Yeast Based Vaccines

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US12/570,947 Continuation US8460919B2 (en) 2005-12-13 2009-09-30 Compositions and methods to elicit immune responses against pathogenic organisms using yeast based vaccines

Publications (1)

Publication Number Publication Date
US20070172503A1 true US20070172503A1 (en) 2007-07-26

Family

ID=38285811

Family Applications (3)

Application Number Title Priority Date Filing Date
US11/610,457 Abandoned US20070172503A1 (en) 2005-12-13 2006-12-13 Compositions and Methods to Elicit Immune Responses Against Pathogenic Organisms Using Yeast Based Vaccines
US12/570,947 Active 2028-11-05 US8460919B2 (en) 2005-12-13 2009-09-30 Compositions and methods to elicit immune responses against pathogenic organisms using yeast based vaccines
US13/800,094 Active 2027-07-31 US9333246B2 (en) 2005-12-13 2013-03-13 Compositions and methods to elicit immune responses against pathogenic organisms using yeast-based vaccines

Family Applications After (2)

Application Number Title Priority Date Filing Date
US12/570,947 Active 2028-11-05 US8460919B2 (en) 2005-12-13 2009-09-30 Compositions and methods to elicit immune responses against pathogenic organisms using yeast based vaccines
US13/800,094 Active 2027-07-31 US9333246B2 (en) 2005-12-13 2013-03-13 Compositions and methods to elicit immune responses against pathogenic organisms using yeast-based vaccines

Country Status (1)

Country Link
US (3) US20070172503A1 (en)

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20090098154A1 (en) * 2002-12-16 2009-04-16 Globelmmune, Inc. Yeast-Based Vaccines As Immunotherapy
US20090118750A1 (en) * 2006-01-30 2009-05-07 Warsaw Orthopedic, Inc. Chisel System for Osteochondral Implants and A Surgical Procedure Involving Same
US20090304741A1 (en) * 2000-11-15 2009-12-10 Globelmmune, Inc. Yeast-Dendritic Cell Vaccines and Uses Thereof
US20100034840A1 (en) * 2005-07-11 2010-02-11 David Apelian Compositions and methods for eliciting an immune response to escape mutants of targeted therapies
US20100104604A1 (en) * 2005-12-13 2010-04-29 Globelmmune, Inc. Compositions and Methods to Elicit Immune Responses Against Pathogenic Organisms Using Yeast Based Vaccines
US20100111912A1 (en) * 2007-03-19 2010-05-06 David Apelian Compositions and methods for targeted ablation of mutational escape of targeted therapies for cancer
US20100189749A1 (en) * 2007-02-02 2010-07-29 Alex Franzusoff Methods for producing yeast-based vaccines
US20110150909A1 (en) * 2002-12-16 2011-06-23 Alex Franzusoff Yeast-Based Vaccines As Immunotherapy
US8722054B2 (en) 2011-02-12 2014-05-13 Globeimmune, Inc. Compositions and methods for the treatment or prevention of hepatitis B virus infection
US8911722B2 (en) 2009-09-14 2014-12-16 The Regents Of The University Of Colorado, A Body Corporate Modulation of yeast-based immunotherapy products and responses
US9198941B2 (en) 2011-03-17 2015-12-01 The United States Of America, As Represented By The Secretary, Dept. Of Health And Human Services Yeast-Brachyury immunotherapeutic compositions
US9254320B2 (en) 2010-12-17 2016-02-09 Globeimmune, Inc. Compositions and methods for the treatment or prevention of human adenovirus-36 infection
US9533031B2 (en) 2011-08-17 2017-01-03 Globeimmune, Inc. Yeast-MUC1 immunotherapeutic compositions and uses thereof
US9579377B2 (en) 2011-06-14 2017-02-28 Globeimmune, Inc. Yeast-based compositions and methods for the treatment or prevention of hepatitis delta virus infection
US9653272B2 (en) 2012-06-26 2017-05-16 Biodesix, Inc. Mass-spectral method for selection, and de-selection, of cancer patients for treatment with immune response generating therapies
US9782473B2 (en) 2013-03-26 2017-10-10 Globeimmune, Inc. Compositions and methods for the treatment or prevention of human immunodeficiency virus infection
US10226522B2 (en) 2013-08-30 2019-03-12 Globeimmune, Inc. Compositions and methods for the treatment or prevention of tuberculosis
US10383924B2 (en) 2009-04-17 2019-08-20 Globeimmune, Inc. Combination immunotherapy compositions against cancer and methods
US10507235B2 (en) 2013-03-19 2019-12-17 Globeimmune, Inc. Yeast-based immunotherapy for Chordoma
EP3481417A4 (en) * 2016-07-11 2020-04-01 Rutgers, The State University of New Jersey Fungal vaccine compositions and methods of use thereof
US10799567B2 (en) 2014-04-11 2020-10-13 Globeimmune, Inc. Yeast-based immunotherapy and type I interferon sensitivity
US11065313B2 (en) 2015-08-03 2021-07-20 Globeimmune, Inc. Modified yeast-brachyury immunotherapeutic compositions

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10092632B2 (en) 2014-05-15 2018-10-09 Washington University Vaccination against Cryptococcus
EP3687629A4 (en) 2017-09-27 2021-10-06 University of Georgia Research Foundation Treatment and detection of infection and disease associated with different fungal pathogens
US11273190B2 (en) * 2019-05-06 2022-03-15 Kenda Rigdon Composition of heat-killed yeast and taurine or a phytomedicine for the treatment of chronic inflammation
US20220401533A1 (en) * 2019-11-14 2022-12-22 University Of Georgia Research Foundation, Inc. Treatment and protection against aspergillus infection and aspergillosis disease

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5830463A (en) * 1993-07-07 1998-11-03 University Technology Corporation Yeast-based delivery vehicles
US5858378A (en) * 1996-05-02 1999-01-12 Galagen, Inc. Pharmaceutical composition comprising cryptosporidium parvum oocysts antigen and whole cell candida species antigen
US20040156858A1 (en) * 2002-12-16 2004-08-12 Alex Franzusoff Yeast-based vaccines as immunotherapy
US7083787B2 (en) * 2000-11-15 2006-08-01 Globeimmune, Inc. Yeast-dendritic cell vaccines and uses thereof

Family Cites Families (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
FR2486400B1 (en) 1980-07-09 1986-05-30 Univablot MEDICINAL PRODUCTS BASED ON YEAST OR THEIR INSOLUBLE EXTRACTS
NZ199722A (en) 1981-02-25 1985-12-13 Genentech Inc Dna transfer vector for expression of exogenous polypeptide in yeast;transformed yeast strain
US4775622A (en) 1982-03-08 1988-10-04 Genentech, Inc. Expression, processing and secretion of heterologous protein by yeast
US5310654A (en) 1985-07-31 1994-05-10 The Board Of Trustees Of The Leland Stanford Junior University Method for determining virulence of Yersinia
IL95019A0 (en) 1989-08-09 1991-06-10 Mycogen Corp Process for encapsulation of biologicals
US5413914A (en) 1993-07-07 1995-05-09 The Regents Of The University Of Colorado Yeast assay to identify inhibitors of dibasic amino acid processing endoproteases
US20020044948A1 (en) 2000-03-15 2002-04-18 Samir Khleif Methods and compositions for co-stimulation of immunological responses to peptide antigens
AU2001219510B2 (en) 2000-04-06 2007-06-14 Allertein Therapeutics, Llc Microbial delivery system
US7914799B2 (en) 2001-08-27 2011-03-29 Immunitor USA, Inc. Anti-fungal composition
US7439042B2 (en) 2002-12-16 2008-10-21 Globeimmune, Inc. Yeast-based therapeutic for chronic hepatitis C infection
US8343502B2 (en) 2002-12-16 2013-01-01 Globeimmune, Inc. Yeast-based vaccines as immunotherapy
JP2008516610A (en) 2004-10-18 2008-05-22 グローブイミューン,インコーポレイテッド Treatment of chronic hepatitis C infection using yeast
CA2614884A1 (en) 2005-07-11 2007-01-18 Globeimmune, Inc Compositions and methods for eliciting an immune response to escape mutants of targeted therapies
WO2007035930A2 (en) 2005-09-22 2007-03-29 Prosci Incorporated Glycosylated polypeptides produced in yeast mutants and methods of use thereof
US20070172503A1 (en) 2005-12-13 2007-07-26 Mycologics,Inc. Compositions and Methods to Elicit Immune Responses Against Pathogenic Organisms Using Yeast Based Vaccines
SG10201402236VA (en) 2006-02-02 2014-08-28 Globeimmune Inc Yeast-based vaccine for inducing an immune response
CN101448848B (en) 2006-03-27 2013-12-04 全球免疫股份有限公司 Ras mutation and compositions and methods related thereto
MX2009008175A (en) 2007-02-02 2009-08-12 Globeimmune Inc Improved methods for producing yeast-based vaccines.
EP2134358A1 (en) 2007-03-19 2009-12-23 Globeimmune, Inc. Compositions and methods for targeted ablation of mutational escape of targeted therapies for cancer

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5830463A (en) * 1993-07-07 1998-11-03 University Technology Corporation Yeast-based delivery vehicles
US5858378A (en) * 1996-05-02 1999-01-12 Galagen, Inc. Pharmaceutical composition comprising cryptosporidium parvum oocysts antigen and whole cell candida species antigen
US7083787B2 (en) * 2000-11-15 2006-08-01 Globeimmune, Inc. Yeast-dendritic cell vaccines and uses thereof
US20040156858A1 (en) * 2002-12-16 2004-08-12 Alex Franzusoff Yeast-based vaccines as immunotherapy

Cited By (58)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8221763B2 (en) 2000-11-15 2012-07-17 Globeimmune, Inc. Yeast-antigen compositions and methods of using the same
US20090304741A1 (en) * 2000-11-15 2009-12-10 Globelmmune, Inc. Yeast-Dendritic Cell Vaccines and Uses Thereof
US8337830B2 (en) 2002-12-16 2012-12-25 Globeimmune, Inc. Yeast-based vaccines as immunotherapy
US8734778B2 (en) 2002-12-16 2014-05-27 Globeimmune, Inc. Yeast-based vaccines as immunotherapy
US8153136B2 (en) 2002-12-16 2012-04-10 Globeimmune, Inc. Yeast-based vaccines as immunotherapy
US8067559B2 (en) 2002-12-16 2011-11-29 Globeimmune, Inc. Yeast-based vaccines as immunotherapy
US20110150909A1 (en) * 2002-12-16 2011-06-23 Alex Franzusoff Yeast-Based Vaccines As Immunotherapy
US20090142367A1 (en) * 2002-12-16 2009-06-04 Globeimmune, Inc. Yeast-Based Vaccines As Immunotherapy
US20090098154A1 (en) * 2002-12-16 2009-04-16 Globelmmune, Inc. Yeast-Based Vaccines As Immunotherapy
US20090142366A1 (en) * 2002-12-16 2009-06-04 Globeimmune, Inc. Yeast-Based Vaccines As Immunotherapy
US8343502B2 (en) 2002-12-16 2013-01-01 Globeimmune, Inc. Yeast-based vaccines as immunotherapy
US20100034840A1 (en) * 2005-07-11 2010-02-11 David Apelian Compositions and methods for eliciting an immune response to escape mutants of targeted therapies
US8460919B2 (en) 2005-12-13 2013-06-11 Globeimmune, Inc. Compositions and methods to elicit immune responses against pathogenic organisms using yeast based vaccines
US20100104604A1 (en) * 2005-12-13 2010-04-29 Globelmmune, Inc. Compositions and Methods to Elicit Immune Responses Against Pathogenic Organisms Using Yeast Based Vaccines
US9333246B2 (en) 2005-12-13 2016-05-10 Globeimmune, Inc. Compositions and methods to elicit immune responses against pathogenic organisms using yeast-based vaccines
US20090118750A1 (en) * 2006-01-30 2009-05-07 Warsaw Orthopedic, Inc. Chisel System for Osteochondral Implants and A Surgical Procedure Involving Same
US20100189749A1 (en) * 2007-02-02 2010-07-29 Alex Franzusoff Methods for producing yeast-based vaccines
US9066893B2 (en) 2007-02-02 2015-06-30 GlobelImmune, Inc. Yeast-based vaccines
US9549970B2 (en) 2007-02-02 2017-01-24 Globeimmune, Inc. Methods for producing yeast-based vaccines
US20100111912A1 (en) * 2007-03-19 2010-05-06 David Apelian Compositions and methods for targeted ablation of mutational escape of targeted therapies for cancer
US8501167B2 (en) 2007-03-19 2013-08-06 Globeimmune, Inc. Compositions and methods for targeted ablation of mutational escape of targeted therapies for cancer
US10874729B2 (en) 2009-04-17 2020-12-29 Globeimmune, Inc. Combination immunotherapy compositions against cancer and methods
US11590216B2 (en) 2009-04-17 2023-02-28 GlobelImmune, Inc. Combination immunotherapy compositions against cancer and methods
US10383924B2 (en) 2009-04-17 2019-08-20 Globeimmune, Inc. Combination immunotherapy compositions against cancer and methods
US11596675B2 (en) 2009-04-17 2023-03-07 Globeimmune, Inc. Combination immunotherapy compositions against cancer and methods
US8911722B2 (en) 2009-09-14 2014-12-16 The Regents Of The University Of Colorado, A Body Corporate Modulation of yeast-based immunotherapy products and responses
US10117915B2 (en) 2009-09-14 2018-11-06 The Regents Of The University Of Colorado, A Body Corporate Modulation of yeast-based immunotherapy products and responses
US9254320B2 (en) 2010-12-17 2016-02-09 Globeimmune, Inc. Compositions and methods for the treatment or prevention of human adenovirus-36 infection
US9428556B2 (en) 2011-02-12 2016-08-30 Globeimmune, Inc. Compositions and methods for the treatment or prevention of hepatitis B virus infection
US9290547B2 (en) 2011-02-12 2016-03-22 Globeimmune, Inc. Yeast-based therapeutic for chronic hepatitis B infection
US8877205B2 (en) 2011-02-12 2014-11-04 Globeimmune, Inc. Yeast-based therapeutic for chronic hepatitis B infection
US10441650B2 (en) 2011-02-12 2019-10-15 Globeimmune, Inc. Compositions and methods for the treatment or prevention of hepatitis B virus infection
US8961988B2 (en) 2011-02-12 2015-02-24 Globeimmune, Inc. Compositions and methods for the treatment or prevention of hepatitis B virus infection
US8722054B2 (en) 2011-02-12 2014-05-13 Globeimmune, Inc. Compositions and methods for the treatment or prevention of hepatitis B virus infection
US9943591B2 (en) 2011-02-12 2018-04-17 Globeimmune, Inc. Compositions and methods for the treatment or prevention of hepatitis B virus infection
US9198941B2 (en) 2011-03-17 2015-12-01 The United States Of America, As Represented By The Secretary, Dept. Of Health And Human Services Yeast-Brachyury immunotherapeutic compositions
US11938175B2 (en) 2011-03-17 2024-03-26 The Usa, As Represented By The Secretary, Dept. Of Health And Human Services Yeast-brachyury immunotherapeutic compositions
US11944673B2 (en) 2011-03-17 2024-04-02 The Usa, As Represented By The Secretary, Dept. Of Health And Human Services Yeast-brachyury immunotherapeutic compositions
US10363294B2 (en) 2011-03-17 2019-07-30 Globeimmune, Inc. Yeast-brachyury immunotherapeutic compositions
US9623097B2 (en) 2011-03-17 2017-04-18 Globeimmune, Inc. Yeast-brachyury immunotherapeutic compositions
US9987352B2 (en) 2011-06-14 2018-06-05 Globeimmune, Inc. Yeast-based compositions and methods for the treatment or prevention of hepatitis delta virus infection
US9579377B2 (en) 2011-06-14 2017-02-28 Globeimmune, Inc. Yeast-based compositions and methods for the treatment or prevention of hepatitis delta virus infection
US10188714B2 (en) 2011-08-17 2019-01-29 Globeimmune, Inc. Yeast-MUC1 immunotherapeutic compositions and uses thereof
US9533031B2 (en) 2011-08-17 2017-01-03 Globeimmune, Inc. Yeast-MUC1 immunotherapeutic compositions and uses thereof
US11065318B2 (en) 2011-08-17 2021-07-20 Globeimmune, Inc. Yeast-MUC1 immunotherapeutic compositions and uses thereof
US9653272B2 (en) 2012-06-26 2017-05-16 Biodesix, Inc. Mass-spectral method for selection, and de-selection, of cancer patients for treatment with immune response generating therapies
US10593529B2 (en) 2012-06-26 2020-03-17 Biodesix, Inc. Mass-spectral method for selection, and de-selection, of cancer patients for treatment with immune response generating therapies
US10507235B2 (en) 2013-03-19 2019-12-17 Globeimmune, Inc. Yeast-based immunotherapy for Chordoma
US10849971B2 (en) 2013-03-26 2020-12-01 Globeimmune, Inc. Compositions and methods for the treatment or prevention of human immunodeficiency virus infection
US10265393B2 (en) 2013-03-26 2019-04-23 Globeimmune, Inc. Compositions and methods for the treatment or prevention of human immunodeficiency virus infection
US9782473B2 (en) 2013-03-26 2017-10-10 Globeimmune, Inc. Compositions and methods for the treatment or prevention of human immunodeficiency virus infection
US10722567B2 (en) 2013-08-30 2020-07-28 Globeimmune, Inc. Compositions and methods for the treatment or prevention of tuberculosis
US10226522B2 (en) 2013-08-30 2019-03-12 Globeimmune, Inc. Compositions and methods for the treatment or prevention of tuberculosis
US10799567B2 (en) 2014-04-11 2020-10-13 Globeimmune, Inc. Yeast-based immunotherapy and type I interferon sensitivity
US11304996B2 (en) 2014-04-11 2022-04-19 The Regents Of The University Of Colorado, A Body Corporate Yeast-based immunotherapy and type I interferon sensitivity
US11065313B2 (en) 2015-08-03 2021-07-20 Globeimmune, Inc. Modified yeast-brachyury immunotherapeutic compositions
US10987422B2 (en) 2016-07-11 2021-04-27 Rutgers, The State University Of New Jersey Fungal vaccine compositions and methods of use thereof
EP3481417A4 (en) * 2016-07-11 2020-04-01 Rutgers, The State University of New Jersey Fungal vaccine compositions and methods of use thereof

Also Published As

Publication number Publication date
US20100104604A1 (en) 2010-04-29
US8460919B2 (en) 2013-06-11
US20130344110A1 (en) 2013-12-26
US9333246B2 (en) 2016-05-10

Similar Documents

Publication Publication Date Title
US9333246B2 (en) Compositions and methods to elicit immune responses against pathogenic organisms using yeast-based vaccines
Schweizer et al. The TEA/ATTS transcription factor CaTec1p regulates hyphal development and virulence in Candida albicans
Alberti‐Segui et al. Identification of potential cell‐surface proteins in Candida albicans and investigation of the role of a putative cell‐surface glycosidase in adhesion and virulence
RU2559537C2 (en) Hyr1 as target for active and passive immunisation against candida
JP2002526082A (en) Human complement C3 degrading polypeptide from Streptococcus pneumoniae
JP7035063B2 (en) Recombinant BCG overexpressing phoP-phoR
JP5462164B2 (en) Immunogenic streptococcal protein
Porta et al. An Homologue of the Human 100-kDa Protein (p100) Is Differentially Expressed byHistoplasma capsulatumduring Infection of Murine Macrophages
Rooney et al. Selective expression of the virulence factor BAD1 upon morphogenesis to the pathogenic yeast form of Blastomyces dermatitidis: evidence for transcriptional regulation by a conserved mechanism
US7910351B2 (en) Mutant F. turlarensis strain and uses thereof
Fernandes et al. The combined use of Paracoccidioides brasiliensis Pb40 and Pb27 recombinant proteins enhances chemotherapy effects in experimental paracoccidioidomycosis
US20040147719A1 (en) Type III bacterial strains for use in medicine
US20210046169A1 (en) Fungal immunogens and related materials and methods
Meinhardt et al. Yeast killer toxins: Fundamentals and applications
Hester et al. Cross-reactivity between vaccine antigens from the chitin deacetylase protein family improves survival in a mouse model of cryptococcosis
He et al. Tex, a putative transcriptional accessory factor, is involved in pathogen fitness in Streptococcus pneumoniae
Areitio et al. Identification of Mucor circinelloides antigens recognized by sera from immunocompromised infected mice
KR101642499B1 (en) A recombinant fusion protein, an immunogenic composition, comprising water-soluble Pasteurella multocida toxin (PMT), and a method for preparing thereof
US20220111018A1 (en) Compositions and methods for inhibiting yeast infections
Kandel Killer systems and pathogenic yeasts
WO2006020898A2 (en) Methods of screening for inhibitors and compositions thereof for treating or preventing mycobacterium tuberculosis infection
WO2020257625A1 (en) Compositions and methods for inhibiting yeast infections
JP2003504030A (en) Novel antifungal and fungicides, their production and use
KR101803088B1 (en) Vaccine composition using streptococcus parauberis cytoplasmic membrance protein in fishes
Travassos et al. Paracoccidioidomycosis: advance towards a molecular vaccine

Legal Events

Date Code Title Description
AS Assignment

Owner name: MYCOLOGICS, INC., COLORADO

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SELITRENNIKOFF, CLAUDE P.;MILLER, TAMARA K.;REEL/FRAME:019078/0713;SIGNING DATES FROM 20070306 TO 20070309

AS Assignment

Owner name: GLOBEIMMUNE, INC., COLORADO

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNOR:MYCOLOGICS, INC.;REEL/FRAME:023260/0625

Effective date: 20090914

STCB Information on status: application discontinuation

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