CA2756760A1 - Compositions and methods for screening and using compounds antagonizing spore-surface interactions - Google Patents

Compositions and methods for screening and using compounds antagonizing spore-surface interactions Download PDF

Info

Publication number
CA2756760A1
CA2756760A1 CA2756760A CA2756760A CA2756760A1 CA 2756760 A1 CA2756760 A1 CA 2756760A1 CA 2756760 A CA2756760 A CA 2756760A CA 2756760 A CA2756760 A CA 2756760A CA 2756760 A1 CA2756760 A1 CA 2756760A1
Authority
CA
Canada
Prior art keywords
seq
spore
composition
protein
microbial
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
CA2756760A
Other languages
French (fr)
Inventor
David R. Macinga
James Edmund Bingham
Sarah L. Edmonds
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.)
Go-Jo Industries Inc
Original Assignee
Go-Jo Industries 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 Go-Jo Industries Inc filed Critical Go-Jo Industries Inc
Publication of CA2756760A1 publication Critical patent/CA2756760A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/33Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Clostridium (G)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/32Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Bacillus (G)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/12Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from bacteria
    • CCHEMISTRY; METALLURGY
    • C11ANIMAL OR VEGETABLE OILS, FATS, FATTY SUBSTANCES OR WAXES; FATTY ACIDS THEREFROM; DETERGENTS; CANDLES
    • C11DDETERGENT COMPOSITIONS; USE OF SINGLE SUBSTANCES AS DETERGENTS; SOAP OR SOAP-MAKING; RESIN SOAPS; RECOVERY OF GLYCEROL
    • C11D3/00Other compounding ingredients of detergent compositions covered in group C11D1/00
    • C11D3/16Organic compounds
    • C11D3/38Products with no well-defined composition, e.g. natural products
    • CCHEMISTRY; METALLURGY
    • C11ANIMAL OR VEGETABLE OILS, FATS, FATTY SUBSTANCES OR WAXES; FATTY ACIDS THEREFROM; DETERGENTS; CANDLES
    • C11DDETERGENT COMPOSITIONS; USE OF SINGLE SUBSTANCES AS DETERGENTS; SOAP OR SOAP-MAKING; RESIN SOAPS; RECOVERY OF GLYCEROL
    • C11D3/00Other compounding ingredients of detergent compositions covered in group C11D1/00
    • C11D3/48Medical, disinfecting agents, disinfecting, antibacterial, germicidal or antimicrobial compositions
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/02Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving viable microorganisms
    • C12Q1/18Testing for antimicrobial activity of a material

Abstract

The present invention provides genes encoding novel microbial proteins (i.e., for example, exosporium genes and proteins) that mediate the attachment of microbial spores to surfaces.
Specific fragments of these microbial spore proteins may be utilized to inhibit the attachment of microbial spores to surfaces, thereby providing an infection control agent. The invention pro-vides recombinant expression vectors comprising genes encoding exosporium proteins, as well as host cells containing these ex-pression vectors. Further provided herein are screening methods for identifying infection control compositions comprising ex-osporium proteins that inhibit bacterial spore attachment to either bodily tissues or solid surfaces. Additionally, the invention pro-vides for the use of nucleic acid inhibitors of exosporium protein expression by hybridizing with nucleic acid sequences encoding exosporium protiens as well as with exosprorium mRNA. The invention further describes monoclonal and polyclonal antibodies having affinity for exosporium protiens.

Description

Compositions And Methods For Screening And Using Compounds Antagonizing Spore-Surface Interactions Field Of Invention The present invention is related to the field of infection control.
Particularly, the invention is related to compositions and methods for maintenance of antiseptic conditions, bacterial decontamination, and/or bacterial exposure prophylaxis.
In one embodiment, the invention contemplates infection control compositions that inhibit microbial spore binding to a substrate surface. For example, the inhibition disrupts and/or displaces an interaction, attachment, and/or stabilization of a microbial spore to a substrate surface.

Background Spore-forming bacteria may be responsible for contaminations including, but not limited to, hospital infections, food spoilage and/or food-borne illness problems. As the production of minimally processed refrigerated products becomes more efficient and aseptic, background microflora are eliminated, and it is the spore-forming organism which eventually may limit the shelf-lives of these products. Spore-forming bacteria are suspected of being responsible for the spoilage of canned foods, bread, vacuum packed meats, pasteurized dairy products, and fruit juices. The presence of bacterial spores at high levels in ingredients going into any of these types of products may increase the potential for spoilage of the finished product.
In the healthcare environment, spore-forming bacteria frequently cause infections including nosocomial diarrhea, tetanus, and gangrene. Clostridium difficile is a spore forming anaerobic bacterium which causes antibiotic associated diarrhea and pseudomembraneous colitis. The recent emergence of a highly pathogenic epidemic strain with increased attributable morbidity and mortality has further increased the importance of this nosocomial pathogen. Reductions in hospital staff-to-patient infectious transmission would be expected to drastically improve recovery times thereby resulting in accelerated patient release. This is beneficial from both patient health and societal cost viewpoints. Despite efforts by the medical community to increase the use of antiseptics to prevent microbial transmission, the occurrence of patient infection and reinfection leads to unnecessary morbidity and mortality.

Bacteria that form spores that can reside in a dormant form for decades before germinating in a host cell and may be responsible for unexpected contamination, whether related to the food industry or in the practice of medicine. Furthermore, bacterial endospores are resistant to skin and surface disinfectants and thus current antimicrobial measures are largely ineffective. What is needed is a method to screen for infection control compositions that remove and/or prevent the attachment to, and subsequent infection of, a host by a microbial spore.

Summary The present invention is related to the field of infection control.
Particularly, the invention is related to compositions and methods for maintenance of antiseptic conditions, bacterial decontamination, and/or bacterial exposure prophylaxis.
In one embodiment, the invention contemplates infection control compositions that inhibit microbial spore binding to a substrate surface. For example, the inhibition antagonizes, disrupts, and/or displaces an interaction, attachment, and/or stabilization of a microbial spore to a substrate surface.
In one embodiment, the present invention contemplates an infection control composition capable of blocking and/or inhibiting the binding of a microbial spore to a substrate surface. In one embodiment, the composition comprises a small organic molecule. In one embodiment, the composition comprises a protein. In one embodiment, the peptide comprises a spore surface peptide.. In one embodiment, the peptide is derived from a Clostridia spore surface protein. In one embodiment, the Clostridia spore surface protein comprises a Clostridia difficile spore surface protein. In one embodiment, the peptide is derived from a Bacillus spore surface protein. In one embodiment, the Bacillus spore surface protein comprises a Bacillus anthracis spore surface protein. In one embodiment, the Clostridium difficile spore surface peptide comprises SEQ ID
NO: 1 or a homologue of SEQ ID NO: 1. In one embodiment, the spore surface peptide is selected from the group comprising SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, and SEQ ID
NO: 15. In one embodiment, the composition comprises an antibody. In one embodiment, the antibody is a polyclonal antibody. In one embodiment, the antibody is a monoclonal antibody. In one embodiment, the composition comprises a synthetic peptide. In one embodiment, the composition comprises an inorganic ion complex. In one embodiment, the composition comprises a protein mimetic. In one embodiment, the substrate surface comprises an inanimate surface. In one embodiment, comprises an
2 animate surface. In one embodiment, the animate surface comprises a body tissue surface. In one embodiment, the body tissue surface comprises an epithelial surface. In one embodiment, the epithelial surface comprises a mammalian skin surface. In one embodiment, the inanimate surface comprises stainless steel. In one embodiment, the inanimate surface comprises ceramic. In one embodiment, the inanimate surface comprises vinyl. In one embodiment, the inanimate surface comprises tile. In one embodiment, the composition further comprises at least one antimicrobial drug.
In one embodiment, the composition further comprises a carrier. In one embodiment, the carrier further comprises a detergent. In one embodiment, the carrier is sterile and suitable for human administration. In one embodiment, the carrier comprises an antimicrobial skin cleaner. In one embodiment, the skin cleaner comprises a handwash cleaner. In one embodiment, the carrier comprises an antimicrobial surface cleaner.
In one embodiment, the present invention contemplates an infection control composition comprising an amino acid sequence capable of blocking and/or inhibiting the binding of a microbial spore to a substrate surface, wherein the amino acid sequence has substantial homology to a microbial spore surface protein. In one embodiment, the substantial homology comprises approximately 75% homology, preferably 85%
homology, but most preferably 90% or 95% homology. In one embodiment, the surface protein comprises a spore surface protein. In one embodiment, the spore surface protein is a Clostridium spore surface protein. In one embodiment, the Clostridium spore surface protein is a C. difficile spore surface protein. In one embodiment, the spore surface protein is a Bacillus spore surface protein. In one embodiment, the Bacillus spore surface protein is a B. anthracis spore surface protein. In one embodiment, the spore surface protein comprises a spore surface peptide.. In one embodiment, the peptide is derived from a Clostridia surface protein. In one embodiment, the Clostridia surface protein comprises a Clostridia difficile surface protein. In one embodiment, the peptide is derived from a Bacillus surface protein. In one embodiment, the Bacillus surface protein comprises a Bacillus anthracis surface protein. In one embodiment, the Clostridium difficile spore surface peptide comprises SEQ ID NO: 1 or a homologue of SEQ
ID NO:
1. In one embodiment, the spore surface peptide is selected from the group comprising SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, and SEQ ID NO: 15. In one embodiment, the amino acid sequence is derived from a C-terminal end of the spore surface protein. In one embodiment, the amino acid sequence is derived from an N-terminal end of a spore surface protein. In one embodiment, the composition further
3 comprises a carrier. In one embodiment, the carrier comprises a detergent. In one embodiment, the composition further comprises at least one antimicrobial drug.
In one embodiment, the carrier is suitable for human administration. In one embodiment, the composition further comprises an antimicrobial skin cleaner. In one embodiment, the skin cleaner is a handwash cleaner. In one embodiment, the composition further comprises an antimicrobial surface cleaner. In one embodiment, the composition further comprises a skin barrier. In one embodiment, the composition further comprises a surface barrier.
In one embodiment, the present invention contemplates an infection control composition comprising an antagonistic compound, wherein the compound has affinity for a microbial spore surface protein amino acid sequence. In one embodiment, the affinity is directed at a substrate surface specific binding site region. In one embodiment, the compound comprises an antibody. In one embodiment, the compound comprises a small organic molecule. In one embodiment, the spore surface protein amino acid sequence comprises a peptide fragment. In one embodiment, the peptide fragment is derived from a C-terminal end of the spore surface protein. In one embodiment, the peptide fragment is derived from an N-terminal end of the spore surface protein. In one embodiment, the spore surface protein is a Clostridium spore surface protein.
In one embodiment, the Clostridium spore surface protein is a C. difficile spore surface protein.
In one embodiment, the spore surface protein is a Bacillus spore surface protein. In one embodiment, the Bacillus spore surface protein is a B. anthracis spore surface protein. In one embodiment, the composition further comprises at least one antimicrobial drug. In one embodiment, the composition further comprises a carrier. In one embodiment, the carrier is suitable for human administration. In one embodiment, the composition further comprises an antimicrobial hand cleaner. In one embodiment, the composition further comprises an antimicrobial surface cleaner.
In one embodiment, the present invention contemplates a method, comprising: a) providing: i) a subject at risk for exposure to a microbe, wherein the exposure is likely to result in a microbial infection; and ii) an infection control composition; b) administering the composition to the subject before the microbial exposure, under conditions such that a microbial infection barrier is formed. In one embodiment, the microbial infection barrier reduces the microbial infection. In one embodiment, the microbial infection barrier prevents the microbial infection. In one embodiment, the microbe comprises a bacteria.
In one embodiment, the bacteria is selected from the group comprising Clostridia,
4 Bacillus, Desulfotomaculum, Sporolactobacillus, Sporosarcina, or Thermoactinomyces.
In one embodiment, the bacteria comprises a bacterial spore. In one embodiment, the bacterial spore comprises at least one spore surface protein. In one embodiment, the Clostridium spore surface protein is a C. diffcile spore surface protein. In one embodiment, the Bacillus spore surface protein is a B. anthracis spore surface protein. In one embodiment, the infection control composition comprises an amino acid sequence.
In one embodiment, the amino acid sequence is derived from a microbial spore surface protein. In one embodiment, the amino acid sequence has substantial homology to a microbial spore surface protein. In one embodiment, the infection control composition comprises is capable of inhibiting, antagonizing, disrupting, displacing, and/or blocking the binding of a microbial spore to a substrate surface. In one embodiment, the infection control composition further comprises an antagonistic compound, wherein the compound has affinity for a microbial spore surface protein amino acid sequence. In one embodiment, the composition further comprises a carrier. In one embodiment, the infection control composition further comprises at least one antimicrobial drug. In one embodiment, the subject at risk is a healthcare worker. In one embodiment, the subject at risk is a medical first responder. In one embodiment, the subject at risk is a medical patient. In one embodiment, the patient is in a healthcare setting. In one embodiment, the medical patient has been administered at least one antibiotic. In one embodiment, the subject at risk is a fireman. In one embodiment, the subject at risk is a policeman. In one embodiment, the subject at risk is a military personnel. In one embodiment, the subject at risk is a food handler. In one embodiment, the subject at risk is an inanimate surface and/or object. In one embodiment, the administering is selected from the group comprising topical, oral, parenteral, pulmonary, anal, vaginal, ocular, or intranasal.
In one embodiment, the present invention contemplates a method, comprising: a) providing: i) a subject having been exposed to a microbe, wherein the exposure resulted in a microbial infection; ii) an infection control composition; b) administering the composition to the subject after the microbial exposure, under conditions such that the microbial infection is reduced. In one embodiment, the microbe comprises a bacteria. In one embodiment, the bacteria is selected from the group comprising Clostridia, Bacillus, Desulfotomaculum, Sporolactobacillus, Sporosarcina, or Thermoactinomyees. In one embodiment, the bacteria comprises a bacterial spore. In one embodiment, the bacterial spore comprises at least one spore surface protein. In one embodiment, the Clostridium spore surface protein is a C. diff cile spore surface protein. In one embodiment, the Bacillus spore surface protein is a B. anthracis spore surface protein. In one embodiment, the infection control composition comprises an amino acid sequence. In one embodiment, the amino acid sequence is derived from a microbial spore surface protein.
In one embodiment, the amino acid sequence has substantial homology to a microbial spore surface protein. In one embodiment, the infection control composition is capable of inhibiting, antagonizing, disrupting, displacing and/or blocking the binding of a microbial spore to a substrate surface. In one embodiment, the infection control composition further comprises an antagonistic compound, wherein the compound has affinity for a microbial spore surface protein amino acid sequence. In one embodiment, the composition further comprises a carrier. In one embodiment, the infection control composition further comprises at least one antimicrobial drug. In one embodiment, the exposed subject is a healthcare worker. In one embodiment, the exposed subject is a medical first responder. In one embodiment, the exposed subject is a fireman.
In one embodiment, the exposed subject is a policeman. In one embodiment, the exposed subject is a military personnel. In one embodiment, the administering is selected from the group comprising topical, oral, parenteral, pulmonary, anal, vaginal, ocular, or intranasal.
In one embodiment, the exposed subject is an inanimate surface and/or object.
In one embodiment, the present invention contemplates a screening method comprising: a) providing; i) an isolated peptide, wherein the peptide is derived from a microbial spore surface protein; and ii) an infection control test compound suspected of having an interaction with the peptide; b) contacting the peptide with the infection control test compound; and c) detecting the interaction of the peptide with the infection control test compound. In one embodiment, the method further comprises a substrate capable of attaching the isolated peptide. In one embodiment, the substrate surface comprises pigskin. In one embodiment, the peptide is derived from a Clostridia spore surface protein. In one embodiment, the Clostridia spore surface protein comprises a Clostridia diffcile exosporium. In one embodiment, the peptide is derived from a Bacillus spore surface protein. In one embodiment, the Bacillus exosporium comprises a Bacillus anthracis spore surface protein. In one embodiment, the peptide comprises a spore surface peptide. In one embodiment, the Clostridium difficile spore surface peptide comprises SEQ ID NO: 1 or a homologue of SEQ ID NO: 1. In one embodiment, the spore surface peptide is selected from the group comprising SEQ ID NO: 3, SEQ
ID NO:
5, SEQ ID NO: 7, and SEQ ID NO: 15. In one embodiment, the infection control test compound comprises a small organic molecule. In one embodiment, the infection control
6 test compound comprises a synthetic peptide. In one embodiment, the infection control test compound comprises a protein mimetic. In one embodiment, the infection control test compound comprises an antibody.
In one embodiment, the present invention contemplates a method, comprising: a) providing; i) an isolated labeled microbial spore surface peptide, wherein said peptide creates a first fluorescent intensity pattern; and ii) an infection control test compound capable of interacting with said peptide; b) contacting said peptide with said compound to produce a contacted peptide having a second fluorescent intensity pattern; c) comparing said first intensity pattern with the second fluorescence intensity pattern.
In one embodiment, the method further comprises step d) detecting a difference between the first intensity pattern and the second intensity pattern. In one embodiment, the difference indicates that said compound binds to said peptide. In one embodiment, the peptide is derived from a bacterial spore surface protein. In one embodiment, the bacterial spore surface protein is derived from a Clostridia spore surface protein. In one embodiment, the Clostridia spore surface protein comprises Clostridia difficile spore surface protein.
In one embodiment, the bacterial spore surface protein is derived from a Bacillus spore surface protein. In one embodiment, the Bacillus spore surface protein comprises Bacillus anthracis spore surface protein. In one embodiment, the spore surface protein comprises a peptide fragment. In one embodiment, the peptide is selected from the group comprising SEQ ID NO: I, SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, or SEQ ID
NO: 9. In one embodiment, the label is selected from the group consisting of 4'-6-diamidino-2-phenylindole (DAPI), anilinonapthalene sulfonate (ANS), bis-ANS
(Bis-anilinonapthalene sulfonate), N-phenyl- I -naphthalene (NPN), ruthenium red, cresol violet, and 4-(dicyanovinyl)julolidine (DCVJ).
In one embodiment, the present invention contemplates a method of screening for a compound, comprising: a) providing; i) a bacteria comprising a recombinant expression vector, wherein said vector comprises at least a portion of a spore surface oligonucleotide sequence; and ii) a compound suspected of having infection control activity;
b) expressing said vector such that a spore surface protein is displayed on the bacterial membrane; c) contacting said bacteria with said compound; and d) detecting infection control activity of said compound. In one embodiment, the infection control activity comprises forming a binding pair of said compound with said displayed protein.
In one embodiment, the oligonucleotide sequence comprises SEQ ID NO: 2. In one embodiment, the oligonucleotide sequence selected from the group comprising SEQ ID
7 NO: 4, SEQ ID NO: 6, SEQ ID NO: 8, or SEQ ID NO: 16. In one embodiment, the recombinant expression vector further comprises a fusion sequence, wherein the fusion sequence comprises reporter sequence. In one embodiment, the reporter sequence is a galactosidase sequence.

In one embodiment, the present invention contemplates a method for detecting the presence of polynucleotide sequences encoding at least a portion of a microbial spore surface protein gene in a sample, the method comprising: a) providing; i) at least a portion of SEQ ID NO: 2; and ii) a sample suspected of containing a spore surface oligonucleotide sequence; b) combining the SEQ ID NO: 2 portion and the sample under conditions such that a hybridization complex is formed between the SEQ ID NO:

portion and the spore surface oligonucleotide sequence; and c) detecting the hybridization complex. In one embodiment, the spore surface oligonucleotide is derived from a microbial spore. In one embodiment, the spore is derived from a Clostridia species. In one embodiment, the spore is derived from a Bacillus species. In one embodiment, the oligonucleotide sequence is RNA. In one embodiment, the oligonucleotide sequence is DNA.
In one embodiment, the present invention contemplates a method comprising;
providing an infection control composition and a substrate surface comprising bacterial spores; applying the composition to the surface, wherein at least 50%, preferably 60%, more preferably 70%, even more preferably 80%, most preferably 90%, or 100% of the spores are removed from the surface. In one embodiment, the surface may be selected from the group consisting of skin, body tissues, inorganic surfaces (i.e., for example, stainless steel, granite, plastic, tile, ceramic etc), gloves, laboratory coats, or medical instruments.
In one embodiment, the present invention contemplates a method comprising;
providing an infection control composition and a substrate surface at high risk of bacterial spore binding; applying the composition to the surface, wherein at least 50%, preferably 60%, more preferably 70%, even more preferably 80%, most preferably 90%, or 100% of bacterial spore binding is prevented. In one embodiment, the surface may be selected from the group consisting of skin, body tissues, inorganic surfaces (i.e., for example, stainless steel, granite, plastic, tile, ceramic etc.) or medical instruments.
In one embodiment, the present invention contemplates a method of screening a plurality of infection control compositions for binding to a microbial spore protein,
8 comprising: a) providing a chromatography column comprising beads carrying said microbial spore proteins; b) contacting said plurality of infection control compositions with said beads under conditions such that at least one of said infection control compositions form a binding pair with said beads; c) controllably releasing said at least one bound infection control composition from said bead; d) detecting said at least one bound infection control composition. In one embodiment, said binding pair comprises a microbial spore surface protein. In one embodiment, the detecting of the at least one bound infection control composition comprises the production of a fluorescent signal. In one embodiment, the detecting of the at least one bound infection control composition comprises the formation of an enzyme-substrate product. In one embodiment, the step of controllably releasing comprises exposing the beads to a releasing agent. In one embodiment, the releasing agent is selected from a group consisting of light, acid and base.
In one embodiment, the present invention contemplates an infection control composition comprising a protein mimetic compound and a carrier, wherein the protein mimetic compound has a first specific binding site pattern on a substrate surface wherein the first binding pattern has at least 50% identity, preferably 60% identity, more preferably 70% identity, even more preferably 80% identity, most preferably 90%
identity, or 100% identity to a second specific binding site pattern of an amino acid sequence, wherein the sequence has substantial homology to a microbial spore surface protein. In one embodiment, the protein mimetic compound comprises a small organic molecule. In one embodiment, the protein mimetic compound comprises a synthetic peptide. In one embodiment, the protein mimetic compound comprises an inorganic ion complex. In one embodiment, the surface protein comprises a spore surface protein. In one embodiment, the microbial spore surface protein is derived from a Clostridium spore surface protein. In one embodiment, the Clostridium spore surface protein is a C. difficile spore surface protein. In one embodiment, the spore surface protein is derived from a Bacillus spore surface protein. In one embodiment, the Bacillus spore surface protein is a B. anthracis spore surface protein. In one embodiment, the amino acid sequence is derived from a C-terminal end of the microbial spore surface protein. In one embodiment, the amino acid sequence is derived from a N-terminal end of the microbial spore surface protein. In one embodiment, the substrate surface comprises an animate surface. In one embodiment, the substrate surface comprises an inanimate surface. In one embodiment, the animate surface comprises a body tissue surface. In one
9 embodiment, the body tissue surface comprises a mammalian skin surface. In one embodiment, the inanimate surface comprises stainless steel. In one embodiment, the inanimate surface comprises ceramic. In one embodiment, the inanimate surface comprises vinyl. In one embodiment, the inanimate surface comprises tile. In one embodiment, the carrier comprises a detergent. In one embodiment, the composition further comprises at least one antimicrobial drug. In one embodiment, the carrier is sterile and suitable for human administration. In one embodiment, the composition further comprises an antimicrobial skin cleaner. In one embodiment, the composition further comprises an antimicrobial surface cleaner.

Definitions The term "pharmaceutically" or "pharmacologically acceptable", as used herein, refer to molecular entities and compositions that do not produce adverse, allergic, or other untoward reactions when administered to an animal or a human.
The term, "pharmaceutically acceptable carrier", as used herein, includes any and all solvents, or a dispersion medium including, but not limited to, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils, coatings, isotonic and absorption delaying agents, liposome, commercially available cleansers, and the like.
Supplementary bioactive ingredients also can be incorporated into such carriers.
The term, "purified" or "isolated", as used herein, may refer to a peptide composition that has been subjected to treatment (i.e., for example, fractionation) to remove various other components, and which composition substantially retains its expressed biological activity. Where the term "substantially purified" is used, this designation will refer to a composition in which the protein or peptide forms the major component of the composition, such as constituting about 50%, about 60%, about 70%, about 80%, about 90%, about 95% or more of the composition (i.e., for example, weight/weight and/or weight/volume). The term "purified to homogeneity" is used to include compositions that have been purified to 'apparent homogeneity" such that there is single protein species (i.e., for example, based upon SDS-PAGE or HPLC
analysis). A
purified composition is not intended to mean that some trace impurities may remain.
As used herein, the term "substantially purified" refers to molecules, either nucleic or amino acid sequences, that are removed from their natural environment, isolated or separated, and are at least 60% free, preferably 75% free, and more preferably 90% free from other components with which they are naturally associated. An "isolated polynucleotide" is therefore a substantially purified polynucleotide.
"Nucleic acid sequence" and "nucleotide sequence" as used herein refer to an oligonucleotide or polynucleotide, and fragments or portions thereof, and to DNA or RNA of genomic or synthetic origin which may be single- or double-stranded, and represent the sense or antisense strand.
The term "an isolated nucleic acid", as used herein, refers to any nucleic acid molecule that has been removed from its natural state (e.g., removed from a cell and is, in a preferred embodiment, free of other genomic nucleic acid).
The term "functionally equivalent codon", as used herein, refers to different codons that encode the same amino acid. This phenomenon is often referred to as "degeneracy" of the genetic code. For example, six different codons encode the amino acid arginine.
The terms "amino acid sequence" and "polypeptide sequence" as used herein, are interchangeable and to refer to a sequence of amino acids.
The term "spore surface protein" as used herein, refers to any protein or peptide fragment that is derived from, or has substantial similarity (i.e., is homologous) to, any bacterial spore exoporium protein. Microbial spore surface proteins are believed responsible for attaching, and immobilizing, a microbial spore to an inanimate and/or animate surface until conditions are sufficient to induce germination.
The term "derived from" as used herein, refers to the source of a compound or sequence. In one respect, a compound or sequence may be derived from an organism or particular species. In another respect, a compound or sequence may be derived from a larger complex or sequence.
As used herein, "BcIA" or "Bc1A polypeptide" or "BelA protein" or "Bc1A
homologue" are used interchangeably to refer to the amino acid sequence of substantially purified Bacillus-like protein, obtained from any species, particularly bacterial species which include gram negative, gram positive, aerobic, and anaerobic bacteria, and obtained from any source whether natural, synthetic, semi-synthetic or recombinant. For example, a Clostridium difficile BcIA homologue would have the same function and activity as a Bacillus anthracis BcIA peptide.
A "variant" of a protein is defined as an amino acid sequence which differs by one or more amino acids from a polypeptide sequence (i.e., for example, SEQ ID NO:
1) or any homolog of the polypeptide sequence. The variant may have "conservative"
changes, wherein a substituted amino acid has similar structural or chemical properties, e.g., replacement of leucine with isoleucine. More rarely, a variant may have "nonconservative" changes, e.g., replacement of a glycine with a tryptophan.
Similar minor variations may also include amino acid deletions or insertions (i.e., additions), or both. Guidance in determining which and how many amino acid residues may be substituted, inserted or deleted without abolishing biological or immunological activity may be found using computer programs including, but not limited to, DNAStara' software.
A "variant" of a nucleotide is defined as a novel nucleotide sequence which differs from a reference oligonucleotide by having deletions, insertions and substitutions. These may be detected using a variety of methods (e.g., sequencing, hybridization assays etc.).
Included within this definition are alterations to the genomic DNA sequence which encodes a Clostridium spore surface protein (i.e., for example, by alterations in the pattern of restriction enzyme fragments capable of hybridizing to SEQ ID NO: 2 (RFLP
analysis), the inability of a selected fragment of SEQ ID NO: 2 to hybridize under high stringency conditions to a sample of genomic DNA (e.g., using allele-specific oligonucleotide probes), and improper or unexpected hybridization, such as hybridization to a locus other than the normal chromosomal locus for a Clostridium spore surface gene (e.g., using fluorescent in situ hybridization (FISH)).
A "deletion" is defined as a change in either nucleotide or amino acid sequence in which one or more nucleotides or amino acid residues, respectively, are absent.
An "insertion" or "addition" is that change in a nucleotide or amino acid sequence which has resulted in the addition of one or more nucleotides or amino acid residues, respectively, as compared to, for example, the naturally occurring Clostridium spore surface protein.
A "substitution" results from the replacement of one or more nucleotides or amino acids by different nucleotides or amino acids, respectively.
The term "derivative" as used herein, refers to any chemical modification of a nucleic acid or an amino acid. Illustrative of such modifications would be replacement of hydrogen by an alkyl, acyl, or amino group. For example, a nucleic acid derivative would encode a polypeptide which retains essential biological characteristies.
As used herein the term "portion" when in reference to a protein (as in "a portion of a given protein") refers to fragments of that protein. The fragments may range in size from four amino acid residues to the entire amino acid sequence minus one amino acid.

For example, a protein "comprising at least a portion of the amino acid sequence of SEQ
ID NO: 1" encompasses a full-length Clostridium spore surface protein and fragments thereof of four (4) amino acids or more.
The term "portion" when used in reference to a nucleotide sequence refers to fragments of that nucleotide sequence. The fragments may range in size from 5 nucleotide residues to the entire nucleotide sequence minus one nucleic acid residue.
The term "biologically active" refers to any molecule having structural, regulatory or biochemical functions. For example, spore surface protein biological activity may be determined, for example, by restoration of wild-type growth in cells lacking spore surface protein activity (i.e., for example, spore surface protein null cells and/or "knock out"
cells). Cells lacking spore surface protein activity may be produced by many methods (i.e., for example, point mutation and frame-shift mutation). Complementation is achieved by transfecting cells which lack spore surface protein activity with an expression vector which expresses spore surface protein, a derivative thereof, or a portion thereof.
The term "immunologically active" defines the capability of a natural, recombinant or synthetic peptide (i.e., for example, a spore surface protein), or any oligopeptide thereof, to induce a specific immune response in appropriate animals or cells and/or to bind with specific antibodies.
The term "antigenic determinant" as used herein refers to that portion of a molecule that is recognized by a particular antibody (i.e., an epitope). When a protein or fragment of a protein is used to immunize a host animal, numerous regions of the protein may induce the production of antibodies which bind specifically to a given region or three-dimensional structure on the protein; these regions or structures are referred to as antigenic determinants. An antigenic determinant may compete with the intact antigen (i.e., the immunogen used to elicit the immune response) for binding to an antibody.
The terms "immunogen," "antigen," "immunogenic" and "antigenic" refer to any substance capable of generating antibodies when introduced into an animal. By definition, an immunogen must contain at least one epitope (the specific biochemical unit capable of causing an immune response), and generally contains many more. Proteins are most frequently used as immunogens, but lipid and nucleic acid moieties complexed with proteins may also act as immunogens. The latter complexes are often useful when smaller molecules with few epitopes do not stimulate a satisfactory immune response by themselves.

The term "antibody" refers to immunoglobulin evoked in animals by an immunogen (antigen). It is desired that the antibody demonstrates specificity to epitopes contained in the immunogen. The term "polyclonal antibody" refers to inimunoglobulin produced from more than a single clone of plasma cells; in contrast "monoclonal antibody" refers to immunoglobulin produced from a single clone of plasma cells.
The terms "specific binding" or "specifically binding" when used in reference to the interaction of an antibody and a protein or peptide means that the interaction is dependent upon the presence of a particular structure (i.e., for example, an antigenic determinant or epitope) on a protein; in other words an antibody is recognizing and binding to a specific protein structure rather than to proteins in general.
For example, if an antibody is specific for epitope "A", the presence of a protein containing epitope A (or free, unlabelled A) in a reaction containing labeled "A" and the antibody will reduce the amount of labeled A bound to the antibody.
The term "recombinant DNA molecule" as used herein refers to a DNA molecule which is comprised of segments of DNA joined together by means of molecular biological techniques.
The term "recombinant protein" or "recombinant polypeptide" as used herein refers to a protein molecule which is expressed using a recombinant DNA
molecule.
As used herein, the terms "vector" and "vehicle" are used interchangeably in reference to nucleic acid molecules that transfer DNA segment(s) from one cell to another.
The term "expression vector" or "expression cassette" as used herein refers to a recombinant DNA molecule containing a desired coding sequence and appropriate nucleic acid sequences necessary for the expression of the operably linked coding sequence in a particular host organism. Nucleic acid sequences necessary for expression in prokaryotes usually include a promoter, an operator (optional), and a ribosome binding site, often along with other sequences. Eukaryotic cells are known to utilize promoters, enhancers, and termination and polyadenylation signals.
The terms "in operable combination", "in operable order" and "operably linked"
as used herein refer to the linkage of nucleic acid sequences in such a manner that a nucleic acid molecule capable of directing the transcription of a given gene and/or the synthesis of a desired protein molecule is produced. The term also refers to the linkage of amino acid sequences in such a manner so that a functional protein is produced.

The term "transfection" as used herein refers to the introduction of foreign DNA
into cells. Transfection may be accomplished by a variety of means known to the art including calcium phosphate-DNA co-precipitation, DEAE-dextran-mediated transfection, polybrene-mediated transfection, electroporation, microinjection, liposome fusion, lipofection, protoplast fusion, retroviral infection, biolistics (i.e., particle bombardment) and the like.
As used herein, the terms "complementary" or "complementarity" are used in reference to "polynucleotides" and "oligonucleotides" (which are interchangeable terms that refer to a sequence of nucleotides) related by the base-pairing rules.
For example, the sequence "C-A-G-T," is complementary to the sequence "G-T-C-A."
Complementarity can be "partial" or "total." "Partial" complementarity is where one or more nucleic acid bases is not matched according to the base pairing rules. "Total" or "complete"
complementarity between nucleic acids is where each and every nucleic acid base is matched with another base under the base pairing rules. The degree of complementarity between nucleic acid strands has significant effects on the efficiency and strength of hybridization between nucleic acid strands. This is of particular importance in amplification reactions, as well as detection methods which depend upon binding between nucleic acids.
The terms "homology" and "homologous" as used herein in reference to nucleotide sequences refer to a degree of complementarity with other nucleotide sequences. There may be partial homology or complete homology (i.e., identity). A
nucleotide sequence which is partially complementary, i.e., "substantially homologous,"
to a nucleic acid sequence is one that at least partially inhibits a completely complementary sequence from hybridizing to a target nucleic acid sequence. The inhibition of hybridization of the completely complementary sequence to the target sequence may be examined using a hybridization assay (Southern or Northern blot, solution hybridization and the like) under conditions of low stringency. A
substantially homologous sequence or probe will compete for and inhibit the binding (i.e., the hybridization) of a completely homologous sequence to a target sequence under conditions of low stringency. This is not to say that conditions of low stringency are such that non-specific binding is permitted; low stringency conditions require that the binding of two sequences to one another be a specific (i.e., selective) interaction.
The absence of non-specific binding may be tested by the use of a second target sequence which lacks even a partial degree of complementarity (e.g., less than about 30% identity);
in the absence of non-specific binding the probe will not hybridize to the second non-complementary target.
The terms "homology" and "homologous" as used herein in reference to amino acid sequences refer to the degree of identity of the primary structure between two amino acid sequences. Such a degree of identity maybe directed a portion of each amino acid sequence, or to the entire length of the amino acid sequence. Two or more amino acid sequences that are "substantially homologous" may have at least 50% identity, preferably at least 75% identity, more preferably at least 85% identity, most preferably at least 95%, or 100% identity.
An oligonucleotide sequence which is a "homolog" of a Clostridium spore surface protein nucleic acid sequence (i.e., for example, SEQ ID NO: 2) is defined herein as an oligonucleotide sequence which exhibits greater than or equal to 50% identity to the sequence of SEQ ID NO: 2 when sequences having a length of 100 bp or larger are compared. Alternatively, a homolog of SEQ ID NO: 2 is defined as an oligonucleotide sequence which encodes a biologically active Clostridium spore surface protein amino acid sequence. For example, a Clostridium spore surface protein homolog may comprise a portion of an oligonucleotide sequence encoding a spore surface protein.
Low stringency conditions comprise conditions equivalent to binding or hybridization at 42 C in a solution consisting of 5 x SSPE (43.8 g/l NaCl, 6.9 g/1 NaH2PO4'H20 and 1.85 g/l EDTA, pH adjusted to 7.4 with NaOH), 0.1% SDS, 5x Denhardt's reagent {50x Denhardt's contains per 500 ml: 5 g Ficoll (Type 400, Pharmacia), 5 g BSA (Fraction V; Sigma)} and 100 g/ml denatured salmon sperm DNA
followed by washing in a solution comprising 5x SSPE, 0.1 % SDS at 42 C when a probe of about 500 nucleotides in length. is employed. Numerous equivalent conditions may also be employed to comprise low stringency conditions; factors such as the length and nature (DNA, RNA, base composition) of the probe and nature of the target ( DNA, RNA, base composition, present in solution or immobilized, etc.) and the concentration of the salts and other components (e.g., the presence or absence of formamide, dextran sulfate, polyethylene glycol), as well as components of the hybridization solution may be varied to generate conditions of low stringency hybridization different from, but equivalent to, the above listed conditions. In addition, conditions which promote hybridization under conditions of high stringency (e.g., increasing the temperature of the hybridization and/or wash steps, the use of formamide in the hybridization solution, etc.) may also be used.

As used herein, the term "hybridization" is used in reference to the pairing of complementary nucleic acids using any process by which a strand of nucleic acid joins with a complementary strand through base pairing to form a hybridization complex.
Hybridization and the strength of hybridization (i.e., the strength of the association between the nucleic acids) is impacted by such factors as the degree of complementarity between the nucleic acids, stringency of the conditions involved, the T,,, of the formed hybrid, and the G:C ratio within the nucleic acids.
As used herein the term "hybridization complex" refers to a complex formed between two nucleic acid sequences by virtue of the formation of hydrogen bounds between complementary G and C bases and between complementary A and T bases;
these hydrogen bonds may be further stabilized by base stacking interactions. The two complementary nucleic acid sequences hydrogen bond in an antiparallel configuration. A
hybridization complex may be formed in solution (e.g., Co t or Ro t analysis) or between one nucleic acid sequence present in solution and another nucleic acid sequence immobilized to a solid support (e.g., a nylon membrane or a nitrocellulose filter as employed in Southern and Northern blotting, dot blotting or a glass slide as employed in in situ hybridization, including FISH (fluorescent in situ hybridization)).
As used herein, the term "T,,, " is used in reference to the "melting temperature."
The melting temperature is the temperature at which a population of double-stranded nucleic acid molecules becomes half dissociated into single strands. As indicated by standard references, a simple estimate of the T,,, value may be calculated by the equation:
Tm = 81.5 + 0.41 (% G+C), when a nucleic acid is in aqueous solution at lM
NaCl.
Anderson et al., "Quantitative Filter Hybridization" In: Nucleic Acid Hybridization (1985). More sophisticated computations take structural, as well as sequence characteristics, into account for the calculation of T,,,.
As used herein the term "stringency" is used in reference to the conditions of temperature, ionic strength, and the presence of other compounds such as organic solvents, under which nucleic acid hybridizations are conducted. "Stringency"
typically occurs in a range from about T,,, to about 20 C to 25 C below T. A "stringent hybridization" can be used to identify or detect identical polynucleotide sequences or to identify or detect similar or related polynucleotide sequences. For example, when fragments of SEQ ID NO: 2 are employed in hybridization reactions under stringent conditions the hybridization of fragments of SEQ ID NO: 2 which contain unique sequences (i.e., regions which are either non-homologous to or which contain less than about 50% homology or complementarity with SEQ ID NOs: 2) are favored.
Alternatively, when conditions of "weak" or "low" stringency are used hybridization may occur with nucleic acids that are derived from organisms that are genetically diverse (i.e., for example, the frequency of complementary sequences is usually low between such organisms).
As used herein, the term "amplifiable nucleic acid" is used in reference to nucleic acids which maybe amplified by any amplification method. It is contemplated that "amplifiable nucleic acid" will usually comprise "sample template."
As used herein, the term "sample template" refers to nucleic acid originating from a sample which is analyzed for the presence of a target sequence of interest.
In contrast, "background template" is used in reference to nucleic acid other than sample template which may or may not be present in a sample. Background template is most often inadvertent. It may be the result of carryover, or it may be due to the presence of nucleic acid contaminants sought to be purified away from the sample. For example, nucleic acids from organisms other than those to be detected may be present as background in a test sample.
"Amplification" is defined as the production of additional copies of a nucleic acid sequence and is generally carried out using polymerase chain reaction.
Dieffenbach C.
W. and G. S. Dveksler (1995) In: PCR Primer, a Laboratory Manual, Cold Spring Harbor Press, Plainview, N.Y.
As used herein, the term "polymerase chain reaction" ("PCR") refers to the method of K. B. Mullis U.S. Pat. Nos. 4,683,195 and 4,683,202, herein incorporated by reference, which describe a method for increasing the concentration of a segment of a target sequence in a mixture of genomic DNA without cloning or purification.
The length of the amplified segment of the desired target sequence is determined by the relative positions of two oligonucleotide primers with respect to each other, and therefore, this length is a controllable parameter. By virtue of the repeating aspect of the process, the method is referred to as the "polymerase chain reaction" (hereinafter "PCR").
Because the desired amplified segments of the target sequence become the predominant sequences (in terms of concentration) in the mixture, they are said to be "PCR
amplified". With PCR, it is possible to amplify a single copy of a specific target sequence in genomic DNA
to a level detectable by several different methodologies (e.g., hybridization with a labeled probe; incorporation of biotinylated primers followed by avidin-enzyme conjugate detection; incorporation of 32P-labeled deoxynucleotide triphosphates, such as dCTP or dATP, into the amplified segment). In addition to genomic DNA, any oligonucleotide sequence can be amplified with the appropriate set of primer molecules. In particular, the amplified segments created by the PCR process itself are, themselves, efficient templates for subsequent PCR amplifications.
As used herein, the term "primer" refers to an oligonucleotide, whether occurring naturally as in a purified restriction digest or produced synthetically, which is capable of acting as a point of initiation of synthesis when placed under conditions in which synthesis of a primer extension product which is complementary to a nucleic acid strand is induced, (i.e., in the presence of nucleotides and an inducing agent such as DNA
polymerase and at a suitable temperature and pH). The primer is preferably single stranded for maximum efficiency in amplification, but may alternatively be double stranded. If double stranded, the primer is first treated to separate its strands before being used to prepare extension products. Preferably, the primer is an oligodeoxyribonucleotide.
The primer must be sufficiently long to prime the synthesis of extension products in the presence of the inducing agent. The exact lengths of the primers will depend on many factors, including temperature, source of primer and the use of the method.
As used herein, the term "probe" refers; to an oligonucleotide (i.e., a sequence of nucleotides), whether occurring naturally as in a purified restriction digest or produced synthetically, recombinantly or by PCR amplification, which is capable of hybridizing to another oligonucleotide of interest. A probe may be single-stranded or double-stranded.
Probes are useful in the detection, identification and isolation of particular gene sequences. It is contemplated that any probe used in the present invention will be labeled with any "reporter molecule," so that is detectable in any detection system, including, but not limited to enzyme (e.g., ELISA, as well as enzyme-based histochemical assays), fluorescent, radioactive, and luminescent systems. It is not intended that the present invention be limited to any particular detection system or label.
As used herein, the terms "restriction endonucleases" and "restriction enzymes"
refer to bacterial enzymes, each of which cut double-stranded DNA at or near a specific nucleotide sequence.
DNA molecules are said to have "5' ends" and "3' ends" because mononucleotides are reacted to make oligonucleotides in a manner such that the 5' phosphate of one mononucleotide pentose ring is attached to the 3' oxygen of its neighbor in one direction via a phosphodiester linkage. Therefore, an end of an oligonucleotide is referred to as the "5' end" if its 5' phosphate is not linked to the 3' oxygen of a mononucleotidc pentose ring. An end of an oligonucleotide is referred to as the "3' end" if its 3' oxygen is not linked to a 5' phosphate of another mononucleotide pentose ring. As used herein, a nucleic acid sequence, even if internal to a larger oligonucleotide, also may be said to have 5' and 3' ends. In either a linear or circular DNA molecule, discrete elements are referred to as being "upstream" or 5' of the "downstream" or 3' elements. This terminology reflects the fact that transcription proceeds in a 5' to 3' fashion along the DNA strand. The promoter and enhancer elements which direct transcription of a linked gene are generally located 5' or upstream of the coding region. However, enhancer elements can exert their effect even when located 3' of the promoter element and the coding region. Transcription termination and polyadenylation signals are located 3' or downstream of the coding region.
As used herein, the term "an oligonucleotide having a nucleotide sequence encoding a gene" means a nucleic acid sequence comprising the coding region of a gene, i.e. the nucleic acid sequence which encodes a gene product. The coding region may be present in a cDNA, genomic DNA or RNA form. When present in a DNA form, the oligonucleotide maybe single-stranded (i.e., the sense strand) or double-stranded.
Suitable control elements such as enhancers/promoters, splice junctions, polyadenylation signals, etc. may be placed in close proximity to the coding region of the gene if needed to permit proper initiation of transcription and/or correct processing of the primary RNA
transcript. Alternatively, the coding region utilized in the expression vectors of the present invention may contain endogenous enhancers/promoters, splice junctions, intervening sequences, polyadenylation signals, etc. or a combination of both endogenous and exogenous control elements.
As used herein, the term "regulatory element" refers to a genetic element which controls some aspect of the expression of nucleic acid sequences. For example, a promoter is a regulatory element which facilitates the initiation of transcription of an operably linked coding region. Other regulatory elements are splicing signals, polyadenylation signals, termination signals, etc.
Transcriptional control signals in eukaryotes comprise "promoter" and "enhancer"
elements. Promoters and enhancers consist of short arrays of DNA sequences that interact specifically with cellular proteins involved in transcription. Maniatis, T. et al., Science 236:1237 (1987). Promoter and enhancer elements have been isolated from a variety of eukaryotic sources including genes in plant, yeast, insect and mammalian cells and viruses (analogous control elements, i.e., promoters, are also found in prokaryotes). The selection of a particular promoter and enhancer depends on what cell type is to be used to express the protein of interest.
The presence of "splicing signals" on an expression vector often results in higher levels of expression of the recombinant transcript. Splicing signals mediate the removal of introns from the primary RNA transcript and consist of a splice donor and acceptor site. Sambrook, J. et al., In: Molecular Cloning: A Laboratory Manual, 2nd ed., Cold Spring Harbor laboratory Press, New York (1989) pp. 16.7-16.8. A commonly used splice donor and acceptor site is the splice junction from the 16S RNA of SV40.
The term "poly A site" or "poly A sequence" as used herein denotes a DNA
sequence which directs both the termination and polyadenylation of the nascent RNA
transcript. Efficient polyadenylation of the recombinant transcript is desirable as transcripts lacking a poly A tail are unstable and are rapidly degraded. The poly A signal utilized in an expression vector may be "heterologous" or "endogenous." An endogenous poly A signal is one that is found naturally at the 3' end of the coding region of a given gene in the genome. A heterologous poly A signal is one which is isolated from one gene and placed 3' of another gene. Efficient expression of recombinant DNA
sequences in eukaryotic cells involves expression of signals directing the efficient termination and polyadenylation of the resulting transcript. Transcription termination signals are generally found downstream of the polyadenylation signal and are a few hundred nucleotides in length.
The term "transfection" or "transfected" refers to the introduction of foreign DNA
into a cell.
As used herein, the terms "nucleic acid molecule encoding", "DNA sequence encoding," and "DNA encoding" refer to the order or sequence of deoxyribonucleotides along a strand of deoxyribonucleic acid. The order of these deoxyribonucleotides determines the order of amino acids along the polypeptide (protein) chain. The DNA
sequence thus codes for the amino acid sequence.
As used herein, the term "antisense" is used in reference to RNA sequences which are complementary to a specific RNA sequence (e.g., mRNA). Antisense RNA may be produced by any method, including synthesis by splicing the gene(s) of interest in a reverse orientation to a viral promoter which permits the synthesis of a coding strand.
Once introduced into a cell, this transcribed strand combines with natural mRNA
produced by the cell to form duplexes. These duplexes then block either the further transcription of the mRNA or its translation. In this manner, mutant phenotypes may be generated. The term "antisense strand" is used in reference to a nucleic acid strand that is complementary to the "sense" strand. The designation (-) (i.e., "negative") is sometimes used in reference to the antisense strand, with the designation (+) sometimes used in reference to the sense (i.e., "positive") strand.
The term "Southern blot" refers to the analysis of DNA on agarose or acrylamide gels to fractionate the DNA according to size, followed by transfer and immobilization. of the DNA from the gel to a solid support, such as nitrocellulose or a nylon membrane. The immobilized DNA is then probed with a labeled oligodeoxyribonucleotide probe or DNA
probe to detect DNA species complementary to the probe used. The DNA may be cleaved with restriction enzymes prior to electrophoresis. Following electrophoresis, the DNA
may be partially depurinated and denatured prior to or during transfer to the solid support.
Southern blots are a standard tool of molecular biologists. J. Sambrook et al.
(1989) In:
Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press, NY, pp 9.31-9.58.
The term "Northern blot" as used herein refers to the analysis of RNA by electrophoresis of RNA on agarose gels to fractionate the RNA according to size followed by transfer of the RNA from the gel to a solid support, such as nitrocellulose or a nylon membrane. The immobilized RNA is then probed with a labeled oligodeoxyribonucleotide probe or DNA probe to detect RNA species complementary to the probe used. Northern blots are a standard tool of molecular biologists, J.
Sambrook, J.
et al. (1989) supra, pp 7.39-7.52.
The term "reverse Northern blot" as used herein refers to the analysis of DNA
by electrophoresis of DNA on agarose gels to fractionate the DNA on the basis of size followed by transfer of the fractionated DNA from the gel to a solid support, such as nitrocellulose or a nylon membrane. The immobilized DNA is then probed with a labeled oligoribonuclotide probe or RNA probe to detect DNA species complementary to the ribo probe used.
As used herein the term "coding region" when used in reference to a structural gene refers to the nucleotide sequences which encode the amino acids found in the nascent polypeptide as a result of translation of a mRNA molecule. The coding region is bounded, in eukaryotes, on the 5' side by the nucleotide triplet "ATG" which encodes the initiator methionine and on the 3' side by one of the three triplets which specify stop codons (i.e., TAA, TAG, TGA).

As used herein, the term "structural gene" refers to a DNA sequence coding for RNA or a protein. In contrast, "regulatory genes" are structural genes which encode products which control the expression of other genes (e.g., transcription factors).
As used herein, the teen "gene" means the deoxyribonucleotide sequences comprising the coding region of a structural gene and including sequences located adjacent to the coding region on both the 5' and 3' ends for a distance of about I kb on either end such that the gene corresponds to the length of the full-length mRNA. The sequences which are located 5' of the coding region and which are present on the mRNA
are referred to as 5' non-translated sequences. The sequences which are located 3' or downstream of the coding region and which are present on the mRNA are referred to as 3' non-translated sequences. The term "gene" encompasses both cDNA and genomic forms of a gene. A genomic form or clone of a gene contains the coding region interrupted with non-coding sequences termed "introns" or "intervening regions" or "intervening sequences." Introns are segments of a gene which are transcribed into heterogeneous nuclear RNA (hnRNA); introns may contain regulatory elements such as enhancers.
Introns are removed or "spliced out" from the nuclear or primary transcript;
introns therefore are absent in the messenger RNA (mRNA) transcript. The mRNA
functions during translation to specify the sequence or order of amino acids in a nascent polypeptide.
In addition to containing introns, genomic forms of a gene may also include sequences located on both the 5' and 3' end of the sequences which are present on the RNA transcript. These sequences are referred to as "flanking" sequences or regions (these flanking sequences are located 5' or 3' to the non-translated sequences present on the mRNA transcript). The 5' flanking region may contain regulatory sequences such as promoters and enhancers which control or influence the transcription of the gene. The 3' flanking region may contain sequences which direct the tennination of transcription, posttranscriptional cleavage and polyadenylation.
The term "sample" as used herein is used in its broadest sense and includes environmental and biological samples. Environmental samples include material from the environment such as soil and water. Biological samples may be animal, including, human, fluid (e.g., blood, plasma and serum), solid (e.g., stool), tissue, liquid foods (e.g., milk), and solid foods (e.g., vegetables). A biological sample suspected of containing nucleic acid encoding a spore surface protein may comprise a cell, tissue extract, body fluid, chromosomes or extrachromosomal elements isolated from a cell, genomic DNA (in solution or bound to a solid support such as for Southern blot analysis), RNA
(in solution or bound to a solid support such as for Northern blot analysis), eDNA (in solution or bound to a solid support) and the like.
The teen "infection control composition" refers to any compound or compounds which prevent and/or reduce the rate of growth of an organism (i.e., for example, a microbial organism, including spores derived from a microbial organism) compared to the rate of growth of the organism in the absence of the composition. An infection control composition can be natural (e.g., derived from a microbe such as a bacteria, virus, fungi, algae, mold etc.), synthetic, or recombinant. An infection control composition may be a competitive inhibitor of a microbial spore protein and/or a microbial spore surface protein. Alternatively, an infection control composition may form stable and/or transient binding pairs with microbial spore proteins and/or microbial spore surface proteins. An `infection control composition" may also include antibacterial and/or antimicrobial agents and/or drugs (i.e., for example, antibiotics). Consequently, "infection control activity" is interpreted to mean any reduction and/or prevention of microbial growth as a result of the application of an infection control composition to a surface.
The terms "antibacterial", and "antimicrobial" are used interchangeably to refer to a composition which prevents and/or reduces the rate of growth of an organism compared to the rate of growth of the organism in the absence of the composition. An antibacterial and/or antimicrobial composition can be bacteriostatic, bactericidal, both, or neither. An antibacterial and/or antimicrobial composition is bacteriostatic if it inhibits cell division without affecting the viability of the inhibited cell. An antibacterial and/or antimicrobial composition is bactericidal if it causes cell death. Cell death is commonly detected by the absence of cell growth in liquid growth medium (e.g., absence of turbidity) or on a solid surface (e.g., absence of colony formation on agar). An antibacterial and/or antimicrobial composition may be effective to reduce the growth rate and/or cause cell death for viruses, molds, and fungi. A composition which is bacteriostatic at a given concentration may be bactericidal at a higher concentration, while other certain bacteriostatic compositions are not bactericidal at any concentration.
The term "bacteria" and "bacterium" refer to all prokaryotic organisms, including those within all of the phyla in the Kingdom Procaryotae. It is intended that the tern encompass all microorganisms considered to be bacteria including, but not limited to, Clostridia, Bacillus, Mycoplasma, Chlamydia, Actinomyces, Streptomyces, and Rickettsia.
All forms of bacteria are included within this definition including cocci, bacilli, spirochetes, spheroplasts, protoplasts, spores etc. Also included within this term are prokaryotic organisms which are gram negative or gram positive. "Gram negative" and "gram positive" refer to staining patterns with the Gram-staining process.
Finegold and Martin, In: Diagnostic Microbiology, 6th Ed. (1982), C. V. Mosby St. Louis, pp 13-15.
"Gram positive bacteria" are bacteria which retain the primary dye used in the Gram stain, causing the stained cells to appear dark blue to purple under the microscope.
"Gram negative bacteria" do not retain the primary dye used in the Gram stain, but are stained by a counter stain. Thus, gram negative bacteria appear red.
The term "test agent" refers to an agent that is to be screened in one or more of the assays described herein. The agent can be virtually any chemical compound. It can exist as a single isolated compound or can be a member of a chemical (e.g., combinatorial) library. In a particularly preferred embodiment, the test agent will be a small organic molecule.
The term "small organic molecule" as used herein, refers to any molecule of a size comparable to those organic molecules generally used in pharmaceuticals. The term excludes biological macromolecules (e.g., proteins, nucleic acids, etc.).
Preferred small organic molecules range in size from approximately 10 Da up to about 5000 Da, more preferably up to 2000 Da, and most preferably up to about 1000 Da.
The term "label" or "detectable label" are used herein, to refer to any composition detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical, optical or chemical means. Such labels include biotin for staining with labeled streptavidin conjugate, magnetic beads (e.g., Dynabeads ), fluorescent dyes (e.g., fluorescein, texas red, rhodamine, green fluorescent protein, and the like), radiolabels (e.g., 3H 1251, 35S, '4C, or 32P), enzymes (e.g., horse radish peroxidase, alkaline phosphatase and others commonly used in an ELISA), and calorimetric labels such as colloidal gold or colored glass or plastic (e.g., polystyrene, polypropylene, latex, etc.) beads. Patents teaching the use of such labels include, but are not limited to, U.S. Pat.
Nos. 3,817,837; 3,850,752; 3,939,350; 3,996,345; 4,277,437; 4,275,149; and 4,366,241 (all herein incorporated by reference). The labels contemplated in the present invention may be detected by many methods. For example, radiolabels may be detected using photographic film or scintillation counters, fluorescent markers may be detected using a photodetector to detect emitted light. Enzymatic labels are typically detected by providing the enzyme with a substrate and detecting, the reaction product produced by the action of the enzyme on the substrate, and calorimetric labels are detected by simply visualizing the colored label.
The term "primary surfactant" as used herein, means any surfactant which acts upon or in conjunction with a substituted phenol to enhance further, or at the least, to not significantly reduce infection control activity of a substituted phenol.
The term "inhibit", "disrupt", "displace", "antagonize", or "block" as used herein refer to any means wherein a microbial spore and/or microbial spore surface protein is removed from, and/or prevented from attaching to, a substrate surface.
The term "antagonist" as used herein, refers to any compound that is capable of preventing the attachment of a microbe (i.e., for example, bacteria, virus, mold, fungi etc) and/or a reproductive component of the microbe (i.e., for example, a spore) to a surface.
The `antagonist' may prevent attachment by either competitive displacement or by forming a binding pair (i.e., for example, stable or transient) with the microbe and/or reproductive component of the microbe. For example, an antagonist may prevent the attachment of a bacterial spore to a surface by binding to a spore surface protein.
The term "binding antagonist" as used herein, refers to any compound that is capable of forming a binding pair with either a microbial spore nap layer protein (i.e., for example, a Clostridium spore surface protein) or a substrate surface. A
formation of either binding pair prevents and/or displaces the attachment of a microbial spore to a substrate surface.
The tern "substrate surface" as used herein, refers to any material comprising a binding site for a microbial spore surface protein attachment. A substrate surface may including but not limited to, an animate surface and/or an inanimate surface.
The term "inanimate surface" as used herein, refers to any non-living material.
For example, an inanimate surface may include but is not limited to, vinyl, stainless steel, plastic, wood, ceramic, glass, chrome, tile etc. Such surfaces commonly encountered including but not limited to countertops, chairs, tables, surgical equipment, medical devices, floors, walls, windows, sinks, cabinets etc.
The term "animate surface" as used herein, refers to any living material. For example, an animate surface may include but is not limited to, epidermal skin surfaces, mucosal epithelial tissue surfaces, and/or internal organ surfaces. As used herein, the term "animate surface" also comprises vegetative surfaces including but not limited to, fruit surfaces (i.e., for example, strawberries, apples, oranges, tangerines etc.), vegetable surfaces (eggplant, squash, tomatoes, potato etc), legume surfaces (i.e., for example, peanuts, cashews, beans etc), grain surfaces (i.e., for example, wheat, soy, rice, rye etc), flower surfaces, stem surfaces, stalk surfaces, leaf surfaces, bark surfaces, limb surfaces, branch surfaces, etc.
The term "interaction antagonist" as used herein, refers to any compound that is capable of interfering with the formation of a microbial spore nap layer protein/substrate surface binding pair.
The term "binding" as used herein, refers to any interaction between an infection control composition and a surface. Such as surface is defined as a "binding surface".
Binding may be reversible or irreversible. Such binding may be, but is not limited to, non-covalent binding, covalent bonding, ionic bonding, Van de Waal forces or friction, and the like. An infection control composition is bound to a surface if it is impregnated, incorporated, coated, in suspension with, in solution with, mixed with, etc.
The term "infection control test composition", as used herein, refers to the collection of compounds that are to be screened for their ability to prevent microbial spore binding to a surface. Such test compositions may include, but are not limited to, a wide variety of different compounds, including chemical compounds, mixtures of chemical compounds, e.g., polysaccharides, small organic or inorganic molecules, biological macromolecules, e.g., peptides, proteins, nucleic acids, or an extract made from biological materials such as bacteria, plants, fungi, or animal cells or tissues, naturally occurring or synthetic compositions.
The term ''at risk" as used herein, refers to any person or inanimate surface having a more than average probability (i.e., greater than 50%) of becoming contaminated with a microbe. 'At risk' probabilities of exposure may be found in vocations such as first responders including, but not limited to, fireman, policeman, emergency medical personnel. 'At risk' probabilities of exposure may also be found in vocations such as healthcare workers including, but not limited to, doctors, nurses, orderlies, hospital janitorial staff and the like. 'At risk' probabilities of exposure may also be found in vocations such as military personnel including, but not limited to, Army personnel, Navy personnel, Air Force personnel, Marine personnel, National Guard personnel, Coast Guard personnel. 'At risk' probabilities of exposure may also be found in medical patients, including, but not limited to, immunocompromised patients, burn patients, gunshot patients etc.
The term "specific binding site pattern", as used herein refers to any reproducible set of binding sites on either a body surface or inanimate surface. Such binding sites have inherent affinity for compositions including, but not limited to, microbial spores, infection control compositions, or small organic molecules as discussed herein. Although it is not necessary to understand the mechanism of an invention, it is believed that such a binding pattern is a result of unique arrangements of surface chemical groups and/or topography that is three-dimensionally compatible (i.e., for example, via stereospecific chemical group interactions) with a composition. It is further believed that a specific binding site pattern predicts an arrangement of non-specific binding (i.e., that binding not mediated via a dedicated receptor) within a designated surface area.

Brief Description Of The Figures Figure 1 presents an exemplary scanning electron micrograph of a C. sporogens endospores (S) having long, sinuous, or filiform exosporial membrane projections (arrows). Projections facing the surface form tenacious contacts.
Figure 2 presents an exemplary electron micrograph showing C. difficile 43594 smooth-surfaced, elliptical endospore before activation (insert), and covered with exosporial projections (arrows) following activation.
Figure 3 presents an exemplary electron micrograph showing a C. difficile spore as germination proceeds wherein an exosporial membrane is covered with low bumps and a thick anchoring structure at one pole, designated the tail (T) region.
Figure 4 presents exemplary data showing an SDS-PAGE gel identifying the total spore proteins present in the 18 kDa band (see arrow). This 18 kDa band corresponds with Western Blot 18kDa bands capable of hybridizing C. difficile antibodies.
Figure 5 presents exemplary data showing an SDS-PAGE gel identifying the total spore proteins present in the 140, 150, and 160 kDa bands (see pl, p2, and p3, respectively). These 140, 150, and 160 kDa bands correspond with Western Blot 140, 150, and 160 kDa bands capable of hybridizing C. difficile antibodies.
Figure 6A presents exemplary data showing an SDS-PAGE gel used to identify C.
difficile exosporium proteins at approximately 300 kDa (arrow 1); 275 kDa (arrow 2);
250 kDa (arrow 3); 160 kDa (arrow 4); 150 kDa (arrow 5); 140 kDa (arrow 6); 35 kDa (arrow 7); 28 kDa (arrow 8); and 15 kDa (arrow 9). Six other non-hybridizing bands (arrows 10-15) were also identified to determine the non-immunoreactive exosporium proteins.
Figures 6B and 6C present exemplary data showing Western blots demonstrating C. difficile antibody hybridization to 300 kDa (arrow 1); 275 kDa (arrow 2);
250 kDa (arrow 3); 160 kDa (arrow 4); 150 kDa (arrow 5); 140 kDa (arrow 6); 35 kDa (arrow 7);
28 kDa (arrow 8); and 15 kDa (arrow 9) bands corresponding to those bands shown in Figure 6A.
Figure 7 presents one embodiment of a structure for: BclA (green) and Clq (mauve) monomers (A and B), respectively and their superposition (C). The N
and C
termini and the O-strands are labeled.
Figure 8 presents one embodiment of BelA (A) and Cl q (B) trimers viewed from the side and from above. The three monomers comprising each trimer are colored blue, green, and red.
Figure 9A presents an exemplary transmission electron micrographs of a B.
cereus ATCC 10876 spore showing the exosporium.
Figure 9B presents an exemplary transmission electron micrograph of a B.
anthracis spore showing a nap layer projecting from the exosporium.
Figure9C presents an exemplary close-up of a B. anthracis exosporium layer showing the Bc1A nap layer and the interior basal layer.
Figure 10 presents an illustration of embodiments showing microbial spore interactions with surface substrates (i.e., for example, skin surfaces).
Figure IOA illustrates the attachment of a microbial spore nap layer (i.e., for example, by spore surface protein projections) to the epithelial layer of mammalian skin.
Figure I OB illustrates the attachment of a microbial spore surface protein (i.e., for example a native to a surface substrate and various infection control compositions capable of disrupting the attachment of the microbial spore surface proteins to a surface substrate.
Figure 11 presents a schematic of one embodiment of a pET-19b plasmid comprising a spore surface protein gene.
Figure 12 presents exemplary data showing an SDS-PAGE electrophoretic separation of recombinant CD 1067 (red arrow) expression from a pET-19b vector. Lane 1: MW
standards ladder. Lane 2: 1 hour sample. Lane 3: 2 hour sample. Lane 4: 3 hour sample:
Lane 5: 4 hour sample: Lane 6: 5 hour sample. Lane 7: 6 hour sample. Protein Stain:
Coomassie Blue.
Figure 13 presents exemplary data showing Western Blot analysis of recombinant CD1067 (red arrows) expression from a pET-19b vector. Lane 1: MW standards ladder.

Lane 2: 1 hour sample. Lane 3: 2 hour sample. Lane 4: 3 hour sample: Lane 5: 4 hour sample: Lane 6: 5 hour sample. Lane 7: 6 hour sample.
Figure 13A: Detection using F1373 anti-C. difficile antibody.
Figure 13B: Detection using anti-HIS antibody.
Figure 14 presents exemplary data showing an SDS-PAGE electrophoretic separation of recombinant CD3620 (red arrow) expression from a pET-19b vector.
Lane 1: MW standards ladder. Lane 2: 0 hour sample. Lane 3: 1 hour sample. Lane 4:
2 hour sample: Lane 5: 3 hour sample: Lane 6: 4 hour sample. Lane 7: 4.5 hour sample.
Protein Stain: Coomassie Blue.
Figure 15 presents exemplary data showing Western Blot analysis of recombinant CD3620 (red arrows) expression from a pET-19b vector. Lane 1: MW standards ladder.
Lane 2: 0 hour sample. Lane 3: 1 hour sample. Lane 4: 2 hour sample: Lane 5: 3 hour sample: Lane 6: 4 hour sample. Lane 7: 4.5 hour sample.
Figure 15A: Detection using F1373 anti-C. difficile antibody.
Figure 15B: Detection using anti-HIS antibody.
Figure 16 presents exemplary data showing antibody detection control data for recombinant CD3620 (red arrows) expression from a pET-19b vector. Lane 1: MW
standards ladder. Lane 2: 6 hour pJEB02 sample. Lane 3: 4.5 hour pJEB03 sample.
Figure 16A: SDS-PAGE Coomassie Blue reference electrophoresis separation identifying recombinant CD 1067 (top red arrow) and recombinant CD3620 (bottom red arrow).
Figure 16B: Western Blot analysis using preimmune F1373 detection.
Figure 16C: Western Blot analysis using preimmune F 1997 detection.
Figure 16D: Western Blot analysis using F1997 anti-C. difficile antibody detection.
Figure 17 illustrate several embodiments of a method to remove a C. difficile exosporium coat (see, Example IV).
Figure 18A depicts a representative C. difficile 630 spore that may be used as starting material.
Figure 18B depicts a representative C. difficile 630 spore after exosporium stripping.
Figure 18C depicts a purified and concentrated representative C. difficile 630 spore exoporium after stripping.

Detailed Description The present invention is related to the field of infection control.
Particularly, the invention is related to compositions and methods for maintenance of antiseptic conditions, bacterial decontamination, and/or bacterial exposure prophylaxis.
In one embodiment, the invention contemplates infection control compositions that inhibit microbial spore binding to a substrate surface. For example, the inhibition disrupts and/or displaces an interaction, attachment, and/or stabilization of a microbial spore to a substrate surface.
For approximately 70 years, the first line of defense against bacterial infection has been antibiotics. The first antibiotics were used clinically in the 1940s and 1950s, and their use has been increasing significantly since this period. Although an invaluable advance, antibiotic and antimicrobial therapy suffers from several problems.
First, antibiotics are ineffective against spores. Second, the development of antibiotic resistance is a serious and life-threatening event of worldwide importance.
For example, some microbial strains are known to be immune to a majority of available antibiotics.
(Travis, Science 264:360-362 (1994). Among other drug resistant organisms are:
pneumococci that cause pneumonia and meningitis; Cryptosporidium and E. coli that cause diarrhea; and enterococci that cause blood-stream, surgical wound and urinary tract infections. (Berkelman et. al., JlnfectDis. 170:272-277 (1994). The present invention contemplates compositions and methods directed towards solving problems regarding contamination control of microbial spores and the clinical resistance of microbial infection to conventional antibiotic administration.

1. Bacterial Spores Some microbes shed spores that enter a dormant phase when exposed to environments that do not favor growth and proliferation. Spore survival is enhanced when attachment and/or adhesion to a solid surface is made. When the environmental conditions again change to favor growth and proliferation the microbial spore germinates and proceeds to contaminate the surrounding area. A person or animal coming into contact with the proliferating new microbial growth then becomes infected from an area previously unknown to be contaminated. In some cases, a person and/or animal may come into contact with the microbial spore wherein germination and infection proceeds after contact. Alternatively, such undetected spore germination can result in food spoilage thereby initiating food-borne illness outbreaks. Because of their relative resistance to chemical and physical sterilization agents, spores of these bacteria are used as indicators of sterilization efficiency for treatments involving moist and dry heat, UV
irradiation, and hydrogen peroxide. Ito et al., "Sterilization of packaging materials using aseptic systems" Food Technol. 38:60-62 (1984).
An understanding the surface properties of bacterial spores and their interactions with inanimate and animate substrates is important for selection of packaging materials and for evaluation of the surface sterilization procedures used in the packaging of food, pharmaceutical agents, and medical supplies. For example, the role of hydrophobic interactions in the adhesion of bacteria (i.e., the vegetative cell) to the surfaces of inert materials is well accepted. Relatively few studies, however, have thoroughly examined the surface hydrophobicity of bacterial spores or the adhesion of spores to inanimate substrata. The hydrophobicity of bacterial spores can be determined by applying methodologies used for measuring vegetative cell hydrophobicity. Established techniques for measuring surface hydrophobicity include, but are not limited to: i) adherence to hydrocarbons (Beck et al., "Effect of growth on surface charge and hydrophobicity of Staphylococcus aureus" Ann. Inst. Pasteur/Microbiol. (Paris) 139:655-664 (1988)); ii) hydrophobic interaction chromatography (HIC) (Doyle et al., "Hydrophobic characteristics of Bacillus spores" Curr. Microbiol. 10:329-332 (1984); iii) salt aggregation (Takubo et al., "Isolation and characterization of outermost layer deficient mutant spores of Bacillus megaterium" Microbiol. Immunol. 9:973-979 (1988);
iv) contact angle measurements (Minagi et al., "Cell-surface hydrophobicity of Candida species as determined by the contact-angle and hydrocarbon-adherence methods"
J. Gen.
Microbiol. 132:1111-1115 (1986)); and v) bacterial adherence to hexadecane (BATH) (Wienek et al., Hydrophobicity of Bacillus and Clostridium spores" Appl Environ Microbiol 56:2600-2605 (1990).
Studies demonstrate that spore hydrophobicity varies among species and strains.
However, for each organism, the hydrophobicity of spores is believed to be greater than the vegetative cell hydrophobicity. Further, moderate heat treatment increases the hydrophobicity of most Bacillus spores. One hypothesis suggests that increases in spore hydrophobicity following heat treatment may result from the disruption of outer coat or exosporium proteins.
Although it is not necessary to understand the mechanism of an invention, it is believed that the increased hydrophobicity of bacterial spores is due to the relative abundance of protein in the outer coats and exosporium as compared with peptidoglycan on vegetative cells. Doyle et al., "Hydrophobic characteristics of Bacillus spores"' Curr.
Microbiol. 10:329-332 (1984); Matz et al., "Chemical composition of exosporium from spores of Bacillus cereus" J. Bacteriol. 101:196-201 (1970); and Takumi et al., "Isolation and partial characterization of exosporium from spores of a highly sporogenic mutant of Clostridium botulinum type A" Microbiol. Immunol. 28:443-454 (1979). An association between spore hydrophobicity and the presence of an exosporium has been reported recently for several Bacillus species. Kjelleberg, S. "Adhesion to inanimate surfaces" In:
Microbial adhesion and aggregation, pp. 51-70. K. C. Marshall (ed.), Springer-Verlag KG, Berlin (1984). Decreased spore hydrophobicity of B. cereus I spores was observed of when spore surfaces were removed by chemical treatments. Kutima et al., "Involvement of the spore surface in germination of Bacillus cereus T spores"
Appl.
Environ. Microbiol. 53:47-52 (1987). Decreases in spore hydrophobicities have also been seen in "knock-out" outer-coat-negative mutants of B. megaterium (QMB 1551:
ATCC
12872). Takubo et al., "Isolation and characterization of outermost layer deficient mutant spores of Bacillus megaterium" Microbiol. Immunol. 9:973-979 (1988); and.
Koshikawa et al., "Surface hydrophobicity of spores of Bacillus spp.".I. Gen. Microbiol.
135:2717-2722 (1989). These observations suggest that the outer coats of the exosporium play a role in spore hydrophobicity.
In one embodiment, the present invention contemplates compositions and methods to improve surface sanitation, sterilization of equipment and/or packaging materials used in the medical, pharmaceutical, and/or food industries. In one embodiment, the present invention contemplates compositions and methods to improve skin sanitation.
A. Clostridium difficile Spore Surface Proteins Bacteria from the genus Clostridium, are thought to produce such diseases including, but not limited to, gas gangrene, wound infections, botulism, tetanus, pseudomembranous colitis, septicemia, or nosocomial diarrhea. This genus survives well under adverse conditions by forming dehydrated endospores that are highly resistant to heat, chemicals, and radiation. These spores can be found in soil, carried by the wind, recovered from mammalian digestive and urogenital tracts, and have the ability to remain dormant for very extended periods of time until conditions for growth support spore activation and germination.
C. difficile spores are prevalent within healthcare and hospital environments.
C.
difficile spores survive on animate and/or inanimate surfaces for a prolonged period, are resistant to many disinfectants and antibiotics, and are easily transmitted from patient to patient. Understanding C. difficile spore attachment to surfaces, as well as sporulation and germination, will help in the prevention, control, and treatment of C.
difficile-associated disease. For example, C. difficile lacks several genes that are involved in the phospho-relay system that triggers the sporulation process in Bacillus species; this, and other aspects of the sporulation system in Clostridia spp have recently been reviewed.
Paredes et al., "A comparative genomic view of clostridial sporulation and physiology"
Nat. Rev. Microbiol. 3:969-978 (2005). Further, C. difficile also lacks a tricistronic ger operon that are usually present in most Bacillus and Clostridium species..
Moir et al., "Spore germination" Cell !'Viol Life Sci. 59:403-409 (2002); and Broussolle et al., "Molecular and physiological characterisation of spore germination in Clostridium botulinum and C. sporogenes" Anaerobe 8:89-100 (2002). These observations suggest that the C. difficile genninant receptors are substantially different from, and should not be identified on the basis of sequence similarity to, other Clostridium spp. and Bacillus spp.
Microbial spore resistance to heat, solvents, enzymes, detergents, ultraviolet and ionizing radiation, sodium hydroxide, ethanol, performic acid, phenol, cold, and most sporicides, as well as their ability to grow aggressively when conditions are optimum, make them a serious threat to animal and especially human health. Tipper et al., "Structure of the bacterial endospore" In: Spores V, Ed. H. Halvorson, R.
Hanson and L.
Campbell. Amer. Soc. for Microbiol., Bethesda, Maryland, 3-12 (1972); and Russell, A., "Bacterial spores and chemical sporicidal agents" Clin. Microbiol. Rev., 3, 99-119 (1990).
Nosocomial acquisition of Clostridium difficile infection in hospitals and nursing homes presents a serious epidemiologic problem because spores are found on walls, floors, utensils, bed sheets, and on the hands of caregivers in health facilities. The spores easily seem to infect individuals including, but not limited to, those who have: i) an altered intestinal flora from antibiotic therapy; ii) a compromised immune system, or iii) require multiple enemas in relation to diagnostic testing or treatment. Zaleznik, D.
"Clostridium difficile: an important nosocomial pathogen for the 1990s" Clin. Microbiol.
Newsletter 13, 145-152 (1991).
The exosporia from two species of Clostridium spores (i.e., for example, Clostridium sporogenes ATCC 3584 and C. difficile ATCC 9689 and ATCC 43594) may play a role in attachment, germination, and/or colonization. Panessa-Warren et al., "Exosporial membrane plasticity of Clostridium sporogens and Clostridium difficile"
Tissue & Cell 29:449-461 (1997). Ultrastructural change was associated with germination and outgrowth of C. bifermentans spore appendages but no specific function was determined. Samsonoff et al., "Ultrastructural changes associated with germination and outgrowth of an appendage-bearing clostridia] spore" J. Bacteriol., 101, (1970). Examination of C. sporogenes spores by thin-section transmission electron microscopy did reveal an exosporial outer layer, with hair-like projections, but no discussion regarding distribution, function, or appearance in relation to Clostridium colonization was offered. Hoeniger et al., "Ultrastructural aspects of spore germination and outgrowth in Clostridium sporogenes" Can. J. Microbiol., 15, 1061-1065 (1969).
Transmission electron microscopy thin-section reconstruction is known to have significant problems when observing and interpreting an exosporial morphology of numerous spores at one time, and the problems associated with obtaining thin-section slices of such small, delicate, and poorly staining, spore attachments to a nutritive substrate.
C. sporogenes spores may exhibit specific morphological stages once dormancy is broken, and that the exosporium seems to play an active role in successful germination, outgrowth, vegetative proliferation, and colonization of a substrate surface binding site.
Panessa-Warren et al., "Electron microscopy of C. sporogenes endospore attachment and germination" Scanning 16, 227-240 (1994). A Clostridium exosporium is believed to be an outer 'membranous' layer of a spore, has been described as a passive participant in germination. However, once dormancy is broken, C. sporogenes ATCC 3584 spores appear to produce very specific exosporial morphological structures visible by scanning electron microscopy (SEM), which seem to facilitate attachment of a spore to a substrate or to other spores (i.e., for example, spore co-aggregation). Exosporial projections are later replaced by a single stalk extending from the distal (i.e., for example, the tail region) of the spore, which appears to anchor the spore during the final stages of vegetative cell development and outgrowth.
During the initial stages of C..sporogenes attaclunent, delicate filiform projections of the exosporial membrane extend outward with some of the shorter projections attaching to a surface. See, Figure 1, white arrows. Usually long filamentous projections from the exosporial membrane originate from the top surface or sides of the spore, and extend to either the surface or to a nearest neighboring spores, binding the spores together (co-aggregation). Exosporial membrane projections appear all over the outer surface of a C. sporogenes activated spore eventually developing into a single, thick, attachment structure extending to a surface, appearing to anchor the spore.

Unlike C. sporogenes, C difficile has a reduced number of attached spores and requires a longer incubation time to allow attachment to take place. Dormant C. difficile spores are small (approximately 2.0 gm long x 1.1 gm wide), elliptical, and predominantly smooth. See, Figure 2, insert. Once activation had begun, spores exhibited low prominences covering the exosporium. See, Figure 3. In the early stage of attachment, C. difficile 43594 developed exosporial bumps and knobs covering the entire surface of the elongate spore. These spores developed more elongate exosporial projections at the tail region, which were replaced later during the germination cycle by a thickened attachment structure. Figure 3, black arrows. Unlike C. sporogenes, C.
difticile exhibits fewer exosporial membrane extensions but produces a thickened exosporial membrane extension that developed at the tail end of the spore.
The ability of C. sporogenes and C. difficile spores to remain attached to an agar surface during agitation (i.e., for example, a 300 rpm centrifugation) in water or buffer, strongly suggests that these spores have a formidable means of adhesion and/or attachment to surfaces. Under ideal conditions, the attachment of C.
sporogenes to an agar surface has been observed to take place within 27 min following inoculation. Under anaerobic conditions on pre-reduced agar, maximal attachment of C. sporogenes spores has been observed to take place within 88 to 105 min. C. difficile spore attachment took considerably longer, for example, 105 min for C. difficile 9689 and 180 min for C.
difficile ATCC 43594.
Once attached, the spores can remain tenaciously anchored to a solid surface.
C.
difficile 43594 appears unique in that the spores attach very slowly. However, C. difficile may have an adaptive response that accelerates attachment following exposure to an animate surface. For example, an increased incidence of C. difficile nosocomial infection have been reported in patients having increased intestinal motility and those who have undergone multiple enemas. Bartlett, J. "Introduction" In: Clostridium Difficile: Its Role in Intestinal Disease, Ed. R. Rolfe and S. Finegold). Academic Press, New York, pp. 1-13 (1988); Silva, J. Jr., "Prevention of Clostridium difficile-associated intestinal disease"
In: Clostridium difficile: Its Role in Intestinal Disease, Ed. R. Rolfe and S.
Finegold.
Academic Press, New York, pp. 368-378 (1988).; and McFarland et al., "Nosocomial acquisition of Clostridium difficile infection. New Engl. J. Med., 320:204-210 (1989). C.
difficile spores may be routinely found in the digestive tract of healthy, non-symptomatic individuals. Consequently, a triggering mechanism that initiates attachment to the gut and begins the infective process may be a combination of the loss of normal flora (i.e., for example, caused by antibiotics, disease, or repeated enemas) and/or hypermobility of the colon.
Clostridium spore exosporium attachment outgrowths typically have a diameter of approximately 7 nm and may vary in length between 0.1 and 2.9 mn. These outgrowths comprise fibrils that are usually less than 200 nm in length and can be extremely delicate.
Hancock, I, "Microbial cell surface architecture" In: Microbial Cell Surface Analysis , Ed. N. Mozes, P. Handley, H. Busscher and P. Rouxhet, VCH Publishers, New York, pp.
23-59 (1991). Usually. exosporiurn outgrowths of C. sporogenes measure 85 to 1015 nm in width, however, some outgrowths, especially those also comprising co-aggregation attachments, can be as long as 2000 nm in length. Knob-like protrusions from C.
sporogenes exosporium can range from 68 to 75 nm in diameter, and swollen projections seen in the late stages of germination can be as large as 180 to 240 urn in diameter.
Clearly, the spore exosporium, which has always been thought to be 'a passive participant in germination', appears to play an active role in spore attachment and colonization of C. sporogenes and C. difficile. In one embodiment, a C.
difficile protein is derived from an exosporium surface layer, wherein the exosporium layer comprises the outermost protein of a C. difficile spore. In one embodiment, the present invention contemplates a composition comprising at least one outermost surface protein of the Clostridium difficile spore. In one embodiment, the surface protein comprises a spore surface protein. Although it is not necessary to understand the mechanism of an invention, it is believed that these spore surface proteins present an interface between the bacterial spore and a substrate surface binding site. It is also believed that these proteins can he used to define molecular attachment interactions including, but not limited to: i) between the spore and animate surfaces; and ii) between the spore and inanimate surfaces.
Size characterizations of Clostridium difficile exosporium proteins are presented in Table 1.

Table I. Size Characterizations Of Representative C. difficile Exosporium Proteins.
Protein (alternative name) Size (kDa) BAS gene no GI

Putative spore surface protein 22 598 126698176 Putative spore surface protein 22 2401 126700016 Superoxide dismutase 27 1631 126699235 Putative proline racemase 24 3237 126700857 60 kDa chaperonin 58 0194 126697767 Chaperone protein 67 2461 126700078 kDa chaperonin 10 0193 126697766 Pyruvate-flavodoxin oxidoreductase 130 2682 126700296 Cell surface protein 76 2793 126700409 3-hydroxybutyryl-CoA dehydrogenase 31 1058 126698642 Putative spore cortex-lytic enzyme 47 0551 126698130 Putative aliphatic sulfonates ABC transporter, 36 1484 126699088 substrate binding lipoprotein Putative fructose-bisphosphate aldolase 33 0403 126697972 Dipicolinate synthase, subunit B 21 2967 126700588 Putative indolepyruvate oxidoreductase 21 2380 126699995 subunit B
Electron transfer flavoprotein alpha-subunit 36 1056 126698640 Putative phage-related cell wall hydrolase 28 1898 126699508 Cell surface protein 66 2791 126700407 Formate-tetrahydrofolate ligase 60 0718 126698298 Oligopeptide ABC transporter, substrate- 58 2672 126700286 binding protein Gamma-aminobutyrate metabolism 56 2341 126699959 dehydratase/isomerase NAD-specific glutamate dehydrogenase 46 0179 126697752 Putative aminoacyl-histidine dipeptidase 53 0708 126698287 Proline reductase subunit proprotein 68 3244 126700863 Glycine/sarcosine/betaine reductase complex 55 2349 126699967 component C beta subunit Glycine dehydrogenase subunit 2 54 1658 126699262 Acetyl CoA acetyltransferase 41 1059 126698643 Aspartate aminotransferase 45 1339 126698938 Putative bifunctional protein: 82 1433 126699037 peroxidoredoxin/chitinase Glyceraldehydes-3-phosphate dehydrogenase 36 3174 126700794 Butyryl-CoA dehydrogenase 41 1054 126698638 d-ribose ABC transporter, substrate-binding 35 0300 126697872 protein Putative ruberythrin 20 1474 126699078 Succinate-semialdehyde dehydrogenase 51 2342 126699960 Oxidoreductase, thiamine diP-binding subunit 39 0116 126697688 In one embodiment, Clostridium difficile proteins may be identified by probing total spore protein preparations with antibodies directed against purified, intact, C.
difficile spores, to created a hybridized protein-antibody complex. In one embodiment, a C. difficile spore surface protein includes, but is not limited to, CD1067, CD3620, CD
1581, CD 598, or CD 2401. In one embodiment, C. difficile enzymes are associated with C. difficile spore surface protein preparations.
In one embodiment, a Clostridium hypothetical protein (CD 1581) may be isolated as a hybridized antibody-protein complex comprising a molecular weight including, but not limited to, 18, 140, 150, or 160 kDa. See, Figures 4-6. In one embodiment, the hypothetical protein (-S 20 kDa) comprises an amino acid sequence of SEQ ID
NO: I
(Accession Numbers YP001088081, GI: 126699184):

encoded by a Clostridium hypothetical nucleic acid SEQ ID NO: 2:
1 atggaaaata aaaaatgtta ttcagaagat tggtatgaaa gaggagaatc tacagctaaa 61 tggttccaaa atgatagaga agaatatgaa agagaagcat atgatgaaga tagagaaaga 121 agaggttcaa actgtggatg ttcagattca ggagaaaata gacctagaaa ctgtgaaaga 181 tttagaagag aagctgagat aagagaaaga gaagcaagag aagcattctg tgaatcttca 241 gagaaaaaga aagaggcatt agcatatgaa tgtgaageta gaaaattatg ggaagaagca 301 gaaaaataat gggatgaata ttcaaaatac aactataaag gaatcgaata tttageagaa 361 gctgctagat tatttgatga aggtatggaa tgtgaagcta gaagaaatgg aaataatgga 421 ggaaacaata ataattgttg ecataaatgc cataaatgta attgtaactg ctgtagaaaa 481 taa In one embodiment, a Clostridium hypothetical protein (CD 1067) may be isolated as a hybridized antibody-protein complex comprising a molecular weight including, but not limited to, 140, 150, and 160 kDa. See, Figures 5 ,6 . In one embodiment, the hypothetical protein (- 47 kDa) comprises an amino acid sequence of SEQ ID NO:

(Accession Numbers YP_001087551; GI: 126698654):

201 DFDLDPLGDI VGRNCETrFE FAVCGERNSE CCRQGKGKSV AYKQRGLTVA

encoded by a Clostridium hypothetical nucleic acid SEQ ID NO: 4:

1 atgcaagatt ataaaaaaaa taaaagaaga atgatgaatc agccaatgtc tacaatgaat 61 gaagaagaag tgtatacaga tgaaataaat tcagaagaca tgagaggttt taaaaaatca 121 caccatcata atggatgtaa tactgataat aagtgtgagt gccatgatga ttgcaatcca 181 tgcaaeceat gtaateeatg taaacetaac ceatgcaatc catgcaaacc taatecatgt 241 gatgacaatt gtggatgcca tgacaattgt aaatgtgatt gtgaaccatg tgaaatggat 301 tcagatgaat gttttgaaaa caaatgtgga ccagaatgct gtaatcctat atctccaaga 361 aacttctctg tatcaaatgc agtgccattt gcaatagagg ctaatagaat atttgatact 421 atgcaattcc aaacatttac agatgcaaca ggaccaaatg gagagccatt aacttttgaa 481 acagaagtag tagaagtatt tggttcagtt ccaagtgcag gtcaagcaag tgtaactata 541 gaaaaaatat gcttaagtaa tgatggaatc gttatagaca caggaatgac aactttagaa 601 gatttcgatt tagacccatt aggagatata gtaggaagaa actgtgaaac aacttttgaa 661 tttgcagttt gtggagaaag aaactctgag tgctgtagac aaggaaaagg caaatcagta 721 gcttataaac aaagaggatt aactgtagca gttcgtaatt tagtactaga gctaagaggt 781 agatgtggat gtacagagtt cgttgcatta gctttcccag cagttagagc aggaggtgga 841 tgtaagagaa gagttgatta tgtagaattt acttttaaca cactttcagc accaatatgc 901 ttgccagctg acggaagagc tgttacttta agacaagaat atcaaactaa ettaactgta 961 gattgtatag gaaaatctat attaaaatta gaatgcaacg aatgttgtga acetttetat 1021 gaattaatta taccaaatga tatagattta gtactttgct tacaagaaac agttagcaca 1081 ttaataagtg aacaaatagt agttttagca tcaccaaatc caatccaacc aagacttgtt 1141 gatactttct ctaaagtatg tgatttttcg caatgtggac ctaatcatgg aagtggaaag 1201 ccaagttgcc acagatag In one embodiment, a Clostridium hypothetical protein (CD 3620) may be isolated as a hybridized antibody-protein complex comprising a molecular weight including, but not limited to, 140 kDa. See, Figures 5 & 6. In one embodiment, the hypothetical protein (- 14 kDa) comprises an amino acid sequence of SEQ ID NO: 5 (Accession Numbers YP001090143; GI: 126701246):

encoded by a Clostridium hypothetical nucleic acid SEQ ID NO: 6:

1 ttatactaaa ggtgcttcta tttgagtagt aaaaaggaat ttatgacaat gaccatcatt 61 aacagtagtt tctccacata ttaagtgaat gtgtttatca gttccaggta ttctaattgc 121 tggccctgtt gtaacacata tcttatgaaa atggtcaagg aaatctacat tatcatttat 181 tttatgaaca tgatttgtac ctccatttat tggtatagct tcaccagtaa cccctgctgc 241 acggtggtta tgtctatcct cacattcttc tgctaactta acactttctg aatattcatg 301 aacatgtgtc atttcctcat aatcattata gcacttgttg cacttattgt atttataatc 361 atcatacac In one embodiment, a Clostridium hypothetical protein (CD 1613) maybe isolated as a hybridized antibody-protein complex. In one embodiment, the hypothetical protein comprises an amino acid sequence of SEQ ID NO: 7 (Accession Number YP001088114):

1 mennkcredf rftgeyeedy pntneryyen yqvadryyny pnkykepkik qccckksmre 61 alellrydal rpfimfngfa fisdffivga nlvgidlsap pkdnlsgldg tferfsacnc 121 dlidiagrvs ypipvpltle glintigtip gvaelialid avipptidlg aildailaai 181 idfilaastp lanvdlaslc nikavafdit padyedfias lgyyldkkhy kecncncdcd 241 deccnkgild nlyrnsninnq vtvvagslvl tgvevlgkkn dvivlgnsnd sriyfvcvds 301 idyia encoded by a Clostridium hypothetical nucleic acid SEQ ID NO: 8:

1 ttatgcaata taatetatag aatetacaca tacaaagtat attettgaat cattagaatt .61 tccaagtact ataacatcat ttttcttacc tagaacttca acaccagtta gaaccaaact 121 accagctact acagtaactt gattatttat atttgacata taaagattat ctaggatacc 181 tttattacaa cagcaatcat cacaatcgca gttacaatta cattctttgt aatgtttttt 241 atcaagatag taacctaaag atgctatgaa atcttcataa tctgcaggtg taatatcaaa 301 tgcaacagct ttaagattac acaatgatgc taaatctacg tttgctaatg gagtagatgc 361 agcaagtata aaatcaatta tagcagcaag tattgcatct aatatagccc caaggtctat 421 cgtaggagga ataactgcat caataagtgc aattaattca gctactcctg gtatagttcc 481 tatagtatta attaatcect caagagttaa agggactgga ataggataag atactetacc 541 agetatatct attaaatcac agttacaagc agaaaatctt tcaaaagtac catcaagtcc 601 agataaatta tcttttggag gagctgaaag atctatacct accaaattag cacetactat 661 aaagaaatct gagataaaag caaattgatt aaagtttaca aaaggtctta gagcatcata 721 tcttagaagt tctaaggcct ctctcatact ttttttacaa caacattgtt ttattttagg 781 ttctttatat ttatttggat aattatagta tctatcagct acttgataat tttcatagta 841 tctttcattt gtatttggat aatettcctc atattcttgt gtaaatctaa agtcctetet 901 acatttatta ttttccac In one embodiment, a Clostridium hypothetical protein (CD 1711) may be isolated as a hybridized antibody-protein complex. In one embodiment, the hypothetical protein comprises an amino acid sequence of SEQ ID NO: 9:

1 mlivttekve gkkiskvlgl vrgstirakh vgkdigasfk nlvggeltgy 51 nemltearqi aigrmvedae akganaviaf rlssasvmgg aaemlaygta 101 vvleddnsil ek:
encoded by a Clostridium hypothetical nucleic acid SEQ ID NO: 10:

I atgttaatag taactacaga aaaagtagaa ggtaaaaaga tatcaaaggt tttaggatta gtgagaggaa 71 gtacaataag agcaaaacat gttggaaaag atataggagc aagttttaaa aatcttgttg gaggagaact 141 tactggatat aatgaaatgc tcactgaagc aagacaaatt gccataggta gaatggttga agatgcagaa 211 getaaaggtg caaacgeagt aatagcattt agactgtctt cagcttcagt tatgcaaggg gcagcagaaa 281 tgcttgctta tggaacagca gttgttttag aagatgataa tagtattctt gaaaaataa In one embodiment, a Clostridium hypothetical protein (CD 0881) may be isolated as a hybridized antibody-protein complex. In one embodiment, the hypothetical protein comprises an amino acid sequence of SEQ ID NO: 11:

1 mgtkivlsiv livvvvaisl tcirvikgsk vgiimrlgkf gkvaetgvhf lipfldkmay 61 vidireivid fppgpvitkd nvtmqidtvv yykvtdpvry vfeianpiaa ienltattlr 121 niigeldlde tltsrdiinv kmrtildeat dkwgikvnrv elknimppqd iqvamekqmr 181 aererreail qaegnksaai lqaegekqsa iltaeakkea mvrvaegeke sailvaegea 241 eairgtaiak aqgeaemikr tqmataeglk lvfsamkead idnnilalks mealekmaeg 301 kstklvlpse avnflgtfkg ikevmsddnk evldikevln dneslkk encoded by a Clostridium hypothetical nucleic acid SEQ ID NO: 12 1 atgggtacta aaattgtttt aagtattgta ttaatagtag tagtcgtagc aataagttta 61 acttgtataa gagttataaa acaatctaaa gtaggtataa taatgagact tggtaagttt 121 caaaaagtgg ctgaaacagg tgtacatttc ttaataccat ttttagataa aatggcatat 181 gtaattgacc ttagagaaat tgtaatagat tttccacctc aaccagttat aactaaagat 241 aatgtaacta tgcagattga tacagttgta tattataaag ttacagaccc agttagatat 301 gtgtttgaga tagctaatcc aatagctgct attgaaaatt taacagctac aacactaaga 361 aatataattg gtgaacttga tttagatgaa acattaacat caagagatat aataaatgta 421 aaaatgagaa ctattcttga tgaagcaaca gataaatggg gtataaaagt aaatagagta 481 gagttaaaaa atataatgcc tcctcaggat attcaagttg caatggaaaa acaaatgaga 541 gcagaaagag aaagaagaga ggcaatactt caageagaag gtaataagtc agctgcaata 601 cttcaagctg aaggtgaaaa acaatctgct atattaacag cagaagccaa aaaagaagct 661 atggtacgtg tagcagaagg tgaaaaagaa tctgctatat tagtggcaga aggtgaagct 721 gaagctataa gacagacagc tattgctaag gcacaaggtg aagctgaaat gataaaaaga 781 actcaaatgg caacagcaga aggtttaaaa ttagtttttt cagcaatgaa agaggctgat 841 atagacaata atattctagc attaaaatct atggaagctc ttgaaaaaat ggctgaaggt 901 aagtcaacaa aactggtttt accttcagaa gcagttaatt tcctaggaac attcaaagga 961 ataaaggaag ttatgagtga tgataacaaa gaagtacttg atataaaaga agttttaaat 1001 gataatgaat cattaaaaaa ataa In one embodiment, a Clostridium hypothetical protein (CD 1511) may be isolated as a hybridized antibody-protein complex. In one embodiment, the hypothetical protein comprises an amino acid sequence of SEQ ID NO: 13:

1 midnqkyvil slelhlffsr imkehalfle agftnknynl ameadhykkq fedllsytvs 61 asngiirpdi lyseelvttl tsvaeqktee ftgieinkni ttrelnlqsg vnpgvggdly 121 nyvaqlnsda irlldglinf kervldgvls ctiftsnypl llehiihean lyrsyvvdle 181 nkidieskna keielfwdhi mmehalfmrg lldpsegeli ntsndfaikf neliektnem 241 tdsniknite etlnetvefk dfkeagasgi egckiksiil plladhvlre anhyiriles 301 yknm encoded by a Clostridium hypothetical nucleic acid SEQ ID NO: 14:

1 ttacatgttt ttataactct ccaatattct aatataatga tttgcctctc ttaaaacatg 61 gtctgctaaa agtggtaata ttatagattt tatcttacac tgttctattc ctgatgctcc 121 tgcttcttta aaatctttaa actcaacagt ttcatttaga gtttcttctg taatgttctt 181 gatattagaa tcagtcattt cgtttgtttt ttcaattaat tcattaaatt ttatagcaaa 241 atcatttgaa gtatttatta gttcaccttc tgaggggtct agtaatcctc tcataaacag 301 agcatgttcc atcataatat ggtcccagaa taattctatt tctttagcgt tttttgactc 361 aatatctatt ttattttcaa ggtcaactac ataagaacga tataaatttg cttcatgtat 421 tatatgttca agaagtagag ggtagtttga tgtaaatata gtacatgata gtacaccatc 481 taagactctt tcttaaaat taataagccc atcaagtaat cttatgcat cagagttaag 541 ttgagctaca tagttcacta aatcttgacc aacttgtggg tttacaccac tttgtagatt 601 taattctctt gtagtgatgt ttttgtttat ttctatccct gtaaactctt ctgttttttg 661 ttctgcaact gatgtgagag tagtacaag ttcttctgaa tataatatat caggtctaat 721 tataccatta ctagcactaa cagtgtatga taataaatct tcaaattgct ttttatagtg 781 gtcagcttcc atagcaagat tataattttt atttgtgaat cctgcttcta aaaaaagagc 841 atgctctttc ataattcttg aaaaaaataa atgtaattct agtgataaaa taacatattt 901 ttgattatct atcat B. Clostridium difficile Collagen-Like Proteins The B. anthracis exosporium protein BelA was compared to the C. diff cile genome, wherein three C. difficile genomic areas were identified as homologous to B.
anthracis Bc1A (i.e., for example, spore surface genomic areas).
In one embodiment, a Clostridium spore surface homolog protein comprises an amino acid sequence of SEQ ID NO:15 (Accession Number CAJ67154):

mmiilylnd dtfiskkypd knfsnldycl igskcsnsfv keklitffkv ripdilkdks ilkaelfihi dsnknhifke kvdieikris eyynlrtitw ndrvsmenir gylpigisdt snyiclnitg tikawamnky pnyglalsln ypyqilefts srgcnkpyil vtfedriidn cypkcecppi ritgpmgprg atgstgpmgv tgptgstgat gsigptgptg ntgatgsigp tgvtgptgst gatgsigptg vtgptgntgv tgsigptgat gptgntgvtg sigptgvtgp tgntgeigpt gatgptgvtg sigptgatgp tgeigptgat gatgsigptg atgptgatgv tgeigptgei gptgatgptg vtgsigptga tgptgatgei gptgatgptg vtgsigptga tgptgatgei gptgatgptg vtgeigptga tgptgntgvt geigptgatg ptgntgvtge igptgatgpt gvtgeigptg ntgatgsigp tgvtgptgat gsigptgatg atgvtgptgp tgatgnssqp vanflvnaps pqtlnngdai tgwqtiigns ssitvdtngt ftvqengvyy isvsvalqpg sssinqysfa ilfpilggkd laglttepgg ggvlsgyfag flfggttfti nnfssttvgi rnggsagtaa tltifriadt vmt encoded by a Clostridium spore surface homolog nucleic acid SEQ ID NO: 16:
atgagaaata ttatacttta tttaaatgat gatactttta tatetaaaaa atatccagat aaaaacttta gtaatttaga ttattgetta ataggaagta aatgttcaaa tagttttgta aaagaaaagt tgattacttt ttttaaagtg agaataccag atatattaaa agacaaaagt atattaaaag cagagttatt tattcatatt gattcaaata agaatcatat ttttaaagaa aaagtagata ttgaaattaa aagaataagt gaatattata atttacgaac tataacatgg aatgatagag tgtctatgga aaatatcagg ggatatttac caattgggat aagtgataca tccaactata tttgtttaaa tattacggga actataaaag catgggcaat gaataaatat eetaattatg ggttagettt atctttaaat tacccttatc agattettga atttacatct agtagaggtt gtaacaaacc gtatatactt gtaacatttg aagatagaat tatagataat tgttateeta aatgtgagtg tcctccaatt agaattacag gtecaatggg accaagagga gegacaggaa gtacaggacc aatgggagta acaggcccaa ccggaagtac aggagcgaca ggaagcatag gaccaacagg cccaaccgga aatacaggag caacaggaag tatagggcca acgggagtaa caggcccaac eggaagtaca ggagcgacag gaagtatagg accaacagga gtaacaggtc cgacaggaaatacgggagtg acaggaagta taggaccaac gggagcaaca ggcccgacag gaaatacggg agtgacagga agtataggac caacaggagt aacaggccca acaggaaata caggagaaataggaccaacg ggagcaacag gtccaacagg agtgacagga agtataggac caacaggagc aacaggacca acaggagaaa taggaccaac gggagcaaca ggagcgacag gaagtatagg accaacagga gcaacaggtc caacaggagc gacaggagtg acaggagaaa tagggccaac aggagaaata ggaccaacgg gagcaacagg cccaacagga gtgacaggaa gtataggacc aacgggagca acaggcccaa caggagcgac aggagaaata ggaccaacag gagcaacagg cccaacagga gtgacaggaa gtataggacc aacgggagca acaggeccaa caggagcgac aggagaaata ggaccaacgg gagcaacagg cccaacagga gtaacaggag aaataggacc aacgggagca acaggeccaa caggaaatac aggagtaaca ggagaaatag gaccaacggg agcaacgggtcegacaggaa atacaggagt gacaggagaa ataggaccaa cgggagcaac aggaccaaca ggagtgacag gagaaatagg gccaacagga aatacaggag cgacaggaag tatagggcca acgggagtaa caggtccaac aggagcgaca ggaagtatag gaccaacggg agcaacagga gcgacaggag taacaggacc aacaggtcca aeaggagcaa caggcaattc ctctcagcca gttgctaact tcctcgtaaa tgcaccatct ccacaaacac taaataatgg agatgctata acaggttggc aaacaataat aggaaatagt tcaagtataa cagtagatac aaatggtacg tttacagtac aagaaaatgg tgtgtattat atatcagttt cagtagcatt acaaccaggt tcatcaagta taaatcaata ttctttcgct atcctattcc caattttagg aggaaaagat ttggcagggc ttactactga gecaggagge ggaggagtac tttctggata ttttgctggt tttttatttg gtgggactac ttttacaata aataattttt catctacaac agtagggata cgaaatgggc aatcagcagg aactgcggct actttgacga tatttagaat agctgatact gttatgactt as In one embodiment, a Clostridium spore surface homolog protein comprises an amino acid sequence of SEQ ID NO:17 (Accession Number CAJ70248):

mllimsrnky fgpfddndyn ngydkyddcn ngrddynscd chhceppscv gptgpmgprg rtgptgptgp tgpgvgatgp tgptgptgpt gntgntgatg lrgptgatga tgptgatgai gfgvtgptgp tgatgatgad gvtgptgptg atgadgitgp tgatgatgfg vtgptgptga tgvgvtgatg ligptgatgt pgatgptgai gatgigitgp tgatgatgad gatgvtgptg ptgatgadgv tgptgatgat gigitgptgp tgatgigitg atgligptga tgtpgatgpt gatgptgvgv tgatgatgat gadgatgvtg ptgatgatga nglvgptgat gaagtpgatg ptgatgptgv gitgatgatg atgptgadga tgptgatgnt gadgvagptg atgntgadga tgptgatgat gadgatgptg atgatgvaga tgatgptgat gadgatgptg atgatgadga tgptgatgat gvtgatgptg ptgatgatga tgasaiipfa sgiplsltti agglvgtpgf vgfgssapgl sivggvidlt naagtltnfa fsmprdgtit sisayfstta alslvgstit itatlyqsta pnnsftavpg atvtlapplt gilsvgsiss givtglniaa taqtpdrqyai encoded by a Clostridium spore surface homolog nucleic acid SEQ ID NO: 18:
ttatatggca tactgtctgt ctggagtttg tgctgttgct gctatattta atectgttac aattecacta gaaattgaac caactgataa tatacctgta agtggtggag ctagtgtaac tgtcgctcct ggtacagctg taaatgagtt atttggtgca gtagattggt aaagtgttgc tgtaattgta attgttgaac caacaagtga aagtgctgct gttgtactga agtatgctga aatagatgtt attgttccat ctcttggcat tgaaaatgca aagttagtca atgttcctgc tgcgtttgta aggtctatta ctccaccaac tatacttaat cctggagccg aactaccaaa tccaacaaat ccaggtgtac ctactaatcc tccagctata gttgtaagtg atagtggtat acctgatgca aaaggtatta ttgcactagc acctgttgct cctgttgctc ctgttgctcc tgttgggcct gttgggcctg ttgctcctgt aactcctgtt geccctgttg ctcctgttgg acctgttgct ccatctgccc ctgttgctcc tgttgctcct gttggacctg ttgctccatc tgctcctgtt gctcctgttg gacctgttgc tcctgttgct cctgccactc ctgttgctcc tgttgctcct gttggacctg ttgctccatc tgctcctgtt gcccctgttg ctcctgttgg acctgttgct ccatctgctc ctgtatttcc tgttgctcet gttggacctg ctactccatc tgctcctgta tttcctgttg ctcctgttgg acctgttgct ccatctgctc ctgttggacc tgtcgetcct gttgcccctg ttgctcctgt tattcctact cctgttgggc ctgttgctcc tgttggacct gttgctccag gtgttcctgc tgctectgtg getectgttg ggcctactaa tccatttgct cctgttgccc ctgttgctcc tgttggacct gttactcctg ttgctccgtc tgctcctgtt gctcctgttg ctcctgttgc tcctgttact cctactcctg ttgggcctgt tgctcctgtt ggacctgttg ctccaggtgt tcctgttgcc cctgttggac etattaatcc tgttgcccet gttatcccta ttcctgttgc tcctgttgga cctgttgggc ctgttattcc tattcctgtt gctcctgttg ctcctgttgg gcctgttact ecatctgctc ctgttgcccc tgttgggcct gttgggcctg ttactcctgt tgctccatct gctcctgttg ctectgttgc tcctgttgga cctgttattc ctattcctgt tgctcctatt gcccctgttg gacctgttgc tccaggtgtt cctgtcgctc ctgttggacc tattaatcct gttgctcctg ttactectac tcctgttgct cctgttgggc ctgttggacc tgttactcca aatcctgttg cccctgttgc tcctgttgga cctgttattc catctgctcc tgttgctccc gttggacctg ttggacctgt tactccatct gctcctgttg ctcctgtcgc tcctgttggg cctgttggac ctgttactec aaaccctata gctcctgtcg ctcctgttgg gcctgttgcc cctgttgctc ctgttggacc tcttaatcct gttgctcctg tattecctgt atttcctgtt ggaccagtcg gaceggttgg tcctgttggg cctgttgccc ctactcctgg acctgttgga cccgttggac ctgttgggcc ggttctacct cttggaccca ttgggcctgt tggacctaca catgatggtg gacagcaatg atggcaatca cagctattat aatcatcacg accattgtta caatcatcat atttatcata gccattgttg taatcattat catcaaatgg tccaaaatat ttatttetac tcattattaa aagcac In one embodiment, a Clostridium spore surface homolog protein comprises an amino acid sequence of SEQ ID NO:19 (Accession Number CAJ70128):
msdisgpsly qdvgptgptg atgptgptgp rgatgatgan gitgptgntg atgangitgp tgnmgatgpn gttgstgptg ntgatgangi tgptgntgat gangitgptg nkgatgangi tgstgptgnt gatgangitg ptgntgatga tgptgltgat gatgangitg ptgntgatga ngvtgatgpt gntgatgptg sigatgatgt tgatgpigat gatgadgevg ptgavgatgp dglvgptgpt gptgatgang lvgptgptga tganglvgpt gatgatgvag aigptgavga tgptgadgav gptgatgatg angatgptga vgatgangva gpigptgptg engvagatga tgatgangat gptgavgatg angvagaigp tgptgangat gatgatgatg angatgptga tgatgvlaan naqftvssss lvnntlvtfn ssfingtnit fptsstinla vggiynvsfg iratlslagf msittnfngv tgnnfiakav ntltssdvsv slsflvdara aavtlsftfg sgttgtsaag yvsvyriq encoded by a Clostridium spore surface homolog nucleic acid SEQ ID NO: 20:
ctattgtatt etataaactg atacatatcc agctgcagaa gtacctgtcg tgcctgaacc aaatgtaaaa cttaaagtaa cagctgctgc tctagcatca actaaaaagc ttaaacttac acttacatct gatgaagtaa gtgtatttac tgcttttgca ataaagttat tttgagttac tccattaaag ttagtagtaa ttgacataaa tcctgcaagt gaaagtgtgg cacgtatacc gaaagataca ttgtatatcc ctccaactgc aagatttata gtactacttg ttggaaaagt tatattagtt ccatttataa atgatgaatt aaatgtcact aatgtattat tcactaaact tgaagaggac actgtaaatt gtgcattgtt tgctgctaac actcctgttg ctcctgtcgc tccggttgga cctgttgctc catttgctcc tgttgctcct gtcgcccctg ttgctcctgt cgctccattt getceggttg ggcctgttgg tcctatcgct cctgctactc catttgctcc cgttgctcct actgctcctg ttgggcctgt tgctccattt gcccctgttg ctcctgtcgc tcctgttgct cctgctactc cattttctcc cgttggacct gttggaccta ttggacctgc tactccattc gctcccgttg ctcctactgc tcccgttggg cctgttgctc catttgcccc tgttgctcct gtcgctcctg ttggacetac tgetecatet gctcccgttg ggcctgttgc tcctactgct cccgttggac ctattgcccc agctactcct gttgctcctg tcgctcctgt tggacctacc aaaccatttg ctccggtgc tccggttggg cctgttgggc ctaccaaacc atttgetceg gttgctccgg ttgggcctgt tgggcctgtt ggacctacca aaccatctgg acctgttgct cctactgctc ctgttggacc tacctctcca tctgctcctg ttgctcctgt tgctcctatt gggcctgttg ccccagttgt tcctgttgct ccagtcgctc ctatacttcc tgttggacct gttgctcctg tatttcctgt tgggcctgta gcacctgtta ctecatttgc tccggttget cctgtatttc ctgttggtcc tgttatteca tttgctccgg ttgctcctgt tgctcctgtt agtccggttg gacctgttgc tcctgttgct cctgtattcc ctgttggacc tgttatteca tttgctccag tcgctectgt atttcctgtt ggtcctgtag aacctgttat tccatttgct ccggttgctc ctttgtttcc tgttggtcct gttattccat ttgetccggt tgctcctgta ttccctgtcg gacctgttat tccattcgct ccagtcgetc ctgtatttcc tgttggtcct gtagaacctg ttgttccatt tggtccagtc gctcccatat ttcctgttgg acccgttatt ccattcgctc cagttgcacc tgtattgct gttggtcctg ttattccatt tgctccggtc gctccagttg cacctetagg ccccgtcggt cctgttggac cagtagcacc tgttggccct gttggaceta catcttgata taaacttgga cctgaaatat cactcat C. Clostridium diff cle Spore Proteins 1. Putative Clostridum Spore Proteins In one embodiment, a Clostridium putative spore protein (CD 2401) may be isolated as a hybridized antibody-protein complex comprising a molecular weight including, but not limited to, 140, 150, or 160 kDa. See, Figures 5 & 6. In one embodiment, the hypothetical protein (- 22 kDa) comprises an amino acid sequence of SEQ ID NO: 21 (Accession Numbers YP_001088913; GI: 126700016):

encoded by a Clostridium putative spore protein nucleic acid SEQ ID NO: 22:
1 atgtggatat atcagaaaac tatacaacac ccagttaata taaaaacttg tgaccctaga 61 atggetaaat ttcttataac tcaatttggt gggecaaatg gggaacttge tgcatcttta 121 agatatttaa gccaaagata tacaatgcct actggtaata tgcgtgcact tttaacagat 181 attggtacag aagaactagc tcacgttgag cttatatgta ctatggttta tcagttaact 241 tctgatgcaa gcccagaaga gttaaaagct gcaggtettg gttcaaacta tgctcaaaat 301 ggatatggaa tttatccaac agattcaaat ggtgttecat ttgatgtaag acctatagca 361 gttatgtcaa atcecgtaac cgatttacat gaggatatgg cagctgaaca aaaagcactt 421 gcaacttatt atcaacttat aaacctaaca gatgacgttg atgttataga tgtattaaaa 481 ttcttgggtc aaagagaaat aattcactat caaagatttg gtgaagcttt aatggatget 541 tacgagttag aagaatctca aaaaatgttc taa In one embodiment, a Clostridium putative spore surface protein (CD 589) may be isolated as a hybridized antibody-protein complex comprising a molecular weight including, but not limited to, 140, 150, or 160 kDa. See, Figures 5 & 6. In one embodiment, a hypothetical protein (- 22 kDa) comprises an amino acid sequence of SEQ ID NO: 23 (Accession Numbers YP001087073; GI: 126698176):

encoded by a Clostridium putative spore protein nucleic acid SEQ ID NO: 24:

1 atgtggattt atcaaaaaac actggaacat ccagttaaca taagacaagc agacectaga 61 atggcaaaat atatcatgac tcagttggga ggacctaatg gtgagttggc agctgcaact 121 agatatcttc aacaaagata tactatgcca actggaaaat ctcgtgcact tttaactgat 181 ataggtacag aggaaatggc tcatgttgag ataatttctt cagtgttata tcaattaata 241 ggcaattgta ctccagaaga gcttaaggct gctggacttg gtagtaatta tgctaatttt 301 ggacatggtc ttcagccagt agattctaat ggagtaaact ttactacaag ttatattaat 361 gtctttggeg attcggtaac tgatttacat gaggatatgg ctgctgaaca aaaagcattg 421 getacgtact atcaattaat aaatttaact gatgaccctg atttgaaaga tatattgaga 481 tttttgggtg agagggaagt agttcactat caaagatttg gtgaagcatt aatggatgtt 541 tatgagttta cagagtgcaa gcatcagttt taa 2. Pyruvate-Flavodoxin Oxidoreductases In one embodiment, a Clostridium spore protein comprises a pyruvate-flavodoxin oxidoreductase protein (CD 2682) described by the amino acid sequence of SEQ
ID NO:
(Accession Numbers YP_001089193; GI: 126700296):

101 AGELLPGVFH VSARALASQA I. STFGDHQDV MAARQTGCVL LASGSVQEVA

451 YFDYDSKKSG GITMSHLRFG DTPIRATYLI DEADYIACHK QSYVYQYDLI:

651 7_.NLPVSTFNGI EDGTFPCG'I'A AYEKRGIAVD VPEWIIDNCI QCNQCSFICP

801 GSQFRQPLME FSGACAGCGE TAYIKVVTQI. YGDRMMIANA TGCSSIWGAS

encoded by a Clostridium pyruvate-flavodoxin oxidoreductase nucleic acid SEQ
ID NO:
26:

1 ttattcaget aattttttgt atccttcata tctttcttta gcatcagett cagctttagc 61 aaataateca tcagcttctt ctgggaattg tttagctata gcagaatatc ttacttgtgc 121 aagtatgaaa tctttgaagc ttgcagttgg ttcttttgaa tctaaagtga atgggttttt 181 accttcagct tttacttctg gattgaatct gtataaatgc cagtatccgc aagcaacagc 241 ttgtgcttcg ttagctacag ttcttcccat accttctttt aatccatggt ttatacatgg 301 agcataagct attattaaag atggcccatc atgtttttca gcttctaaaa cggctttgat 361 aaattggttt ttatcagcac ccatagcaac ttgagctaca taacgtttc catatgtcat 421 agccatcatt cctaaatctt tctttctaga ttttttacct gaagcagcaa acttagccat 481 agcagcagct ggagtagctt tagaagcttg acctcctgta tttgaatata cttctgtatc 541 aaatacaagt acgtttacat cttcaccaga tgcaagaaca tggtctaatc caccgtaacc 601 gatatcatat gcccaaccgt ctccacctag tatccattga gatctcttaa ctagataatc 661 ttttctatct tttatagctt ttactaattc tttagctttt tcatctgtac aatcacaagc 721 tggtttttct aataattcaa caaccttagc acttgctact ttagaagctt ctccatcatt 781 tctagaatct aaccactctg taaatactgc tttagcatct tcacagatat ccatagaaac 841 taactctgtc atagcatcta ctattttatt tcttatttgc ttaacagctg tgtacatacc 901 aaatccatat tcagcattat cctggaataa agagtttgcc caagatggac ctttaccttc 961 atggttacaa gtataaggaa ttgatggtgc agaagctccc catatagaag aaaatcaagt 1021 agcattagct atcatcattc tatctccata taattgtgta actactttaa tgtacgcagt 1081 ttcttcacat ccagcacatg ctcctgaaaa ctccattaat ggttgtctaa attgacttcc 1141 tttaacagta tttgcaggca taacatcaac ttttggagct acttcatcag gatttactgc 1201 aaatgcccaa ttatctaact ctgtatcaag ttgagtatct attggcttca taactaaagc 1261 tttttcttta gctggacata tgtcagcaca gtttccacat cctgtacagt caagtggact 1321 aacttgaatt ctatatttta atccttctaa tcctttacct aatgcttttt tagcttcaaa 1381 tccttctgga gcattagcta attcttcttc agtaactaat actggtctta tacatgcatg 1441 aggacatata aaagaacatt ggttacattg aatacagtta tctattatcc attcaggaac 1501 atcaacagct ataccacgtt tttcataagc tgcagtacca caagggaatg ttccatcttc 1561 tataccattg aatgtactta ctggtaagtt gtttccttct tgtgcagtca ttggtcttag 1621 tatatttttt ataaattctg gttctttaac ttctccttta tctgcatcaa cagcattttc 1681 ccaagaagct ggaacttcaa ctttaactaa agcatttatt cctgcatcaa cagcagcata 1741 gttcatatta actatttttt cacctttttt accataagct ttaactatag aatcttttaa 1801 gtatttaaca gcatcttctt ctggtattat ttcagctaat ttaaagaatg cagattgcat 1861 tatcatgttt attctgtttc caagacctat ttcttgtcct agtttaacag cgttaactgt 1921 atagaatttt atatcatttt tagctatata ttgtttcata tgagctggta aatgttcttc 1981 aagtcctgct tggtcccata tagtgttaag tacaaatgta ccaccttttt taagaccttt 2041 taataaatca tattgataaa catatgattg tttatgacag gcaatataat cagcttcatc 2101 tattaaataa gtagctctta ttggagtatc accaaatctt aagtgagaca tagttatacc 2161 accagatttt ttagagtcat aatcaaagta agcttgagca tatttttcag tatggtcacc 2221 gattattttg atagcttgtt tattagctcc aacagtaccg tcagaaccta atccccagaa 2281 cttacatctt atagttcctt tagaagctat ttttattggc tcagatggag ttaaagaagt 2341 attagttaca tcatcaccta tacctactgt aaatccattt ttaggttgtt ctgaaactaa 2401 gttatcaaaa acagttttta tatctgaagg agtagtatct tttgaaccta atccgtatct 2461 tccacctatt atcataggag cattttcttt tccatagaat acatcacgta catctaagta 2521 taatggttca ccagttgaac ctggctcttt agttctatca agtacacata ttctttcaac 2581 agtagatggc atagcatcta agaaatgttt cattgagaat ggtctatata aatgaacttt 2641 aactaaacca tatttttctc ccttagcatt tcagtaatct atagtttctt ctattgtttc 2701 tgttactgaa cccatagcta ttaatacata tttagcatct tctgctccat agtaatcaaa 2761 taaactatgt tttctacctg ttaaattact cattttttcc atgtattttt caactatacc 2821 tactatattt tgatagtatt tattagaagc ttctctagtt tggaaatata tatctgggtt 2881 ttgagcagta ccacgagcaa ctggatgatt tggagataaa gcattatctc taaatgcttg 2941 aactgcttca aagtttaata aacttgcata atcttcattt tctagtaatt caactttttg 3001 gatttcatgt gaagttctga atccatcaaa gaaatgtata aatggtaatc taccttctat 3061 tgcagctaca tgtgcaacag gtgctatatc agctacttct tgaactgaac cagaagctaa 3121 taatacacat ccagtttgtc tagcagccat aacgtcttgg tgatccccaa atattgataa 3181 cgcttgagat gctaaagcac gagcacttac atggaatact cctggtaata actctcctgc 3241 aactttatac atgttaggta tcattaataa taaaccttga gatgctgtaa aagtagaagt 3301 taaagctcct ccttgtaaag aaccgtggaa tgttcctgat gcacctgctt ctgactgcat 3361 ttcaacaaca ttaactgttt gtecaaatat attttttctt ccttgtgatg cccactcatc 3421 aactacttca gecatagttg aagatggtgt gattggatat atagctgcta catctgtaaa 3481 cgcataggca acatgagctg cagctgtatt tccatcaagt gttttcataa acttagecat 3. y-Aminobutryrate Metabolism Dehydratases/Isomerases In one embodiment, a Clostridium spore protein comprises a y-aminobutyrate metabolism dehydratase/isomerase protein (CD 2341) described by the amino acid sequence of SEQ ID NO: 27 (Accession Numbers YP_001088856; GI: 126699959):

encoded by a Clostridium gamma aminobutyrate metabolism dehydratase/isomerase nucleic acid SEQ ID NO: 28:

1 ttataaattt aaagcttcct ctttttttat tcccgcaatt ttcttagcta attctttctt 61 ttgatttatg tttccttgtc ttgaaatcat tattctttga gcttgtggtg aacctgcccc 121 atgcattgac tcagttctat aaccaacagc agatgaacca aggcatatat tttctaagaa 181 tcttaatatt ctcattcttt cttctgttga agccactgaa gaagetttaa aatacttgtt 241 acatacttcg ccaatagtca ttecacttgt tecaacttta aggtctgatt taaagtcttt 301 ttctgaaggc atagtaacca taagecctcc agetatatct tctgcaagtc ttactatttc 361 ataaggaaat ctagttacat tttgtttaca cacatttgca agaagtaaat ctatttgata 421 gtttccagct tctgttttgt gaccttctga tgaacatgct ataccacagc aatataagct 481 ttcattaagg tgagtcattt ctattagttt atettttata tgtgaagctt tgtttgctcc 541 attataatca gctgctaaag ctgctgetcc tattattaca tcacctactc ctactttaca 601 tccaccataa ctttgtctat ggtatcctgc aaatctctct actaacatac cagagaaatc 661 ccattcteca cataagaaaa ttttatcatt tggaacaaat acattatcaa atactactaa 721 agettettga ectecaaact cettatttec taaatcaaca tctgcacctt cttctaattt 781 tctagtgtca caagattgtc ttccataaat cataaacact ecttcagcat ctgaaggaac 841 tgcaaatgat acagcgtagt etttatctgc ttcagtcata cttatagtag gcattataag 901 gtgctcatgt gaatttattg aaccagtttg atgtgccttt gctcctctta caattatcec 961 atcttctctt ctttctacta tatgaaggaa catatctggg tcaggttgag cactaggtga 1021 tagactteta tcteccttag ggtcagtcat agctccatca actactaaat cgttttcttg 1081 tatataagtt aagtatttta caaaattatc atgataatta gttccatggg etttatcaca 1141 ctcaaatgtt gttgaataca ctgcattaaa cgcatccata ccaacacatc tttggaaaca 1201 agaagctgtt ttttgaccac atagtctttg catttttact ttttttatca aatcatcaac 1261 actttgatga agatgaccaa atctatttat tttttcacct gtgatatttg aagtaactgt 1321 cattaaatct gcatattctg ggtcatgagc taagtcataa gttgctgcta cacaatttat 1381 agatggtcta atcataggat ggtcgaccca attcttaacc tcttcaccaa acatataaac 1441 cttagtattt aattttctca aactttcaat atattgagct cctgtcatta atgccat 4. Butyryl-CoA Dehydrogenases In one embodiment, a Clostridium spore protein comprises a butyryl-CoA
dehydrogenase protein (CD 1054) described by the amino acid sequence of SEQ ID
NO:
29 (Accession Numbers YP_001087535; GI: 126698638)-51 GIPYPKEYGG EGGDTVGYIM AVEELSRVCG "I'1'GVILSAHT SLGSWPIYQY

encoded by a Clostridium butyryl-CoA dehydrogenase nucleic acid SEQ ID NO: 30:
1 atggatttaa attctaaaaa atatcagatg cttaaagagc tatatgtaag cttcgctgaa 61 aatgaagtta aacctttagc aacagaactt gatgaagaag aaagatttcc ttatgaaaca 121 gtggaaaaaa tggcaaaagc aggaatgatg ggtataccat atccaaaaga atatggtgga 181 gaaggtggag acactgtagg atatataatg gcagttgaag aattgtctag agtttgtggt 241 actacaggag ttatattatc agctcataca tctcttggct catggcctat atatcaatat 301 ggtaatgaag aacaaaaaca aaaattctta agaccactag caagtggaga aaaattagga 361 gcatttggtc ttactgagcc taatgctggt acagatgcgt ctggccaaca aacaactgct 421 gttttagacg gggatgaata catacttaat ggctcaaaaa tatttataac aaacgcaata 481 gctggtgaca tatatgtagt aatggcaatg actgataaat ctaaggggaa caaaggaata 541 tcagcattta tagttgaaaa aggaactcct gggtttaget ttggagttaa agaaaagaaa 601 atgggtataa gaggttcagc tacgagtgaa ttaatatttg aggattgcag aatacctaaa 661 gaaaatttac ttggaaaaga aggtcaagga tttaagatag caatgtctac tcttgatggt 721 ggtagaattg gtatagctgc acaagcttta ggtttagcac aaggtgctct tgatgaaact 781 gttaaatatg taaaagaaag agtacaattt ggtagaccat tatcaaaatt ccaaaataca 841 caattccaat tagctgatat ggaagttaag gtacaagcgg ctagacacct tgtatatcaa 901 gcagctataa ataaagactt aggaaaacct tatggagtag aagcagcaat ggcaaaatta 961 tttgcagctg aaacagctat ggaagttact acaaaagctg tacaacttca tggaggatat 1021 ggatacactc gtgactatcc agtagaaaga atgatgagag atgetaagat aactgaaata 1081 tatgaaggaa ctagtgaagt tcaaagaatg gttatttcag gaaaactatt aaaatag 5. Amino Acid Amino transferases In one embodiment, a Clostridium spore protein comprises an amino acid aminotransferase protein (CD 3664) described by the amino acid sequence of SEQ
ID
NO: 31 (Accession Numbers YP_001090189, GI: 126701292):

encoded by a Clostridium amino acid aminotransferase nucleic acid SEQ ID NO:
32:
1 ttaagcttet acacccatag cttcttttaa tacagctgct atatttttta taccttctat 61 tattttttct tctggcatat ttgaatagtt aagtctgaaa gtattttctc taccaccatt 121 agggaagaat cctcctccag caacataagc aacattctta tctaaacatt ttggcattaa 181 ttctttagca tttaaattac ttggaagctc tacccatgta aataatccac ettctggatg 241 agtaaatact aatccttctg ggaactcttc ttccatagtc ttaagcatta catctctacg 301 tttaacataa actgctttta ttttgtcaac atgtgcatct aggtcataca tatccataaa 361 tttgcttact tccatttgag atatagttga tgcttgtaag tctgcacctt gttttgcaaa 421 gttaaactta gataaaattt etggagaagc acaagtecat cctaatctgt atcetgggca 481 gaatatttta gagaaagttc ctaagaatat tactaatect tttgtatcca ttgattttaa 541 agatggtaaa gtctctectt caaatcttaa atctecatat ggattatctt ctattactgg 601 tatttcaaat ttatttacta tttccatgaa ttttttacgt ctttcaagtg gccaagttct 661 tcctgttgga ttttggaagt caggtattac gtaaatcatt tttattctat cagttgtttc 721 tagtatettt tetaattett ccattatcat tecateagaa tctgttggaa catctatgaa 781 tttaggttga taagatttaa atgcgtttat agctcctatg taagatggac tttcacataa 841 aattacatca ccctcatcaa tgaatacttt tectgcaaaa tctagacctt gttgagaacc 901 acttgtaact aatatatcat etttgtttac attagttttg tttttatcat tcattctagc 961 tgctattttt tctcttaatg gttcatatcc ttctgttgta gtatattgca tagctgatct 1021 tecattttct tcaagcactg caactgatac ttttttcatt tcctcaactg gaaataactc 1081 tggagccggc attccaccag caaaagatat tatttgtggt tgttgagtaa gttttaaaag 1141 ctcacgtatt tctgatcctt gtaatccttg cattcttttt gcatatttaa ctgecat 6. Succinic-Semialdehyde Dchydrogenascs In one embodiment, a Clostridium spore protein comprises a succinic-semialdehyde dehydrogenase protein (CD 2342) described by the amino acid sequence of SEQ ID NO: 33 (Accession Numbers YP001088857; GI: 126699960):

encoded by a Clostridium succinic-semialdehyde dehydrogenase nucleic acid SEQ
ID
NO: 34:

1 ttatccccaa agttcttcat ctgtagggac ttcattttct ggcatataat aagcaattct 61 agaaatatta attagatgct tataatcaag attttcagag atgctgttat ttccccaaga 121 accacaacct agagtattag taggtgcaag accattaaag aaacttcctc cagcactagt 181 agcacaacat tgattaataa caaatcttga tacttctata ttttctcctg catattctat 241 atttttaact gtatttgaat gtatagatac actatgtcct tttccttcta catttaaatt 301 tgcttttgca atagctacac cttcttcaaa acttttatac ttgtatgcag atataactgg 361 acacattttt tcttttgcaa taatatcctc ttctcctggt ccatcagctt ctattactat 421 tatttttgta tcttctggta tctcaacacc tgcaattttt gcaattgtat gaactgattg 481 acctactgca tctttattca tagatttatt tacaaacata gcatttctga aagcttcttt 541 ttgtacttta tctctaacta taaaaccttt gttattttta aattcatcca ttattttatc 601 aaacatttct tctgctacta ttactgactg ttcacctgaa caaatgatac cattatcaaa 661 aattcttcct gctatgattt taggtactgc ttctttaatg tctacatctc tatctatgat 721 acattgaaca tttcctgccc ctacaccaag tgctggtttt ccacttgagt acgcagcttt 781 taccattecc attcctccag tggcaattac tacatctgct gatgaaatta aatttcttgt 841 attttctctt gattgttggt caagtatttg tattaaattt tctggagctc ctattttttc 901 taattcttca ttaatcatct ctacagtttt tgtactacat ccaattgctt tatgatgagg 961 tgtgataata attgcatttc ttcccttaag tgcaaacatt gcattgctca taggtgtaac 1021 gattggattt gtacaaggag ttattgccgc tactactcca actggtttag ctacctttgt 1081 tattccagtt tctctatcta tatcaattat accaactgat tttttatctt ttaaattgtt 1141 atatataact tttgctttgc ttttattttt agctacctta tcttcatata ctcccattcc 1201 agtctcttct acagcaagtt tagctaaata ttcagcatta taatacacta cttttccaac 1261 tatttttact attttatcta cttgttcttg agtataacat tcaaactctc tttgagcttt 1321 tcttgctctc tcaatataac cattaatgta ttccttactt aaatcttcaa agttttccac 1381 tgctttttcc at 7. Rubrerythrins In one embodiment, a Clostridium spore protein comprises a rubrerythrin protein (CD 1524) described by the amino acid sequence of SEQ ID NO: 35 (Accession Numbers YP001088025; GI: 126699128):

Si QGVDAEIIEG LRANFTGECT EVGMYL.AMSR QADREGYPEV AEAYKRIAFE

151 DAIHDTVHEM CKDEARHGKA FLGL,LNRHFG K

encoded by a Clostridium rubrerythrin nucleic acid SEQ ID NO: 36:

1 atgaaaaaat ttgtttgtac agtatgtgga tatatacatg aaggagatgc tgcaccagca 61 caatgtccag tatgtaaagt tggagctgat aaatttgaag aaatgaaagg cgaaatggtt 121 tgggctgatg aacatagaat aggagtagct caaggtgtag atgcagaaat aatcgaagga 181 ttaagagcta actttactgg tgagtgtaca gaagtaggaa tgtatttagc aatgagtaga 241 caagctgata gagaaggtta tccagaagta gctgaagcgt ataagagaat agcttttgaa 301 gaggctgaac atgctgctaa atttgcagaa cttcttggag aagttgtagt tgcagataca 361 aaagaaaact taagagttag agttgatgct gagtatggtg caactgatgg aaaattaaaa 421 ttagctaaga gagetaaaga attaggatta gatgctatac atgatacagt agatgaaatg 481 tgtaaagacg aagctagaca tggtaaagca ttcttaggat tattaaacag acattttgga 541 aaataa 8. Flavoprotein Alpha Subunits In one embodiment, a Clostridium spore protein comprises a flavoprotein alpha subunit protein (CD 1056) described by the amino acid sequence of SEQ ID NO:

(Accession Numbers YP 001087535; GI: 126698640):

1 MGNVLVVIEQ RF,NVIQTVSL ELLGKATEIA KDYDTKVSAL LLGSKVEGLI

encoded by a Clostridium flavoprotein alpha subunit nucleic acid SEQ ID NO:
38:
1 atgggtaacg ttttagtagt aatagaacaa agagaaaatg taattcaaac tgtttcttta 61 gaattactag gaaaggctac agaaatagca aaagattatg atacaaaagt ttctgcatta 121 cttttaggta gtaaggtaga aggtttaata gatacattag cacactatgg tgcagatgag 181 gtaatagtag tagatgatga agctttagca gtgtatacaa ctgaaccata tacaaaagca 241 gcttatgaag caataaaagc agctgaccct atagttgtat tatttggtgc aacttcaata 301 ggtagagatt tagctcctag agtttctgct agaatacata caggtcttac tgctgactgt 361 acaggtettg cagtagctga agatacaaaa ttattattaa tgacaagacc tgcatttggt 421 ggaaatataa tggcaacaat agtttgtaaa gatttcagac ctcaaatgtc tacagttaga 481 ccaggggtta tgaagaaaaa tgaacctgat gaaactaaag aagctgtaat taaccgtttc 541 aaggtagaat ttaatgatgc tgataaatta gttcaagttg tacaagtaat aaaagaaact 601 aaaaaacaag ttaaaataga agatgctaag atattagttt ctgctggacg tggaatgggt 661 ggaaaagaaa acttagacat actttatgaa ttagctgaaa ttataggtgg agaagtttct 721 ggttetcgtg ccactataga tgcaggttgg ttagataaag caagacaagt tggtcaaact 781 ggtaaaactg taagaccaga cctttatata gcatgtggta tatctggagc aatacaacat 841 atagctggta tggaagatgc tgagtttata gttgctataa ataaaaatcc agaagetcca 901 atatttaaat atgctgatgt tggtatagtt ggagatgttc ataaagtgct tccagaactt 961 atcagtcagt taagtgttgc aaaagaaaaa ggtgaagttt tagctaacta a 9. 2-Ketoisovalerate Ferrodoxin Reductases In one embodiment, a Clostridium spore protein comprises a 2-ketoisovalerate ferrodoxin reductase protein (CD 116) described by the amino acid sequence of SEQ ID
NO: 39 (Accession Numbers YP001086585; GI: 126697688):

encoded by a Clostridium flavoprotein alpha subunit nucleic acid SEQ ID NO:
40::
1 atggctaaga tacttatgaa aggtaacgaa gcatttggaa aagcagetat agaagctggt 61 tgcaaatatt tcttcggtta cccaataact ccacaaagtg aattaccaga atatttatca 121 agagagttac ctaaaatagg tggagcgttc gttcaagctg aatcagaagt ttctgcgata 181 aacatggttt atggtggagc aggtgcagga gcaagagtta tgacttcttc atcttcacca 241 ggagttgett taaagcaaga aggtataaca tatgcagtag gagcagaagt tccttgtgtt 301 gtacttaatg taatgagagg tggtccagga cttggaagta tacaaccttc acaagctgac 361 tactttatgt ctacaagagg tggaggaaat ggagactaca gaactccagt attcgcacca 421 gctacagttc aagaagctgt tgatatgata atggaagctt ttgacgttgc agattattac 481 agatcaccag ttatggtagt tgcagatggt atgataggtc aaatgatgga gccagttgaa 541 tttagagctc ctgagaagaa aagagaattg cctcctaaag attgggcaac tgttggaact 601 aaaggaaaaa gaaaacctaa tgtaataaac tctctatact tagagecaga agtactagaa 661 gaccattgtt ggcatttaca agaaaaattt gatgcaatgg aaaagaatga agtacaatac 721 gaaatgtaca aaacagaaga tgctgaattt gtatttgcag cttatggaac tacttcagga 781 gtagtaaaaa gcgcaattga tatattaaga gaagaaggaa taaaagcagg tattataaga 841 cctaaagtat tatggccatt cccattegaa gcattcaacc aaattcctaa tgctagaaac 901 atattgactg ttgaaatgag tatgggacaa atggtggaag atgttaaaat ggcagtagaa 961 ggtaaattac cagtttactt ecatggaaga ccaggaggaa tgactccaac accagcagaa 1021 atagttgaaa aagctaaaaa aattattgec ggagaattgg ttgcaggagg tgctagataa D. Bacillus BcIA Proteins And Homologs The genus Bacillus is composed of rod-shaped, gram-positive, aerobic or (under some conditions) anaerobic bacteria widely found in soil and water. Most strains of Bacillus are not pathogenic for humans and only infect them incidentally in their role as soil organisms. A notable exception, however, is Bacillus anthracis. B.
anthracis is believed to cause anthrax in humans and domestic animals. In addition to its role as a naturally occurring pathogen, Bacillus anthracis may also be used as a biological weapon.
The spore form of B. anthracis is one of the most likely organisms to be used as a biological weapon. B. anthracis spores are well known to be highly resistant to most disinfectants, wherein effective killing usually requires the use of toxic and irritating chemicals such as formaldehyde or sodium hypochlorite (i.e., bleach).
Consequently, improvements to current decontamination procedures for both domestic and military applications are urgently needed, the present invention contemplates methods and compositions to provide effective and non-toxic Bacillus contamination control, and especially B. anthracis decontamination procedures for both animate and inanimate surfaces.
1. BcIA
In one embodiment, a B. anthracis Bc1A spore surface protein comprises an amino acid sequence (Accession Number AAY15450; SEQ ID NO:41):

msnnnysngl npdeslsasa fdpnlvgptl ppippftlpt gptgptgptg ptgptgptgp tgptgptgpt gdtgttgptg ptgptgptgp tgdtgttgpt gptgptgptg ptgptgptgp tgptgptgpt gptgptgptg ptgptgdtgt tgptgptgpt gptgptgptg ptgptgptgp tgptgptgpt gdtgttgptg ptgptgptgp tgdtgttgpt gptgptgptg ptgptgptga tgltgptgpt gpsglglpag lyafnsggis ldlgindpvp fntvgsqfgt aisqldadtf visetgfyki tviantatas vlggltiqvn gvpvpgtgss lislgapivi qaitqitttp slvevivtgl glslalgtsa siiiekva encoded by the B. anthracis Bc1A nucleic acid (Accession Number AY995120; SEQ
ID
NO: 42):

atgtcaaata ataattattc aaatggatta aacccegatg aatctttatc agctagtgca tttgacccta atcttgtagg acctacatta ccaccgatac caccatttac ccttcctacc ggaccaactg ggccgactgg accgactggg ccgactgggc caactggacc aactgggccg actgggccaa ctggaccaac tgggccaact ggagacactg gtactactgg accaactggg ccgactgggc caactggacc aactgggcca actggagaca ctggtactac tggaccaact gggccaactg gaccaactgg gccgactggg ccaactggac caactgggcc gactgggeca actggaccaa ctgggccaac tggaccaact ggaccaactg ggccaactgg accaactgga ccaactgggc caactggaga cactggtact actggaccaa ctgggccaac tggaccaact ggaccaactg ggccgactgg accgactggg ccgactgggc caactggacc aactgggccg actgggccaa ctggaccaac tgggccaact ggagacactg gtactactgg accaactggg ccaactggac caactggacc aactgggcca actggagaca ctggtactac tggaccaact gggccaactg gaccaactgg accaactggg ccaactggac caactgggcc aactggtgct accggactga ctggaccgac tggaccgact gggccatccg gactaggact tccagcagga ctatatgcat ttaactccgg tgggatttct ttagatttag gaattaatga tccagtacca tttaatactg ttggatctca gtttggtaca gcaatttctc aattagatgc tgatactttc gtaattagtg aaactggatt ctataaaatt actgttateg ctaatactgc aacagcaagt gtattaggag gtcttacaat ccaagtgaat ggagtacctg taccaggtac tggatcaagt ttgatttcac tcggagcacc tatcgttatt caagcaatta cgcaaattac gacaactcca tcattagttg aagtaattgt tacagggctt ggactatcac tagctcttgg cacgagtgca tccattatta ttgaaaaagt tgcttaa The BcIA protein contains multiple spore surface GXX repeats in its central region and appears to be a structural component of the hair-like nap of the exosporium identified by a major immunoblot band separating exosporium proteins probed with mouse anti-spore polyclonal antibodies. Sylvestre et al., "A collagen-like surface glycoprotein is a structural component of the Bacillus anthracis exosporium"
Mol.
Micro biol. 45:169-178 (2002).
The BeIA protein is believed to be highly glycosylated and an immunodominant antigen of the B. anthracis spore surface providing a structural component of exosporium filaments. Daubenspeck et al., J. Biol. Chem. 279:30945-30953 (2004). The length of the gene bola differs between strains of B. anthracis, encoding proteins of different sizes.
Most BcIA proteins possess an internal spore surface region (CLR) of GXX
repeats of variable length (i.e., for example, from approximately 17 to 91 GXX repeats), which include a large proportion of GPT triplets. It has been shown that the length of the Bc1A
spore surface region is responsible for the variation in filament length.
Boydston et al., "Orientation within the exosporium and structural stability of the collagen-like glycoprotein BeIA of Bacillus anthracis" J. Bacteriol. 187, 5310-5317 (2005).
One B.
anthracis Bc1A comprises 70 GPT repeats and has a 21 amino acid sequence ((GPT)5GDTGTT; SEQ ID NO: 43) repeated six times. BeIA polymorphisms may map to sequences encoding CLRs with a number of contiguous GXX triplets varying from 17 - 91. In one report, bcla genes were exchanged between strains having different CLRs and the produced exosporium was examined by electron microscopy. The data supports a conclusion that the BcIA protein CLR may control nap filament length.
Sylvestre et al., "Polymorphism in the collagen region of the Bacillus anthracis Bc1A protein leads to variation in exosporium filament length" JBacteriol. 185:1555-1563 (2003).
CLRs are rare in microorganisms, but have been identified in some bacteria and viruses. Rasmussen et al., (2003) J. Biol. Chem. 278, 32313-32316 (2003).
Their function is generally not clear, but they are found in surface proteins.
Bacillus cereus, and Bacillus thuringiensis, species close to B. anthracis, both possess a protein related to BeIA in their the exosporia. Todd et al., J. Bacteriol. 185, 3373-3378 (2003).
There is no sequence similarity, however, between the C-terminal portion of BeIA and proteins of known structure.
The C-terminal two-thirds of a Bc1A protein chain folds into an all-fl structure with a jelly roll topology. See, Figure 7. Similar folds in the TNF-like of proteins, ectodysplasin Eda-Al (DALI; Z-score 16.1) and Clq (DEJAVU; DALI Z-score 14.6) indicate a strong structural relationships. Holm et al., J. Mol. Biol. 233:123-138 (1993);
and Kleywegt et al., In: International Tables for Crystallography (Rossmann, M. G., and Arnold, E., eds) Vol. F, pp. 353-356, Kluwer Academic Publishers, Dordrecht, The Netherlands (2001). The twelve (3-strands, numbered sequentially A, A", A', B', B, C, D, E, F, G, G', and H, according to the TNF-like conventions, are connected with the Greek key topology and form two (3-sheets, each containing five strands, A'AHCF and B'BGDE, respectively. The main difference with other members of the TNF-like is the presence of the extra A" and G' strands that form a two-strand antiparallel (3-sheet, packing together the AA' and GH loops.
Three monomers related by crystallographic symmetry form a tight, globular trimer of -50 A in diameter. The total surface buried by the trimer formation is 1411 A2/monomer, with two small cavities filled with solvent molecules at the center. The C
and N termini come together on the bottom side of the trimer, with the N-terminal residues of the non-collagenous part intertwining and then pointing outwards.
See, Figure 8. Circular dichroism spectroscopy shows the entire trimer structure, except for the very N- and C-terminal residues, where for the latter, only the first two His residues of the affinity tag are visible. Seven residues per monomer have two alternate conformations; i) four that lie on the surface of the trimer close to the crystallographic 2-fold axis; and ii) three that lie at the trimer interface within one of the two cavities.
Clearly, BelA trimer assembly is very similar to other TNF-like trimers. Because of the shorter protruding loops, the overall shape of the BelA trimer is somewhat more globular than that of TNF, and it is slightly smaller than the globular head of Clq (by -5 A in diameter). Upon trimer formation, the buried surface of 4234 A2 is less than the equivalent surface areas in Clq (5490 A2) or collagen VIII NCI (6150 A). This is due both to its slightly smaller overall size but principally to the existence of a solvent-filled cavity within the trimer center.
In one embodiment, a BelA protein (BAS 1130) comprises an amino acid sequence of SEQ ID NO:44 (Accession Number YP027402):
1 msnnnysngl npdeslsasa fdpnlvgptl ppippftlpt gptgptgptg ptgptgptgp 61 tgptgptgpt gptgptgptg dtgttgptgp tgptgptgpt gdtgttgptg ptgptgptgp 121 tgptgptgpt gptgptgdtg ttgptgptgp tgptgptgdt gttgptgptg ptgptgptgp 181 tgptgptgpt gptgptgptg ptgdtgttgp tgptgptgpt gptgdtgttg ptgptgptgp 241 tgptgptgpt gatgltgptg ptgpsglglp aglyafnsgg isldlgindp vpfhtvgsgf 301 gtaisqldad tfvisetgfy kitviantat asvlggltiq vngvpvpgtg sslislgapi 361 viqaitqitt tpslvevivt glglslalgt sasiiiekva In one embodiment, a modified Bacillus anthracis protein (BelA) comprises an amino acid sequence of SEQ ID NO: 45, wherein the modification comprises a SEQ
ID
NO: 41 fragment:

MSNNNYSNGLNPDESLSASAFDPNLVGPTLPPIPPFTLPTGPTGPTGPTGPT
GPTGDTGTTGPTGPTGPTGPTGATGLTGPTGPTGPSGLGLPAGLYAFNSGG
ISLDLGINDPVPFNTVGSQFGTAISQLDADTI'FVISETGFYKITVIANTATASV
LGGLTIQVNGVPVPGTGSSLISLGAPIVIQAITQITTTPSLVEVIVTGLGLSLA
LGTSASIIIEK.VA

2. BcIB
In one embodiment, a Bc1B protein (BAS2281) comprises an amino acid sequence of SEQ ID NO:46 (Accession No. YP028542):

1 mkqndklwld kgiigpenig ptfpvlppih iptgitgatg atgitgatgp tgttgatgat 61 gitgvtgatg itgvtgatgi tgvtgatgit gvtgatgitg vtgptgitga tgptgttgvt 121 gptgdtglag atgptgatgl agatgptgdt gatgptgatg lagatgptga tgltgatgat 181 gatgggaiip fasgttpall vnavlantgt llgfgfsqpg iapgvggtlt ilpgvvgdya 241 fvaprdgiit slagffsata alapltpvgi gmgifiapaa sntftpvapp llltpalpai 301 aigttatgiq aynvpvvagd kilvyvsltg aspiaavagf vsaglniv D. Bacillus Exosporium Proteins The genus Bacillus also includes a diverse collection of gram positive, rod-shaped, aerobic bacteria that form a spore upon deprivation of an essential nutrient.
Enington, "Bacillus subtilis sporulation: regulation of gene expression and control of morphogenesis' Microbiol. Rev. 57:1-33 (1993); and Priest, F. G., "Systemics and ecology of Bacillus" In: Bacillus subtilis and other gram-positive bacteria.
Biochemistry, physiology, and molecular genetics. pp. 3-16., A. L. Sonenshein, J. A. Hoch, and R.
Losick (ed.), American Society for Microbiology, Washington, D.C. (1993). In this process, an asymmetric septation of the starved vegetative cell produces a large and a small genotne-containing compartment called the mother cell and forespore, respectively.
The mother cell then engulfs the forespore, thereby surrounding it with two opposing cell membranes. A thick layer of modified peptidoglycan called the cortex is synthesized between the two membranes, and proteins synthesized in the mother cell form multiple layers of a spore surface that covers the cortex. While the coat forms the outermost detectable layer for spores of some species (e.g., Bacillus subtilis), in others (e.g., Bacillus anthracis), the spore is enclosed by an additional layer called the exosporium, a prominent, loose-fitting, balloon-like layer also synthesized by the mother cell. Henriques et at., "Structure and assembly of the bacterial endospore surface" Methods 20:95-110 (2000).
Bacillus cereus, Bacillus anthracis, and Bacillus thuringiensis are representative of Bacillus bacteria believed to possess an exosporium. Ohye et al., "Exosporium and spore surface formation in Bacillus cereus TV Bacteriol. 115:1179-1190 (1973);
and Andersen G., "Genome differences that distinguish Bacillus anthracis from Bacillus cereus and Bacillus thuringiensis" Appl. Environ. Microbiol. 69:2755-2764 (2003). See, Figure 9A, 9B. B. subtilis, however, lacks a distinct, separate exosporium, although there has been a report that a very outermost tightly fitting layer of the spore surface can be visualized after extracting some of the coat material from spores with urea-mercaptoethanol and might be considered an exosporium. Sousa et al., "An exosporiurn-like outer layer in Bacillus subtilis spores" Nature 263:53-54 (1976). The exosporium of Bacillus cereus is first observed as a small lamellar structure in the mother cell cytoplasm in proximity to, but not in contact with, the outer forespore membrane; it is synthesized concurrently with the spore surface, although the two structures are clearly separate within the mature spore. Exosporia usually contain a hexagonal crystal-like basal layer comprising 53% protein, 20% amino and neutral polysaccharide, 18% lipids, and approximately 4% ash. The exosporium comprises approximately 2% of the dry weight of the spore. The hydrophobic properties of Bacillus megaterium QMB 1551 spores are also believed to be dictated by the exosporium, as spores with a defective or absent exosporium show greatly reduced affinity for hexadecane. Koshikawa et al., "Surface hydrophobicity of spores of Bacillus spp" J. Gen. Microbiol. 135:2717-2722 (1989).
After a final stage of maturation, the mother cell lyses to release the mature spore, which is dormant and capable of persisting in the soil for many years until it encounters a germination signal. Most Bacillus species are not pathogenic to humans. The most notable exception is B. anthracis, the causative agent of anthrax. Mock et al., "Anthrax"
Annu. Rev. Microbiol. 55:647-671 (2001). In light of the recent use of B.
anthracis spores as a terrorist weapon in the United States and the development of these spores as a weapon of mass destruction by a number of countries, there is a pressing need to learn more about spore components that can be used for rapid detection and targeted for decontamination, treatment and prevention of anthrax. Inglesby et al., "Anthrax as a biological weapon, 2002: updated recommendations for management" JAMA 287:2236-2252 (2002).

One such component is the exosporium, which serves as a primary permeability barrier for the spore and as the source of spore surface antigens. Gerhardt, P., "Cytology of Bacillus anthracis" Fed. Proc. 26:1504-1517 (1967). As the outermost surface of the spore, the exosporium will interact with the soil environment, with collection and detection devices, with spore-binding target cells in a mammalian host, and with host defenses. Presently, detailed information about exosporium structure and function is limited. Previous studies of the exosporium focused on spores of B. anthracis and the opportunistic human pathogen Bacillus cereus. These species, along with Bacillus thuringiensis, an insect pathogen, and Bacillus mycoides make up the closely related B.
cereus group. All members of this group produce spores with a structurally similar exosporium composed of a basal layer and an external hair-like nap extending up to 600 A in length. Gerhardt et al.,"Ultrastructure of the exosporium enveloping spores of Bacillus cereus" J. Bacteriol, 88:1774-1789 (1964); and Hachisuka et al., "Fine filaments on the outside of the exosporium of Bacillus anthracis spores" J. Bacteriol.
91:2382-2384 (1966). The basal layer contains four paracrystalline sublayers, each exhibiting a hexagonal, perforate lattice structure. The hair-like nap varies in length from species to species and even from strain to strain. Beaman et al., "Paracrystalline sheets reaggregated from solubilized exosporium of Bacillus cereus" J. Bacteriol. 107:320-324 (1971); and Kramer et al., "Ultrastructural differences in the exosporium of the Sterne and Vollum strains of Bacillus anthracis" Can. J. Microbiol. 14:1297-1299 (1968).
The exosporium reacts with lectins, which appear to bind polysaccharides located in the hair-like nap. Cole et al., "Differentiation of Bacillus anthracis and other Bacillus species by lectins" J. Clin. Microbiol. 19:48-53 (1984); Matz et al., "Chemical ultrastructure of the exosporium enveloping spores of Bacillus cereus"
Bacteriol. Proc., p. 31 (1965). The exosporium constitutes about 2% of the mass of the spore and contains approximately 50% protein, 20% lipid, 20% neutral polysaccharides, and 10%
other components. Matz et al., "Chemical composition of exosporium from spores of Bacillus cereus" J. Bacteriol. 101:196-201 (1970).
The exosporium contains multiple proteins that are synthesized concurrently with the cortex and coat. Desrosier et al., "Synthesis of the exosporium during sporulation of Bacillus cereus" J. Gen. Microbial. 130:935-940 (1984); and Ohye et al., "Exosporium and spore surface formation in Bacillus cereus T' J. Bacteriol. 115:1179-1190 (1973).

More recent studies have attempted to identify individual exosporium proteins.
An apparent 72-kDa glycoprotein was identified as a component of either the exosporium or the coat of B. thuringiensis spores. Garcia-Patrone et al., "A glycoprotein multimer from Bacillus thuringiensis sporangia: dissociation into subunits and sugar composition" Mol.
Cell. Biochem. 145:29-37 (1995). Three other proteins (GroEL and homologues of InA
and RocA) were found to he associated with, although probably not structural components of, the B. cereus exosporium. Charlton et al., "Characterization of the exosporium of Bacillus cereus" J. Appl. Microbiol. 87:241-245 (1999).
The Bacillus spore itself is believed responsive to environmental stimuli. In one report, atomic force microscopy was used to identify species-specific alterations in spore morphology when exposed to and versus humid environments. For example, dimensional changes appeared dependent upon species, growth regimes, and environmental conditions. In general, spore widths may fluctuate between 500 -mn depending upon the interaction of these factors. Plomp et al., "The high-resolution architecture and structural dynamics of Bacillus spores" Biophys J 88:603-608 (2005).
Additionally, in addition to BelA, the B. anthracis exosporium is also believed to contain four other primary proteins: i) alanine raccmase; ii) iron/manganese superoxide dismutase; iii) a 29,174-Da protein having an unknown function; and iv) a 17,331-Da protein having an unknown function. Steichen et al., "Identification of the immunodominant protein and other proteins of the Bacillus anthracis exosporium" J
Bacteriol. 185:1903-1910 (2003).
As the outer surface layer of mature bacterial spores mature, the exosporium may represent a contact surface between a spore, the environment, and/or a host.
The exosporium comprises proteins that are responsible for functions including, but not limited to, immunogenicity, environment/host attachment and stability, or germination responses. Redmond et al., "Identification of proteins in the exosporium of Bacillus anthracis" Microbiol 150:355-363 (2004).
For example, a bacterial species exosporium protein may include, but are not limited to, putative coat proteins (i.e., for example, genes cotJC1 or cotJC2), hypothetical exosporium proteins, Bc1A proteins, BcIB proteins, Bel Sterne proteins, modified BelA or Be1B proteins, ExsF proteins; CotB proteins; ExsY proteins; CotY proteins;
ExsK
proteins, inosine hydrolases, alanine racemases, pyruvate-flavodoxin oxidoreductases (i.e.,. for example, gene nifJ), gamma-aminohutyrate metabolism dehydratases/isomerases (i.e., for example, gene abID), butyryl-CoA
dehydrogenases (i.e., for example, gene bcd2), amino acid aminotransferases, succinate-semialdehyde dehydrogenases (i.e., for example, gene sucD), rubrerythrins, flavoprotein alpha-subunits (i.e., for example, gene etfA2), or 2-ketoisovalerate ferredoxin reductases.
Size characteristics of representative exosporium proteins are summarized in Table II:
Table 11: Size Characteristics Of Various Exosporium Proteins rotein (alternative BAS gene ame) Size (Da) o.a Alanine racemase 13,662 238 BelA 36,836 1130 BxpA 27,702 2008 PxpB (ExsF) 17,331 1144 xpC (BA2332) 14,379 2174 CotB-1 19,422 340 CotB-2 16,830 341 CotY (CotZ-2) 16,842 1145 ~ExsK 12,034 2377 ExsY (CotZ, CotZ-1) 16,146 1141 Inosine-uridine 6,276 2693 ucleoside hydrolase Superoxide dismutase 36,162 1378 1. Alanine Racemase In one embodiment, a Bacillus thuringiensis serovar kurstaki strain 1.175 alanine racemase (i.e., for example, gene alrA) (Accession No. DQ402058) comprises an amino acid sequence (SEQ ID NO: 47):

MEEAPFYRDTW VEVDLDAIYNNVTHIKEFIPSNVEIFAV VKANAYGHDYV
PVAKTALEAGATRLAVAFLDEALVLRRAGITEPILVLGPSPPRDVNVAAEN
DVALTVFQKEWVEEAIKLWDGSSVMKFHINFDSGMGRIGIRERKELKEFL
KSLEGAPFLELEGVYTHFATADEVETSYFDKQYNTFLEQLSWLKEFGVDP
KFVHTANSAATLRFQGITFNAVRIGIAMYGLSPSVEIRPFLPFELEPALSLHT
KVAHIKQVIKGDGISYNVTYRTKAEEWIATVAIGYADG WLRRLQGFEVLI
NGKRVPIVGRVTMDQFMIHLPCEVPLGTKVTLIGRQGDEYISATEVAEYS
GTINYEIIATISFRVPRIFIRNGKVVEIINYLNNI
encoded by the nucleic acid sequence (SEQ ID NO: 48):

1 cgtgcgatac tcacatacac tggaaataag aaatccttta etttaataca agaaaaggca 61 aaagttgcag aggcatcatc agcggtaagt gtaagtggag aaccagttga tettgggttt 121 acgattggtg cattgacgaa agactctgta acgtggtcge ataacagagt agaatatatg 181 ctcgtgtcta aaggtttaga gccgaaggag ctgttaatgg ttgctcgttc agttacageg 241 aagcaggtga agtaaacttc ttagacgtgg tgatatatgt gcaccaegtc ttttcttagt 301 ttggaggtgg atttcataaa agaagcatat aaaagaataa gcttcgcata tcgtgtataa 361 ggaagtgtat ttatggaaga agcaccattt taccgtgaca cttgggtgga agtggattta 421 gatgecattt ataacaacgt tacacatatt aaagagttca tcccaagtaa tgtagagatt 481 tttgctgtag ttaaagcaaa cgcatatggg cacgattatg taccggtggc taaaacagca 541 ttagaagcgg gggcaacaag gttagctgtt gcttttttag atgaagettt agtgcttcgg 601 agggctggta ttactgaacc gattttagtg ttaggtccct cgccgccgcg tgatgtaaat 661 gtagctgctg aaaatgatgt agegetaact gtttttcaaa aagaatgggt ggaagaagca 721 attaaacttt gggatggttc atctgtaatg aaattccata ttaactttga tagtggtatg 781 gggagaattg gaatacgtga aagaaaagag ctaaaagaat ttttaaagag tttagagggt 841 gcaccgtttt tagagttaga aggagtatac acgcattttg caacggcaga tgaggttgag 901 acttcgtatt ttgataagca atataacacg ttcttggagc agttaagttg gttgaaagaa 961 ttcggagtgg atcctaagtt tgttcataca gctaatagtg ctgcgacgtt acgttttcaa 1021 gggattacat ttaatgcagt gcgaattggg attgcaatgt atggattatc tccttctgta 1081 gaaatacgtc cttttttacc atttgaatta gaaccggcgc tatcacttca tacgaaagtt 1141 gctcatatta aacaggtaat taaaggtgat ggaattagtt ataacgtcac ctatcgaacg 1201 aaagctgaag aatggattgc gactgtagcg attggctatg cagatgggtg gcttagaagg 1261 ttacaaggat ttgaagtgct tataaatggt aaaagggtac cgattgtagg gcgagtaaca 1321 atggatcagt tcatgattca tcttcettgt gaagtgecac ttggtacgaa agttacgctt 1381 attggaaggc aaggagatga gtatattagc gctacagagg ttgcggaata ctcagggact 1441 attaattatg aaattattgc aacgatcagt ttccgtgtgc caagaatatt tatacggaat 1501 ggtaaggttg tagagataat taattacttg aacaatatat agaaggtagt atgattgctt 1561 ettgtcattg gagtgactgt tcattgctgg ataataagct ctttttgatg agaagctttt 1621 tacttataat cattttttat ataagttgca cgtaagtttt gaagaggagt tctattgttt 1681 ttagttagtt atgagataaa aaatttaaga agatgaatgt tttgtttgtg aattggaaaa 1741 cataagcaag gaataaggaa gtctttgcaa caggctccat acaatggtat tattacaata 1801 ggtgtcatat atatatttgg gtgtgtagtt gacggtggag gtgtattttt gtgtccgaat 1861 caagtgtaac tactgaaatc gtggttcggt tgccaaagca aatggtaacg gaattggaag 1921 gaattggaaa acaagagaat aagaatcgcc atgaactaat ttgccaggca acacaattgt 1981 tattgcgtca acataagacg aagaaacgct accaacatga atcaatgcga cgtgggtaca 2041 ttgaaatggg aaaaattaat cttggtattg catctgaagc tttcttagca gagtatgaag 2101 cagctcatac agtagaacgc ttagttagc 2. Sterne Proteins In one embodiment, a Bacillus anthracis str. Sterne protein (BAS3290) comprises an amino acid sequence of SEQ ID NO :49 (Accession No. YP029547):
1 msekyiilhg talepnligp tlppippftf pngptgitgp tgatgftgig itgptgvtgp 61 tgigitgptg atglgilpvf gtittdvgig fsvivntnin ftlpgpvsgt tlnpvdnsii 121 inttgvysvs fsivfviqai sssilnltin dsigfaiesr igggpgvrat sartdllsln 181 ggdvlrvrir eatgdiiysn aslvvskvd In one embodiment, a Bacillus anthracis sir. Sterne protein (BAS3557) comprises an amino acid sequence of SEQ ID NO: 50 (Accession No. YP029811):

1 msryddsgnk fskpcfpssa gripntpsip vtkaglrtfr aiiidltkii pklfanpspq 61 niedlidtln llskficsld aasslkaggl aiiknlitil knptfvasav fielgnliny 121 llsitklfri dpctlqellk liaalgtaly nsasfiqgpt gptgptgptg pagatgatgp 181 qgvqgpagat gatgpqgvqg pagatgatgp qgaqgpagat gatgpqgaqg pagatgatgp 241 ggiggpagat gatgpqgvqg ptgatgigvt gptgpsggpa gatgpqgpqg ntgatgpggi 301 qgpagatgat gpqgaqgpag atgatgpqgv qgptgatgig vtgptgpsgp sfpvativvt 361 nniqqtvlqf nnfifntain vnniifngtd tvtvinagiy visvsistta pgcaplgvgi 421 singavatdn fssnligdsl sfttietlta ganisvgstl neitipatgn tnirltvfri 481 a In one embodiment, a Bacillus anthracis str. Sterne protein having a collagen triple helix repeat domain comprises an amino acid sequence of SEQ ID NO: 51 (Accession No. YP030670):
1 minmsswrnn ingycscnnq ngvhvdsccf scdgtvpklg ptgptgatga tgatgvtgvt 61 gvtgatgitg atgitgatgi tgvtgptgat gitgatgatg ptgatgptga tgitgatgat 121 gatgatgvtg vtgatgitga tgitgvtgpt gatgitgatg ptgatgitga tgptgvtgts 181 itatyafann tsgtaisvll ggtnvplpnn qnigpgitvs ggntvftvan agnyyisyti 241 nitasllvss ritingapla gtinspavat tsfsatiitt laagsaislq lfgllavatl 301 stttpgavlt iirls In one embodiment, a Bacillus anthracis str. Sterne protein having a collagen triple helix repeat domain comprises an amino acid sequence of SEQ ID NO: 52 (Accession No. YP_030869):

1 mvkvvegngg kskiksplns nfkilsdlvg ptfppvptgm tgitgstgat gntgptgetg 61 atgsagitgs tgptgntggt gstgptgntg atgstgvtgs tgvtgstgvt gstgvtgstg 121 ptgetggtgs tgvtgstgat gstgvtgntg ptgstgatgn tgsigetggt gsmgptgetg 181 vtgstggtgs tgvtgntgpt gstgvtgstg vtgstgptgs tgvtgstgpt gstgvtgstg 241 vtgnmgptgs tgvtgntgpt gttgatgetg pmgstgatgt tgptgetget getggtgstg 301 ptgntgatgs tgvtgstgvt gstgvtgetg ptgstgatgn tgptgetggt gstgatgstg 361 vtgntgptgs tgvtgntgat getgptgntg atgntgptge tgvtgstgpt getgvtgstg 421 ptgntgatge tgatgstgvt gntgstgetg ptgstgptgs tgatgvtgnt gptgstgatg 481 atgstgptgs tgttgntgvt gdtgptgatg vsttatyafa iultsgsvisv llggtniplp 541 nngnigpgit vsggntvftv anagnyyiay tinltaglly ssritvngsp lagtinsptv 601 atgsfsatii aslpagaavs Iglfgvvala tlstatpgat Itiirls In one embodiment, a Bacillus anthracis str. Sterne protein (BAS2280) comprises an amino acid sequence of SEQ ID NO: 53 (Accession No. YP_028541):

1 mkkqknllsl aleiilpyrl tgptgitgpt gitgptgitg ptgftgpnit tnsmfanntl 61 ggpisvilgg tniplsnnqs lgnftvnasn diftipvtgr yyltyqvntt tallagtrll 121 lnsstplsgs ifspaistsn ynmiliinli agntislglf gvlsivnlvg ggstgaslti 181 irid 3. Modified Proteins In one embodiment, a modified Bacillus anthracis str CEB protein comprises an amino acid sequence of SEQ ID NO: 54, wherein the modification comprises a truncated leader sequence and the addition of a His-6 Tag:

MAFDPNLVGPTLPPIPPFTLPTGPTGPTGPTGPTGPTGPTGPTGDTGTTGPT
GPTGPTGPTGPTGATGLTGPTGPTGPSGLGLPAGLYAFNSGGISLDLGINDP
VPFNTVGSQFGTAIS QLDADTFVISETGFYKITVIANTATAS V LGGLTIQV N
GVPVPGTGSSLISLGAPIVIQAITQITTTPSLVEVIVTGLGLSLALGTSASIIIE
KVA

4. ExsA Proteins In one embodiment, the present invention contemplates a composition comprising a Bacillus spore surface protein, wherein the protein comprises an ExsA
protein. The ExsA protein is localized in both the exosporium and bacterial coat and may be related to the SafA (SpoVID-associated factor) protein of B. subtilis. SafA interacts with SpoVID
during the early stages of coat assembly. Ozin et al., "SpoVID guides SafA to the spore surface in Bacillus subtilis" J. Bacteriol. 183:3041-3049 (2001). Both SpoIVA
and SpoVID are believed for SafA localization, and homologues of both are encoded in the B.
cereus genome. By analogy, one would predict that they might also assist in the localization of ExsA. ExsA might play a larger role in B. cereus than SafA
does in B..
subtilis, in that a less extreme coat assembly defect has been reported for B.
subtilis safA
detection mutants. Like the exsA mutants, safA mutant spores were sensitive to lysozyme, but the exsA spore structure appeared rather less disturbed.
The ExsA protein is very different in amino acid sequence from SafA, apart from N-terminal and C-terminal regions. Given the large differences in sequence between SafA and ExsA, local regions of conservation are likely to be significant for functions conserved in both SafA and ExsA, and could be explored by directed mutagenesis in B.
subtilis. The multiple proline-rich tandem repeat sequences in ExsA are likely to be in an extended conformation, but their function is not known- they either could be repetitive regions for binding of other coat proteins or could act as spacers between regions that bind other coat proteins. Speculatively, this could be a mechanism to provide some distance between integument layers, but it would not be obvious why there should be three such regions, each with its own sequence repeat. The number of repeats in genome sequences of different species and strains in the B. cereus/B. anthracis/B.
thuringiensis cluster vary slightly, but the sequences are otherwise almost identical. In both the Bacillus halodurans and Oceanobacillus iheyensis genome sequences, encoded SafA
homologues may also contain multiple short repeats, whose sequence is not conserved between species, but they are not as extensive as in B. cereus, and again, their significance is unknown. Bailey-Smith et al., "The ExsA protein of Bacillus cereus is required for assembly of coat and exosporium onto the spore surface" J.
Bacteriol.
187:3800-3806 (2005).
5. ExsF Proteins In one embodiment, the present invention contemplates a composition comprising a Bacillus spore surface protein, wherein said protein comprises an ExsF
protein. ExsF
proteins (i.e., for example, ExsF, ExsFA, and ExsFB) are exosporium components believed involved in the proper localization of BeIA filaments on a Bacillus spore surface and for the stability of the crystalline layers of the exosporium. These proteins arc probably organized in multimeric complexes with other exosporium components, including BelA. The protein ExsFA may interact with BcIA and ensures that it is localized on the spore surface. The contribution of ExsF is less clear, but one hypothesis is that ExsFB and ExsFA interact with the components of both arrays of the crystalline basal layer and ensure its stability. Alternatively, the absence of two spore components (35 to 37 kDa) in mutant ExsF knock-out strains suggest that ExsFs proteins may be involved in the assembly of exosporium structural components or with maintenance on the spore. Sylvestre et al., "Contribution of ExsFA and ExsFB proteins to the localization of BelA on the spore surface and to the stability of the Bacillus anthracis exosporium" J
Bacteriol 187:5122-5128 (2005). Such a role would then resemble that described for some morphogenetic coat proteins, such as SpoIVA and SpoVID which are involved in B.
subtilis coat assembly. Driks A., "Maximum shields: the assembly and function of the bacterial spore surface" Trends Microbiol. 10:251-254 (2002).
6. BxpB Proteins In one embodiment, the present invention contemplates a composition comprising a Bacillus spore surface protein, wherein said protein comprises a BxpB
protein. BxpB
was discovered as one of several proteins present in a purified sample of exosporium that had been isolated from the Sterne strain of B. anthracis. Steichen et al., "Identification of the immunodominant protein and other proteins of the Bacillus anthracis exosporium" J.

Bacteriol. 185:1903-1910 (2003). Specifically, BxpB was found in high levels in samples of chemically deglycosylated exosporium. This result suggested that BxpB was either glycosylated or associated with glycosylated material. However, BxpB is not a glycoprotein but forms a high-molecular-mass complex with a BelA glycoprotein and other proteins including, but not limited to, ExsY or CotY both of which are found in Bacillus exosporium. An anti-BxpB MAb studies suggests that BxpB may be an integral basal layer protein. For example, binding of the anti-BxpB MAb to Sterne spores was weaker than that detected with Obcla spores that lack BclA. The reduced MAb binding to Slerne spores was due primarily to the protein component of BelA, perhaps reflecting a close BxpB-Bc1A interaction. Spores that lack BxpB (i.e., for example, AbxpB
mutants) are enclosed by an exosporium that are devoid of hair-like nap despite the presence of a normal Bc1A level. However, very little of this Bc1A was associated with AbxpB
spores (i.e., for example, - 2% of the level found on Sterne spores). Steichen et al., "Characterization of the exosporium basal layer protein BxpB of Bacillus anthracis" J
Bacteriol 187:5868-5876 (2005).
Therefore, it appears that BxpB may mediate attachment of BcIA to the basal spore layer and assembly of the hair-like nap. Recent studies indicate that this attachment occurs through the 38-residue amino-terminal domain (NTD) of Bc1A, with the rest of the spore surface BelA protein extending away from the spore. Boydston et al., "Orientation within the exosporium and structural stability of the collagen like glycoprotein Bc1A of Bacillus anthracis" J. Racteriol. 187:5310-5317 (2005). Apparently, the first 19 amino acids of this NTD are absent in mature BelA isolated from spores, most likely due to a proteolytic processing event. Steichen et al., "Identification of the immunodominant protein and other proteins of the Bacillus anthracis exosporium" J. Bacteriol.
185:1903-1910 (2003); and Sylvestre et al., "A collagen-like surface glycoprotein is a structural component of the Bacillus anthracis exosporium" Mol. Microbiol. 45:169-178 (2002).
Although it is not necessary to understand the mechanism of an invention, it is believed that that this processing event may be linked to the attachment of BelA to the basal layer of the exosporium, perhaps through a covalent linkage directly to BxpB.
In one embodiment, a BxpB exosporium protein comprises an amino acid sequence of SEQ ID NO: 55:

mfssdceftkidceakpastlpafgfafnasapgfaslftplllpsvspnpnitvpvind tvsvgdgirilragiygisytltisldnspvapeagrfflslgtpaniipgsgtavrsnv igtgevdvssgvilinlnpgdlirivpveligtvdiraaaltvagi s 7. ExsJ Proteins In one embodiment, an ExsJ exosporium protein (Bacillus cereus) comprises an amino acid sequence of SEQ ID NO: 56 (Accession No. AY 183116.1):

M KHNDCFDI INNCNPIVFSADCCKNPQS VPITREQLSQLITLLNSLVSAISAFF
ANPSNANRLVLLDLFNQFLIFLNSLLPSPEVNFI,KQLTQSTIVLLQSPAPNLG
QLSTLLQQFYSALAQFFFALDLIPISCNSNVDSATLQLLFNLLIQLINATPGAT
GPTGPTGPTGPTGPAGTGAGPTGATGATGATGPTGATGPAGTGGATGATG
ATGVTGATGATGATGPTGPTGATGPTGATGATGATGPTGATGPTGATGLT
GATGAAGGGAIIPFAS GTTPSALVNALVANTGTLLGFGFSQPGVALTGGTSI
TLALGVGDYAFVAPRAGTITSLAGFFSATAALAPISPVQVQIQILTAPAASNT
FTVQGAPLLLTPAFAAIAIGSTASGIIAEAIP VAAGDKILLYV SLTAASPIAAV
AGFVSAGINIV

encoded by the nucleic acid sequence SEQ ID NO: 57:

1 gaaaacactc aactataaga aaagtaagga ggaaataaat gaaacataat gattgttttg 61 atcataataa ctgcaatccg attgtttttt cagcagattg ttgtaaaaat ccacagtcag 121 ttcctattac tagggaacaa ttaagtcaat taattacttt actaaactca ttagtatcag 181 ctatttcagc attttttgca aatccaagta atgcaaacag attagtgtta ctcgatttat 241 ttaatcaatt tttaattttc ttaaattcct tattaccttc cccagaagtt aattttttga 301 aacaattaac tcaaagtatt atagttttat tacaatctec agcacctaat ttaggacaat 361 tgtcaacatt attgcaacaa ttttatagcg cccttgcaca attcttcttc gctttagatc 421 ttatccctat atcctgcaae tcaaatgttg attctgcaac tttacaactt ctttttaatt 481 tattaattca attaatcaat gctactccag gggcgacagg tccaacaggt ccaacaggtc 541 caacaggtcc aacgggccca gcaggaaccg gagcaggtcc aacgggagca acgggagcaa 601 caggagcaac aggcccaaca ggagcgacag gtccagcagg tactggtgga gcaacaggag 661 caacaggagc aacaggagta acaggagcaa caggagcaac aggcgcaaca ggtecaacag 721 gtccaacagg ggcaacaggt ccaacagggg caacaggagc aacaggagca acaggtccaa 781 caggagcaac aggtccaaca ggggcaacgg gettaacagg agcaactggt gcagctggtg 841 gcggagctat tattccattt gcttcaggta caacaccatc tgcgttagtt aacgcgttag 901 tagctaatac aggaactett ettggatttg gatttagtca gcctggtgta gcattaacag 961 gtggaactag tatcacatta gcattaggtg taggtgatta tgcatttgta gcaccacgtg 1021 caggaactat cacgtcatta gcaggtttct ttagtgcaac agctgcatta gctccaatat 1081 cacctgttca agtgcaaata caaatattaa ctgcacctgc agcaagcaat acgtttacag 1141 tacaaggcgc acctctttta ttaacaccag catttgccgc aatagcgatt ggttctacag 1201 catcaggtat catagctgaa gctattccag tagctgctgg agataaaata ctactgtatg 1261 tttcattaac agcagcaagt ccaatagctg cagttgctgg atttgtaagt gcaggtatta 1321 atatcgttta atttgttaca atgtttagta ggaaattaaa caggggtttg tatatagtcc 1381 ctcctaaata aatttaggaa ggactatatt t 8. ExsH Proteins In one embodiment, an ExsH exosporium protein (Bacillus cereus) comprises an amino acid sequence of SEQ ID NO: 58:

MKHNDCFDHNNCNPIVFSADCCKNPQSVPITREQLSQLITLLNSLVSAISAFF
ANPSNANRLVLLDLFNQFLIFLNSLLPSPEVNFLKQLTQSIIVLLQSPAPNLG
QLSTLLQQFYSALAQFFFALDLIPISCNSNVDSATLQLLFNLLIQLINATPGAT
GPTGPTGPTGPTGPAGTGAGPTGATGA'1'GATGPTGATGPAGTGGATGATG
ATGVTGATGATGATGPTGPTGATGPTGATGATGATGPTGATGPTGATGLT
GATGAAGGGAIIPFASGTTPSALVNALVANTGTLLGFGFSQPGVALTGGTSI
TLALGVGDYAFVAPRAGTITSLAGFFSATAALAPISPVQVQIQILTAPAASNT
FTVQGAPLLLTPAFAAIAIGSTASGIIAEAIPVAAGDKILLYVSLTAASPIAAV
AGFVSAGINIV

encoded by a nucleic acid sequence comprising SEQ ID NO: 59:

1 gtttcctatt ttaagtcaat atataacata tttttttaca actttatgat ccttataata 61 ggaaaaactc aaatataaaa caagtaagga ggacataaat gacaaacaat aattgttttg 121 gtcataacca ctgcaataat ccgattgttt tcactccaga ttgctgtaac aatccacaaa 181 cagttccaat tactagtgag caattaggta gattaattac tttactaaac tctttaatag 241 cggctattgc agcgtttttt gcaaatccaa gtgatgcaaa cagattagct ttactcaatt 301 tgtttactca actattgaac ttactaaatg aattagcacc ttccccagaa gggaatttet 361 taaaacaatt aattcaaagt attattaatt tactacaatc tcctaaccca aatctaagtc 421 aattactttc tttattacaa caattctaca gtgctcttgc accattettc ttctctttaa 481 ttattgaccc tgcaagttta caacttttat taaacttatt aactcaatta attggtgcta 541 ctccaggagg cggagcaaca ggtccaacag gtccaacaggtccaggaggc ggagcaacag 601 gtccaacagg tccaacaggt ccaggaggcg gagcgacaggtccaacaggc ccaacaggag 661 cgacaggtcc agcaggtact ggtggagcaa caggtttaac aggagcaaca ggtttaacag 721 gagcaacagg cttaacagga gcgacaggcc caacgggagc aacaggttta acaggagcaa 781 caggtttaac aggagcaaca ggcttaacag gagcgacagg tccaacagga gcaacaggtc 841 caacaggagc aacaggttta acaggagcaa ctggtgcaac tggtggegga gctattattc 901 catttgcttc aggtacaaca ccatctgcgt tagttaacgc gttaatagct aatacaggaa 961 ctcttcttgg atttggattt agtcagcctg gtgtagcttt aactggtgga acaagtatca 1021 cattagcatt aggtgtaggt gattatgcat ttgtagcacc acgcgcaggg gttattacgt 1081 cattagctgg tttctttagt gcaacagctg cattagctcc attatcacct gttcaagtgc 1141 aaatacaaat attaactgca cctgcagcaa gcaatacgtt tacagtacaa ggcgcacctc 1201 ttttattaac accagcattt gccgcaatag cgattggttc tacagcatca ggaatcatac 1261 ctgaagctat tccagtagca gctggggata aaatactgtt atatgtttca ttaacagcag 1321 caagtccaat agctgcagtt gctggatttg taagtgcagg tattaatatc gtttaatttg 1381 ttacaatgtt to 9. CotB Proteins In one embodiment, the present invention contemplates a CotB exosporium protein (Bacillus subtilis; Accession No. AY 186996) comprising an amino acid sequence of SEQ ID NO: 60:

MSKRRMKYHSNNEISYYNFLHSM
encoded by the nucleic acid sequence of SEQ ID NO: 61:

I gatcatttaa gtattttatg aatgcgtgaa aatgggtatt cgeggaaaaa gcgacaatta 61 ggctattgaa ttagttcaac aaataaatgt gacacgtata tatgcagtat gtttatcatc 121 tatgtataag tgactaggag gaatttgaat gagcaagagg agaatgaaat atcattcaaa 181 taatgaaata tcgtattata actttttgca ctcaatg.
10. CotY Proteins In one embodiment, the present invention contemplates a CotY exosporiumn protein (Bacillus cereus) comprising an amino acid sequence of SEQ ID NO: 62:
MSCNCNEDHQHECDFNCVSNVVRFIHELQECATTTCGSGCEVPFLGAHN
NASVANTRPFILYTKTGEPFEAFAPSSSLTSCRSPIFRVESIDDDDCAVLRVL
TVVLGDGTAVPPGDDPICTFLAVPNARLISTSTCITVDLSCFCAIQCI.RDVSI
encoded by the nucleic acid sequence of SEQ ID NO: 63:

1 caatttttca gaggaaagaa attagatata tctcgttttt tatatactgt gcgaaaagat 61 ttacctgaaa agacatccac taaataagga tgtctttttt tatatcacgc tatgtacatc 121 cctcctatat aaattccttg tttttatcgt gagaattaac gtaatatcag ccatatecca 181 ctcatattgt aatagtgtat gtcagaactc acgagaagga gtgaacataa tgagctgcaa 241 ttgtaatgaa gatcatcaac atgagtgtga tttcaactgt gtatcgaatg tcgttcgttt 301 tatacatgaa ctacaagaat gtgcaacgac aacatgtgga tctggttgcg aagttccatt 361 tttaggtgca cataacaatg catcagtagc gaatacacgt ccttttattt tatacacaaa 421 aaccggagaa cctttcgaag catttgcacc atcttcaagc cttactagtt gccgatctcc 481 cattttccgc gtagaaagta tagatgatga cgactgtgct gtattgcgtg tattaactgt 541 agttttaggt gatggtaccg ctgtaccacc tggtgacgac ccaatctgta cgtttttagc 601 tgtaccaaat gcaagactaa tatcaacctc cacttgcatt actgttgatt taagttgctt 661 ctgcgctatt caatgettac gtgatgtttc tatttaa
11. ExsY Proteins In one embodiment, an ExsY exosporium protein (Bacillus cereus) comprises an amino acid sequence of SEQ ID NO: 64 (Accession No. AY121973):

MSCNENKHHGS SHCV VDVVKFINELQDCSTTTCGSGCEIPFLGAHNTASV
ANTRPFILYTKTGEPFEAFAPSASLTSCRSPIFRVESVDDDSCAVLRVLTVV
LGDSSPVPPGDDPICTFLAVPNARLISTTTCITVDLSCFCAIQCLRD VSIVK

S encoded by a nucleic acid sequence SEQ 1D NO: 65:

1 aaatagtatg agagatatta ggttgtetat agatgatatc caatcgatta gtatggccgg 61 tttttaaata tgtaaaggtg gtagaaagct tctaccaccc caatagtttt ttagtttact 121 ttacgataga aacgtcgcgt aagcattgaa tcgcacagaa acagettaaa tcaacagtaa 181 tgcaagtagt tgtagatatt aatcttgcat ttggtacagc taaaaacgta caaattggat 241 cgtcaccagg tggtactgga gaactgtcac ctaatactac agttaataca cgtagcacag 301 cacagctatc atcatctaca ctttccacac ggaaaattgg agateggcag ctagtaaggc 361 ttgctgatgg tgcgaatggt tcaaaaggtt ctccagtttt tgtgtataaa ataaaagggc 421 gtgtatttgc tactgatgca gtattgtgtg cacctaaaaa tgggatttca caaccagatc 481 cacatgttgt tgtagaacaa tcttgtaatt cattgatgaa ttttacaacg tcaactacac 541 aatgagaaga gccatggtgt ttattttcgt tacaactcat taatgtcact cettatcttc 601 ttgtttgtat ttacattaat aagatattgg agtcgaggag atttggtcac aatetcaaga 661 cctttttttt taaataggcg aaagaggata agggaaggtg aaattatget gtttacaagc 721 tggcttttat tttttatttt cgcgttagca gcttttaggc tcactcgttt aattgtatat 781 gataaaataa cagccttttt gcgaagacca tttattgatg as In one embodiment, a Clostridium perfringes spore surface homolog protein comprises a triple helix protein comprising an amino acid sequence of SEQ ID
NO: 66 (Accession No. YP_695648):
MIRKIYNPNRYYDDYNRYNCYDRYNCYDDEYCQDDYYCKEDCYCKDDC
YLEINCNCCDCCKPGPRGPRGPQGPRGPQGPRGPMGCQGERGPIGPMGPM
GPIGPQGPQGDQGLTGPQGPAGPQGEQGPQGDQGPVGPIGPQGPQGEQGL
TGPQGPAGSQGPEGPTGPQGATGPQGPEGPTGAQGDQGPVGPQGAQGPQ
GPQGPQGATGPTGPQGPQGNQGPAGPQGPVGPQGPQGEPGVDFDDTLLV
SYS SLTSQN VNANGIFTYNIQN PNGSTFI'AITANIANGTFTINEPGRYLFMW
SFNLDNTNNTTASAIVSLFRNGSRVFLSGTPRVAPGEIGVVNGSIAVNANA
GDVFALVNNSTRNVLSQIISSPISVTPAILGESTGINSGIGSW VQIVRVSD

II. Antibody Inhibition Of Microbial Spore Binding In one embodiment, the present invention contemplates inhibiting microbial spore binding and/or attachment to inanimate and/or animate surfaces using antibodies. In one embodiment, the antibodies inhibit Clostridium spore binding. These antibodies include, but are not limited to, polyclonal and monoclonal antibodies. In addition, chimeric antibodies may be produced, for example, by splicing mouse antibody genes to human antibody genes. Single chain antibodies are also contemplated as well as antibody fragments generated by pepsin digestion of the antibody molecule or by reduction of the disulfide bridges of the Fab fragments.
A. Antibody Generation Methods The present invention provides isolated antibodies or fragments thereof.
(i.e., for example, polyclonal, monoclonal, and/or Fab's). In one embodiment, the present invention provides monoclonal antibodies that specifically inhibit the binding of microbial spore surface proteins to various surfaces (i.e., for example, animate surfaces and/or inanimate surfaces).
An antibody against a protein of the present invention may be any monoclonal or polyclonal antibody, as long as it can recognize the protein. Antibodies can be produced by using a protein of the present invention as the antigen according to a conventional antibody or antiserum preparation process. Any suitable method may be used to generate the antibodies used in the methods and compositions of the present invention, including but not limited to, those disclosed herein. For example, for preparation of a monoclonal antibody, protein, as such, or together with a suitable carrier or diluent is administered to an animal (e.g., a mammal) under conditions that permit the production of antibodies (i.e., for example, immunization). Normally, the protein is administered once every 2 weeks to 6 weeks, in total, about 2 times to about 10 times. Animals suitable for use in such methods include, but are not limited to, primates, rabbits, dogs, guinea pigs, mice, rats, sheep, goats, etc. and may be immunized by injection with a full-length protein or any portion, fragment or oligopeptide which retains immunogenic properties.
Depending on the host species, various adjuvants may be used to increase immunological response. For enhancing the antibody production capability, complete or incomplete Freund's adjuvant may be administered. Such adjuvants include, but are not limited to, Freund's, mineral gels such as aluminum hydroxide, and surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanin, and dinitrophenol. BCG (Bacillus Calmette-Guefin) and Comebactenum parrum are potentially useful adjuvants.
Monoclonal antibodies may be prepared using any technique which provides for the production of antibody molecules by continuous cell lines in culture.
These include, but are not limited to hybridoma techniques. Koehler et al. Nature 256:495-497(1975);
Kosbor et al., Immunol Today 4:72 (1983); Cote et al., Proc Natl Acad Sci 80:2026.-2030 (1983); Cole et al., In: Monoclonal Antibodies and Cancer Therapy, Alan R Liss Inc, New York, N.Y., pp 77-96 (1985)(all references incorporated herein by reference).

For preparing monoclonal antibody-producing cells, an individual animal (e.g., a mouse) whose antibody titer has been confirmed is selected, and 2 days to 5 days after the final immunization, its spleen or lymph node is harvested and antibody-producing cells contained therein are fused with myeloma cells to prepare the desired monoclonal antibody producer hybridoma. Measurement of the antibody titer in antiserum can be carried out, for example, by reacting the labeled protein, as described hereinafter and antiserum and then measuring the activity of the labeling agent bound to the antibody.
The cell fusion can be carried out according to known methods, for example, the method described by Koehler and Milstein (Nature 256:495 [1975]). As a fusion promoter, for example, polyethylene glycol (PEG) or Sendai virus (HVJ), preferably PEG is used.
Examples of myeloma cells include NS-1, P3U 1, SP2/0, AP-1 and the like. The proportion of the number of antibody producer cells (i.e., for example, spleen cells) and the number of myeloma cells to be used is preferably about 1:1 to about 20:1.
PEG
(preferably PEG 1000-PEG 6000) is preferably added in concentration of about 10% to about 80%. Cell fusion can be carried out efficiently by incubating a mixture of both cells at about 20 C to about 40 C, preferably about 30 C to about 37 C for about 1 minute to 10 minutes.
Various methods may be used for screening for a hybridoma producing the antibody (e.g., against a tumor antigen or autoantibody of the present invention). For example, where a supernatant of the hybridoma is added to a solid phase (e.g., microplate) to which antibody is adsorbed directly or together with a carrier and then an anti-immunoglobulin antibody (if mouse cells are used in cell fusion, anti-mouse immunoglobulin antibody is used) or Protein A labeled with a radioactive substance or an enzyme is added to detect the monoclonal antibody against the protein bound to the solid phase. Alternately, a supernatant of the hybridoma is added to a solid phase to which an anti-immunoglobulin antibody or Protein A is adsorbed and then the protein labeled with a radioactive substance or an enzyme is added to detect the monoclonal antibody against the protein bound to the solid phase.
Selection of the monoclonal antibody can be carried out according to any known method or its modification. Normally, a medium for animal cells to which HAT
(hypoxanthine, aminopterin, thymidine) are added is employed. Any selection and growth medium can be employed as long as the hybridoma can grow. For example, RPMI

medium containing I% to 20%, preferably 10% to 20% fetal bovine serum, GIT
medium containing 1% to 10% fetal bovine serum, a serum free medium for cultivation of a hybridoma (SFM-101, Nissui Seiyaku) and the like can be used. Normally, the cultivation is carried out at 20 C to 40 C., preferably 37 C for about 5 days to 3 weeks, preferably I week to 2 weeks under about 5% CO2 gas. The antibody titer of the supernatant of a hybridoma culture can be measured according to the same manner as described above with respect to the antibody titer of the anti-protein in the antiserum.
Separation and purification of a monoclonal antibody can he carried out according to the same manner as those of conventional polyclonal antibodies such as separation and purification of immunoglobulins, for example, salting-out, alcoholic precipitation, isoelectric point precipitation, electrophoresis, adsorption and desorption with ion exchangers (e.g., DEAE), ultracentrifugation, gel filtration, or a specific purification method wherein only an antibody is collected with an active adsorbent such as an antigen-binding solid phase, Protein A or Protein G and dissociating the binding to obtain the antibody.
Polyclonal antibodies may be prepared by any known method or modifications of these methods including obtaining antibodies from patients. For example, a complex of an immunogen (an antigen against the protein) and a carrier protein is prepared and an animal is immunized by the complex according to the same manner as that described with respect to the above monoclonal antibody preparation. A material containing the antibody against is recovered from the immunized animal and the antibody is separated and purified.
As to the complex of the immunogen and the carrier protein to be used for immunization of an animal, any carrier protein and any mixing proportion of the carrier and a hapten can be employed as long as an antibody against the hapten, which is crosslinked on the carrier and used for immunization, is produced efficiently.
For example, bovine serum albumin, bovine cycloglobulin, keyhole limpet hemocyanin, etc.
may be coupled to an hapten in a weight ratio of about 0.1 part to about 20 parts, preferably, about 1 part to about 5 parts per 1 part of the hapten.
In addition, various condensing agents can be used for coupling of a hapten and a carrier. For example, glutaraldehyde, carhodiimide, maleirnide activated ester, activated ester reagents containing thiol group or dithiopyridyl group, and the like find use with the present invention, The condensation product as such or together with a suitable carrier or diluent is administered to a site of an animal that permits the antibody production. For enhancing the antibody production capability, complete or incomplete Freund's adjuvant may be administered. Normally, the protein is administered once every 2 weeks to 6 weeks, in total, about 3 times to about 10 times.
The polyclonal antibody is recovered from blood, ascites and the like, of an animal immunized by the above method. The antibody titer in the antiserum can be measured according to the same manner as that described above with respect to the supernatant of the hybridoma culture. Separation and purification of the antibody can be carried out according to the same separation and purification method of immunoglobulin as that described with respect to the above monoclonal antibody.
The protein used herein as the immunogen is not limited to any particular type of immunogen. For example, a protein expressed resulting from a virus infection (further including a gene having a nucleotide sequence partly altered) can be used as the immunogen. Further, fragments of the protein may he used. Fragments may be obtained by any methods including, but not limited to expressing a fragment of the gene, enzymatic processing of the protein, chemical synthesis, and the like.
For the production of antibodies in the context of the present invention, any antigenic portion of a microbial spore protein amino acid sequence contemplated herein (i.e., for example, SEQ ID NOs:1, 3, 5, 7, or 9) can he used either alone or fused with amino acids of another protein (i.e., for example, glutathione to produce an antibody against a chimeric molecule). Such antibodies include, but are not limited to, polyclonal, monoclonal, chimeric, single chain, Fab fragments and fragments produced by a Fab expression library. Neutralizing antibodies, i.e., those which inhibit dimer formation, are especially preferred for diagnostics and therapeutics.
Polypeptides to be used for antibody induction need not retain biological activity;
however, the protein fragment, or oligopeptide should be antigenic. Peptides used to induce specific antibodies may have an amino acid sequence comprising at least five amino acids, preferably at least 10 amino acids. Preferably, they should mimic a portion of the amino acid sequence of the natural protein and may contain the entire amino acid sequence of a small, naturally occurring molecule. Short stretches of an amino acid sequence may be fused with those of another protein including, but not limited to, keyhole limpet hemocyanin and antibody produced against a chimeric molecule.
Antibodies can also be used to detect polypeptides in samples including, but not limited to, body fluids, cell extracts, tissue extracts, or surface swipes using techniques including, but not limited to, enzyme linked immunosorbent assay (ELISA), radioimmunoassay (RIA) and fluorescent activated cell sorting (FACS). Reporter molecules can be joined to the polypeptides and antibodies to facilitate detection of polypeptide-antibody binding.
B. Methods For Antibody Inhibition Of Microbial Spore Surface Protein Binding In one embodiment, the present invention contemplates a screening method comprising a monoclonal and/or polyclonal antibody having affinity to a Clostridium spore surface protein.
It has been reported that mice immunized with a 1:1 mixture of B. cereus T
spores and Clostridium sporogenes PA3679 spores resulted in the generation of 397 hybridomas that were screened for their reactivities with 107 B. cereus spores and for their reactivities with 107 C. sporogenes spores. Apparently, nine hybridomas produced antibodies specific for B. cereus T spores while fifteen hybridomas produced antibodies specific for C. sporogenes spores. Seven hybridomas produced antibodies which cross-reacted with B. cereus T and C. sporogenes spores. Quinlan et al., "Monoclonal antibodies for use in detection of Bacillus and Clostridium spores" Appl Environ Microbial. 63:482-(1997). Two of the hybridomas producing antibodies specific for C. sporogenes spores were subcloned and the resulting IgM antibodies were designated C33 and C225.
The antibodies were examined qualitatively (visually) for their reactivities with a range of Bacillus, Clostridium, and Desulfotomaculum spores by using a dot blot format.
Antibody C33 reacted strongly with C. sporogenes spores and weakly with C.
perfringens spores. Antibody C225 reacted weakly with B. megaterium spores and strongly with spores of B. stearothermophilus, C. perfringens, and C. sporogenes.
Immunocytochemical localization of the antigen in C. sporogenes indicated that it is present on the exosporium and the outer cortex region of the spore.
While less information is available about C. sporogens spore antigens, it has been demonstrated that specific spore antigens are distinct from vegetative antigens. Norris J., "Bacterial spore antigens: a review" J. Gen. Microbiol. 28:393-408 (1962); and Princewell, T., "Spore antigens of Clostridium sporogenes" J. Med. Microbial.
12:29-41 (1979). Although it is not necessary to understand the mechanism of an invention, it is believed that polyclonal antibodies raised to Clostridium spore antigens are common to more than one specific of Clostridium. Foegeding et al., "Polyclonal antibodies in an enzyme-linked immunosorbent assay (ELISA) to detect bacterial spores" J. Rapid Methods Automat. Microbial. 2:135-150 (1993).

In one embodiment, the present invention contemplates a method whereby a monoclonal and/or a polyclonal antibody has affinity to a Clostridium spore surface protein (i.e., for example, CD 1581) is used to disrupt the binding of a Clostridium spore to a surface. In one embodiment, the surface may be selected from the group including, but not limited to, plastic, ceramic, stainless steel, iron steel, vinyl, tile, wood laminate, skin (i.e., for example, pigskin), internal body tissues, clothing, or environmental protection suits.
C. Antibody Inhibition Of Microbial Spore Binding A modified cup scrub method was used to evaluate the ability of C. difficile spores to bind to a human skin surrogate (i.e., for example, pigskin). See, Example X. The pigskin was first washed using a non-antimicrobial soap for 30 s followed by water rinse for 30 s. After washing the pigskin, up to 50 l of spores were added to a 20 mm diameter area on the surface of the skin. The spores were allowed to dry for up to an hour and then removed according to the cup scrub method. This experiment determined the ability of C. difficile spores to bind to the skin surrogate. Preliminary data has shown that up to 80% of spores can be removed from the surface of the pigskin. In subsequent experiments, spores were first reacted with C. difficile spore surface antibodies prior to being applied to the skin surface. The ability to remove the spores from the skin was again determined using the cup scrub method. Preliminary data has shown the ability to remove greater than 80% of the spores from the pigskin, suggesting the antibody is preventing the spores from binding to the skin.

III. Microbial Spore Surface Protein Inhibition Of Microbial Spore Binding In one embodiment, the present invention contemplates a method comprising inhibiting microbial spore binding using an infection control agent, wherein the agent comprises at least a portion of a microbial spore protein. Although it is not necessary to understand the mechanism of an invention, it is believed that the infection control agent competes for surface binding sites to which the microbial spore has similar affinity. It is further believed that the resultant competitive binding either displaces previously bound microbial spores from the surface attachment site and/or is sterically prevented from binding due to the presence of the at least a portion of the microbial spore surface protein.
In one embodiment, the spore surface protein may be derived from microbial organisms selected from the group comprising bacteria, viruses, fungi, algae, or molds.
In one embodiment, the method further comprises preventing attachment of a microbial spore to a surface with the spore surface protein. In one embodiment, the spore surface proteins are formulated with non-spore surface protein compounds are selected from the group comprising small molecules, proteins, or nucleic acids. In one embodiment, the spore surface proteins are formulated with surface acting agents which may include, but are not limited to, surfactants, soaps, detergents, foaming agents, wetting agents, and aerosolizing agents. In one embodiment, the compounds are selected form the class of compounds known as solvents.
A. Full Length Spore Surface Proteins In one embodiment, at least a portion of a microbial spore surface protein comprises a full length microbial spore surface protein. In one embodiment, the full length microbial spore surface protein comprises a Clostridium spore surface protein. In one embodiment, the Clostridium full length spore surface protein comprises a C. diffacile full length spore surface protein. In one embodiment, the C. difficile full length spore surface proteins maybe selected from the group comprising SEQ ID NO: 1, SEQ ID
NO:3, SEQ ID NO:5, SEQ ID NO: 7 SEQ ID NO: 9 .
In one embodiment, the full length microbial spore surface protein comprises a Bacillus spore surface protein. In one embodiment, the Bacillus full length spore surface protein comprises a B. anthracis full length spore surface protein. In one embodiment, the B. anthracis full length spore surface proteins may be selected from the group comprising SEQ ID NO:41, SEQ ID NO: 43 SEQ ID NO: 45 SEQ ID NO: 47 SEQ ID
NO:49 .
B. Truncated Spore Surface Proteins In one embodiment, the at least a portion of a microbial spore surface protein comprises a truncated microbial spore surface protein. Although it is not necessary to understand the mechanism of an invention, it is believed that such truncated microbial spore surface proteins may be constructed by recombinant protein expression platforms (infra) wherein a portion of a nucleic acid sequence encoding a microbial spore surface protein is integrated within a vector and placed in operable combination with a compatible promoter. Alternatively, such truncated microbial surface proteins may be created by digesting full length microbial spore surface proteins using any one, or more, of peptidase enzymes previously reported. Such peptidase enzymes may comprise enzymes including but not limited to endopeptidases, exopeptidases, amidopeptidases, and/or carboxypeptidases.

In one embodiment, the truncated microbial spore surface protein is derived from a Clostridium spore surface protein. In one embodiment, the Clostridium truncated spore surface protein comprises a C. difficile truncated spore surface protein. In one embodiment, the C. difficile truncated spore surface protein may be derived from a Clostridium hypothetical protein (CD 1581) as listed in Table III:

Table III. Clostridium difficile Spore Surface Protein Truncated Peptide Fragments Peptide Fragment Sequence Identification Number MENKKCYSED SEQ ID NO: 67 WYERGESTAK SEQ ID NO: 68 WFQNDREEYE SEQ ID NO: 69 REAYDEDRER SEQ ID NO: 70 RGSNCGCSDS SEQ ID NO: 71 GENRPRNCER SEQ ID NO: 72 FRREAEIRER SEQ ID NO: 73 EAREAFCESS SEQ ID NO: 74 EKKKEALAYE SEQ ID NO: 75 CEARKLWEEA SEQ ID NO: 76 EKYWDEYSKY SEQ ID NO: 77 NYKGIEYLAE SEQ ID NO: 78 AARLFDEGME SEQ ID NO: 79 CEARRNGNNG SEQ ID NO: 80 GNNNNCCHKC SEQ TD NO: 81 HKCNCNCCRK SEQ ID NO: 82 In one embodiment, the C. difficile truncated spore surface protein may be derived from a Clostridium hypothetical protein (CD 1067) as listed in Table IV:

Table IV. Clostridium difficile Spore Surface Protein Truncated Peptide Fragments Peptide Fragment Sequence Identification Number MQDYKKNKRR SEQ ID NO: 83 MMNQPMSTMN SEQ ID NO: 84 EEEVYTDEIN SEQ ID NO: 85 SEDMRGFKKS SEQ ID NO: 86 HHHNGCNTDN SEQ ID NO: 87 KCECHDDCNP SEQ ID NO: 88 CNPCNPCKPN SEQ ID NO: 89 PCNPCKPNPC SEQ ID NO: 90 DDNCGCHDNC SEQ ID NO: 91 KCDCEPCEMD SEQ ID NO: 92 SDECFENKCG SEQ ID NO: 93 PECCNPISPR SEQ ID NO: 94 NFSVSNAVPF SEQ ID NO: 95 AIEANRIFDT SEQ ID NO: 96 MQFQTFTDAT SEQ ID NO: 97 GPNGEPLTFE SEQ ID NO: 98 TEVVEVFGSV SEQ ID NO: 99 PSAGQASVTI SEQ ID NO: 100 EKICLSNDGI SEQ ID NO: 101 VIDTGMTTLE SEQ ID NO: 102 DFDLDPLGDI SEQ ID NO: 103 VGRNCETTFE SEQ ID NO: 104 FAVCGERNSE SEQ ID NO: 105 CCRQGKGKSV SEQ ID NO: 106 AYKQRGLTVA SEQ ID NO: 107 VRNLVLELRG SEQ ID NO: 108 RCGCTEFVAL SEQ ID NO: 109 AFPAVRAGGG SEQ ID NO: 110 CKRRVDYVEF SEQ ID NO: 111 TFNTLSAPIC SEQ ID NO: 112 LPADGRAVTL SEQ ID NO: 113 RQEYQTNLTV SEQ ID NO: 114 DCIGKSILKL SEQ ID NO: 115 ECNECCEPFY SEQ ID NO: 116 ELIIPNDIDL SEQ ID NO: 117 VLCLQETVST SEQ ID NO: 118 LISEQIVVLA SEQ ID NO: 119 SPNPIQPRLV SEQ ID NO: 120 DTFSKVCDFS SEQ ID NO: 121 QCGPNHGSGK SEQ ID NO: 122 PSCHR SEQ ID NO: 123 In one embodiment, the C. difficile truncated spore surface protein may be derived from a Clostridium hypothetical protein (CD 3620) as listed in Table V:
Table V. Clostridium difficile Spore Surface Protein Truncated Peptide Fragments Peptide Fragment Sequence Identification Number MYDDYKYNKC SEQ ID NO: 124 NKCYNDYEEM SEQ ID NO: 125 THVHEYSESV SEQ ID NO: 126 KLAEECEDRH SEQ ID NO: 127 NHRAAGVTGE SEQ ID NO: 128 AIPINGGTNH SEQ ID NO: 129 VHKrNDNVDF SEQ ID NO: 130 LDHFHKICVT SEQ ID NO: 131 TGPA1RIPGT SEQ ID NO: 132 DKHIHLICGE SEQ ID NO: 133 TTVNDGHCHK SEQ ID NO: 134 FLFTTQIEAPLV SEQ ID NO: 135 In one embodiment, the C. difficile truncated spore surface protein may be derived from a Clostridium hypothetical protein (CD 1613) as listed in Table VI:

Table VI. Clostridium difficile Spore Surface Protein Truncated Peptide Fragments Peptide Fragment Sequence Identification Number MENNKCREDF SEQ ID NO: 136 RFTQEYEEDY SEQ ID NO: 13 )7 PNTNERYYEN SEQ ID NO: 138 YQVADRYYNY SEQ ID NO: 139 PNKYKEPKIK SEQ ID NO: 140 QCCCKKSMRE SEQ ID NO: 141 ALELLRYDAL SEQ ID NO: 142 RPFVNFNQFA SEQ ID NO: 143 FISDFFIVGA SEQ ID NO: 144 NLVGIDLSAP SEQ ID NO: 145 PKDNLSGLDG SEQ ID NO: 146 TFERFSACNC SEQ ID NO: 147 DLIDIAGRVS SEQ ID NO: 148 YPIPVPLTLE SEQ ID NO: 149 GLINTIGTIP SEQ ID NO: 150 GVAELIALID SEQ ID NO: 151 AVIPPTIDLG SEQ ID NO: 152 AILDAILAAI SEQ ID NO: 153 IDFILAASTP SEQ ID NO: 154 LANVDLASLC SEQ ID NO: 155 NLKAVAFDIT SEQ ID NO: 156 PADYEDFIAS SEQ ID NO: 157 LGYYLDKKHY SEQ ID NO: 158 KECNCNCDCD SEQ ID NO: 159 DCCCNKGILD SEQ ID NO: 160 NLYMSNINNQ SEQ ID NO: 161 VTVVAGSLVL SEQ ID NO: 162 TGVEVLGKKN SEQ ID NO: 163 DVIVLGNSND SEQ TD NO: 164 SRIYFVCVDS SEQ ID NO: 165 IDYIA SEQ ID NO: 166 In one embodiment, a Clostridium truncated spore surface protein may be derived from a hypothetical protein (CD 1711) as listed in Table VII:

Table VII. Clostridium diffacile Spore Surface Protein Truncated Peptide Fragments Peptide Fragment Sequence Identification Number MLIVTTEEKVE SEQ ID NO: 167 GKKISKVLGL SEQ ID NO: 168 VRGSTIRAKH SEQ ID NO: 169 VGKDIGASFK SEQ ID NO: 170 NLVGGELTGY SEQ ID NO: 171 NEMLTEARQI SEQ ID NO: 172 A.IGRMVEDAE SEQ ID NO: 173 AKGANAVIAF SEQ ID NO: 174 RLSSASVMQG SEQ ID NO: 175 AAEMLAYGTA SEQ ID NO: 176 VVLEDDNSILEK SEQ ID NO: 177 In one embodiment, a Clostridium truncated spore surface protein may be derived from a hypothetical protein (CD 0881) as listed in Table VIII:

Table VIII, Clostridium diffacile Spore Surface Protein Truncated Peptide Fragments Peptide Fragment Sequence Identification Number MGTKIVLSIV SEQ ID NO: 178 LIVwvAISL SEQ ID NO: 179 TCIRVIKQSK SEQ ID NO: 180 VGIIMRLGKF SEQ ID NO: 181 QKVAETGVHF SEQ ID NO: 182 LIPFLDKMAY SEQ ID NO: 183 VIDLREIVID SEQ ID NO: 184 FPPQPVITKD SEQ ID NO: 185 NVTMQIDTVV SEQ ID NO: 186 YYKVTDPVRY SEQ ID NO: 187 VFEIANPIAA SEQ ID NO: 188 IENLTATTLR SEQ ID NO: 189 NIIGELDLDE SEQ ID NO: 190 TLTSRDIINV SEQ ID NO: 191 KMRTILDEAT SEQ ID NO: 192 DKWGIKVNRV SEQ ID NO: 193 ELKNIMPPQD SEQ ID NO: 194 IQVAMEKQMR SEQ ID NO: 195 AERERREAIL SEQ ID NO: 196 QAFGNKSAAI SEQ ID NO: 197 LQAEGEKQSA SEQ ID NO: 198 ILTAEAKKEA SEQ ID NO: 199 MVRVAEGEKE SEQ ID NO: 200 SAILVAEGEA SEQ ID NO: 201 EAIRQTAIAK SEQ ID NO: 202 AQGEAEMIKR SEQ ID NO: 203 TQMATAEGLK SEQ ID NO: 204 LVFSAMKEAD SEQ ID NO: 205 IDNNILALKS SEQ ID NO: 206 MEALEKMAEG SEQ ID NO: 207 KSTKLVLPSE SEQ ID NO: 208 AVNFLGTFKG SEQ ID NO: 209 IKEVMSDDNK SEQ ID NO: 210 EVLDIKEVLN SEQ ID NO: 211 DNESLKK SEQ ID NO: 212 In one embodiment, a Clostridium truncated spore surface protein may be derived from a hypothetical protein (CD 1511) as listed in Table IX:

Table IX. Clostridium difficile Spore Surface Protein Truncated Peptide Fragments Peptide Fragment 1 Sequence Identification Number MIDNQKYVIL SEQ ID NO: 213 SLELHLFFSR SEQ ID NO: 214 IMKEHALFLE SEQ ID NO: 215 AGFTNKNYNL SEQ ID NO: 216 LEADHYKKQ SEQ ID NO: 217 FEDLLSYTVS SEQ ID NO: 218 ASNGIIRPDI SEQ ID NO: 219 LYSEELVTTL SEQ ID NO: 220 TSVAEQKTEE SEQ ID NO: 221 FTGIEINKNI SEQ ID NO: 222 TTRELNLQSG SEQ ID NO: 223 VNPQVGQDLV SEQ ID NO: 224 NYVAQLNSDA SEQ ID NO: 225 IRLLDGLINF SEQ ID NO: 226 KERVLDGVLS SEQ ID NO: 227 CTIFTSNYPL SEQ ID NO: 228 LLEIIIIHEAN SEQ ID NO: 229 LYRSYVVDLE SEQ ID NO: 230 NKIDIESKNA SEQ ID NO: 231 KEIELFWDHI SEQ ID NO: 232 MMEHALFMRG SEQ ID NO: 233 LLDPSEGELI SEQ ID NO: 234 NTSNDFAIKF SEQ ID NO: 235 NELIEKTNEM SEQ ID NO: 236 TDSNIKNITE SEQ ID NO: 237 ETLNETVEFK SEQ ID NO: 238 DFKEAGASGI SEQ ID NO: 239 EQCKIKSIIL SEQ ID NO: 240 PLLADHVLRE SEQ ID NO: 241 ANHYIRILES SEQ ID NO: 242 YKNIvl SEQ ID NO: 243 In one embodiment, the C. difficile truncated spore surface protein may be derived from a collagen-like protein as listed in Table X.

Table X. Clostridium difficile Spore Surface Protein Truncated Peptide Fragments Peptide Fragment Sequence Identification Number MRNIILYLND SEQ ID NO: 244 DTFISKKYPD SEQ ID NO: 245 KNFSNLDYCL SEQ ID NO: 246 IGSKCSNSFV SEQ ID NO: 247 KEKLITFFKV SEQ ID NO: 248 RIPDILKDKS SEQ 1D NO: 249 ILKAELFIHI SEQ ID NO: 250 DSNKNHIFKE SEQ ID NO: 251 KVDIEIKRIS SEQ ID NO: 252 EYYNLRTITW SEQ ID NO: 253 NDRVSMENIR SEQ ID NO: 254 GYLPIGISDT SEQ ID NO: 255 SNYICLNITG SEQ ID NO: 256 TIKAWAMNKY SEQ ID NO: 257 PNYGLALSLN SEQ ID NO: 258 YPYQILEFTS SEQ ID NO: 259 SRGCNKPYIL SEQ ID NO: 260 VTFEDRIIDN SEQ ID NO: 261 CYPKCECPPI SEQ ID NO: 262 RITGPMGPRG SEQ ID NO: 263 ATGSTGPMGV SEQ ID NO: 264 TGPTGSTGAT SEQ ID NO: 265 GSIGPTGPTG SEQ ID NO: 266 NTGATGSIGP SEQ ID NO: 267 TGVTGPTGST SEQ ID NO: 268 GATGSIGPTG SEQ ID NO: 269 VTGPTGNTGV SEQ ID NO: 270 TGSIGPTGAT SEQ ID NO: 271 GPTGN'I'GVTG SEQ ID NO: 272 SIGP'I'GVTGP SEQ ID NO: 273 TGNTGEIGP'I' SEQ ID NO: 274 GATGPTGVTG SEQ ID NO: 275 SIGPTGATGP SEQ ID NO: 276 TGEIGPTGAT SEQ ID NO: 277 ATGPTGATGV SEQ ID NO: 278 TGEIGPTGEI SEQ ID NO: 279 GPTGATGPTG SEQ ID NO: 280 VTGEIGPTGA SEQ ID NO: 281 TGPTGNTGVT SEQ ID NO: 282 GEIGPTGATG SEQ ID NO: 283 PTGNTGVTGE SEQ ID NO: 284 IGPTGATGPT SEQ ID NO: 285 GVTGEIGPTG SEQ ID NO: 286 NTGATGSIGP SEQ ID NO: 287 TGVTGPTGAT SEQ ID NO: 288 GSIGPTGATG SEQ ID NO: 289 ATGVTGPTGP SEQ ID NO: 290 TGATGNSSQP SEQ ID NO: 291 VANFLVNAPS SEQ ID NO: 292 PQTLNNGDAJ SEQ ID NO: 293 TGWQTIIGNS SEQ ID NO: 294 SSITVDTNGT SEQ ID NO: 295 FTVQENGVYY SEQ ID NO: 296 ISVSVALQPG SEQ ID NO: 297 SSSINQYSFA SEQ ID NO: 298 ILFPILGGKD SEQ ID NO: 299 LAGLTTEPGG SEQ ID NO: 300 GGVLSGYFAG SEQ ID NO: 301 FLFGGTTFTI SEQ ID NO: 302 NNFSSTTVGI SEQ ID NO: 303 RNGQSAGTAA SEQ ID NO: 304 TLTIFRIADTVMT SEQ ID NO: 305 In one embodiment, the C. difficile truncated spore surface protein may be derived from a collagen-like protein as listed in Table XI.

Table XI. Clostridia difficile Spore Surface Protein Truncated Peptide Fragments Peptide Fragment Sequence Identification Number MLLIMSRNKY SEQ ID NO: 306 FGPFDDNDYN SEQ ID NO: 307 NGYDKYDDCN SEQ ID NO: 308 NGRDDYNSCD SEQ ID NO: 309 CHHCCPPSCV SEQ ID NO: 310 GPTGPMGPRG SEQ ID NO: 311 RTGPTGPTGP SEQ III NO: 312 TGPGVGATGP SEQ ID NO: 313 TGPTGPTGPT SEQ ID NO: 314 GNTGNTGATG SEQ ID NO: 315 LRGPTGATGA SEQ ID NO: 316 TGPTGATGAI SEQ ID NO: 317 GFGVTGPTGP SEQ ID NO: 318 TGATGATGAD SEQ ID NO: 319 GVTGPTGPTG SEQ ID NO: 320 ATGADGITGP SEQ ID NO: 321 TGATGATGFG SEQ ID NO: 322 VTGPTGPTGA SEQ ID NO: 323 TGVGVTGATG SEQ ID NO: 324 LRGPTGATGT
SEQ ID NO: 325 PGATGPTGAT SEQ ID NO: 326 GATGIGITGP SEQ ID NO: 327 TGATGATGAD SEQ ID NO: 328 GATGVTGPTG SEQ ID NO: 329 PTGA'I'GADGV SEQ ID NO: 330 TGPTGATGAT SEQ ID NO, 331 GIGITGPTGP SEQ ID NO: 332 TGATGJGITG SEQ ID NO: 333 ATGLIGPTGA SEQ ID NO: 334 TGTPGATGPT SEQ ID NO: 335 GATGPTGVGV SEQ ID NO: 336 TGATGATGAT SEQ ID NO: 337 GADGATGVTG SEQ ID NO: 338 PTGATGATGA SEQ ID NO: 339 NGLVGPTGAT SEQ ID NO: 340 GAAGTPGATG SEQ ID NO: 341 PTGATGPTGV SEQ ID NO: 342 GITGATGATG SEQ ID NO: 343 ATGPTGADGA SEQ ID NO: 344 TGPTGATGNT SEQ ID NO: 345 GADGVAGPTG SEQ ID NO: 346 ATGNTGADGA SEQ ID NO: 347 'I'GPTGATGAT SEQ ID NO: 348 GVTGATGPTG SEQ ID NO: 349 PTGATGATGA SEQ ID NO: 350 TGASAIIPFA SEQ ID NO: 351 SGIPLSLTTI SEQ ID NO: 352 AGGLVGTPGF SEQ ID NO: 353 VGFGSSAPGI, SEQ ID NO: 354 SIVGGVIDLT SEQ ID NO: 355 NAAGTLTNFA SEQ ID NO: 356 FSMPRDGTIT SEQ ID NO: 357 SISAYFSTTA SEQ ID NO: 358 ALSLVGSTIT SEQ ID NO: 359 ITATLYQSTA SEQ ID NO: 360 PNNSFTAVPG SEQ ID NO: 361 ATVTLAPPLT SEQ ID NO: 362 GILSVGSISS SEQ ID NO: 363 GIVTGLNIAA SEQ ID NO: 364 TAQTPDRQYAI SEQ ID NO: 365 In one embodiment, the C. difficile truncated spore surface protein may be derived from a collagen-like protein as listed in Table XII.

Table XII. Clostridia difficile Spore Surface Protein Truncated Peptide Fragments Peptide Fragment Sequence Identification Number MSDISGPSLY SEQ ID NO: 366 QDVGPTGPTG SEQ ID NO: 367 ATGPTGPTGP SEQ ID NO: 368 RGATGATGAN SEQ ID NO: 369 GITGPTGNTG SEQ ID NO: 370 ATGANGITGP SEQ ID NO: 371 TGNMGATGPN SEQ ID NO: 372 GTTGSTGPTG SEQ ID NO: 373 NTGATGANGI SEQ ID NO: 374 TGPTGNTGAT SEQ ID NO: 375 GANGITGPTG SEQ ID NO:-376 NXGATGANGI SEQ ID NO: 377 TGSTGPTGNT SEQ ID NO: 378 GATGANGITG SEQ ID NO: 379 PTGNTGATGA SEQ ID NO: 380 TGPTGLTGAT SEQ ID NO: 381 GATGANGITG SEQ ID NO: 382 PI'GNTGA'rGA SEQ ID NO: 383 NGVTGATGP'I' SEQ ID NO: 384 GNTGATGPTG SEQ ID NO: 385 SIGATGATGT SEQ ID NO: 386 TGATGPIGAT SEQ ID NO: 387 GATGATGVVG SEQ ID NO: 388 PTGAVGATGP SEQ ID NO: 389 DGLVGPTGPT SEQ ID NO: 390 GPTGATGANG SEQ ID NO: 391 LVGPTGPTGA SF,Q ID NO: 392 TGANGLVGPT SEQ ID NO: 393 GATGATGVAG SEQ ID NO: 394 A-IGPTGAVGA SEQ ID NO: 395 TGPTGADGAV SEQ ID NO: 396 GPTGATGATG SEQ ID NO: 397 ANGATGPTGA SEQ ID NO: 398 VGATGANGVA SEQ ID NO: 399 GPIGPTGPTG SEQ ID NO: 400 ENGVAGATGA SEQ ID NO: 401 TGATGANGAT SEQ ID NO: 402 GPTGAVGATG SEQ ID NO: 403 ANGVAGAIGP SEQ ID NO: 404 TGPTGANGAT SEQ ID NO: 405 GATGATGAAG SEQ ID NO: 406 ANGATGPTGA SEQ ID NO: 407 TGATGVLAAN SEQ ID NO: 408 NAQFTVSSSS SEQ ID NO: 409 LVNNTLVTFN SEQ ID NO: 410 SSFINGTNIT SEQ ID NO: 411 FPTSSTINLA SEQ ID NO: 412 \ GG1YNVSFG SEQ ID NO: 413 IRATLSLAGF SEQ ID NO: 414 MSITT"NFNGV SEQ ID NO: 415 TQNNFIAKAV SEQ ID NO: 416 NTLTSSDVSV SEQ ID NO: 417 SLSFLVDARA SEQ ID NO: 418 AAVTLSFTFG SEQ ID NO: 419 SGTTGTSAAG SEQ ID NO: 420 YVSVYIUQ SEQ ID NO: 421 In one embodiment, the truncated microbial spore surface protein is derived from a Bacillus full length spore surface protein. In one embodiment, the Bacillus truncated spore surface protein comprises a B. anthraces truncated spore surface protein. In one embodiment, the B. anthracis truncated spore surface protein may be derived from a BelA protein as listed in Table XIII.

Table XIII. Bacillus anthracis Spore Surface Protein Truncated Peptide Fragments Peptide Fragment Sequence ID Number MSNNNYSNGL SEQ ID NO: 422 NPDESLSASA SEQ ID NO: 423 FDPNLVGPTL SEQ ID NO: 424 PPIPPFTLPT SEQ ID NO: 425 GPTGPTGPTG SEQ ID NO: 426 PTGPTGPTGP SEQ ID NO: 427 TGPTGPTGPT SEQ ID NO: 428 GPTGTTGPTG SEQ ID NO: 429 TGDTGTTGPT SEQ ID NO: 430 PTGPTGDTGT SEQ ID NO: 431 PTGPTGPTGA SEQ ID NO: 432 TGLTGPTGPT SEQ ID NO: 433 GPSGLGLPAG SEQ ID NO: 434 LYAFNSGGIS SEQ ID NO: 435 FNTVGSQFGT SEQ ID NO: 436 AISQLDADTF SEQ ID NO: 437 VISETGFYKI SEQ ID NO: 438 TVIANrATAS SEQ ID NO: 439 GVPVPGTGSS SEQ ID NO: 440 LISLGAPIVI SEQ ID NO: 441 QAITQITTTP SEQ ID NO: 442 SLVEVIVTGL SEQ ID NO: 443 GLSLALGTSA SEQ ID NO: 444 S111EKVA SEQ ID NO: 445 C. Spore Surface Protein Mimetics In one embodiment, the present invention contemplates compositions and methods for making and using infection control compositions comprising microbial spore surface protein mimetics. In one embodiment, the protein mimetic comprises an amino acid sequence. In one embodiment, the amino acid sequence comprises a circular tertiary structure. In one embodiment, the protein mimetic comprises a small organic molecule.
Small organic molecules mimicking the necessary conformation for recognition and docking to the active site binding to the peptides of the present invention are contemplated as within the scope of this invention. For example, mimetics of microbial spore surface proteins and/or peptide fragments are contemplated. A variety of designs for such mimetics are possible. For example, cyclic and/or linear proteins and/or peptide fragments, in which the necessary conformation for binding is stabilized by nonpeptides, are specifically contemplated. United States Patent No. 5,192,746 to Lobl, et at., United States Patent No. 5,169,862 to Burke, Jr., et al., United States Patent No.
5,539,085 to Bischoff, et al., United States Patent No. 5,576,423 to Aversa, et al., United States Patent No. 5,051,448 to Shashoua, and United States Patent No. 5,559,103 to Gaeta, et al., all hereby incorporated by reference, describe multiple methods for creating such compounds.
Synthesis of non-peptide compounds that mimic peptide sequences is also known in the art. For example, some non-peptide antagonists mimic the Arg-Gly-Asp sequence.
Eldred, et al., (J. Med. Chem. 37:3882 (1994)). Likewise, Ku, et al., (J. Med.
Chem. 38:9 (1995)) give further elucidation of the synthesis of a series of such compounds. Such non-peptide compounds that mimic microbial spore surface proteins and/or peptide fragments are specifically contemplated by the present invention.
The present invention also contemplates synthetic mimicking compounds that are multimeric compounds that repeat relevant peptide sequences. As is known in the art, peptides can be synthesized by linking an amino group to a carboxyl group that has been activated by reaction with a coupling agent, such as dicyelohexylcarbodiimide (DCC).
The attack of a free amino group on the activated carboxyl leads to the formation of a peptide bond and the release of dicyclohexylurea. It can be necessary to protect potentially reactive groups other than the amino and carboxyl groups intended to react.
For example, the a-amino group of the component containing the activated carboxyl group can be blocked with a tertbutyloxycarhonyl group. This protecting group can be subsequently removed by exposing the peptide to dilute acid, which leaves peptide bonds intact.
With this method, peptides can be readily synthesized by a solid phase method by adding amino acids stepwise to a growing peptide chain that is linked to an insoluble matrix, such as polystyrene beads. The carboxyl-terminal amino acid (with an amino protecting group) of the desired peptide sequence is first anchored to the polystyrene beads. The protecting group of the amino acid is then removed. The next amino acid (with the protecting group) is added with the coupling agent. This is followed by a washing cycle. The cycle is repeated as necessary.
In one embodiment, the mimetics of the present invention are peptides having sequence homology to the above-described microbial spore surface proteins and/or peptide fragments. One common methodology for evaluating sequence homology, and more importantly statistically significant similarities, is to use a Monte Carlo analysis using an algoritlun written by Lipman and Pearson to obtain a Z value.
According to this analysis, a Z value greater than 6 indicates probable significance, and a Z
value greater than 10 is considered to be statistically significant. W.R. Pearson and D.J.
Lipman, Proc.
Natl. Acad. Sci. (USA), 85:2444-2448 (1988); D.J. Lipman and W.R. Pearson, Science, 227:1435-1441 (1985). In the present invention, synthetic polypeptides useful in solving the problems of microbial decontamination and microbial infection are those peptides with statistically significant sequence homology and similarity (Z value of Lipman and Pearson algorithm in Monte Carlo analysis exceeding 6).
IV. Screening For Infection Control Compositions In one embodiment, the present invention contemplates methods comprising screening for infection control compositions that inhibit, antagonize, interfere, displace, and/or disrupt the binding of a microbial spore to an inanimate and/or animate surface.
In one embodiment, the present invention contemplates methods comprising screening for infection control compositions that inhibit, antagonize, interfere, displace, and/or disrupt the binding of a microbial spore surface protein to an inanimate and/or animate surface.

In one embodiment, the animate surface may be selected from the group comprising skin, human body tissues, and/or human cells. In one embodiment, the inanimate surface may comprise hard surfaces including, but not limited to, ceramic, tile, granite, and/or stainless steel. In one embodiment, the microbial spore comprises a Clostridium spore. In one embodiment, the Clostridium spore comprises a C.
diffacile spore. In one embodiment, the screened infection control compositions are capable of binding to a microbial spore surface protein. Although it is not necessary to understand the mechanism of an invention, it is believed that the infection control compositions bind to a surface thereby blocking binding of a microbial spore surface protein.
Although it is not necessary to understand the mechanism of an invention, it is believed that such infection control compositions may include, but are not limited to, natural compounds isolated from viral, bacterial, fungal, plant, and animal sources, as well as synthetic compounds. It is further believed that neither the source, chemical structure, or mechanism of action of the infection control composition is essential to this invention.
A. Assays For Detecting Microbial Spore Binding To A Surface 1. Standard Growth Assays In one embodiment, the present invention contemplates methods for detecting microbial spores and/or microbial spore proteins comprising: a) providing; i) a substrate surface suspected of contamination by at least one microbial spore; ii) a spore collection device comprising a plurality of infection control compositions capable of attaching the at least one microbial spore; and iii) a spore growth plate capable of supporting growth of the at least one microbial spore; b) contacting the spore collection device with the substrate surface wherein the at least one microbial spore becomes attached to the device, c) contacting the microbial collection device with the spore growth plate; and d) detecting at least one microbial spore on the spore growth plate. In one embodiment, the infection control composition comprises a microbial spore protein peptide fragment. In one embodiment, the microbial spore protein peptide fragment is selected from the group comprising a Clostridium spore protein peptide fragment, or a Bacillus spore protein peptide fi-agment. In one embodiment, the microbial spore is a Clostridium difficile spore. In one embodiment, the microbial spore is a Bacillus anthracis spore.
Although it is not necessary to understand the mechanism of an invention, it is believed that a growth assay can be performed using any microbial spore. By way of illustration, C. sporogenes spores are used below in a method to detect the growth of spores after their collection from a contaminated surface, but is should be understood that any microbial spore can be substituted wherein collection and detection each respective microbial spore will be successfully performed (i.e., for example, Clostridium difficile antibodies).
a. Cup Scrub Assays One described technique is suitable for the recovery of resident and transient microorganisms from human or animal skin. For example, the technique may be used in situ within clinical protocols or in vitro for studies using isolated skin or skin equivalents.
ASTM International, "Standard Test Method for Recovery of Microorganisms From Skin using the Cup Scrub Technique" El 874 - 09 (2009). Briefly, resident microorganisms or transient microorganisms (i.e., for example, microorganisms previously applied to a test site), are recovered from the site by pressing a rigid cylinder against the skin with sufficient pressure to form a seal and instilling recovery liquid into the cylinder. The surface of the skin is then mechanically `scrubbed' with a glass rod, rubber policeman, or some other suitable device for a prescribed period of time. The fluid is pipetted from the cylinder into a test tube, or other suitable receptacle, for further analysis.
A cub-scrub procedure can be incorporated into any method describes herein to evaluate test materials containing infection control compositions that are intended to reduce significantly the number of microbial spores on a test surface (i.e., for example, intact skin). It also may be used to provide an indication of residual antimicrobial activity.
b. Cell Culture Assays Aliquots of C diff cile endospore suspension are anaerobically incubated at 35 C
on prereduced (i.e., for example, oxygen removed) BCDC, TCCFA, and TSA.
Control and experimental plates are inoculated identically with a spore suspension, and one set of control plates are allowed to incubate and grow undisturbed for the duration of each experiment.
Attachment experiments are done by placing a circle (i.e., for example, a target area) on the bottom of the agar-filled Petri dish, and inoculating the plate with 5 or 10 l of C. difficile spore suspensions directly within a target area, incubating these spores in the anaerobic hood (Forma Scientific, Marietta, Ohio) on pre-reduced plates for 27 min, 99 min, 150 min, and 360 min. After timed incubation, the plates are removed, and 4 ml of sterile distilled water added to each culture plate. The plates are then shaken at 300 rpm for 10 min, after which the agitation solution is aseptically pipetted off the agar surface, and the plates re-incubated anaerobically for 24, 48, and 72 h. To determine if the plates had become contaminated, light microscopy smears on glass slides are made from randomized colonies and photographed on a photomicroscope, and plates are inoculated and incubated under aerobic conditions to identify any aerobic contaminants.
To determine if spores are not yet attached to the agar, and establish if they could be easily detached and washed off the agar surface, the water agitation solution (removed after shaking the plate) is also tested for the presence of spores. Aliquots of the water is:(i) screened by phase microscopy; and (ii) used to streak agar plates; and (iii) injected into BacTec NR6A broth (anaerobic blood culture broth, BD Microbiology Systems Cockeyesville, MD) and the plates and broth read at 24 and 48 h for the presence of bacterial growth.
2. Anti-Spore Surface Antibody Assays In one embodiment, the present invention contemplates methods for detecting microbial spores comprising: a) providing; i) a substrate surface suspected of contamination by at least one microbial spore; and ii) an anti-spore surface antibody capable of binding to the at least one microbial spore; b) contacting the anti-spore surface antibody with the substrate surface or spore surface under conditions such that the antibody binds to the at least one microbial spore, wherein an antibody/spore complex is created; and c) detecting the antibody/spore complex. In one embodiment, the antibody comprises a polyclonal antibody (i.e.., for example, F1997) In one embodiment, the anti-spore surface antibody comprises a label. In one embodiment, the label comprises a fluorescent label. In one embodiment, the microbial spore is a Clostridium difficile spore.
In one embodiment, the microbial spore is a Bacillus anthraces spore.
In one embodiment, the present invention contemplates methods for detecting microbial spores comprising: a) providing; i) a substrate surface suspected of contamination by at least one microbial spore; ii) an anti-spore surface antibody capable of binding to the at least one microbial spore; and iii) a growth assay plate;
b) combining the anti-spore surface antibody with the antibody, wherein an antibody/spore complex is created; c) contacting the antibody/spore complex with the substrate surface;
d) removing a sample of the antibody/spore complex (i;e., for example, by the cup-scrub method) from the surface; e) streaking the sample on the growth assay plate. In one embodiment, the antibody comprises a polyclonal antibody (i.e.., for example, F1997). In one embodiment, the substrate surface comprises a pigskin surface. In one embodiment, the label comprises a fluorescent label. In one embodiment, the microbial spore is a Clostridium difficile spore. In one embodiment, the microbial spore is a Bacillus anthraces spore.

a. Antibody Development Although it is not necessary to understand the mechanism of an invention, it is believed that an anti-spore surface antibody can be directed towards any epitope on a spore surface (i.e., whether a protein or other antigenic structure). By way of illustration, C. sporogenes spores are used below in a method to generate anti-spore surface monoclonal and polyclonal antibodies, but is should be understood that any microbial spore can be substituted wherein antibodies against each respective microbial spore will be successfully generated (i.e., for example, Clostridium difficile antibodies).
Mouse polyclonal antiserum and monoclonal antibodies against C. sporogenes spores were obtained by several injections of 108 inactivated spores (incubated in 4%
(v/v) formaldehyde for 4 h at 37 C). Monoclonal antibodies were produced according to the procedures of Kohler and Milstein (Kohler and Milstein, 1975). Briefly, the mice were boosted on days 21 and 42 and bled on day 50 for polyclonal antibodies, or boosted on day 50 and sacrificed for fusion on day 53 for monoclonal antibodies. Spore stocks are prepared by growing C. sporogenes cells in cooked meat medium at 35 C, and Clostridium perfringens cells in fluid thioglycolate medium at 37 C under anaerobic conditions Antigen prepared by the above formalin inactivation of C. sporogenes PA3679 spore stocks were free of vegetative cell material as observed by phase-contrast microscopy. Phillips et al., "Assessment of immunofluorescence measurements of individual bacteria in direct and indirect assays for Bacillus anthracis and Bacillus cereus spores" J. Appl. Bacteriol. 53:223-231 (1982). A total of 1 x 10' C.
sporogenes spores are used as the antigen.
Monoclonal antibody production is done according one of many procedures.
Kohler et al., "Continuous cultures of fused cells secreting antibody of predefined specificity" Nature 296:495-497 (1975)(herein incorporated by reference in its entirety).
Hybridomas are screened for reactivity with C. sporogenes spores. Hybridomas found to be reactive are also screened for reactivity with the vegetative cells.
Isotyping of monoclonal antibodies are performed by using a dot blot immunoassay format. Spores are applied to an Immobilon-P membrane (Millipore Corp., Bedford, Mass.) in a dot blot apparatus. The spores are then incubated with 100 ml of the appropriate tissue culture supernatants. Monoclonal antibody bound to spores are isotyped by using rabbit anti-mouse subclass specific antiserum obtained from Bio-Rad` (Hercules, Calif).

Clostridium sporogenes spores were then applied to an Immobilon-P membrane, followed by incubation with above generated antibodies. Similarly, 106 of the Clostridium vegetative cells are applied to the membrane, followed by incubation with the indicated antibodies. Reactivity is detected with a goat anti-mouse immunoglobulin G (IgG)-horseradish peroxidase conjugate (Sigma A4416) or a goat anti-mouse Ig'M-horseradish peroxidase conjugate (Sigma A8786). Color development may be performed with the insoluble substrate3-amino-9-ethyl-carbazole (Biomeda Corp.).
Culture supernatants are then diluted 1:32 and 1:128 with Tns-buffered saline (TBS) (0.01 M Tris, 0.15 M NaCl, pH 7.4). Aliquots of diluted culture supernatants are mixed with an equal volume of the spore antigen (4 x 109 spores/ml) by gentle inversion overnight at 48 C. Diluted culture supernatants mixed with sterile distilled water serve as a positive control. Antibodies adsorbed to spores are removed by centrifugation (ca.
4,000 3g, 10 min). The supernatant fluid is used in immunoblot assays as detailed above.
Antibodies are concentrated by ammonium sulfate precipitation of tissue culture supernatants. Halow et al., "Storing and purifying antibodies" In: Antibodies:
A
Laboratory Manual. p. 283--318. E. Halow and D. Lane (ed.), Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y. (1988). The concentrated protein is biotinylated by incubation with N-hydroxysuccinimidobiotin (Sigma Chemical Co.) (ca. 100 mg/mg of antibody in the sample) at room temperature for 4 h. One molar ammonium chloride (20 ml/250 mg of biotin) is added to stop the reaction, and the biotinylated protein is dialyzed extensively against phosphate-buffered saline (0.1 M sodium phosphate, 0.8%
NaCl) (PBS), pH 7Ø For immunocytochemical localization studies.
IgG antibodies are purified out of tissue culture supernatants by using a protein A
column (Pierce Chemical Co., Rockford, Ill.) according to the manufacturer's directions.
IgM antibodies are purified from the culture supernatant by using a goat anti-mouse IgM
antibody column (Pierce Chemical Co.). The tissue culture supernatant is diluted 1:1 in PBS and applied to the goat anti-mouse IgM column, which is equilibrated with PBS.
Washing and elution of antibody off the column is performed according to the manufacturer's directions. Fractions containing protein are pooled and concentrated with a Centricon 10 micro concentrator (Amicon, Danvers, Mass.) and centrifugation at 3,000 3 g for 30 min.
3. Anti-Spore Surface Protein Antibody Assays In one embodiment, the present invention contemplates methods for detecting microbial spores comprising: a) providing; i) a substrate surface suspected of contamination by at least one microbial spore, said spore comprising at least one surface protein; and ii) a anti-surface protein antibody capable of binding to the at least one surface protein; b) contacting the anti- surface protein antibody with the substrate surface under conditions such that the antibody binds to the at least one surface protein, wherein an antibody/protein complex is created; and c) detecting the antibody/protein complex. In one embodiment, the spore surface protein includes, but is not limited to, CD
1067, CD
1581, CD 3520, CD 0372, or CD 1613. In one embodiment, the anti- surface protein antibody comprises a label. In one embodiment, the label comprises a fluorescent label.
In one embodiment, the surface protein is a Clostridium difficile surface protein. In one embodiment, the surface protein is a Bacillus anthracis surface protein.
Although it is not necessary to understand the mechanism of an invention, it is believed that a microbial surface protein antibody can be directed towards a specific epitope on a spore surface protein (i.e., whether an amino acid sequence or other antigenic structure). By way of illustration, a specific C. diffcile spore surface protein is used below in a method to generate polyclonal and monoclonal antibodies, but is should be understood that any microbial spore surface protein can be substituted wherein polyclonal and/or monoclonal antibodies directed against each respective microbial spore surface protein will be successfully generated.
Mouse polyclonal antiserum and monoclonal antibodies against C. difficile hypothetical protein (SEQ ID NO: 1) are obtained by several injections of isolated hypothetical protein (i.e., for example, approximately 108 proteins per injection).
Monoclonal antibodies are produced according to the procedures of Kohler and Milstein (Kohler and Milstein, 1975). Briefly, the mice can be boosted on days 21 and 42 and bled on day 50 for polyclonal antibodies, or boosted on day 50 and sacrificed for fusion on day 53 for monoclonal antibodies. Hybridomas may be screened for reactivity with C.
difficile spores. Hybridomas found to be reactive are also screened for reactivity with the vegetative cells.
Isotyping of monoclonal antibodies are performed by using a dot blot immunoassay format. Spores are applied to an Immobilon-P`P membrane (Millipore Corp., Bedford, Mass.) in a dot blot apparatus. The spores are then incubated with 100 ml of the appropriate tissue culture supernatants. Monoclonal antibody bound to spores are isotyped by using rabbit anti-mouse subclass specific antiserum obtained from Bio-Rad` ' (Hercules, Calif.).

In one embodiment, the present invention contemplates methods for detecting microbial spores comprising: a) providing; i) a substrate surface suspected of contamination by at least one microbial spore, said spore comprising at least one surface protein; and ii) a anti-surface protein monoclonal antibody capable of binding to the at least one surface protein; b) contacting the anti-surface protein monoclonal antibody with the substrate surface under conditions such that the monoclonal antibody binds to the at least one surface protein, wherein a monoclonal antibody/protein complex is created; and c) detecting the monoclonal antibody/protein complex. In one embodiment, the spore surface protein includes, but is not limited to, CD 1067, CD 1581, CD 3520, CD
0372, or CD 1613. In one embodiment, the anti-surface protein monoclonal antibody comprises a label. In one embodiment, the label comprises a fluorescent label. In one embodiment, the surface protein is a Clostridium difficile surface protein. In one embodiment, the surface protein is a Bacillus anthracis surface protein.
B. Assays For Detecting Microbial Spore Surface Protein Binding To A
Surface Several methods are provided herein for detecting spore surface protein binding in order to screen potential infection control compositions.
In order to provide methods and devices for screening compounds for developing infection control compositions, the present invention generally incorporates model in vitro systems which mimic a given microbial system in vitro for which infection control compositions are desired. The range of systems against which these infection control compositions can be screened is extensive. For example, potential infection control compositions are optionally screened for effects in blocking, slowing or otherwise inhibiting key events associated with infectious microbial organisms. For example, potential infection control test compositions are optionally screened for their ability to block systems that are responsible, at least in part, for sporulation or for the binding of spores to a surface, including, e.g., skin, bodily tissues, granite, stainless steel, ceramic etc.. Infection control test compositions which show promising results in these screening assay methods can then be subjected to further testing to identify effective pharmacological agents for the treatment of disease or symptoms of microbial infections 1. Binding Screens In their simplest forms, the infection control composition screening models employed in the methods and apparatuses of the present invention will screen for an interaction of a infection control composition with a component of a biochemical system, e.g., receptor-ligand interaction, enzyme-substrate interaction, and the like.
In this form, the screening model will typically include two interacting components of the system (i.e., for example, a bacterial spore protein and an infection control composition or a microbial enzyme and an infection control composition).
In one embodiment, the method comprises determining whether an infection control test composition has an effect on microbial spore protein binding interactions with a surface by contacting the microbial spore protein with the infection control test composition and assaying for residual binding of the microbial spore to the surface. The residual surface binding amount is then compared to a control surface binding, e.g., the same binding assay performed in the absence of the infection control composition.
Typically, such binding assays involve the measurement of a parameter of the screening system. By "parameter of the screening system" is meant some measurable evidence, e.g., the presence or absence of a labeled group or a change in molecular weight (e.g., in binding reactions, transport screens), the presence or absence of a reaction product or substrate (in substrate turnover measurements), or an alteration in electrophoretic mobility (typically detected by a change in elution time of a labeled compound).
Although described above in terms of a two-component screening systems, the methods and apparatuses may also he used to screen for effectors of much more complex systems, where the result or end product of the system is known and assayable at some level, e.g., enzymatic pathways, spore surface signaling pathways and the like.
Alternatively, the methods and apparatuses described herein are optionally used to screen for compounds that interact with a single component of a biochemical system, e.g., compounds that specifically bind to a particular biochemical compound, e.g., a receptor, ligand, enzyme, nucleic acid, structural macromolecule, etc.
In some embodiments, assays of this invention may involve the interaction of two or more components. Assays involving interactions between two components can be viewed as assays for enhancers or inhibitors of binding between members of "binding pairs". Preferred binding pairs include, but are not limited to, microbial spore proteins/small organic molecules, microbial spore proteins/nucleic acids, microbial spore proteins/infection control compositions (i.e., for example, Clostridium spore surface protein fragments). In one embodiment, either member of the binding pair can be immobilized (i.e., for example, attached to the surface) while the other member is in a solution contacted to the surface. In another embodiment, both members can be attached to different surfaces which are then juxtaposed to perform the assay.
Accordingly, in one aspect, the present invention will be useful in screening for infection control compositions that form a binding pair with a receptor molecule, wherein the binding pair affects an interaction between the receptor molecule and a surface. As used herein, the tern "receptor" generally refers to one member of the binding pair (i.e., for example, a microbial spore surface protein). The other member of the binding pair is termed "an infection control composition. Thus, a receptor/ligand pair may include a typical protein receptor, usually membrane associated, and its natural ligand, e.g., another protein or small molecule. Receptor/ligand pairs may also include antibody/antigen binding pairs, complementary nucleic acids, nucleic acid associating proteins and their nucleic acid ligands.
Traditionally, methods for screening for effectors of a receptor/ligand interaction (i.e., for example, a microbial spore surface protein/infection control composition interaction) have involved incubating a receptor/ligand binding pair in the presence of a test compound. The level of binding of the receptor/ligand pair is then compared to negative and/or positive controls. Where a decrease in normal binding is seen, the test compound is determined to be an inhibitor of the receptor/ligand binding.
Where an increase in that binding is seen, the test compound is determined to be an enhancer or inducer of the interaction. In the present invention, however, it is contemplated that the receptor/ligand interaction is assayed by measuring the amount of residual binding of the receptor to a surface in the presence of the infection control composition.
In one embodiment, high throughput screening assays may comprise multiwell reaction plates, e.g., multi-well microplates, which allow for the simultaneous, parallel screening of large numbers of potential infection control compositions.
A similar, and perhaps overlapping, set of biochemical systems may include the interactions between microbial enzymes and their substrates. The term "enzyme"
as used herein, generally refers to a protein which acts as a catalyst to induce a chemical change in other compounds or "substrates." Typically, effectors of an enzyme's activity toward its substrate are screened by contacting the enzyme with a substrate in the presence and absence of the compound to be screened and under conditions optimal for detecting changes in the enzyme's activity. After a set time for reaction, the mixture is assayed for the presence of reaction products or a decrease in the amount of substrate.
The amount of substrate that has been catalyzed is them compared to a control, i.e., enzyme contacted with substrate in the absence of test compound or presence of a known effector. As above, a compound that reduces the enzymes activity toward its substrate is termed an "inhibitor," whereas a compound that accentuates that activity is termed an "inducer."
Depending upon the particular embodiment being practiced, the infection control test compositions are provided in various different media including, but not limited to, by injection, free in solution, attached to a carrier, attached to a solid support (i.e., for example, beads). A number of suitable solid supports are employed for immobilization of the test compounds. Examples of suitable solid supports include agarose, cellulose, dextran (commercially available as, i.e., Sephadex, Sepharose) carboxymethyl cellulose, polystyrene, polyethylene glycol (PEG), filter paper, nitrocellulose, ion exchange resins, plastic films, glass beads, polyaminemethylvinylether maleic acid copolymer, amino acid copolymer, ethylene-maleic acid copolymer, nylon, silk, etc. Additionally, for the methods and apparatuses described herein, test compounds are screened individually, or in groups. Group screening is particularly useful where hit rates for effective test compounds are expected to be low such that one would not expect more than one positive result for a given group. Alternatively, such group screening is used where the effects of different test compounds are differentially detected in a single system.
2. Immunoassays In one embodiment, the present invention contemplates immunological tests where a reaction takes place between two or more related immunological agents (i.e., for example, an antigen and antibody). Thus, for example, when it is sought to determine whether and how much of a particular antigen or antibody resides in a sample one must attempt to react the sample suspected of containing an antigen with its immunological binding pair. Although it is not necessary to understand the mechanism of an invention, it is believed that if a reaction takes place, then the visualization of that reaction is evidence of the presence of the antigen or antibody in the originally tested fluid.
In one embodiment, an antibody/antigen binding pair may be visualized using an indicator or carrier particle. One suitable system comprises a polymeric particle technique in which synthetic resin particles, surfaces, or matrices have been used as an adsorbent onto which the appropriate antigen (i.e., for example, a microbial spore protein fragment) or antibody (i.e., for example, having affinity to a microbial spore protein fragment) has been adsorbed.
Formats for immunoassays include, but are not limited to, competitive (e.g., ELISA, hapten inhibition, etc.) and noncompetitive assays. Various immunoassay formats have been reported. See, U.S. Pat. Nos. 4,366,241; 4,376,110;
4,517,288; and 4,837,168; Asai (1993) Methods in Cell Biology Volume 37: Antibodies in Cell Biology, Academic Press, Inc. New York, Stites & Terr, eds. (1991) Basic and Clinical Immunology 7th Edition, etc.) and the references cited therein)(all reference herein incorporated by reference.
3. Nucleic Acid Binding Assays In another embodiment, the materials and methods of this invention are used to immobilize components in nucleic acid binding protein assays. Typically such assays detect binding of a nucleic acid by one or more binding proteins. The assays are generally used to screen for agents that improve (specificity or affinity) or inhibit nucleic acid binding by one or more particular nucleic acid binding proteins. Nucleic acid binding proteins include, but are not limited to endonucleases, polymerases, nuclear transcription factor receptors, API proteins (i.e., for example,fos andjun).
Nucleic acid binding assays are facilitated by attachment to a surface of either the nucleic acid containing the protein binding site or the binding protein itself.
4. Binding Assays in Physiologically Compatible Solutions It will be appreciated that may of the foregoing assays require particular conditions for the subject assay components (e.g., enzymes) to maintain their desired activity. Typically such assays are optimized to maintain physiologically compatible conditions for elements (e.g., pH, ion composition) critical to component activity.
It is a significant advantage that the infection control compositions of this invention are stable to physiological concentrations of most ionic species (e.g., Na+, Cat+, Mgt+, etc.) Thus, the assays can be run in standard physiologically compatible compositions (e.g., phosphate buffered saline (PBS) standard ringers solutions, and the like.).
The infection control compositions of this invention can also be used to purify the moiety (e.g., polypeptide(s)) to which they for binding pairs. Specifically, the infection control compositions can be used in conjunction with virtually any anion or cation exchange resin. Suitable anion and cation exchange resins are commercially available.
Cation exchange resins, for example include, but are not limited to, carboxymethylcellulose, while anion exchange resins include, but are not limited to, DEAE cellulose, DEAE SEPHAROSE gel filtration ion exchange media, heparin, and the like.

in one preferred embodiment, the interaction of an infection control composition with a microbial spore surface protein can be exploited for purification purposes. This accomplished in a manner analogous to a cation exchange resin or in a manner analogous to the polyhistidine/nickel separation systems substituting mica for the cation exchange resin or the nickel resin, respectively. In one embodiment, the mica is provided as a bed (either of mica flakes or powder) through which the sample is flowed. The mica bed is housed in any of a variety of suitable vessels, including cartridges that are attachable to syringes, chromatography columns, and the like. The microbial spore surface protein binds to the mica bed, while other components of the sample are washed away.
Non-specifically binding components can be eluted away using salt solutions at concentrations sufficient to release undesired components while retaining the microbial spore surface protein. Once the infection control composition is bound to the microbial spore surface protein, the infection control composition can then be released by application of sufficient salt concentrations and/or other appropriate solvent mixtures as desired.
5. Continuous Flow Assay Systems In one embodiment, the methods and apparatuses of the invention are used in screening infection control test compositions using a continuous flow assay system. The use of such continuous flow assay systems are not limited to screening for spore surface protein binding to surfaces but can also be readily used in screening for inhibitors or inducers of enzymatic activity, or for agonists or antagonists of receptor-ligand binding.
In brief, the continuous flow assay system involves the continuous flow of the particular biochemical system along a fluid pathway (i.e., for example, a microarray, a chromatography column, or a microfabricated channel). As used herein, the term "continuous" generally refers to an unbroken or contiguous stream of the particular composition that is being continuously flowed. For example, a continuous flow may include a constant fluid flow having a set velocity, or alternatively, a fluid flow which includes pauses in the flow rate of the overall system, such that the pause does not otherwise interrupt the flow stream.
6. Detectable Labels In one embodiment, a functioning screening system comprises producing a detectable event or signal. Such detectable signals may include, but are not limited to, optically detectable chromophoric or fluorescent signals. For enzyme systems, such signals can he produced by products of the enzyme's catalytic action, e.g., on a chromogenic or fluorogenic substrate. For binding systems, e.g., receptor ligand interactions or microbial spore surface binding to surfaces, signals will typically involve the association of a labeled ligand with the receptor, or vice versa.
A wide variety of other detectable signals and labels can also be used in the embodiments of the presently contemplated invention. In addition to the chromogenic and fluorogenic labels described above, radioactive decay, electron density, changes in pH, solvent viscosity, temperature and salt concentration are also conveniently measured.
More generally, labels are commonly detectable by spectroscopic, photochemical, biochemical, immunochemical, or chemical means. For example, useful nucleic acid labels include 32p 35S, fluorescent dyes, electron-dense reagents, enzymes (e.g., as commonly used in an ELISA), biotin, dioxigenin, or haptens and proteins for which antisera or monoclonal antibodies are available. A wide variety of labels suitable for labeling biological components are known and are reported extensively in both the scientific and patent literature, and are generally applicable to the present invention for the labeling of biological components. Suitable labels include, but are not limited to, radionucleotides, enzymes, substrates, cofactors, inhibitors, fluorescent moieties, chemiluminescent moieties, magnetic particles, and the like. Labeling agents optionally include e.g., monoclonal antibodies, polyclonal antibodies, proteins, or other polymers such as affinity matrices, carbohydrates or lipids.
Detection of labels may proceed by a variety of methods, including but not limited to, spectrophotometric or optical tracking of radioactive or fluorescent markers, or other methods which track a molecule based upon size, charge or affinity. A
detectable moiety can be of any material having a detectable physical or chemical property. Such detectable labels exist in the fields of gel electrophoresis, column chromatography, solid substrates, spectroscopic techniques, and the like, and in general, labels useful in such methods can be applied to the present invention.
Thus, a label may comprise any composition detectable by spectroscopic, photochemical, biochemical, immunochemical, electrical, optical thermal, or chemical means. In some embodiments, labels may include, but are not limited to, fluorescent dyes (e,g., fluorescein isothiocyanate, Texas red, rhodamine, and the like), radiolabels (e.g., 3H, 125I 35S, ' C, 32P or 33P), enzymes (e.g., LacZ, CAT, horse radish peroxidase, alkaline phosphatase and others, commonly used as detectable enzymes, either as marker products or as in an ELISA), nucleic acid intercalators (e.g., ethidium bromide) and colorimetric labels such as colloidal gold or colored glass or plastic (e.g, polystyrene, polypropylene, latex, etc.) beads.

In one embodiment, the present invention contemplates fluorescent labels that are typically characterized by one or more of the following: high sensitivity, high stability, low background, low environmental sensitivity and high specificity in labeling. Such fluorescent moieties, which are incorporated into the labels of the invention may include, but are not limited to, 1- and 2-aminonaphthalene, p,p'-di aminostilbenes, pyrencs, quatemary phenanthridine salts, 9-aminoacridines, p,p'-di am inobenzophenone imines, anthracenes, oxacarbocyanine, merocyanine, 3-aminocquilenin, perylene, his-benzoxazole, bis-p-oxazolyl benzene, 1,2-benzophenazin, retinol, bis-3-aminopyridinium salts, hellebrigcnin, tetracycline, sterophenol, benzimidazolylphenylamine, 2-oxo-3-chromen, indole, xanthen, 7-hydroxycoumarin, phenoxazine, calicylate, strophanthidin, porphyrins, triarylmethanes and flavin. Individual fluorescent compounds which have functionalities for linking to an element detectable by embodiments of the invention, or which can be modified to incorporate such functionalities, include but are not limited to, dansyl chloride; fluoresceins such as 3,6-dihydroxy-9-phenylxanthhydrol;
rhodamineisothiocyanate; N-phenyl 1-amino-8-sulfonatonaphthalene; N-phenyl 2-amino-6-sulfonatonaphthalene; 4-acetamido-4-isothiocyanato-stilbene-2,2'-disulfonic acid;
pyrene-3-sulfonic acid; 2-toluidinonaphthalene-6-sulfonate; N-phenyl-N-methyl-aminoaphthalene-6-sulfonate; ethidium bromide; stebrine; auromine-0,2-(9'-anthroyl)palmitate; dansyl phosphatidylethanolamine; N,N'-dioctadecyl oxacarbocyanine:
N,N'-dihexyl oxacarbocyanine; merocyanine, 4-(3'pyrenyl)stearate; d-3-aminodesoxy-equilenin; 12-(9'-anthroyl)stearate; 2-m ethylanthracene; 9-vinylanthracene;
2,2'(vinylene-p-phenylene)bisbenzoxazole; p-bis(2-(4-methyl-5-phenyl-oxazolyl))benzene; 6-dimethylamino-1,2-benzophenazin; retinol; bis(3'-aminopyridinium) 1, 1 0-decandiyl diiodidc; sulfonaphthylhydrazone of hellibrienin; chlorotetracycline; N-(7-dimethylamino-4-methyl-2-oxo-3-chromenyl)maleimide; N-(p-(2-benzimidazolyl)-phenyl)maleimide; N-(4-fluoranthyl)maleimide; bis(homovanillic acid);
resazarin; 4-chloro-7-nitro-2,1,3-benzooxadiazole; merocyanine 540; resorufin; rose bengal;
and 2,4-diphenyl-3(2H)-furanone.
Many fluorescent tags are commercially available, for example, from SIGMA
chemical company (Saint Louis, Mo.), Molecular Probes, R&D systems (Minneapolis, Minn.), Phannacia LKB Biotechnology (Piscataway, N.J.), CLONTECH Laboratories, Inc. (Palo Alto, Calif.), Chem Genes Corp., Aldrich Chemical Company (Milwaukee, Wis.), Glen Research, Inc., GIBCO BRL Life Technologies, Inc. (Gaithcrsberg, Md.), Fluka Chemica-Biochemika Analytika (Fluka Chemie AG, Buchs, Switzerland), and Applied Biosystems (Foster City, Calif.).
Desirably, fluorescent labels absorb light above about 300 nm, preferably about 350 nm, and more preferably above about 400 nm, usually emitting at wavelengths greater than about 10 nm higher than the wavelength of the light absorbed. It should be noted that the absorption and emission characteristics of the bound label may differ from the unbound label. Therefore, when referring to the various wavelength ranges and characteristics of the labels, it is intended to indicate the labels as employed and not the label which is unconjugated and characterized in an arbitrary solvent.
Fluorescent labels are detectable because by irradiating a fluorescent label with light, one can obtain a plurality of emissions. Thus, a single label can provide for a plurality of measurable events. Detectable signal may also be provided by chemiluminescent and bioluminescent sources.
Chemiluminescent sources include a compound which becomes electronically excited by a chemical reaction and may then emit light which serves as the detectible signal or donates energy to a fluorescent acceptor. A diverse number of families of compounds have been found to provide chemiluminescence under a variety or conditions.
One of compounds is 2,3-dihydro-1,4-phthalazinedione. The most popular compound is luminol, which is a 5-amino compound. Other members of the include the 5-amino-6,7,8-trimethoxy- and the dimethyl amino[ ca]benz analog. These compounds can be made to luminesce with alkaline hydrogen peroxide or calcium hypochlorite and base.
Another of compounds is the 2,4,5-triphenylimidazoles, with lophine as the common name for the parent product. Chemiluminescent analogs include para-dimethylamino and -methoxy substituents. Chem] luminescence may also be obtained with oxalates, usually oxalyl active esters, e.g., p-nitrophenyl and a peroxide, e.g., hydrogen peroxide, under basic conditions. Other useful chemiluminescent compounds are also known and available, including -N-alkyl acridinum esters (basic H202) and dioxetanes.
Alternatively, luciferins may be used in conjunction with luciferase or lucigenins to provide bioluminescence.
A label may be coupled directly or indirectly to a molecule to be detected (a product, substrate, enzyme, or the like) according to many methods. As indicated above, a wide variety of labels are used, with the choice of label depending on the sensitivity required, ease of conjugation of the compound, stability requirements, available instrumentation, and disposal provisions. Non-radioactive labels are often attached by indirect means. Generally, a ligand molecule (e.g., biotin) is covalently bound to a polymer. The ligand then binds to an anti-ligand (e.g., streptavidin) molecule which is either inherently detectable or covalently bound to a signal system, such as a detectable enzyme, a fluorescent compound, or a chemiluminescent compound. A number of ligands and anti-ligands can be used. Where a ligand has a natural anti-ligand, for example, biotin, thyroxine, and cortisol, it can be used in conjunction with labeled, anti-ligands. Alternatively, any haptenic or antigenic compound can be used in combination with an antibody. Labels can also be conjugated directly to signal generating compounds, e.g., by conjugation with an enzyme or fluorophore. Enzymes of interest as labels will primarily be hydrolases, particularly phosphatases, esterases and glycosidases, or oxidoreductases, particularly peroxidases. Fluorescent compounds include fluorescein and its derivatives, rhodamine and its derivatives, dansyl, umbelliferone, etc.
Chemiluminescent compounds include luciferin, and 2,3-dihydrophthalazinediones, e.g., luminol.
Means of detecting labels are dependent upon the type of label. Thus, for example, where the label is a radioactive label, a means for detection may include, but is not limited to, a scintillation counter or photographic film as in autoradiography. Where the label is a fluorescent label, a means for detection may include, but is not limited to, exciting the fluorochrome with the appropriate wavelength of light and detecting the resulting fluorescence, e.g., by microscopy, visual inspection, via photographic film, by the use of electronic detectors such as digital cameras, charge coupled devices (CCDs) or photomultipliers and phototubes, and the like. When using fluorescent labels and detection techniques, microscopy and spectroscopy techniques are often integrated into the detection system. Similarly, enzymatic labels may be detected by providing appropriate substrates for the enzyme and detecting the resulting reaction product.
Finally, simple colorimetric labels are often detected simply by observing the color associated with the label. For example, conjugated gold often appears pink, while various conjugated beads appear the color of the bead.
In one embodiment, a continuous flow screening system generates a constant signal which varies only when a test compound is introduced that affects the system.
Specifically, as the system components flow through the system, they will produce a relatively constant signal level at a detection zone. When test compounds (i.e., for example, infection control compositions) are periodically introduced into the channel and mixed with the system components, the test compounds cause a deviation from the constant signal level at the detection zone. This deviation may then be correlated to the particular test compound screened.
D. Detection Of Microbial Spore Surface Protein Binding To An Epidermal Protein In one embodiment, the present invention contemplates a method for detecting microbial spore surface protein binding to an epidermal layer. In one embodiment, the epidermal layer is derived from skin. In one embodiment, the epidermal layer is derived from an internal organ. In one embodiment, the epidermal layer is derived from a mucosal membrane. In one embodiment, the epidermal layer comprises a skin protein. In one embodiment, the skin protein comprises involucrin. In one embodiment, the skin protein comprises loricrin. In one embodiment, the skin protein comprises cytokeratin.
See, Figure 10.
Enzyme-linked immunosorbent assay plates were coated with 5 mg of human keratin in 100 p1 of PBS/well overnight at 4 C. Wells were then washed three times with PBS and then blocked with I% bovine serum albumin in PBS for 1 h before the addition of spores. Spores (C, difficile prepared as described above) were diluted to a concentration having an absorbance of 1.0 at 600 nm in PBS (approximately 5x107 spores/ml) before being labeled with FITC. 100 l of labeled spores were added per well, and the plates were incubated at 37 C for 1 h. The total fluorescence (Ft tai) per well was measured after a 1-h incubation using a Fluoroskan II fluorescence reader (Labsystems, Beverly, MA), with Xec = 485 nm and Xeii, = 535 nm. The wells were washed with PBS
three times to remove unbound spores and the remaining fluorescence (Ft,,,) measured.
Adherence was calculated as follows: adherence = Ftest/Ftotai. Adherence of labeled cells in the absence of antibodies or infection control compositions was normalized to 100%.
Bovine serum albumin-coated wells were used as negative controls. For inhibition assays, FITC-labeled spores were first incubated with anti- spore antibodies or infection control compositions for 1 h at 37 C before addition of the mixture to the keratin-coated wells.
Alternatively, EpiDermTM Skin Model plates (24-well or 96-well) were blocked with I% bovine serum albumin in PBS for I h before the addition of spores.
Spores (C.
difficile prepared as described above) were diluted to a concentration having an absorbance of 1.0 at 600 nm in PBS (approximately 5x107 spores/ml) before being labeled with FITC. 100 pi of labeled spores were added per well, and the plates were incubated at 37 C for I h. The total fluorescence (Ftotai) per well was measured after a 1-h incubation using a Fluoroskan II fluorescence reader (Labsystems, Beverly, MA), with Xex = 485 rim and k,,, = 535 rim. The wells were washed with PBS three times to remove unbound spores and the remaining fluorescence (FtCS1) measured. Adherence was calculated as follows: adherence = F1es1/Ft tai. Adherence of labeled cells in the absence of antibodies or infection control compositions was normalized to 100%. Bovine serum albumin-coated wells were used as negative controls. For inhibition assays, FITC-labeled spores were first incubated with anti- spore antibodies for 1 h at 37 C
before addition of the mixture to the EpiDermTM Skin Model plates.
E. High-Throughput Screening of Infection Control Compositions In one embodiment, the present invention contemplates methods comprising high throughput assays for screening of infection control compositions. For example, high throughput binding assays for proteins and/or nucleic acids have been developed; i) high throughput screening methods for proteins (U.S. Pat. No. 5,559,410); ii) high throughput screening methods for nucleic acid binding (i.e., in arrays) (U.S. Pat. No.
5,585,639); and iii) high throughput methods of screening for ligand/antibody binding (U.S.
Pat. Nos.
5,576,220 and 5,541,061)(all patents herein incorporated by reference). In addition, high throughput screening systems are commercially available (see, e.g., Zyunark Corp., Hopkinton, Mass.; Air Technical Industries, Mentor, Ohio; Beckman Instruments, Inc.
Fullerton, Calif.; Precision Systems, Inc., Natick, Mass., etc.). These systems typically automate entire procedures including all sample and reagent pipetting, liquid dispensing, timed incubations, and final readings of the microplate in detector(s) appropriate for the assay. These configurable systems provide high throughput and rapid start up as well as a high degree of flexibility and customization. The manufacturers of such systems provide detailed protocols the various high throughput. Thus, for example, Zymark Corp.
provides technical bulletins describing screening systems for detecting the modulation of gene transcription, ligand binding, and the like.
Conventionally, new chemical entities with useful properties are generated by identifying a chemical compound (called a "lead compound") with some desirable property or activity, creating variants of the lead compound, and evaluating the property and activity of those variant compounds. However, the current trend is to shorten the time scale for all aspects of drug discovery. Because of the ability to test large numbers quickly and efficiently, high throughput screening (HTS) methods are replacing conventional lead compound identification methods.

In one preferred embodiment, high throughput screening methods involve providing a library containing a large number of potential therapeutic compounds (candidate compounds). Such "combinatorial chemical libraries" are then screened in one or more assays, as described herein, to identify those library members (particular chemical species or subclasses) that display a desired characteristic activity. The compounds thus identified can serve as conventional "lead compounds" or can themselves be used as potential or actual therapeutics.

1. Combinatorial Chemical Libraries Recently, attention has focused on the use of combinatorial chemical libraries to assist in the generation of new chemical compound leads. A combinatorial chemical library is a collection of diverse chemical compounds generated by either chemical synthesis or biological synthesis by combining a number of chemical "building blocks"
such as reagents. For example, a linear combinatorial chemical library such as a polypeptide library is formed by combining a set of chemical building blocks called amino acids in every possible way for a given compound length (i.e., the number of amino acids in a polypeptide compound). Millions of chemical compounds can be synthesized through such combinatorial mixing of chemical building blocks. For example, a systematic, combinatorial mixing of 100 interchangeable chemical building blocks may result in the theoretical synthesis of 100 million tetrameric compounds or 10 billion pentameric compounds.
Combinatorial chemical libraries include, but are not limited to, peptide libraries.
U.S, Pat. No. 5,010,175, Furka et al., Int. JPept. Prot. Res., 37: 487-493 (1991); and Houghton et al., Nature 354:84-88 (1991 )(all references herein incorporated by reference). Peptide synthesis is by no means the only approach envisioned and intended for use with the present invention. Other chemistries for generating chemical diversity libraries can also be used. Such chemistries include, but are not limited to:
peptoids (PCT
Publication No WO 91/19735, 26 Dec. 1991), encoded peptides (PCT Publication WO
93/20242, 14 Oct. 1993), random bio-oligomers (PCT Publication WO 92/00091, 9 Jan.
1992), benzodiazepines (U.S. Pat. No. 5,288,514)(herein incorporated by reference), diversomers such as hydantoins, benzodiazepines and dipeptides (Hobbs et al., Proc. Nat.
Acad. Sci. USA 90:6909-6913 (1993), vinylogous polypeptides (Hagihara et al., JAmer.
Cliena. Soc. 114:6568 (1992), nonpeptidal peptidomimetics with a Beta-D-Glucose scaffolding (Hirschmann et al., J. Amer. Chem. Soc. 114:9217-9218 (1992), analogous organic syntheses of small compound libraries (Chen et al., J. Amer. Chem.
Soc.
116:2661 (1994), oligocarbamates (Cho et al., Science 261:1303 (1993), and/or peptidyl phosphonates (Campbell et al., J. Org. Chem. 59 658 (1994); nucleic acid libraries, peptide nucleic acid libraries (U.S. Pat. No. 5,539,083)) antibody libraries (Vaughn et al., Nature Biotechnology 14(3): 309-314 (1996), and PCT/US96/10287), carbohydrate libraries (Liang et al. Science 274:1520-1522 (1996), and U.S. Pat. No.
5,593,853), and small organic molecule libraries - benzodiazepines (Baum C&EN, January 18, page 33 (1993); isoprenoids (U.S. Pat. No. 5,569,588)(herein incorporated by reference);
thiazolidinones and metathiazanones, U.S. Pat. No. 5,549,974; pyrrolidines, U.S. Pat.
Nos. 5,525,735 and 5,519,134; morpholino compounds, U.S. Pat. No. 5,506,337;
benzodiazepines, 5,288,514; and the like)(all references herein incorporated by reference).
Devices for the preparation of combinatorial libraries are commercially available (See, e.g., 357 NIPS, 390 NTS, Advanced Chem Tech, Louisville Ky., Symphony, Rainin, Woburn, Mass., 433A Applied Biosystems, Foster City, Calif., 9050 Plus, Millipore, Bedford, Mass.).
A number of robotic systems have also been developed for solution phase chemistries. These systems include automated workstations like the automated synthesis apparatus developed by Takeda Chemical Industries, LTD. Osaka, Japan) and many robotic systems utilizing robotic arms (Zymate 11, Zymark Corporation, Hopkinton, Mass.; Orca, Hewlett-Packard, Palo Alto, Calif.) which mimic the manual synthetic operations performed by a chemist. Any of the above devices are suitable for use with the present invention. In addition, numerous combinatorial libraries are themselves commercially available. ComGenex, Princeton, N.J.; Asinex, Moscow, Ru; Tripos, Inc., St. Louis, Mo.; ChemStar, Ltd, Moscow, RU; 3D Pharmaceuticals, Exton, Pa.; or Martek Biosciences, Columbia, Md..
2. High Throughput Assays of Chemical Libraries Any of the assays for compounds inhibiting the attachment of bacterial spores as described herein are amenable to high throughput screening. As described above, in a preferred embodiment, the assays screen for agents that interact with a microbial spore surface protein.
High throughput implementation of the assays described herein can be implemented with, at most, routine modification of the assays format (e.g., for compatibility with robotic manipulators, large plate readers, and the like).
Various high throughput screening systems (e.g., for protein binding, nucleic acid binding, etc.) are described. U.S. Pat. Nos. 5,559,410, 5,585,639, 5,576,220, and 5,541,061 (all herein incorporated by reference).
In addition, high throughput screening systems are commercially available.
Zymark Corp., Hopkinton, Mass.; Air Technical Industries, Mentor, Ohio;
Beckman Instruments, Inc. Fullerton, Calif.; or Precision Systems, Inc., Natick, Mass.
These systems typically automate entire procedures including all sample and reagent pipetting, liquid dispensing, timed incubations, and final readings of the microplate in detector(s) appropriate for the assay. These configurable systems provide high throughput and rapid start up as well as a high degree of flexibility and customization. The manufacturers of such systems provide detailed protocols the various high throughput. Thus, for example, Zymark Corp. provides technical bulletins describing screening systems for detecting the modulation of gene transcription, ligand binding, and the like.
V. Detection Of Infection Control Test Compounds In one embodiment, the present invention contemplates a method for detecting and/or quantifying of an amount, or rate, of interaction between a microbial spore surface protein and an infection control test compound. In some embodiments, detecting methods include, but are not limited to, video microscopy; DAPI fluorescence changes, fluorescence resonance energy transfer or centrifugation.
A. Microscopy In one embodiment, an infection control test compound is detected by microscopy (i.e., for example, by direct visual observation or using a video or photographic recording device). Assays involving microscopic visualization may utilize Clostridium spore surface peptides and/or another microbial spore surface protein comprising a labeled (e.g., fluorescently labeled). In one embodiment, the peptides and/or proteins are placed adjacent to a solid support (e.g., a glass slide), and exposed to a solution containing an infection control test compound. Attachment of the peptides and/or proteins to the solid support can be directly visualized using a microscope (i.e., for example, a scanning electron microscope).
A microscope can optionally be equipped with a still camera or a video camera and may be equipped with image acquisition and analysis software to quantify the relative abundance of protein and/or peptide attachment.

Any label that can be visualized in a microscope can be used in conjunction with this type of detection. Such labels include, but are not limited to, fluorescent labels (e.g., fluorescein, rhodamine, etc.), calorimetric labels, and radioactive labels (with appropriate scintillation screen), and the like. In some embodiments of the assays of this invention, the microtubules can be visualized without any label (e.g., via differential interference contrast microscopy).
B. DAPI Fluorescence Changes.
In another embodiment, the interaction of an infection control test compound with a Clostridium spore surface peptide and/or another microbial spore surface protein can be determined by changes in fluorescence utilizing a label comprising DAPI. A
decreased rate or amount in DAPT fluorescence is indicative of an interaction between a test compound and a microbial spore surface protein. A change in fluorescence with a test compound is compared to that observed in a negative and/or positive control reaction.
Other labels that can be used include, but are not limited to, anilinonapthalene sulfonate (ANS) (e.g., Molecular Probes Catalogue Nos: A-47, A-50, T-53, etc.), bis-ANS (Molecular Probes Catalogue No: B-153), N-phenyl-l-naphthylene (NPN) (Molecular Probes Catalogue No: P65), 1CVJ (Molecular Probes Catalogue No: D-3923), ruthenium red, and cresol violet.
C. Fluorescence Resonance Energy Transfer The degree of interaction between a Clostridium spore surface peptide and/or another microbial spore surface protein and an infection control test compound can also be determined by fluorescent resonance energy transfer (FRET). Fluorescence resonance energy transfer, a phenomenon that occurs when two fluorophores with overlapping absorption and emission spectra are located close together (e.g., <7 mm apart). Stryer et al., Ann. Rev. Biochem., 47:819-846 (1978). In one embodiment, the present invention contemplates detecting protein-protein interactions using FRE"I'. Taylor et al., J. Cell Biol. 89:362-367 (1981).
In one embodiment, equimolar proportions of differently labeled microbial spore surface peptide and an infection control test peptide compound are combined (e.g., fluorescein labeled and rhodamine-labeled, respectively). Fluorescence may be quenched upon the interaction of the microbial spore surface peptide with the peptide test compound, indicating that the respective bound fluorochromes come in close enough proximity for energy transfer to occur. The rates and/or amount of fluorescence generated by a reaction with a test compound and a control can be compared.

VI. Recombinant Expression Of Microbial Spore Surface Proteins And Peptides In one embodiment, Clostridium spore surface peptides and/or another microbial spore surface proteins are synthesized using recombinant DNA methodology.
Generally this involves creating a DNA sequence that encodes a protein, placing the DNA
in an expression cassette under the control of a particular promoter, expressing the protein in a host, isolating the expressed protein and, if required, renaturing the protein. DNA
encoding a peptide and/or protein as contemplated by this invention can be prepared by many suitable methods, including, for example, cloning and restriction of appropriate sequences or 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: 1859-1862 (1981); and the solid support method of U.S. Pat. No.
4,458,066 (all above reference herein incorporated by reference in their entirety).
Generally, the nomenclature used hereafter and the laboratory procedures in cell culture, molecular genetics, and nucleic acid chemistry and hybridization described below will be understood by those having ordinary skill in the art. Standard techniques are used for recombinant nucleic acid methods, polynucleotide synthesis, and microbial culture and transformation (e.g., electroporation, lipofection). Generally, enzymatic reactions and purification steps are performed according to the manufacturer's specifications supplied with commercial kits. The techniques and procedures are generally performed according to conventional methods in the art and various general references. Sambrook et al., In:
Molecular Cloning: A Laboratory Manual, 2d ed. Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., (1989); and In: Current Protocols in Molecular Biology John Wiley and Sons, Inc., N.Y. (1996), which are incorporated herein by reference in their entirety).
The present invention contemplates any nucleic acid sequence which encodes a spore surface polypeptide sequence or its variants; these nucleic acid sequences are used to make recombinant molecules which express a spore surface protein. For example, the redundancy of the genetic code permits many nucleic acid sequences that encode a spore surface polypeptide. Thus, codons which are different from SEQ ID NOs: 2, 4, 6 or 8 may be used to increase the rate of expression of a spore surface nucleotide sequence.
Additionally, alternative codons may also be used in eukaryotic expression hosts to generate splice variants of recombinant RNA transcripts which have other properties (i.e., for example, longer or shorter half-life). In addition, different codons may also be desirable for the purpose of altering restriction enzyme sites or, in eukaiyotic expression hosts, of altering glycosylation patterns in translated polypeptides.
Variants of microbial spore surface protein nucleotide sequences (i.e., for example, a Clostridium spore surface protein) are also included within the scope of this invention. These variants include, but are not limited to, nucleotide sequences having deletions, insertions or substitutions of different nucleotides or nucleotide analogs as long as the biological activity of the translation product of the nucleotide sequence is maintained. This invention is not limited to the Clostridium spore surface protein nucleic acid sequences (i.e., for example, SEQ ID NO: 2) but specifically includes nucleic acid homologues which are capable of hybridizing to SEQ ID NO: 2 (i.e., for example, Bacillus homologues; SEQ ID NOs: 3, 5, and 7), and to portions, variants and derivatives thereof, under different hybridization stringencies. For example, higher stringencies may reduce or eliminate nonspecific binding between SEQ ID NO:2 and other nucleic acid sequences. Alternatively, lower stringencies may detect a larger number of nucleic acid sequences having different homologies to SEQ ID NO:2. Fragments of SEQ ID NO:2 (i.e., also referred to as a portion) are also specifically contemplated to be within the scope of this invention. In one embodiment, a fragment has a length equal to or greater than 10 nucleotides and shows greater than 50% homology to SEQ ID NO:2.
The present invention further contemplates antisense molecules comprising the nucleic acid sequence complementary to at least a portion of a polynucleotide of SEQ ID
NO:2 or at least a portion of a polynucleotide encoding a spore surface protein. The present invention further contemplates a fusion gene comprising at least a portion, variant, derivative and/or homologue of a nucleic acid selected from the group comprising SEQ ID NO:2 or a polynucleotide encoding a spore surface protein ligated to one or more heterologous sequences. Although it is not necessary to understand the mechanism of an invention, it is believed that such a fusion gene may detect expression of nucleic acid sequences. Examples of a heterologous sequence include, but are not limited to, a reporter sequence encoding enzymes such as P-galactosidase or luciferase. Fusion genes may also facilitate purification of the expressed protein. For example, a heterologous sequence of protein A allows purification of a fusion protein on an immobilized immunoglobulin. Other affinity traps can also be utilized to purify expressed fusion protein. For example, pGEX vectors (Promega, Madison, Wis.) may be used to express a spore surface protein as a fusion protein with glutathione S-transferase (GST). In general, such fusion proteins are soluble and can easily be purified from lysed cells by adsorption to glutathione-agarose beads followed by elution in the presence of free glutathione. Proteins made in such systems are designed to include heparin, thrombin, or factor XA protease cleavage sites so that the cloned polypeptide of interest can he released from the GST moiety at will.
A. Recombinant Expression Of Representative Spore Surface Proteins In one embodiment, the present invention contemplates a method for expression recombinant microbial spore surface proteins. In one embodiment, the method comprises constructing a plasmid encoding at least a portion of a microbial spore surface protein. In one embodiment, the method comprises transfecting a suitable organism with the plasmid. In one embodiment, the suitable organism is F. coli.
The data presented herein demonstrates the successful cloning of a transfected E.
coli with a plasmid comprising nucleic acids encoding at least a portion of a C. difficile spore surface protein. In one embodiment, the spore surface protein is CD1067.
In one embodiment, the spore surface protein is CD3620. The following experiment is merely intended to be illustrative, and is not intended to be limiting, of the present embodiment as similar results may be obtained with other spore surface protein nucleic acid constructs using the basic technique described below.
Cloning of C. difficile suspected surface proteins was performed in E. coli Rosetta-Gami expression strain via an pET-19b plasmid (Novagen, product number 69677-3) having a ten (10) amino acid His tag.. Specifically, pET-19b+CD1067 (pJEB02) orpET-19b+CD3620 (pJEB03) was incubated in an overnight culture (i.e., for example, 12 hr) comprising E. coli Rosetta-Gami cells. See, Figure 11.
Approximately 2 ml of the transfected E. coli cell culture was transferred to an expression culture medium comprising 50 ml Luria Broth (LB) + 100 g/ml adenosine monophosphate (AMP) and 34 gg/ml chloramphenical. After the culture was grown to approximately OD625 to 0.5-0.6, expression of the pET plasmid was induced with 1 mM
IPTG. Following IPTG induction, I ml medium samples were taken at approximate one hour intervals. The samples were centrifuged (i.e., for example, approximately 10,000 x g for 1 min), and the resulting pellet was resuspended in 100 l Novex solution (45 12x SDS-PAGE Sample Buffer + 45 p1 H2O + 10 12M dithiothreitol) and boiled for approximately 5 minutes. Approximately 20 pJ of the boiled pellet suspension was initially run on a 4-12% Tris-Glycine SDS-PAGE gel, whereupon approximately 10 l of the suspected isolated recombinant protein was used for a Western Blot analysis.
The pJEB02 plasmid was expected to express a protein having 405 amino acids with an approximate molecular weight of 44 kDa. SDS-PAGE gel electrophoresis separation confirms this expectation showing that expression of a protein of approximately 50 kDa appeared within one hour after IP'l'G induction and increased in intensity in subsequent samples (i.e., 2 - 6 hour samples). See, Figure 12.
This preliminary separation was confirmed using Western Blot analysis detected by either F1373 anti-C. difficile antibody or anti-His antibody. See, Figure 13A and Figure 13B, respectively.
The pJEB03 plasmid was expected to express a protein having 122 amino acids with an approximate molecular weight of 14 kDa. SDS-PAGE gel electrophoresis separation confirms this expectation showing that expression of a protein of approximately 20 kDa appeared within one hour after IPTG induction and increased in intensity in subsequent samples (i.e., 2 - 4.5 hour samples). See, Figure 14.
This preliminary separation was confirmed using Western Blot analysis detected by either F1373 anti-C. difficile antibody or anti-His antibody. See, Figure 15A and Figure 15B, respectively.
Preimmune Western Blot control samples for both the F 1373 anti-C, difficile antibody or the F1997 anti-C. difficile antibody (e.g., commercial customized preparations directed against spore surfaces; Strategic Diagnostics, Inc.) demonstrated an absence of binding specificity in both pJEB02 and pJEB03 expression profiles.
See, Figure 16A-D. These data show that when CD1067 and CD3620 are cloned into E.
coli, a preimmune control detection is not reactive with either a recombinant CD
1067 protein or a recombinant CD3620 protein. In contrast, a C. difficile spore surface antibody (i.e., for example, F1373 or F1997) reacts specifically with a recombinant CD1067 protein and a recombinant CD3620 protein. Specific expression of these proteins was confirmed by the detection of a Hisip tag attached to the expression plasmid construct that corresponds with the MW of both a recombinant CD1067 protein and a CD3620 recombinant protein.
B. Nucleic Acid Sequence Synthesis Techniques Synthetic chemistry techniques may also be used to generate nucleic acid sequences contemplated by the present invention. Oligonucleotides can be synthesized on an Applied BioSystems oligonucleotide synthesizer according to specifications provided by the manufacturer. Sinha et al., Nucleic Acids Res. 12:4539 (1984).

Following synthesis, complementary oligonucleotides can be annealed by heating them to 90 C in a solution of 10 mM Tris-HCI buffer (pH 8.0) containing NaCl (200 mM) and then allowing them to cool slowly to room temperature. For binding and turnover assays, duplex DNA is purified from native polyactylamide (15% w/v) gels. The band corresponding to double-stranded DNA is excised and soaked overnight in 0.30 M
sodium acetate buffer (pH 5.0) containing EDTA (I mM). After soaking, the supernatant is extracted with phenol/chloroform (1/1 v/v) and precipitated with ethanol.
DNA
substrates are radiolabeled on their 5'-OH group by treatment with 32P-ATP and polynucleotide kinase. Salts and unincorporated nucleotides are removed by chromatography on Sephadex G columns.
A synthesized nucleotide sequence may be confirmed using commercially available kits comprising enzymes including, but not limited to, Klenow fragment of DNA polymerase I, Sequenase*, Taq DNA polymerase, or thermostable T7 polymerase.
Capillary electrophoresis may also be used to analyze a nucleic acid sequence size and confirm sequences nucleic acid synthesis products. Synthesized sequences may also be amplified by: i) polymerase chain reaction (PCR) (Mullis et al., U.S. Pal. No.
4,683,195;
and Mullis et al., U.S. Pat. No. 4,683,202, the ligase chain/amplification reaction (LCR/LAR); Barany et al., Proc. Natl. Acad. Sci., 88:189 (1991); Barany et al., PCR
Methods and Applic., 1:5 (1991); and Wu et al., Genomics 4:560 (1989).
A spore surface protein nucleotide sequence of the present invention may be used in a variety of ways. For example, fragments of a sequence of at least about 10 bp, more usually at least about 15 bp, and up to and including the entire (i.e., full-length) sequence can be used as probes for the detection and isolation of complementary genomic DNA
sequences from Bacillus, Clostridia, as well as other bacteria. Genomic sequences are isolated by screening genomic library containing bacterial DNA with all or portion of a spore surface protein nucleic acid sequence. In addition to screening genomic libraries, a spore surface protein nucleic acid sequence can also be used to screen cDNA
libraries made using bacterial RNA. A spore surface protein nucleic sequence is also useful in directing the synthesis of a spore surface protein. spore surface protein antibodies may also be used to antagonize the interaction of spore surface proteins to mammalian cells, mammalian skin, hard surfaces etc. A spore surface protein finds use in producing a spore surface protein antibodies for diagnostic purposes such as detecting infections with bacteria which express a spore surface protein. A spore surface protein nucleic acid sequence is also useful for the screening of binding antagonists and the detection of microbes which contain spore surface proteins.
Chemical synthesis usually 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. Chemical synthesis of DNA is routinely performed for sequences of about 100 bases, but longer sequences may be obtained by the ligation of shorter sequences.
Alternatively, subsequences may be cloned and the appropriate subsequences cleaved using appropriate restriction enzymes. The fragments may then be ligated to produce the desired DNA sequence.
In one embodiment, a microbial spore surface protein nucleic acid can be cloned using DNA amplification methods such as polymerase chain reaction (PCR). Thus, for example, a nucleic acid sequence or subsequence is PCR amplified, using a sense primer containing one restriction site (e.g., NdeI) and an antisense primer containing another restriction site (e.g., Hindlil). This will produce a nucleic acid encoding the desired protein comprising terminal restriction sites. This nucleic acid can then be easily ligated into a vector containing a nucleic acid encoding the second molecule and having the appropri ate corresponding restriction sites.
Suitable PCR primers can be determined using the sequence information provided herein. Appropriate restriction sites can also be added to the nucleic acid encoding the microtubule depolymerizing or severing proteins by site-directed mutagenesis.
The plasmid containing the microtubule depolymerizing or severing protein encoding nucleic acid is cleaved with the appropriate restriction endonuclease and then ligated into the vector encoding the second molecule according to standard methods.
Nucleic acid sequences encoding Clostridium spore surface peptides and/or other microbial spore surface proteins may be expressed in a variety of host cells, including E.
coli, other bacterial hosts, yeast, and various higher eukaryotic cells such as the COS, CHO and HeLa cells lines and myeloma cell lines. A recombinant protein gene will usually be operably linked to appropriate expression control sequences for each host. For E. coli, preferred promoters include, but are not limited to, T7, trp, or lambda promoters.
Further, a ribosome binding site and a transcription termination signal is part of the expression vector. For eukaryotic cells, the control sequences will include a promoter and preferably an enhancer derived from immunoglobulin genes, SV40, cytomegalovirus, etc., and a polyadenylation sequence, and may include splice donor and acceptor sequences.
The plasmids of the invention can be transferred into the chosen host cell by methods including, but not limited to, calcium chloride, calcium phosphate, or electroporation. Cells transformed by the plasmids can be selected by resistance to antibiotics conferred by genes contained on the plasmids, such as the amp, gpt, neo and hyg genes.
Once expressed, the recombinant proteins can be purified according to standard procedures including, but not limited to, ammonium sulfate precipitation, affinity columns, column chromatography, gel electrophoresis and the like. R. Scopes, In: Protein Purification, Springer-Verlag, N.Y. (1982); and Deutscher et al., In: Methods in Enzymology Vol. 182 (1990). Substantially pure compositions of at least about 90 to 95% homogeneity are preferred, and 98 to 99% or more homogeneity are most preferred.
Once purified, partially or to homogeneity as desired, the polypeptides may then be used (e.g., as immunogens for antibody production).
VII. Clostridium Infections Clostridium difficile, a gram-positive anaerobic bacterium, frequently causes morbidity in hospitalized patients and is believed an etiological agent of antibiotic-associated diarrhea and pseudomembranous colitis. Borriello et al., "Virulence factors of Clostridium difficile" Rev. Infect. Dis. 12:5185-S191 (1990); Cerquetti et al., "Characterization of surface layer proteins from different Clostridium difficile clinical isolates" Microb. Pathog. 28:363-372 (2000); and Kuipers et al., "Quorum sensing-controlled gene expression in lactic acid bacteria" J. Biotechnol. 64:15-21 (1998). Two factors have been evaluated for their involvement in the pathogenesis of these conditions:
i) the suppression of the resident intestinal flora by the administration of antibiotics (Freeman et al., "Antibiotics and Clostridium difficile" Microbes Infect.
1:377-384 (1999); and George W., "Antimicrobial agent-associated colitis and diarrhea:
historical background and clinical aspects" Rev. Infect. Dis. 6:S208-S213 (1984); and ii) the production by the bacterium of two high-molecular-weight (MW) toxins (toxins A
and B) (Pothoulakis et al., "Microbes and microbial toxins: paradigms for microbial-mucosal interactions. II. The integrated response of the intestine to Clostridium difficile toxins"
Am. J. Physiol.-Gastroint. Liver Physiol. 280:G178-G183 (2001); and Wren B., "Molecular characterisation of Clostridium difficile toxins A

and B" Rev. Med. Microbiol. 3:21-27 (1992).
As with other cnterotoxigenic pathogens, delivery of toxins follows C.
difficile colonization of the gut, which requires bacterial adherence to the mucosa. In a Syrian hamster model of clindamycin-induced disease, a correlation was reported between variable efficiencies of gut colonization by different C. difficile strains and their abilities to associate with regions of the gastrointestinal tract spanning from the jejunum to the colon. At least part of the variation was suggested to be due to factors other than toxin production. However, the adherence of a poorly virulent strain to the small bowel mucosa was increased by the administration of a crude toxin preparation, possibly due to the unmasking of receptor sites following cell damage.
Two virulence factors from C. difficile have been shown to be exotoxins, toxin A
(TcdA) and toxin B (TcdB) 3, little is known about other factors involved in the adherence and colonization processes. Voth et al., "Clostridium difficile toxins:
mechanism of action and role in disease" Clin. ,Mcrobiol. Rev. 18, 247-263 (2005). This is in contrast to other enteric organisms such as Escherichia coli, Salmonella, and Shigella spp. Possibly, this might be partly explained by the lack of well-developed mutagenesis systems for studying C. difficile. One approach to possibly understanding C.
difficile involves identifying the genomic locations of potential factors involved in pathogenicity. The complete genome sequence of C. difficile strain 630 (epidemic type X; virulent and multidrug resistant) has been reported. Wust et al., "Investigation of an outbreak of antibiotic-associated colitis by various typing methods" Clin.
Microbiol 16:1096-1101 (1982).
The genome of C. difficile strain 630 comprises a circular chromosome of 4,290,252 bp and a plasmid, pCD630, of 7,881 bp. The chromosome encodes 3,776 predicted coding sequences (CDSs); in common with other low-G+C content Gram-positive bacteria, the chromosome has a strong coding bias, with 82.1 % of the CDSs encoded on the leading strand. The plasmid carries 11 CDSs, none of which has any obvious function. Reciprocal FASTA analysis against the four sequenced clostridial genomes, C. acetobutylicum, C. botulinum, C. peifsringens, and C tetani, showed that only 567 (15%) of C difficile CDSs are shared with all the sequenced clostridia, whereas 1,893 (50%) arc unique to C. difficile. The conserved clostridial CDSs mainly encode essential functions, whereas the C. difficile unique CDSs encode many accessory functions and mobile elements. Sebaihia et al., "The multi-drug-resistant human pathogen Clostridium difficile has a highly mobile, mosaic genome" Nature Genetics 38:779-786 (2006).

Binding of purified C. difficile surface layer proteins (SLPs) has been observed in both human epithelial cells and gastrointestinal tissues. Calabi et al., "Binding of Clostridium difficile surface layer proteins to gastrointestinal tissues"
Infect Immun 70:5770-5778 (2002). Binding to both cells and tissues is observed with acid-extracted SLPs and with the soluble recombinant high-MW subunit. In contrast, much less or no less binding is observed with a soluble recombinant low-MW SLP subunit, expressed and purified under the same conditions as a recombinant high-MW SLP subunit.
Because an anti-high-MW subunit serum partially blocked adherence of C. difficile to HEp-2 cells it is possible that SLPs are involved in the adherence of whole bacteria to epithelial cells. It is believed that the natural conformation of these proteins are retained during the adhesion because; i) both native and recombinant SLP preparations retained peptidoglycan hydrolase activities; ii) acid-extracted SLPs are capable of polymerizing into higher order structures upon the addition of calcium; and iii) antisera raised against both recombinant subunits react at high titers with whole C. difficile cells.
Clostridium perfringens is suspected to cause gas gangrene and is a severe form of gangrene (tissue death; also referred to as necrotizing subcutaneous infection. Gas gangrene can also result from infections from Streptococcus spp. and Vibrio vulnificus.
Gas gangrene usually occurs as a result of infection with a Clostridium perfringens bacteria that, under anaerobic (low oxygen) conditions, produce toxins that cause tissue death and associated symptoms. In the United States, gas gangrene is rare, with only 1,000-3,000 cases yearly. Gas gangrene generally occurs at the site of trauma or a recent surgical wound but may also occur spontaneously. Patients who develop gas gangrene spontaneously often have a high risk factor including, but not limited to, an underlying blood vessel disease (i.e., for example, atherosclerosis or hardening of the arteries), diabetes, or colon cancer.
The onset of gas gangrene is generally sudden and dramatic. Inflammation begins at the site of infection as a pale-to-brownish-red and extremely painful tissue swelling.
Gas may be felt in the tissue as a crackly sensation when the swollen area is pressed with the fingers. The edges of the infected area expand so rapidly that changes are visible over a few minutes. The involved tissue is completely destroyed.
Clostridium perfringens bacteria produce many different toxins, four of which (i.e., for example, alpha, beta, epsilon, and iota) can cause potentially fatal syndromes. In addition, they cause tissue death (i.e., for example, necrosis), destruction of blood cells (i.e., for example, hemolysis), local decrease in circulation (i.e., for example, vasoconstriction), and leaking of the blood vessels (i.e., for example, increased vascular permeability).
Clostridium perfringens toxins are responsible for both the local tissue destruction and the systemic symptoms (the other symptoms that occur throughout the body).
Systemic symptoms develop early in a gas gangrene infection and include, but are not limited to, sweating, fever, and anxiety. If left untreated, the subject develops a shock -like syndrome including, but not limited to, decreased blood pressure (i.e., for example, hypotension), kidney failure, coma, and finally death. Other gas gangrene symptoms may include, but are not limited to, moderate to severe pain around a skin injury, progressive swelling around a skin injury, moderate to high fever, skin color initially pale, later dusky progressing to dark red or purple, jaundice, vesicle formation, coalescent vesicles combining into large blisters, blisters filled with brown-red fluid, drainage from the tissues, foul-smelling brown-red or bloody fluid (i.e., for example, serosanguineous discharge), increased heart rate (i.e., for example, tachycardia), sweating, subcutaneous emphysema (i.e., for example, air under the skin), or crepitus (i.e., for example, air in the tissues).
Alternatively, the subject may be in shock comprising general pallor, cold extremities, low blood pressure, or rapid heart rate. Ultimately a systemic infection may develop involving the entire body (i.e., for example, systemic toxicity or sepsis).
Other methods of determining the presence of gas gangrene include, but are not limited to, a grain stain of fluid from the infected area may shows Gram-positive rods (i.e., for example, a Clostridium species); a culture may grow the bacteria causing the infection;
blood cultures may grow the infecting bacteria; an anaerobic tissue and/or fluid culture may reveal at least on Clostridium species; or an X-ray, computerized tomography scan, or a magnetic resonance image of the affected area may show gas in the tissues Treatment of gas gangrene generally involves a prompt surgical removal of dead, damaged, and infected tissue (i.e., for example, debridement). Amputation of an arm or leg maybe indicated to control the spread of infection. Conventional antibiotics (i.e., for example, of the penicillin ) may be intravenously administered as an initial bolus dose followed by a long term oral regimen.

VIII. Bacillus Infections Anthrax is a disease that was first described over a century ago and one of the first diseases for which a microbial cause was also determined. The infection is propagated by spores of Bacillus anthracis, which can penetrate into the mammalian host by ingestion, inoculation, or inhalation. The latter mode of infection is the most severe, with rapid progression of the disease and a mostly fatal outcome. Dixon et al., N. Engl.
J. Med.
341, 815 --826 (1999). The spore is a donnant cell form of some microorganisms characterized by high resistance to chemicals and to environmental factors, such as heat and drought, which allow the organism to persist in the environment for decades.
After penetrating host tissues, B. anthracis spores germinate into the vegetative, lethal form.
The early stages of B. anthracis infection and the role of the host defenses in the lung are still unclear. A study by confocal microscopy shows that spore germination takes place within the macrophages, where the bacilli start producing toxins, including the lethal factor, protective antigen, and edema factor. Guidi-Rontani et al., Mol.
Microbiol. 31:9-17 (1999). Production of these toxins leads to macrophage apoptosis and the release of vegetative bacilli into the bloodstream, causing septicemia and death. Dixon et al., Cell.
Microbiol. 2:453-463 (2000).
On the other hand, macrophages appear to protect animals against infection due to inhalation of the spores by eliminating them. Cote et al., Microb. Pathog.
37:169-175 (2004). Another protective barrier that B. anthracis spores will encounter upon inhalation by the host is the epithelial surface of lung alveoli, which is coated with a thin layer of pulmonary surfactant. Pulmonary surfactant is a lipid-protein complex that plays two roles by: i) mechanically reducing the surface tension in the alveolus (which prevents it from collapse during expiration); and ii) providing a lung defense against infections.
McCormack et al., J. Clin. Invest. 109:707-712 (2002). This immune function is attributed to surfactant proteins (SPs) including, but not limited to, SP-A
and SP-D. SP-A
and SP-D are hydrophilic members of the collection that can interact with pathogen surfaces, promote their destruction, and thus prevent infections. Crouch et al., Annu. Rev.
Physiol. 63:521-554 (2001). Two other surfactant proteins, SP-B and SP-C, are highly hydrophobic, the latter having been shown to bind bacterial lipopolysaccharides and modulate their cellular effects. Augusto et al., J Biol. Chem. 277:23484-23492 (2002);
and Augusto et al., Am. J. Respir. Crit. Care Med. 168:335-341 (2003).
IX. Prophylactic And Therapeutic Uses A. Hospital Decontamination Hospital pathogens (i.e., for example, C. difficile) are responsible for the vast majority of bacterial infections in both hospital staff and the resident patient population.
Such infections may cause symptoms including, but not limited to, severe diarrhea but may also result in death following the infection of a compromised patient (i.e., for example, an immunocompromised patient). Consequently, improved antiseptic maintenance of both the hospital facility and staff should facilitate patient recovery and reduce overall health care costs due to shorter hospitalization durations.
Some bacteria (i.e., for example, C. difficile) produce spores which have proved highly resistant to common skin and surface disinfectants. Currently available cleansers for both hospital facility surfaces and hospital staff skin (i.e., epidermal layers) are ineffective for killing and/or removing bacterial spores. Anaerobic bacteria (i.e., for example, C. difficile) require specialized equipment and handling techniques to work with and manipulate. Consequently, there are currently no easy ways to develop an anaerobic bacteria specific "spore removal handwash".
In fact C. difficile is responsible for 15-25% of all cases of antibiotic-associated diarrhea. Recurrences of this type of diarrhea is a serious, difficult, and still unresolved management problem and increase both the length and overall cost of hospitalization.
Most studies demonstrate that hospital recurrences of C. difficile infections are split 50:50 between a reinfection with the same, or different, bacterial subtype.
Adequately controlling these types of recurrences depends upon the quality of antimicrobial procedures such as handwashing, environmental decontamination, and enteric isolation.
Contamination of the environment and persistence of spores have been demonstrated and implicated in cross infections. Barbut et al., "Epidemiology of recurrences or reinfections of Clostridium d fficile-Associated diarrhea" J Clin Microbiol 38:2386-2388 (2000).
In one embodiment, the present invention contemplates a composition comprising at least one outermost protein component of an anaerobic bacterial spore. In one embodiment, the anaerobic bacteria comprises a C. difficile spore. In one embodiment, the protein component is derived from an exosporium layer. In one embodiment, the protein component comprises a spore surface protein. Although it is not necessary to understand the mechanism of an invention, it is believed that the outermost protein component of anaerobic bacterial spore directs the attachment of the spores to tissues of an organism (i.e., for example, skin or gut) or to inanimate hard surfaces (i.e., for example, stainless steel, granite, leather, vinyl, ceramic etc).

In one embodiment, the present invention contemplates a method comprising screening for compositions capable of disrupting attachment interactions between anaerobic spore proteins and tissues or inanimate surfaces. In one embodiment, an outermost protein component of an anaerobic bacterial spore serves as a molecular target for screening such compositions.
A spore surface protein derived from a C. difficile exosporium can be expressed and purified by standard methods (supra). Consequently, using purified and isolated spore surface protein, simple screening assays may be used to develop anaerobic spore removing products (e.g., handwashes, inanimate surface cleansers, whole body decontamination solutions) without the need to work with the intact C.
difficile organism.
This eliminates the above mentioned need for large scale anaerobic equipment and effectively miniaturizes the screening process.
B. Emergency First Responders In one embodiment, the present invention contemplates prophylactic and therapeutic treatment of emergency personnel responding to microbial contamination (i.e., for example, first responders). In one embodiment, emergency first responders may include, but are not limited to, emergency medical technicians (EMTs), fireman, firewomen, policeman, policewomen, journalists, police dogs, or police horses.
The collection and identification of unknown substances can be a dangerous and generally time consuming procedure. In particular, microbial analysis typically requires specialized equipment in a laboratory. With the recent increase in global terrorism where there is an increased high risk of encountering toxic substances (i.e., for example anthrax) there is a need for quickly preventing and/or treating emergency first responder contamination. Further, screening and/or testing protocols should be readily available without specialized equipment.
With the recent acceleration of terrorist activities around the world, first responders need a readily available composition comprising compounds that provide specific protection against microbial infection. The need for such protection for emergency first responders extends beyond terrorist attacks to events including, but not limited to, natural disasters, truck crashes, and industrial accidents. These events can disperse microbial substances and make prompt identification nearly impossible by removing or destroying written descriptions, warning labels, etc. of said microbial sources.

Without knowing a microbe's identity, emergency first responders put themselves, their families, and others at high risk of being contaminated as well as the person they are treating. It is also necessary that a first responder know what the person they are treating has been exposed to because they cannot properly treat a person without knowing what is causing the person's condition and if first responders do not know what substance a person has been exposed to they might treat that person with a drug which will react with the unknown substance in a negative way.
Emergency first responders are not the only people who need quick and efficient analysis, protection, and treatment of/from unknown substances. Other examples of people who need to have prophylaxis, treatment, and/or quick analysis of an unknown substances include, but are not limited to, a waste clean up person cleaning up an industrial site, a mail room clerk who sees a package leaking a fluid, a police officer who has found an unknown substance in a suspect's car, and a teacher who has found a child unconscious next to an unmarked bottle of chemicals.
Current methods used to test an unknown substance are often times only available at a specialized laboratory. Precious time is wasted waiting for sample transport, assembly of trained personnel to perform the testing. Consequently, the present invention contemplates a kit comprising an array of labeled microbial DNA, a sample DNA
extraction solution, and a detector capable of quantitating the label. Such a kit would allow an on-site determination of the type of microbial contamination.
Non-specific decontamination is labor intensive and a time consuming task which may not be effective if microbial spores are present. The present invention contemplates a readily available decontamination composition comprising a plurality of peptide fragments derived from at least one spore surface protein. Such fragments would displace bacterial spores from solid surfaces (i.e., for example, building walls, ceiling, staircases, office furnishings etc) as well as from the outer epithelial layers (i.e., for example, skin) of emergency first responders. Further, either parenteral, intranasal, pulmonary, or oral compositions suitable for human and/or animal administration would displace bacterial spores from bodily tissues (i.e., for example, mouth, throat, nasal passages, lungs, stomach, intestine, bladder, or colon). Consequently, the present invention contemplates a composition comprising a compound having an affinity for a spore surface protein, wherein the protein is responsible for the attachment of a microbial spore to a surface (i.e., for example, a solid artificial surface or a skin surface).
C. Military Applications In one embodiment, the present invention contemplates prophylactic and therapeutic treatment of military personnel responding to microbial contamination (i.e., for example, first responders). In one embodiment, military personnel may include, but are not limited to, infantry, pilots, cavalry, naval, marine, special forces, logistics, contractors, or journalists.
The present invention contemplates a readily available decontamination composition comprising a plurality of peptide fragments derived from at least one spore surface protein. Such fragments would displace bacterial spores from solid surfaces (i.e., for example, tents, jeeps, tanks, weapons, uniforms, etc.) as well as from the outer epithelial layers (i.e., for example, skin) of military personnel. Further, either parenteral, intranasal, pulmonary, or oral compositions suitable for human and/or animal administration would displace bacterial spores from bodily tissues (i.e., for example, mouth, throat, nasal passages, lungs, stomach, intestine, bladder, or colon).
Consequently, the present invention contemplates a composition comprising a compound having an affinity for a spore surface protein, wherein the protein is responsible for the attachment of a microbial spore to a surface (i.e., for example, a solid artificial surface o, a skin surface).
In one embodiment, the present invention contemplates a method of routine decontamination of ballistic fabrics. At present, garments made of ballistic fabrics Ore washed by hand with cold water and mild detergent, rinsing thoroughly to removeall traces of detergent. Proper rinsing prohibits the accumulation of residual soap fm, which can absorb water and reduce the ballistic resistance of certain types of ballistic fabric. While such a process is time consuming and laborious, it is necessary$ince machine washing or drying, can be damaging to the fabric, ultimately affecng its ballistic performance. Furthermore, some detergents, dry cleaning solvent, bleach and starch may reduce the garment's level of ballistic resistance and most mtiufacturers strongly recommend against their use. Furthermore, mast manufactures of ballistic fabric strongly recommend that ballistic fabrics never be suhnerged in watr or dried outdoors, even in the shade, because ultraviolet light causes deg-adation of cktain types of ballistic fabric.
Similarly, the care and cleaning of protective apparel, suh as military turnout gear and other protective items such as knee pads, shin guards shoulder pads and the like, is laborious and time consuming. Repeated cleaning can redsse the useful life of such garments.

A problem common to both protective gear and garments made of ballistic fabric is sweat from the wearer. If the sweat is not removed, fungal and/or bacterial growth can result over time. This fungal and/or bacterial growth ultimately degrades the protective properties of the article, results in noxious odors, and possibly even results in long-term health issues for the wearer. With regard to military turnout gear in particular, all fabrics and components used in the construction of military protective clothing must pass minimum performance requirements. Thus, the inner lining of protective garments designed for military personnel where there is a danger of exposure to weapons fire and/or flame and high temperatures are usually made from aramid fibers and yarns.
In one embodiment, the present invention comprises a method to decontaminating ballistic fabric articles and other military protective gear, wherein the articles and gear comprise components including, but not limited to, aramid, polybenzazole, or high performance polyethylene fiber. In one embodiment, the articles and/or gear are decontaminated by a composition comprising a compound having affinity for a spore surface protein. The composition can be applied to the article directly or to the fiber or as a fabric finish easily and in a cost-effective way.

X. Antimicrobial Peptides Infection control research now includes searching for novel anti-microbial peptides as well as anti-microbial organic chemicals. Antimicrobial peptides have been isolated from plants, insects, fish, amphibians, birds, and mammals. Gallo, Jlnvest Dermatol., 111:739 -743 (1998); and Ganz et al, Pharmac. Ther., 66:191-205 (1998).
These peptides are apparently a primary component of innate host protection against microbial pathogenesis functioning to create pores in the cytoplasmic membrane of microorganisms. Oren et al., Biopolymers, 47(6):451-463 (1998). Furthermore, antimicrobial peptides also act on animal cells by stimulating them to change behaviors such as syndecan expression, chernotaxis, and chloride secretion. Gallo, Jlnvest Dermatol., 111:739 -743 (1998). After contact with microorganisms, vertebrate skin, trachea and tongue epithelia have been reported to produce peptide antibiotics. Russell et al., Infect Immun, 64(5):1565-1568 (1996). Antimicrobial peptides, however, have not generally been observed to be effective against microbial spores.
Antimicrobial peptides of higher eukaryotes, though long recognized as components of the innate immune system, were initially considered primitive and of little clinical significance. However, the relative simplicity of these peptides belies their importance, not only in the prevention of primary microbial infection, but also in subsequent immunomodulation. Bowman, "Peptide antibiotics and their role in innate immunity," Annu. Rev. Immunol., 13:61-92 (1995). Further, the small size of the molecules suggests a decreased sensitivity to many of the mechanisms of microbial resistance.
Antimicrobial peptides are generally lethal to bacteria and some fungi. They exhibit differential toxicity towards mammalian cells. Although it is not necessary to understand the mechanism of an invention, it is believed that anti-microbial peptides interact with the lipid bilayer and may thus compromise the integrity of the bacterial membrane. Hwang et al., Biochem Cell Biol, 76:235-246 (1998).
SMAP 29 is an ovine antimicrobial peptide of the cathelicidin originally identified through 3' RACE analysis of sheep bone marrow RNA. Mahoney, FEBS Lett, 377:519-522 (1995). RCAP 18 is a lupine antimicrobial peptide of the cathelicidin originally identified from granulocytes. Hirata et al., Infect. Immun. 62:421-1426 (1994). It is believed that SMAP 29 and RCAP 18 peptides provide protection against some drug resistant bacterial strains. For example, SMAP 29 compositions are capable of controlling antibiotic resistant infections by Pseudomonas aeruginosa, Alcaligenes xvlosoxidans, and Stenotrophomonas maltophila.
The antibacterial potency of the full-length antimicrobial peptides are apparently correlated directly with a hydrophobicity gradient along their backbone, inversely with their relative abundance of anionic residues, but not with the extent of helix formation in tri fluoro ethanol.
This invention thus contemplates methods to screen for, and identify, compositions that are capable of inhibiting microbial spore interaction, attachment, and/or stabilization to a solid surface. It is further contemplated that these screening methods would also identify compounds capable of inhibiting microbial spore interaction, attachment, and/or stabilization to a host organism (i.e., for example, a human, domestic animal, livestock animal, etc).

XI. Combination Vegetative Cell/Spore Antimicrobial Compositions This invention encompasses methods to solve problems related to antimicrobial resistance to common antibiotic drugs. One reason for apparent antimicrobial resistance is that common antimicrobial agents are ineffective against microbial spores.
Consequently, when microbial spores are still present after "decontamination"
with a conventional antimicrobial compound, the spore will convert into an active microbial growth. This can be (mis)-interpreted as "microbial resistance" when, in fact, the recurring microbial presence is due to an incomplete decontamination technique. In one embodiment, the present invention contemplates a composition comprising a compound having affinity for a bacterial spore surface peptide in combination with one or more conventional antimicrobial agents or antibiotics.
Although it is not necessary to understand the mechanism of an invention, it is believed that a composition comprising a compound having affinity for a microbial surface spore peptide and/or one or more conventional antimicrobial agents or antibiotics will not only effectively kill the active microbes (i.e., for example, vegetative cells) but also detach microbial spores from solid surfaces and or bodily tissues. It is further believed that this combination strategy results in an antiseptic level not possible with decontamination with an antimicrobial agent alone.
A list of bacterial strains that have been identified with resistance characteristics are listed in Table XIV.

Table XIV: Mechanisms Of Bacterial Resistance To Antimicrobial Agents Antimicrobial Agent Mechanisms Causing Examples of Organisms Resistance Aminoglycosides Modifying enzymes: Enterobacteriaceae, P.
acetyltransferases, adenylyl- aeruginosa, S. aureus, E.
transferases (nucleotidyl- faecalis, E. faecalis, transferases), Enterobacteriaceae, phosphotransferases M. tuberculosis, Ribosomal resistance P. aeruginosa E. faecalis, P.
(streptomycin, aeruginosa, anaerobes spectinomycin) Inadequate drug transport (3-Lactams Enzymatic inactivation S. aureus, E. faecalis, Enterobacteriaceae, P.
aeruginosa, Neisseria spp., H. influenzae Low affinity PBPs S. pneumoniae, N.
gonorrhoeac, S. aureus, Lack of penetration through P. aeruginosa, outer membrane Enterobacteriaceae Chloramphenicol Acetylation Enterobacteriaceae, S.
aureus, streptococci, Bacteroides uniformis Lack of penetration P. aeruginosa Clindamycin, erythromycin, Ribosomal resistance due to Streptococci, E.
faecalis, lincomycin methylation of rRNA
Inactivation by esterase Enterobacteriaceae Decreased penetration Enterobacteriaceae, S.
hominis Fluoroquinolones Decreased uptake Enterobacteriaceae, P.
Altered target site (DNA aeruginosa, staphylococci gyrase) Enterobacteriaceae, P.
aeruginosa Lincomycin Inactivation S. aureus Sulfonamides Synthesis of an altered or Enterobacteriaceae, alternative target site Neisseria spp., P.
(dihydropteroate synthetase) aeruginosa Lack of penetration Overproduction of PABA Anaerobes Neisseria, S. aureus Tetracycline Drug efflux Enterobacteriaceae, staphylococci, streptococci Protection of ribosome from Streptococci, E. faecalis, tetracycline Neisseria spp., Inactivation Mycoplasma spp. Cryptic gene found in B. fragilis, expressed resistance in E.
coli Trimethoprim Synthesis of an altered or Enterobacteriaceae, V.
alternative target site cholerae, staphylococci (dihydrofolate reductase) Lack of penetration P. aeruginosa Ability to use alternative Enterococci pathway H. influenzae Overproduction of dihydrofolate reductase Vancomycin ? Pediococci, Leuconostoc spp. (intrinsic) ?Blocking of target site Enterococci (acquired) Lorian, In: Antibiotics in Laboratory Medicine, Satterfield (Ed), Williams &
Wilkins, Philadelphia, pp.
558-559 (1991).

In one embodiment, the present invention contemplates a composition comprising a compound having affinity for microbial spore surface peptides, wherein the compound comprises an affinity for peptides derived from different species and strains of bacteria.
In one embodiment, different strains of bacteria are resistant to a plurality of antibiotic drugs. In one embodiment, the compositions of the present invention comprise stable pharmaceutical formulations. In one embodiment, the formulation may further comprise a pharmaceutically acceptable carrier. It is further contemplated that formulations may be administered to humans or animals, included in food preparations, pharmaceutical preparations, medicinal and pharmaceutical products, cosmetic products, hygienic products, cleaning products and cleaning agents, as well as any material to which the compositions could be sprayed on, or adhered to, wherein inhibition of microbial spore attachment and subsequent growth on such a material is desired.
The proper applied amount of compositions of the present invention necessary to prevent microbial attachment and subsequent growth and proliferation depends upon a number of factors including the types of bacteria that might be present, the environment into which the composition is being introduced, and the time that the composition is envisioned to remain in a given area.
It is further contemplated that compositions of the present invention may be used in combination with other antimicrobial agents or antibiotics to enhance their activity.
Combinations of compositions of the present invention with other agents may be useful to allow antibiotics to be used at lower doses due to toxicity concerns, to enhance the activity of antibiotics whose efficacy has been reduced, or to effectuate a synergism between the components such that the combination is more effective than the sum of the efficacy of either component independently.
Antibiotics which may be combined with compositions of the present invention in combination therapy include, but are not limited to, penicillin, ampicillin, amoxycillin, vancomycin, cycloserine, bacitracin, cephalolsporin, imipenem, colistin, methicillin, streptomycin, kanamycin, tobramycin, gentamicin, tetracycline, chlortetracycline, doxycyclinc, chloramphenicol, lincomycin, clindamycin, erythromycin, oleandomycin, polyrnyxin nalidixic acid, rifamycin, rifampcin, gantrisin, trimethoprim, isoniazid, paraminosalicylic acid, and ethambutol.
In one embodiment, the present invention contemplates reducing the resistance of a microorganism to an antimicrobial agent, as exemplified by reducing the resistance of a bacterium to an antibiotic, or to kill a microorganism or bacterium, by generally contacting the microorganism or bacterium with an effective amount of the antibiotic or antimicrobial agent in combination with an amount of a composition having affinity for a microbial spore surface peptide effective to inhibit the attachment and/or subsequent growth of the microorganism or bacterium. The terms "microorganism" and "bacterium"
are used for simplicity and it will be understood that the invention is suitable for use against a population of microorganisms.
The microorganism, e.g., bacterium, or population thereof, may be contacted either in vitro or in vivo. Contacting in vivo may be achieved by administering to an animal (including a human patient) that has, or is suspected to have been contaminated with a microbe or bacteria, a therapeutically effective amount of pharmacologically acceptable formulation comprising a composition having a compound having affinity for a microbial spore surface peptide alone, or in combination with, a therapeutic amount of a pharmacologically acceptable formulation of an antibiotic agent. The invention may thus be employed to treat both systemic and localized microbial and bacterial infections by introducing the combination of agents into the general circulation or by applying the combination, e.g., topically to a specific site, such as a wound or burn, or to the eye, ear or other site of infection.
Where a composition of the present invention is used in combination with other antimicrobial agents or antibiotics, an "effective amount of an antimicrobial agent or antibiotic" means an amount, or dose, within the range normally given or prescribed.
Such ranges are well established in routine clinical practice and will thus be known to those of skill in the art. Appropriate doses and treatment regimens are further detailed herein. See, Table XV. Naturally, in confirming the optimal therapeutic dose for compounds having affinity for microbial spore surface peptides, first animal studies and then clinical trials would be conducted, as is routinely practiced in the art.
See, Example II.

Table XV: Common Antibiotics And Usual Oral Doses Antibiotic Dosage Antibiotic Dosage Penicillin V 2 Rugby (generic) 50 mg qid V-cillin K
Dicloxacillin 250 mg qid Glenlawn (generic) Dynapen Cloxacillin (Tegopen) Amoxicillin Rugby (generic) Polymox Ampicillin Moore (generic) Polycillin Augmentin tid 250-mg tablets chewables (250 mg) 125-mg (suspension) chewables (125 mg) Carbenicillin (Geocillin) 382 mg qid (1 tb) 2 tabs id Cephalexin 250 mg qid Rugby (generic) Keflex Rugby (generic) 500 mg qid Keflex Cefadroxil 1 gm bid Rugby (generic) Duricef Cephradine 250 mg qid Rugby (generic) 500 mg qid Velosef Rugby (generic) Cefaclor 250 mg tid Ceclor Cefuroxime axetil 125 mg bid Ceftin 250 mg bid 500 m bid Cefixime 400 mg q24h Suprax Cefprozil 250 mg ql2h Ccfzil Loracarbef (Lorabid) 200 mg bid Cefpodoxime proxetil 200 mg bid Vantin) Clindamycin 300 mg q8h Cleocin TMP/SMZ 1 double-strength bid Bactrim Sept a (generic) Trimethoprim 100 mg bid Rugby (generic) Proloprim Erythromycin (base) 250 mg qid Abbott E-mycin (delayed release) Erythromycin stearate 250 mg qid Rugby (generic) Azithromycin 1 g once only; 500 mg day 1 plus 250 mg day 2- 5 Zithromax Clarithromycin 250 mg bid Biaxin 500 mg bid Tetracycline hydrochloride 250 mg qid M lan Sumycin 250 Doxycycline 100 mg qd (with 200- mg initial load) Lederle (generic) Vibrarnycin Vancomycin Capsules (oral 125 mg q6h PO soln/powder) Vancocin HCl Metronidazole 250 mg qid Rugby (generic) Flagyl r Norfloxacin 400 mg bid Noroxin Ciprofloxacin 250 mg bid Cipro 500 mg bid 750 mg bid Ofloxacin 200 m bid Floxin 300 mg bid 400 ing bid Lomefloxacin 400 mg once qid Maxaguin J
yid (4 times daily), tid (3 times daily), bid (twice daily), qd (once daily), q4h (every 4 hours around the clock), q6h (every 6 hours around the clock) and q8h (every 8 hours around the clock).

The LD50/ED50 ratio required for safe use of the proposed compositions and/or combinations of the compositions with other antimicrobial agents are assessed by determining the LD50 (median lethal toxic dosage) and the ED50 (median effective therapeutic dosage) in experimental animals. The optimal dose for human subjects is then defined by fine-tuning the range in clinical trials. In the case of LD50, an inhibitor is usually administered to mice or rats (orally or intraperitoneal) at several doses (usually 4-5) in the lethal range. The dose in mg/kg is plotted against % mortality and the dose at 50% represents the LD50. The ED50 is determined in a similar fashion.
In a clinical trial, the therapeutic dose would be determined by maximizing the benefit to the patient, while minimizing any side-effects or associated toxicities. In optimizing a therapeutic dose within the ranges disclosed herein, one would not use the upper limit of the range as the starting point in a clinical trial due to patient heterogeneity.
Starting with a lower or mid-range dose level, and then increasing the dose will limit the possibility of eliciting a toxic or untoward reaction in any given patient or subset of patients. The presence of some side-effects or certain toxic reactions per se would not, of course, limit the utility of the invention, as it is well known that most beneficial drugs also produce a limited amount of undesirable effects in certain patients.
In the treatment of animals or human patients with combination therapy, there are various appropriate formulations and treatment regimens that may be used. For example, a composition comprising a compound having affinity for a spore surface peptide and at least one additional antimicrobial drug may be administered to an animal simultaneously, e.g., in the form of a single formulation that includes the compound and additional drug(s), or by using at least two distinct formulations. The compounds having affinity for a spore surface peptide could also be administered to an animal prior to the additional drug(s) or vice versa.

Further embodiments of the invention include therapeutic kits that comprise, in suitable container means, a pharmaceutical composition of at least one compound having affinity for a spore surface peptide and at least one additional antimicrobial drug and/or antibiotic. The compound, antimicrobial agent and/or antibiotic may be contained within a single container means, or a plurality of distinct containers may be employed.
Depending on the circumstances, antimicrobial agents may be employed in oral or parenteral treatment regimens. Methods to achieve appropriate doses are described in various publications. Reese and Betts, In: A Practical Approach to Infectious Diseases, (3rd ed.), Boston, Little Brown, (1993).
XII. Antibacterial Cleanser Compositions Antibacterial cleansing compositions are typically used to cleanse the arms and hands of the user and to destroy bacteria and any other microorganisms which might be present on the user's arms or hands. These compositions are widely used in the health care industry by hospital staff and other health care personnel as hand washing cleansers to prevent infection ether between staff personnel or between staff personnel and patients.
They are particularly suitable for use by heathcare worker such as surgeons, nurses and other health care professionals who might be subject to contact with bacteria and the like.
They are also suitable for use by personnel in the food service and meat processing industries and, generally are used for antimicrobial cleansing of the hands and arms by the public.
There are several active antimicrobial agents currently available for use in cleansing compositions. For example, many antimicrobial cleansing compositions contain a bisbiguanide bacterial substance such as chlorhexidine digluconate (CHG).
Other cleansing compositions use phenolic compounds. The antimicrobial activity of these substances, however, are often dependent upon the type(s) of surfactants or other ingredients employed therewith, and therefore, the use of the same ingredients with these antimicrobial substances will not necessary derive the same result. For instance, when CHG is employed as an active antimicrobial agent, the cleansing composition may include, but not be limited to, alkyl polyglucosides and nonionic alcohol ethoxylates as the primary surfactants. However, in compositions containing substituted phenols such as para-chloro-meta-xylenol (PCMX; 4-chloro-3,5-dimethyl phenol) as the active antimicrobial agent, most non-ionic alcohol ethoxylates inhibit, rather than enhance, PCMX antibacterial activity. PCMX may yield high initial reductions of Gram-positive and Gram-negative bacteria as well as fungi, and provide good residual activity for several hours between hand washings. Typically, where PCMX is used in cleansing compositions, it is used in concentrations of from about 0.1 to 4 percent by weight.
However, the majority of formulas contain 1 percent or less by weight.
Antimicrobial cleansing compositions containing substituted phenols as the active antimicrobial agent have also included anionic detergents, soaps, surfactants, and the like, as well as other compounds (non-ionic ethoxylated surfactants, polyethylene glycol etc.) which may reduce the antibacterial activity of the phenolic compounds.
Substituted phenolic compounds and anionic surface active detergents or soaps in an alcohol and water-based carrier are also useful. Additionally, chelating agents such as ethylenediaminetetraacetic acid (hereinafter, EDTA) maybe added. Winicov et al., U.S.
Pat. No. 3,824,190 & RE 28,778 (herein incorporated by reference).
Antimicrobial compositions exhibiting mildness characteristics may include, but are not limited to, a substituted phenol, namely PCMX, an anionic detergent, a thickener such as ethylene glycol monostearate, and a foam builder such as a fatty acid alkanol amide. The thickener and fatty acid alkanol amides as well as the anionic surfactant are suspected to hinder antibacterial activity of a phenolic compound. Garabedian et al. U.S.
Pat. No. 4,632,772 (herein incorporated by reference). Other PCMX compositions comprise anhydrous blends in surfactant mixtures. Corti et al., U. S. Pat, No.
5,114, 978 (herein incorporated by reference).
PCMX may be solubilized in high concentrations of water or aqueous compositions. PCMX compositions include, but are not limited to, diethanolamides of fatty acids and anionic surfactants of the class diethanolammonium salts of alkylpolyoxyethylsulfuric acid.
Alcohol-based antimicrobial compositions may include, but are not limited to, PCMX or CHG, hydropropyl cellulose, an alcohol, an emollient, and water.
Although it is not necessary to understand the mechanism of an invention, it is believed that the use of alcohol as a solvent or carrier in the composition is known to defat the skin which may lead to skin dryness and irritation. White et al., U.S. Pat. No. 5,288,486 (herein incorporated by reference).
Consequently, PCMX is similar to that of CHG and other substituted phenols against microorganisms in that their efficacy is highly dependent upon both the carrier and other chemical constituents within the antimicrobial cleanser composition.
While the antimicrobial activity of PCMX may be potentiated against Pseudomonas aeruginosa by the addition of the chelating agent ethylenediaminetetraacetic acid (EDTA) and/or the sequestering agent sodium hexametaphosphate, PCMX may be reduced in the presence of some organic materials, and/or moderate concentrations of anionic surfactants.
Antimicrobial activity may also be lost as a consequence of reversible interactions between PCMX and noninonic surfactants, polyethylene glycol, and polyethylene glycol stearate. Similarly, reduction in the antibacterial efficacy of PCMX has also been found to be the direct result of its interaction with a variety of macromolecules, namely, methyl cellulose, polyethylene glycol 6000, and polysorbate 80.
The present invention is not limited to standard antibiotic cleanser compositions such as PCMX and CGH. In one embodiment, the present invention contemplates an antibacterial composition containing a substituted phenol which does not include conventional anionic surfactants or other chemical ingredients detrimental to substituted phenol antimicrobial activity. In one embodiment, an antimicrobial cleansing composition comprises a substituted phenol and at least one primary surfactant. In one embodiment, the composition comprises a substituted phenol in amounts ranging from about 0.1 to about 4 percent by weight of the total composition, and preferably in amounts ranging from about 0.3 to 1 percent by weight of the total composition. In one embodiment, the composition comprises a substituted phenol in amounts that are more than 4 percent by weight of the antimicrobial agent. Although it is not necessary to understand the mechanism of an invention, it is believed that the upper limit of the antimicrobial agent amount is related to the amount of substituted phenol which is considered to be non-toxic and non-irritating to the skin.
Any substituted phenol which is soluble in water or other non-alkanol solvent (i.e., for example, triethylene glycol, propylene glycol, hexylene glycol etc.) may be used in the compositions of the present invention. In one embodiment, a substituted phenol may include, but is not limited to, alkyl substituted, halogen substituted, phenol substituted, alkyl halo substituted, benzyl alkyl substituted, phenylethyl alkyl substituted and benzyl halo substituted phenols, and alkyl substituted, halogen substituted and alkyl halo substituted bisphenols, bis(hydroxyphenyl) alkanes, amino substituted phenols, resorcinol derivatives, trihydric phenol derivatives, naphthol derivatives and carboxylic acid substituted phenols (e.g., salicylic acid and esters). In one embodiment, a substituted phenol comprises a chloro-substituted phenol, namely para-chloro-meta-xylenol (PCMX), Although it is not necessary to understand the mechanism of an invention, it is believed that PCMX may have 60 times the antimicrobial activity of an unsubstituted phenol against a wide spectrum of bacteria and is also considered to be non-toxic and non-irritating to human skin at levels below about 3 percent by weight. Fendler, et al., "Antimicrobial cleansing compositions" United States Patent 5,635,462 (herein incorporated by reference).
In one embodiment, an infection control cleanser composition of the present invention comprises at least one primary surfactant selected from the group including, but not limited to, amine oxides, phospholipids, partially neutralized carboxylic acids and diacids, betaines, or ethoxylated methylglucosides, and/or mixtures thereof.
Although it is not necessary to understand the mechanism of an invention, it is believed that the amount of primary surfactant(s) to be added to the composition of the present invention is somewhat dependent upon the number of primary surfactants added. In one embodiment, the composition comprises a plurality of primary surfactants wherein the combined amount of the primary surfactants is less than about 20 percent by weight of the composition.
In one embodiment, the composition comprises a primary surfactant including, but not limited to, an amine oxide or an alkyl amine oxide. In one embodiment, an amine oxide is added to the composition in an amount ranging from about 0 (absent) to about 10 percent by weight but more preferably in an amount ranging from about 5 to 7 percent by weight. In one embodiment, a composition comprises an amine oxide as a sole primary surfactant wherein the amine oxide added in amounts ranging from about 1 to about 10 percent by weight. In one embodiment, an amine oxide has a alkyl chain length of from about C8 to about C16, with chain lengths of from C12 to C14.
Although it is not necessary to understand the mechanism of an invention, it is believed that amine oxides contemplated by the present invention are considered to be amphoteric surfactants. Examples of amine oxides found to be suitable for the present invention include, but are not limited to, lauramine oxide a C12 alkyl amine oxide (Mackamine LO ) and cocamine oxide a C12 -C14 alkyl amine oxide (Mackamine CO )(McIntyre Chemical Co., Ltd., Chicago, Ill.), as well as a C14 alkyl amine oxide(Barlox 14 ) and a C12 alkyl amine oxide(Barlox 12 ')( Lonza, Inc., Fair Lawn, N.J).
Alternatively, the compositions contemplated herein are suitable for amidopropyl amine oxides including, but not limited to Standamox LAO or Mackamine CAO`8' (Henkel Corp., Hoboken, N.J.).
In addition, alkylamides such as cocamide may be used in conjunction with the amine oxides or other primary surfactants to boost foaming and add to the viscosity of the composition. Viscosity modifiers and/or foam stabilizers may include, but are not limited to, Standamide CD , Standamide SD , and Cocamide DEA (Henkel Corp., Hoboken, N.J.).
Although it is not necessary to understand the mechanism of an invention, it is believed that amine oxides do not significantly reduce the antibacterial activity of a substituted phenol and have been found to be effective in combination with PCMX for killing microorganisms..
In one embodiment, the present invention contemplates an infection control composition comprising zwitterionic/amphoteric surfactants such as phospholipids and betaines as a primary surfactant. In one embodiment, phospholipids and/or betaines may be used in place of, or in conjunction with, amine oxides. In one embodiment, a composition may comprise phospholipids and betaines in amounts up to about 10 percent by weight.
In one embodiment, an infection control cleanser composition comprises a phospholipid including, but not limited to, an alkyl phosphatidyl PG-dimonium chloride.
In one embodiment, an alkyl phosphatidyl PG-dimonium chloride may be selected from the group comprising cocamidopropyl phosphatidyl PG-dimonium chloride (Phospholipid PTC , MONA Industries, Inc., Paterson, N.J.); stearamidopropyl phosphatidyl PG-dimonium chloride (Phospholipid PTS , MONA Industries, Inc., Paterson, N.J.);
linoleamidopropyl phosphatidyl PG-dimonium chloride (Phospholipid EFA , MONA
Industries, Inc., Paterson, N.J.); and stearamidopropyl phosphatidyl PG-dimonium chloride/cetyl alcohol (Phospholipid SV , MONA Industries, Inc., Paterson, N.J.). In one embodiment, alkyl phosphatidyl PG-dimonium chlorides are incorporated into the composition in an amount ranging from 0 (absent) to about 10 percent by weight of the total composition.
In one embodiment, the present invention contemplates an infection control cleanser composition comprising a betaine lipid as a primary surfactant either alone or in conjunction with other primary surfactants. In one embodiment, a betaine lipid comprises an alkylammonio carboxylate ranging from 8 to 18 carbon atoms. In one embodiment, a composition comprising an alkylammonio carboxylate includes up to 10 percent by weight of the total composition. In one embodiment, an alkylammonio carboxylate comprises cocobetaine (Mackam CB-35 , McIntyre Chemical Co., Chicago, Ill.).
In one embodiment, the present invention contemplates an infection control cleanser composition comprising a mixture of at least one partially neutralized carboxylic acids and/or diacids (i.e., for example, sodium caproyl lactylate) that may be employed as a primary surfactant. In one embodiment, a carboxylic acid and/or diacid is added to the cleansing composition in amounts ranging from 0 (absent) to about 5 percent by weight.
Although it is not necessary to understand the mechanism of an invention, it is believed that although partially neutralized carboxylic acids and diacids might contain some anionic surfactants they have considerable protonated acid character and, therefore, these compounds are expressly excluded from the term "conventional anionic surfactants".
In one embodiment, the present invention contemplates an infection control cleanser composition comprising a nonionic primary surfactant. In one embodiment, the nonionic primary surfactant comprises an ethoxylated nonionic methylglucoside.
In one embodiment, an ethoxylated nonionic methylglycoside may also be added to a composition in amounts ranging from 0 (absent) to about 10 percent by weight.
The composition may also include other additives such as thickeners, emollients, chelating and sequestering agents, fragrances, coloring agents, opacifying agents, pearlizing agents, vitamins and the like. For example, a composition may include a polymer viscosifier or thickener such as hydroxyethyl cellulose to make the composition more aesthetically pleasing. Examples of other suitable polymer viscosifiers include, but are not necessarily limited to, hydroxypropyl cellulose, methyl cellulose, and carboxymethyl cellulose.
A glycol maybe added in order to quicken the rate of dissolution of the substituted phenol and as an emollient and/or humectant. Glycols may include, but are not limited to, propylene glycol, triethylene glycol, and hexylene glycol.
Glycols may be added to a composition in amounts ranging from 0 (absent) to about 10 percent by weight of the total composition, but more preferably in an amount of about 3 percent.
In one embodiment, a PCMX and/or a second substituted phenol may be dissolved in at least a portion of a glycol prior to adding other ingredients to the solution. In one embodiment, a composition comprises a 25 percent/75 percent ratio of PCMX:glycol.
In one embodiment, the present invention contemplates an infection control cleanser composition comprising a chelating and/or sequestering agent ranging up to about 1 percent by weight. Although it is not necessary to understand the mechanism of an invention, it is believed that a chelating agent and/or sequestering agent may be added to soften the water-based composition. In one embodiment, a suitable chelating agent for the present invention comprises ethylenediaminetetra-acetic acid (EDTA) (i.e., for example, disodium EDTA).

The compositions of the present invention may further comprise minor but effective amounts of other conventional additives such as fragrances, color additives, opacifying agents, pearlizing agents, vitamins, etc. An example of a particular pearlizing agent includes, but is not necessarily limited to, ethylene glycol distearate.
Generally, these additives are used in amounts which do not affect the essential nature of the composition with respect to its infection control properties.
For optimal antibacterial efficacy, the pH of the composition should be between 4 and 8, and preferably between 5.5 and 6.5. To adjust the pH of the composition, any acid compatible with the components of the composition can be used. Acids may include, but are not limited to, lactic acid, citric acid, acetic acid, glycolic acid, or gluconic acid.
Typically, less than 1 percent by weight of these acids are used to achieve the proper pH
balance.
The balance of the composition is typically water so as to provide 100 percent by weight of the composition but other water soluble compounds are also suitable (i.e., for example, alcohols).
All percents by weight indicated herein are based upon the percent active composition. Thus, for example, where 5 percent by weight lauramine oxide is employed and the lauraminc oxide is obtained from the manufacturer or has been diluted into solution so as to comprise a 30 percent active solution, 16.7 percent of the solution will have to be used in order to obtain the 5 percent by weight recommended.
The infection control cleansing compositions of the present invention are generally prepared by dissolving substituted phenol, namely PCMX, in glycol as noted hereinabove, and adding this solution to one or more of the primary surfactant discussed hereinabove, in water. More particularly, the primary surfactant or surfactants, and other ingredients (i.e., for example, fragrance, chelating agent, pearlizing agent, etc.) are added to the solution with stirring. The pH of the composition is adjusted with lactic acid or similar acid. The thickener/viscosifier is then preferably added and the solution is mixed until it is completely homogeneous. The pH is checked and adjusted again if necessary.
This process maybe employed with or without the application of heat to enhance hydration of the viscosifier, if required.

XIII. Detecting Microbes Comprising Spore Surface Protein Nucleotide Sequences The present invention provides methods the detection of microbes which contain a spore surface protein nucleotide sequence (i.e., for example, SEQ ID NO: 2) and/or a spore surface protein nucleotide sequence (i.e., for example, SEQ ID NO's: 4, 6, or 8).
Microbes which contain nucleotide sequences may be identified by variety of procedures.
These procedures include, but are not limited to, DNA--DNA or DNA-RNA
hybridization as well as amplification (e.g., PCR) using DNA probes (e.g., oligonucleotide or oligomer probes or amplimers), mRNA probes and fragments of the sequence of interest. These probes and fragments can be made using a wide variety of techniques including, but not limited to, chemical synthesis, restriction digestion and expression of the nucleotide sequence, or any portion of it, in an expression vector.
Labeling of synthesized or expressed probes and nucleotide sequence fragments can be achieved using oligolabeling, end-labeling or PCR amplification using labeled nucleotide.
Having generated labeled probes, microbes present in a biological sample can be tested for the presence of a nucleotide sequence using Southern or reverse Northern analysis of isolated plasmid total cellular DNA, or using Northern analysis of mRNA.
Microbes can be either grown on filters or spotted onto filters either directly or following overnight culture. Moseley et al., J. Infect. Dis. 142:892-898 (1980); Perine et al., J. Infect. Diseases 152(l):59-63 (1985); and Gootz et al., Antimicrob.
Agents and Chemother. 28(l):69-73 (1985). Briefly, a solid support such as nitrocellulose paper or a nylon membrane is inoculated with clinical specimen, or with broth culture of clinical specimen suspected of containing bacteria. The cells lysed onto the nitrocellulose paper and the DNA denatured, for example by treatment with NaOII. The support is treated with pronase and chloroform to lower background non-specific binding of DNA
probes to the colony material. Prehybridization and hybridization of the filters (i.e., support) with oligonucleotides, oligomers, or portions of a nucleotide sequence is then performed using standard techniques. Hybridization is detected using any one of many available methods, such as by imaging radioactive probes using X-ray films (i.e., autoradiography).
Detection of bacteria which harbor a microbial spore surface protein nucleotide sequence in a sample derived from a surface demonstrates that it is beneficial to administer infection control compositions which alter microbial spore surface protein binding to the surface. In addition, such detection also permits monitoring the surface for the presence of the bacteria during and after administration of the infection control composition.
In one embodiment, screening methods (i.e., for example, an assay) contemplated by this invention can be performed in solid phase where one or more components of the assay is attached to a solid surface. In solid phase assays, one or more components of the assay is attached to a solid surface. Virtually any solid surface is suitable, as long as the surface material is compatible with the assay reagents and it is possible to attach the component to the surface without unduly altering the reactivity of the assay components.
Some components might show reduced activity in solid phase, but this is generally acceptable so long as the activity is sufficient to interact with an infection control test compound.
Solid supports include, essentially any solid surface, including, but not limited to, a glass bead, planar glass, controlled pore glass, plastic, porous plastic metal, or resin to which the molecule may be adhered. Solid supports may be derivatized with functional groups (e.g., hydroxyls, amines, carboxyls, esters, and sulfhydryls) to provide reactive sites for the attachment of linkers or the direct attachment of the component(s).
Adhesion of an assay component (e.g., a spore surface protein) to the solid support can be direct or indirect (i.e., a particular compound or compounds are bound to the support, and an assay component binds to this compound or compounds rather than to the solid support). The component can be immobilized either: i) covalently (i.e., for example, by utilizing single reactive thiol groups of cysteine for anchoring protein components;
Colliuod et al., Bioconjugate Chem. 4:528-536 (1993)); ii) non-covalently but specifically (i.e., for example, via immobilized antibodies or other specific binding proteins;
Schuhmann et al. Adv. Mater. 3:388-391 (1991); and Lu et al. Anal. Chem. 67:83-(1995); iii) by the biotin/streptavidin system; Iwane et al., Biophys.
Biochem. Res. Comm.
230:76-80 (1997); iv) by using metal-chelating Langmuir-Blodgett films; Ng et al.
Langmuir 11:4048-4055 (1995); and Schmitt et al., Angew. Chem. Int. Ed. Engl.
35: 317-320 (1996); and v) metal-chelating self-assembled monolayers for binding of polyhistidine fusion proteins; Sigal et al., Analytical Chem. 68:490-497 (1996).
XVI. Detecting Microbes Comprising Spore Surface Proteins In one embodiment, the present invention contemplates methods comprising detecting a microbial spore protein and/or peptide fragment. In one embodiment, the microbial spore protein and/or peptide fragment comprises a microbial spore surface protein and/or peptide fragment. In one embodiment, the microbial spore surface protein and/or peptide fragment comprises a Clostridium spore surface protein and/or peptide fragment. In one embodiment, the Clostridium spore surface protein and/or peptide fragment comprises a Clostridium difficile spore surface protein and/or peptide fragment.

Proteins and/or peptide fragments, from any source, may be detected by a variety of suitable methods. In some embodiments, proteins and/or peptide fragments are detected by immunohistochemistry. In other embodiments, proteins and/or peptide fragments are detected by their binding to an antibody raised against the protein.
Antibody binding may be detected by many different techniques including, but not limited to, (e.g., radioimmunoassay, ELISA (enzyme-linked immunosorbant assay), "sandwich" immunoassays, immunoradiometric assays, gel diffusion precipitation reactions, immunodiffusion assays, in situ immunoassays (e.g., using colloidal gold, enzyme or radioisotope labels, for example), Western blots, precipitation reactions, agglutination assays (e.g., gel agglutination assays, hemagglutination assays, etc.), complement fixation assays, immunofluorescence assays, protein A assays, and immunoelectrophoresis assays, etc.
In one embodiment, antibody binding is detected by detecting a label on the primary antibody. In another embodiment, the primary antibody is detected by detecting binding of a secondary antibody or reagent to the primary antibody. In a further embodiment, the secondary antibody is labeled.
In some embodiments, an automated detection assay is utilized. Methods for the automation of immunoassays include those described in U.S. Pat. Nos.
5,885,530, 4,981,785, 6,159,750, and 5,358,691, each of which is herein incorporated by reference.
In some embodiments, the analysis and presentation of results is also automated. For example, in some embodiments, software that generates a prognosis based on the presence or absence of a series of proteins corresponding to cancer markers is utilized.
In other embodiments, the immunoassay described in U.S. Pat. Nos. 5,599,677 and 5,672,480; each of which is herein incorporated by reference.
In other embodiments, the present invention provides kits for the detection and characterization of proteins and/or nucleic acids. In some embodiments, the kits contain antibodies specific for a protein expressed from a gene of interest, in addition to detection reagents and buffers. In other embodiments, the kits contain reagents specific for the detection of mRNA or eDNA (e.g., oligonucleotide probes or primers). In preferred embodiments, the kits contain all of the components necessary to perform a detection assay, including all controls, directions for performing assays, and any necessary software for analysis and presentation of results.

XV. Pharmaceutical Formulations The active compositions of the present invention may include traditional pharmaceutical formulations. Administration of these formulations according to the present invention will be via any common route so long as the target tissue is available via that route. This includes topical, oral, nasal, buccal, rectal, vaginal or topical.
Alternatively, administration may be by orthotopic, intradermal, subcutaneous;
intramuscular, intraperitoneal or intravenous injection. Such formulations would normally be administered as a pharmaceutically acceptable composition.
A. Solutions For Oral Or Parenteral Administration The formulations, compositions, and other agents and drugs contemplated by the present invention may be active compounds that may also be administered parenterally or orally. Solutions of 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 may contain a preservative.
Pharmaceutical formulations suitable for injectable use include, but are not limited to, sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In one embodiment, a formulation is sterile. In one embodiment, the formulation comprises fluid characteristics such that the formulation is easily manipulated using a syringe. In one embodiment, the formulation is stable under manufacture and/or storage conditions. In one embodiment, the formulation may further comprise a carrier such as a solvent or dispersion medium including, but not limited to, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils. The proper fluidity can be maintained, for example, by 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. The prevention of the action of microorganisms can be brought about by various antibacterial an antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride.
Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.

Sterile injectable solutions may be prepared by incorporating the active compounds in the required amount in the appropriate solvent with various of the other ingredients enumerated above, as required, followed by sterilization (i.e., for example, by filtration, radiation, and/or ethylene oxide exposure). Generally, dispersions may be 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.
For oral administration, compositions having affinity for microbial spore surface proteins of the present invention may be incorporated with excipients and used in the form of non-ingestible mouthwashes and dentifrices. A mouthwash may be prepared incorporating the active ingredient in the required amount in an appropriate solvent, such as a sodium borate solution (i.e., for example, Dobell's Solution).
Alternatively, the active ingredient may be incorporated into an antiseptic wash containing sodium borate, glycerin and potassium bicarbonate. The active ingredient may also be dispersed in dentifrices, including: gels, pastes, powders and slurries. The active ingredient may be added in a therapeutically effective amount to a paste dentifrice that may include water, binders, abrasives, flavoring agents, foaming agents, and humectants.
The compositions of the present invention may be formulated in a neutral or salt form. Pharmaceutically-acceptable salts include the acid addition salts (formed with the free amino groups of the protein) and which are formed with inorganic acids such as, for example, hydrochloric or phosphoric acids, or such organic acids as acetic, oxalic, tartaric, mandelic, and the like. Salts formed with the free carboxyl groups can also be derived from inorganic bases such as, for example, sodium, potassium, ammonium, calcium, or ferric hydroxides, and such organic bases as isopropylamine, trimethylamine, histidine, procaine and the like.
Upon formulation, solutions will be administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective. The formulations are easily administered in a variety of dosage forms such as injectable solutions, drug release capsules and the like. Routes of administration maybe selected from intravenous, intrarterial, intrabuccal, intraperitoneal, intramuscular, subcutaneous, oral, topical, rectal, vaginal, nasal and intraocular.

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 which can be employed will be known to those of skill in the art in light of the present disclosure. For example, one dosage could be dissolved in I ml of isotonic NaCI solution and either added to 1000 ml of hypodennoclysis fluid or injected at the proposed site of infusion. In: Remington's Pharmaceutical Sciences 15th Edition, pages 1035-1038 and 1570-1580 (herein incorporated by reference).
Some variation in dosage will necessarily occur depending on the condition of the subject being treated. The person responsible for administration will, in any event, determine the appropriate dose for the individual subject. Moreover, for human administration, preparations should meet sterility, pyrogenicity, general safety and purity standards as required by FDA Office of Biologies standards.
B. Liposomes For Topical Or Parenteral Administration In one embodiment, the present invention contemplates a formulation comprising a liposoinal preparation comprising a compound having affinity for a microbial spore surface protein. In one embodiment, the compound is encapsulated by a liposome, wherein the encapsulation prolongs the compound half-life when compared to conventional drug delivery systems.
A "liposome" is generally used as a generic term encompassing a variety of single and multilamellar lipid vehicles formed by the generation of enclosed lipid bilayers. For example, phospholipids may be used for preparing liposomes and can carry a net positive charge, a net negative charge, or are neutral. Dicetyl phosphate can be employed to confer a negative charge on the liposomes, and stearylamine can be used to confer a positive charge on the liposomes. Liposomes may be characterized by a phospholipid bilayer membrane and an inner aqueous medium. Multilamellar liposomes have multiple lipid layers separated by aqueous medium. Liposomes usually form spontaneously when phospholipids are suspended in an excess of aqueous solution. Although it is not necessary to understand the mechanism of an invention, it is believed that the lipid components undergo self-rearrangement before the formation of closed structures and entrap water and dissolved solutes between the lipid bilayers. Ghosh et al, In: Liver diseases, targeted diagnosis and therapy using specific receptors and ligands, Wu G. and C. Wu (eds.), New York: Marcel Dekker, pp. 87-104, (1991). Also contemplated are cationic lipid-nucleic acid complexes, such as lipofectamine-nucleic acid complexes. In certain embodiments of the invention, the liposome may be complexed with a hemagglutinating virus (HVJ). HVJ has been shown to facilitate fusion with the cell membrane and promote cell entry. Kaneda et al., JBiol Chem,. 264: 12126-12129 (1989).
Lipids suitable for making liposomes according to the present invention can be obtained from commercial sources. For example, dimyristyl phosphatidylcholine ("DMPC") can be obtained from Sigma Chemical Co., dicetyl phosphate ("DCP") is obtained from K & K Laboratories (Plainview, N.Y.); cholesterol ("Chol") is obtained from Calbiochem-Behring; dimyristyl phosphatidylglycerol ("DMPG") and other lipids may be obtained from Avanti Polar Lipids, Inc. (Birmingham, Ala.). Stock solutions of lipids in chloroform, chloroform/methanol or t-butanol can be stored at about -20 C.
Preferably, chloroform is used as the only solvent since it is more readily evaporated than methanol.
Phospholipids from natural sources, such as egg or soybean phosphatidylcholine, brain phosphatidic acid, brain or plant phosphatidylinositol, heart cardiolipin and plant or bacterial phosphatidylethanolamine are preferably not used as the primary phosphatide, i.e., constituting 50% or more of the total phosphatide composition, because of the instability and leakiness of the resulting liposomes.
Liposomes used according to the present invention can be made by different methods. The size of the liposomes varies depending on the method of synthesis. A
liposome suspended in an aqueous solution is generally in the shape of a spherical vesicle, having one or more concentric layers of lipid bilayer molecules. Each layer consists of a parallel array of molecules represented by the formula XY, wherein X is a hydrophilic moiety and Y is a hydrophobic moiety. In aqueous suspension, the concentric layers are arranged such that the hydrophilic moieties tend to remain in contact with an aqueous phase and the hydrophobic regions tend to self-associate. For example, when aqueous phases are present both within and without the liposome, the lipid molecules will form a bilayer, known as a lamella, of the arrangement XY-YX.
Liposomes within the scope of the present invention can be prepared in accordance with many laboratory techniques. In one embodiment, liposomes are prepared by mixing liposomal lipids, in a solvent in a container, e.g., a glass, pear-shaped flask. The container should have a volume ten-times greater than the volume of the expected suspension of liposomes. Using a rotary evaporator, the solvent is removed at 1,54 approximately 40 C and under negative pressure. The solvent normally is removed within about 5 min to 2 hours, depending on the desired volume of the liposomes. The composition can be dried further in a desiccator under vacuum. The dried lipids generally are discarded after about 1 week because of a tendency to deteriorate with time.
Dried lipids can be hydrated at approximately 25-50 mM phospholipid in sterile, pyrogen-free water by shaking until all the lipid film is resuspended. The aqueous liposomes can be then separated into aliquots, each placed in a vial, lyophilized and sealed under vacuum.
In an alternative method, liposomes can be prepared in accordance with other procedures. Bangham et al., J. Mol. Biol., 13:238-252, (1965); Gregoriadis (Ed), In. Drug Carriers in Biology and Medicine, pp 287-341 (1979); Deamer et al., "Liposome Preparation: Methods and Mechanisms," In. Liposomes, M. Ostro (Ed.) (1983);
and Szoka et al., Proc. Nat'l Acad. Sci. USA, 75:4194-4198 (1978)(the contents of all are incorporated herein by reference). The aforementioned methods may differ in their respective abilities to entrap aqueous material and their respective aqueous space-to-lipid ratios.
The dried lipids or lyophilized liposomes prepared as described above may be reconstituted in a solution of nucleic acid and diluted to an appropriate concentration with an suitable solvent, e.g., DPBS. The mixture is then vigorously shaken in a vortex mixer.
Unencapsulated nucleic acid is removed by centrifugation at 29,000xg and the liposomal pellets washed. The washed liposomes are resuspended at an appropriate total phospholipid concentration, e.g., about 50-200 mM. The amount of nucleic acid encapsulated can be determined in accordance with standard methods. After detern-iination of the amount of nucleic acid encapsulated in the liposome preparation, the liposomes may be diluted to appropriate concentration and stored at 4 C until use.
In one embodiment, a lipid dioleoylphosphatidylchoine is used to create a liposome formulation. In one embodiment, compositions having affinity for microbial spore surface peptides are mixed with lipids in the presence of excess t-butanol. The mixture was vortexed before being frozen in an acetone/dry ice bath. The frozen mixture was lyophilized and hydrated with HEPES-buffered saline (1 mM HEPES, 10 mM
NaCl, ph17.5) overnight, and then the liposomes were sonicated in a bath type sonicator for 10 to 15 min. The size of the peptide-liposomes typically range between 200-300 nm in diameter as determined by the submicron particle sizer autodilute model 370, (Nicomp, Santa Barbara, Calif.).

Purified compositions comprising an infection control composition having affinity for a spore surface peptide may be used without further modifications or it may be diluted in a pharmaceutically acceptable carrier. The infection control composition may be used independently or in combination with other antimicrobial drugs. Because of the stability of the compositions it is contemplated that the invention may be administered to humans or animals, included in food preparations, pharmaceutical preparations, medicinal and pharmaceutical products, cosmetic products, hygienic products, cleaning products and cleaning agents, as well as any material to which the peptides could be sprayed on or adhered to wherein the inhibition of microbial growth is desired.

Experimental The following examples are included to demonstrate preferred embodiments of the invention. These examples are intended to represent techniques that are capable of practicing specific embodiments of the present invention. However, 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.

Example I
Spore Surface Pe to ide Synthesis Spore surface peptides contemplated by the present invention may be synthesized by the solid-phase method employing an Applied Biosystems model 433A peptide synthesizer and Fastmoc strategy at the 0.1 mM scale. Peptide purification may be performed by reversed-phase HPLC on a Waters Delta Prep employing a Vydac 218TP 1022 (22 x 250 mm) column. Separation is performed with a gradient system of aqueous 0.1% trifluoroacetic acid (solvent A) and 100% acetonitrile containing 0.085%
trifluoroacetic acid (solvent B). A linear gradient from 0 to 100% B is applied over 70 min and fractions collected every 0.2 min. Fractions are subsequently monitored by analytical scale reversed-phase HPLC on a Beckman Gold System using a Vydac 218TP54 (4.6x250 mm) column at a flow rate of 0.5 ml/min under isocratic elution conditions. Select fractions are pooled and lyophilized; further characterization of peptides are provided by mass spectrometry and capillary electrophoresis. Mass measurements are performed by flow injection at 0.1 ml/min in 64% acetonitrile containing 0.05% trifluoroacetic acid with a Hewlett-Packard model 1100 MSD
equipped with an electrospray ionization source.

Capillary electrophoresis is performed on a Hewlett-Packard 3D instrument equipped with an extended light path fused-silicate column 75 micrometers (ID) x 80.5 centimeters (total length). Capillary electrophoresis experiments are conducted at 18 C in 100 mM sodium phosphate buffer, pH 2.9 at 20,000 volts. Peptide concentration is determined by quantitative amino acid analysis on a Beckman 6300 Amino Acid analyzer.

Example II
Bacterial Strains And Spore Preparation The following Clostridium difficile strains were used: ATCC 700057, ATCC
BAA-1382 (Strain 630), ATCC 43594, and ATCC 9689. Spore crops were produced by anaerobic growth on Brucella blood agar oxyrase plates (BBAO) at 35 C for 2 days, followed by restreaking on BBAO. Spores were harvested into 15 ml 1 x PBS and 0.1 %
Tween-80 (PBST) after growing for 4 days at 35 C. Spores were centrifuged at 3000 x g for 20 min at 4 C, washed 3 times with 15 ml of (PBST), and resuspended in 10 ml HistoDenz (Sigma). The suspension was layer on top of 15 ml 50% HistoDenz solution to form a gradient. The gradient was centrifuged at 5000 x g for 15 min at 4 C. The pellet was washed three times in 15 ml PBST, previously described, and resuspended in 5 ml PBST. The sample was then sonicated two times for 5 s at power level 0.5 to lyse vegetative mother cells. The HistoDenz procedure, previously described, was then repeated. Final spore suspensions were stored at 4 C.

Example III
Thin Section Transmission Electron Microscopy The following procedure was used for sample prep and Electron Microscopy:
1. Fix samples with 2.5% glutaraldehyde, 2% paraformaldehyde, 0.01 M
phosphate buffer, 2.5% DMSO at room temperature for 2 hours.
2. Wash in 0.1 M phosphate buffer (3 changes over 15-30 minutes elapsed time).
3. 1% aqueous osmium tetroxide+1.5% potassium ferrocyanide for 1 hat RT.
4. Wash in DDW (3-4 changes over 15-30 minutes elapsed time) at RT.
5. Block staining: 0.5% aqueous uranyl acetate for overnight at 4C.

6. Dehydration (i.e., for example, with ethanol), each step for 10- 15 min.:
a) 30%; b) 50%; c) 70% (possible for overnight at 4 C); d) 80%; e) 90%; f) 95 %; g) 99%; and h) 100%
7. Propylene Oxide (PO): each step for 15-30 min a) Ethanol:PO = 50:50 h) Ethanol:PO = 25:75 c) PO = 100%
8. Epon: each step for 2 h a) Epon:PO = 30:70 b) Epon:PO = 50:50 c) Epon:PO = 75:25 d) Epon = 100%
e) Epon = 100%
9. Polymerization, i.e., for example by heating @ 60 ---70 C for 48 hr.
10. Sectioning 11. Uranyl acetate and Lead citrate staining 12. Carbon coating.
13. EM observation (JEOL 1200EX or Zeiss 902) Example IV
Clostridium dif icile Exosporium Preparation Approximately 109 spores were passed once through a chilled (4 C) French press at 20,000 lb/in2. See, Figure 17. Transmission electron microscopic (TEM) examination confirmed that this procedure removed large exosporium fragments without detectable damage to the remainder of the spore when compared to unprocessed control exosporium fragments. The photographs compare spores before and after stripping along with purified exosporium attached.. See, Figure 18A versus Figure 18B. The sample extruded from the French press was centrifuged for 5 min at 9,000 x g and 4 C, and the supernatant was saved. The pellet was resuspended in 10 ml of cold TEP buffer, and centrifuged as described above. The pellet was resuspended in 10 ml of TEP Buffer and stored at -20 C
while the supernatant was combined with the supernatant of the previous spin and centrifuged for 15 min at 9.500 x g and 4 C.
The resulting supernatant was then concentrated by centrifugation for approximately 60 min at 5000 x g and 4 C using Amicon Ultra Centrifugal Filter Devices (Millipore), final volume, 500 l. Electron microscopic examination of this exosporium sample confirmed the virtual absence of spores. See, Figure 18C.

Example V
Exosporium Solubilization. Protein Electrophoresis, And Immunoblotting Exosporium samples, IOj11, were solubilized by boiling for 5 min in 45 l of 2x Novex Tris-Glycine SDS Sample Buffer (Invitrogen), 35 .tl water and 10 l 200 mM
dithiothreitol. Solubilized proteins were separated by SDS-polyacrylamide gel electrophoresis (PAGE) with 4-12% Tris-Glycine gels. For immunoblotting, proteins were electrophoretically transferred from an SDS-polyacrylamide gel to a polyvinylidene difluoride membrane and treated as described in the manual for the Invitrogen Western Breeze assay kit. Briefly, each blot was blocked, probed with 1:1000 dilution of the primary MAb for I h, and washed. The blot was then probed with alkaline phosphatase conjugated, goat anti-rabbit IgG secondary antibody for 30 min, washed, and developed with the alkaline phosphatase developer solution.
Example VI
Amino-Terminal Protein Sequencing And Mass Spectrometry The SDS-PAGE pieces washed with water and dehydrated in acetonitrile. The bands were then alkylated with iodoacetamide prior to the in-gel digestion.
All bands were digested in-gel by incubating overnight at room temperature in trypsin, 5 tL 20 ng/ l trypsin in 50 mM ammonium bicarbonate. Formed peptides were extracted from the polyacrylamide in two aliquots of 30p.L 50% acetonitrile with 5% formic acid. These extracts were combined and evaporated to <10 tl in Speedvac and then resuspended in 1 % acetic acid to make up a final volume of -30 [it for LC-MS analysis.
The LC-MS system (Finnigan LTQ) contained a self-packed 9 cm x 75 m id Phenomenex Jupiter C 18 reversed-phase capillary HPLC column. Ten l volumes of the extract were injected and the peptides eluted were introduced into the source of the mass spectrometer on-line. The digest was analyzed using the data dependent multitask capability of the instrument acquiring full scan mass spectra to determine peptide molecular weights and product ion spectra to determine amino acid sequence in successive instrument scans. This mode of analysis produces approximately 2500 collisionally induced dissociation (CID) spectra of ions ranging in abundance over several orders of magnitude.

The data were analyzed by using all CID spectra collected in the experiment to search the NCBI non-redundant database with the search program Mascot using mammalian taxonomy filter. All matching spectra were verified by manual interpretation.
The interpretation process was aided by additional searches using the programs Sequest and Blast as needed (Cleveland Clinic for Mass Spectrometry and Protein Sequencing).
Example VII
CD Spectroscopy Circular dichroism (CD) spectroscopy may be performed on an AVIV 62 DS
spectrometer (AVIV Associates, Lakewood, N.J.) equipped with a thermoelectric temperature control at 25 C. Samples contained 0.11 - 0.24 mglmL peptide in either 50 mM sodium phosphate, pH 7.0; some samples also contain 40% trifluoroethanol or 0.1%
(0.22 mM) lipopolysaccharide (LPS). Spectra are collected at 0.5 nm intervals with an averaging time of 2 s per datum point using a pathlength of 0.1 cm. The spectra are smoothed once over an interval of five data points prior to plotting with each spectrum representing the average of two scans. The average of two buffer scans are subtracted prior to data smoothing. The fractional helical content are calculated from the ratio of observed mean residue ellipticity to the mean residue ellipticity for a 100%
helical peptide of identical length at 25 C. (Luo and Baldwin (1997).
Example VIII
Calculation of Peptide Hydrophobic Moment and Hydrophobicity Peptide hydrophobicities and hydrophobic moments are calculated by using normalized consensus hydrophobicity scales. Eisenberg et al., J Cell Biochem 31: 11-7 (1986).

Example XI

Clostridium sporogenes Exosporium Removal Exosporia are removed from C. sporogenes spores using homogenization in a Braun homogenizer was for 30-s intervals up to a total of 150 s, with 120 to 150 s required for optimal removal of exosporia as determined by crystal violet staining and light microscopy. Du et al., "Bacillus thuringiensis HD-73 spores have surfaced localized Cry IAe toxin: physiological and pathogenic consequences" Appl. Environ.
Microbiol.
62:3722-3726 (1996).
Spores are extracted to remove spore surface and exosporium components.
Aronson et al., "Comparative structural and functional aspects of spore surfaces" In:
Spores VII. American Society for Microbiology, p. 54-61, G. Chambliss and J.
C. Vary (ed.), Washington, D.C. (1978). Spores of C. sporogenes (1.1 x 109) are extracted in 5 mM cyclohexylaminoethane sulfonic acid-8 M urea-50 mM P-mercaptoethanol-0.8%
sodium dodecyl sulfate (SDS) at pH 9.8 for 90 min at 378C. Spores are centrifuged at 12,000 x g for 10 min, and the supernatant extract boiled for 5 min. Extracts are applied directly to SDS-polyacrylamide gels containing 6 M urea. For molecular weight determination, gels are transferred to 0.2-mm-pore size nitrocellulose. Blots are blocked with 3% bovine serum albumin in TBS, pH 7.4. The blots are then incubated with the appropriate biotinylated antibody (i.e., for example, C33 and/or C225, or D89 for C.
sporogenes) diluted 1:1,000 in TBS containing 0.05% Tween 20. Avidin labeled peroxidase (Sigma A7419) diluted 1:1,000 in TBS-0.05% Twcen 20 is used to detect the antigens with the insoluble substrate 4-naphthol (Bio-Rad) Glycoprotein determination is performed using periodic acid-Schiff staining.
The gels are fixed overnight and treated with 0.7% periodic acid for 2 h and then with 0.2%
metabisulfite for 2 h. Gels are then exposed to a Schiff reagent comprising basic fuchsin, sodium metabisulfite, and hydrochloric acid (Fisher Scientific, Pittsburgh, Pa.) The mobility of proteins that stain positive as glycoproteins from the spore extracts are determined and compared to the mobility of the antigens determined by Western blotting.
Ultrathin (ca. 100-mm) cryosections of spores for immunological labeling with colloidal gold are prepared. Chang et al., "Immunocytochemical localization of antigens in B. cereus T spores" J. Rapid Methods Automat. Microbiol. 2:229-233 (1993).
C.
sporogenes PA3679 spores from the same stock as had been used for the antigen are used for these experiments. The cryosections are floated on PBS containing 5% fetal calf serum, followed in sequence by PBS, PBS containing 0.1 M ammonium chloride, and PBS, each for 5 to 10 min at room temperature. Sections are floated on drops of primary reagent (the appropriate monoclonal antibody diluted 1:100) for I to 2 h, followed by washing in PBS six times for 5 min each. Sections are then floated on drops of secondary reagent for 20 to 30 min. The secondary reagent is goat anti-mouse IgG plus goat anti-mouse IgM conjugated to 10-nm-diameter colloidal gold used at a dilution of 1:10.
Sections are again washed six times in PBS, followed by three washes with distilled water. They are stained and embedded in methylcellulose by floating them on drops of 2.3 M methylcellulose containing about 20% (vol/vol) saturated aqueous uranyl acetate.
Grids were picked up in loops, drained, and air dried. They are viewed in a Philips EM
300 electron microscope. Negative controls are spores treated with monoclonal antibodies of the IgGi and IgM isotypes which did not react with bacterial spores.

Example X
Cup-Scrub Assay Of Clostridium difficile Spores Bound To A Surface This example describes one embodiment of a method that can assay spores that are bound to a surface. In this example, C. difficile spores are bound to a surface and assayed by the cup-scrub method. ATSM International, "Standard Test Method for Recovery of Microorganisms From Skin using the Cup Scrub Technique" E 1874 -(2009).

Materials Sterilizer-Any suitable steam sterilizer capable of producing the conditions of sterilization.
Scrub Cups-Sterile cylinders of suitable composition, preferably with rod handles to facilitate stabilization, height approximately 2.5 cm, inside diameter of convenient size. Useful sizes range from approximately 1.5 to 4.0 cm.
Polished Glass Rod or Rubber Policeman-Can be fashioned in the laboratory or purchased.

Pipettor-With disposable tips to deliver appropriate volume(s).
Sterile Beakers, Test Tubes or other container, to receive the cup scrub fluid.
Microbial Cultures-This test method can be used within a protocol targeting any microbial culture (i.e., for example, a C. difficile microbial culture).
Sampling and Dilution Fluid-Sterile Butterfield's phosphate buffered water or other recovery fluid of suitable composition; this should contain an antimicrobial inactivator specific for any antimicrobial that might be on the test site;
inactivator efficacy should be determined by Test Method Test Control and Baseline Skin Sites Skin sites are selected that are appropriate for C. difficile growth, providing contralateral sample sites for use as controls. A number of subjects (human or animal) selected for the assay depends on the statistical confidence needed for the expected test results, the variability encountered in the study, and the relative efficacy of any antibacterial agent that may be evaluated. There may be multiple sites available on subjects; randomization is required to suppress sample bias. The number of replicates required to discriminate effects will depend in part on the appropriateness and design of controls within the protocol. The use of this technique on isolated skin or equivalents is dependent on securing the test site in order to effectively perform the procedure.
In Vivo Sample Collection The area to be sampled is delineated by a sterile sampling cylinder. The cylinder is pressed firmly against the skin surface during sampling to ensure that the sampling fluid does not leak from the sampling site. A minimum 1.5-mL aliquot of sterile sampling fluid, with or without product neutralizers, is pipetted into the cylinder. The entire area is then scrubbed with moderate pressure for 60 f 6 s using a sterile polished glass rod or policeman. After scrubbing, the sampling fluid is transferred by pipette into a sterile sample tube. This procedure is repeated once more with a fresh aliquot of sampling fluid. The sampling fluids are pooled. This procedure is repeated for each sampling site. The same pipettes, cylinders, glass rods, and policeman are used for both washes of a site, but new sterile equipment is used for each site. After samples are collected, paper toweling is used to blot the site dry. Care must be taken during this process to prevent the sampling fluid from spilling into an adjacent site that has not been sampled. Following all sampling, when using marker organisms, the sampling site should be decontaminated using 70 to 90 % isopropanol or equivalent, followed by a 4 %
chlorhexidine scrub.

Isolated Skin or Skin Equivalent Sample Collection Quantitative microbial counts are obtained by the cup scrub technique. This procedure is used for test and control samples. Samples are positioned and secured as necessary to enable placement and effective use of the sampling cylinder. The area to be sampled is delineated by a sterile sampling cylinder. The cylinder is pressed firmly against the sample surface during sampling to ensure that the sampling fluid does not leak from the sampling site. A minimum 1.5-mL aliquot of sterile sampling fluid, with or without product neutralizers, is pipetted into the cylinder. The entire area is then scrubbed with moderate pressure for 60 6 s using a sterile polished glass rod or policeman. After scrubbing, the sampling fluid is transferred by pipet into a sterile sample tube. This procedure is repeated once more with a fresh aliquot of sampling fluid. The sampling fluids are pooled. This procedure is repeated for each sampling site. The same pipettes, cylinders, glass rods, and policeman are used for both washes of a site, but new sterile equipment is used for each site. If there are multiple sample sites on the same piece of isolated tissue, care must be taken during this process to prevent the sampling fluid from spilling into an adjacent site that has not been sampled.
Microbial Counts Each sample is mixed thoroughly. Tenfold serial dilutions of each sample are prepared in dilution fluid. Duplicate quantitative pour or spread plates using soybean-casein digest agar with suitable neutralizer are prepared. Incubate plated samples at suitable growth temperature, 62 C for 24 to 72 h, or until colonies are visible on the plates.

Example XI
Spore Adherence Assays - Skin Proteins This example provides one embodiment to assay spore adherence to a skin protein. For purposes of illustration, the example describes the generation of a involucrin skin protein to study adherence of C difficile.

Plasmids Compatible plasmids for this assay may including but not limited to, plsdA IsdA overexpression vector, pMK4 E. coli-S. aureus shuttle vector, pSR0001 rlsdA complementation vector, pNZ8148 L. lactis expression plasmid, pLIsdA
IsdA
expression vector, pL NEAT IsdA_NEAT expression vector, pL_C IsdA_C expression vector, pQE30 Overexpression plasmid (Qiagen), pHurnan K, pQE30 human cytokeratin expression, pCMC-SPORT6-INV Involucrin cDNA clone 4749952 (IMAGE
consortium, MRC Geneservice), pQE30-INV 6 His-tagged involucrin, or pET11a-LOR
Loricrin cDNA clone. Clarke et al., "IsdA of Staphylococcus aureus is a broad spectrum, iron-regulated adhesin" Mol. Microbiol. 51:1509-1519 (2004); Clarke et at., "The 5 Staphylococcus aureus surface protein IsdA mediates resistance to innate defenses of human skin"
Cell Host Microbe 1:199-212 (2007); Sullivan et al., "New shuttle vectors for Bacillus subtilis and Escherichia coli which allow rapid detection of inserted fragments" Gene 29:21-(1984); Kuipers, O. P., P. G. G. A. de Ruyter, M. Kleerebezem, and W. M. de Vos.
10 1998. Controlled overproduction of proteins by lactic acid bacteria. J.
Biotechnol.
64:15-21; Sullivan et al., "New shuttle vectors for Bacillus subtilis and Escherichia coli which allow rapid detection of inserted fragments" Gene 29:21-26 (1984); and Walsh et al., "Clumping factor B, a fibrinogen binding MSCRAMM (microbial surface component recognizing adhesive matrix molecules) adhesin of Staphylococcus aureus also binds the tail region of type I cytokeratin 10" J. Biol. Chem. 279:50691-50699 (2004).

Clostridium difficile is grown in a Luria-Bertani medium, using selection with kanamycin (50 g/ml) where appropriate. C. difficile strains are grown in brain heart infusion medium (Oxoid) or chemically defined CL medium. Horsburgh et al., "In Staphylococcus aureus, Fur is an interactive regulator with PerR, contributes to virulence, and is necessary for oxidative stress resistance through positive regulation of catalase and iron homeostasis" J. Bacteriol. 183:468-475 (2001).. When included, antibiotics are added at the following concentrations: erythromycin, 5 g/ml; lincomycin, 25 tg/ml; and tetracycline, 2.5 g/ml. C. difficile cultures may be grown at 37 C.

Cloning the involucrin gene The coding sequence for involucrin is amplified from pCMV-SPORT6-INV by PCR employing Pfu polymerase and the forward primer CGCGGATCCATGTCCCAGCAA CACACAC (SEQ ID NO: 446), incorporating a BamHI site (underlined), and the reverse primer CCCAAGCTTTATTTATGTTTGGG

TGGCCAC (SEQ ID NO: 447), incorporating a HindIIl site (underlined). The fragment is cloned into pQE30 cut with BamHI and HindIII, forming pQE30-INV.

Expression and purification of recombinant proteins.
The expression of hexahistidine-tagged recombinant IsdA (rlsdA), cytokeratin 10, and involucrin is induced by the addition of 100 M isopropyl-(3-D-thiogalactopyranoside (IPTG) to growing cells. Purification is achieved by using nickel chelate chromatography using the Hi-Trap system (Amersham Biosciences). Recombinant loricrin may be affinity tagged or purified from lysed E. coli T4037.
ELISA analysis of ligand binding.
An enzyme-linked immunosorbent assay (ELISA) is used to analyze the ability of rlsdA to bind ligands. Clarke et al., "'Analysis of Ebh, a 1. 1 -megadalton cell wall-associated fibronectin-binding protein of Staphylococcus aureus" Infect. Immun 70:6680-6687 (2002). Briefly, 100 l of appropriate ligand or bovine serum albumin (BSA) in phosphate-buffered saline (PBS) (5 g/ml) is added to wells of a 96-well microtiter plate (Nunc) overnight at 4 C. Plates are washed three times with PBST (PBS
containing 0.05% [vol/vol] Tween 20), and the remaining protein-binding sites are blocked with 5%
(wt/ vol) BSA in PBS for 2 h at room temperature. The plates are again washed three times in PBST, and different concentrations of purified rlsdA (i.e., for example, 0 to 15 M) diluted in PBS-0.1% (wt/vol) BSA were added. The plates are incubated for a further I h before being washed three times, and anti-IsdA (diluted 1:1,000) is added in PBS-0.1 % (wt/vol) BSA, followed by 1 h of incubation.
Mouse antibodies used for the detection of bound IsdA may be raised in accordance with reported methods. Clarke et al., "IsdA of Staphylococcus aureus is a broad spectrum, iron-regulated adhesin" Mol. Microbiol. 51:1509-1519 (2004).
The plates are washed an additional three times, and alkaline phosphatase conjugated anti-mouse antibodies diluted 1:30,000 in PBS-0.1% (wt/vol) BSA is added and the plates incubated for I h. Finally, bound antibodies are detected by using the Sigma Fast p-nitrophenyl phosphate system (Sigma). Plates are read at 405 rim, in a Victor microtiter plate reader (Wallac).
Studies of the inhibition of binding are performed by the incubation of rlsdA
with various concentrations of inhibitor or BSA (0 to 10 M) for I h at room temperature in PBS-0.1% (wt/vol) BSA. The reaction mixtures then are added to ligand-coated wells, and bound protein is detected.

Atomic Force Microscopy Atomic force microscopy (AFM) tips and supports are functionalized with TsdA
and loricrin molecules as follows. AFM cantilevers (Microlevers; Veeco Metrology Group, Santa Barbara, CA) and silicon wafers (Siltronix, France) are coated using electron beam thermal evaporation with a 5-rim-thick Cr layer followed by a 30-nm-thick Au layer. Before use, the gold-coated surfaces are rinsed with ethanol, dried with a gentle nitrogen flow, and cleaned for 15 min by UV/ozone treatment (Jelight Co., Irvine, CA).
Gold tips and gold supports are immersed overnight in ethanol solutions containing 0.05 mM nitrilotriacetate-terminated (5%) (Prochimia) and tri(ethylene glycol) [tri(EG)]-tenninated (95%) alkanethiols and rinsed with ethanol. Sonication is briefly applied to remove alkanethiol aggregates that can be adsorbed. The SAM (self-assembled mono layer) -coated surfaces then are immersed in a 40 mM aqueous solution ofNiSO4 (pH 7.2) for 1 h and rinsed with PBS. Finally, the samples are incubated in PBS with 2 ELM His-tagged IsdA for 2 h and further rinsed several times with PBS. For loricrin and BSA, gold supports are immersed for 36 h in an ethanol solution containing a 20 p.M
mixture (95:5, mol/mol) of alkanethiols terminated with oligo(EG) (OEG) and OEG
propionic acid. Following a rinse with ethanol, sonication is briefly applied to remove alkanethiol aggregates that may be adsorbed. The supports are immersed for 30 min into MilliQ water (Millipore) containing 20 g/liter N-hydroxysuccinimide (NHS) (Aldrich) and 50 g/liter 1-ethyl-3-(3-dimethylaminopropyl)- carbodiimide (EDC) (Sigma), rinsed with MilliQ water, incubated with 10 kg/liter loricrin or BSA (sigma) in PBS
for 3 h, further rinsed, and then used immediately.
AFM images and force-distance curves were obtained in PBS at room temperature using a Picoforcc Multimode AFM (Veeco Metrology Group, Santa Barbara, CA).
The supports were immobilized on a steel sample puck using a small piece of adhesive tape.
The mounted samples were immediately transferred into the AFM liquid cell while avoiding dewetting. All force curves were recorded with a maximum applied force of 450 pN. The spring constants of the cantilevers were measured using the thermal noise method (Picoforce; Veeco Metrology Group), yielding values (0.011 N/m) that were slightly larger than those announced by the manufacturer (0.01 Nhn). To account for the flexibility of the biomolecules, loading rates (in piconewtons per second) were estimated by multiplying the tip retraction velocity (in nanometers per second) by the slope of the rupture peaks (in piconewtons per nanometer).

Microbial interactions with immobilized ligands The binding of mid-log growth-phase cells to immobilized ligands may be measured using a previously described method. Ni Eidhin et al., Clumping factor B
(C1fB), a new surface-located fibrinogenbinding adhesin of Staphylococcus aureus" Mol.
Microbiol. 30:245-257 (1998). Further, squamous cells may be harvested from the anterior nares of healthy donors, and the binding of C. difficile grown in iron-free medium is assayed. O'Brien et al., "Staphylococcus aureus clumping factor B (ClfB) promotes adherence to human type I cytokeratin 10: implications for nasal colonization"
Cell.
Microbiol. 4:759-770 (2002). Comparisons between strains were performed using Student's t test.

Example XII
Expression and Purification of Recombinant Exosporium Proteins Expression constructs were created containing recombinant exosporium proteins with a N-terminus poly-Histidine tag and transformed in E. coli strains.
Recombinant proteins were produced by inoculating 1-liter cultures of Luria broth (supplemented with 100 g/ml ampicillin) with 10 ml of an overnight culture of the expression construct described above. Following 2.5 h of growth at 37 C, isopropyl -P -D-thiogalactoside was added to a final concentration of 0.2 mM to induce protein expression and the cultures were allowed to grow for another 3 h. Bacteria were harvested by centrifugation, the supernatant decanted, and the cell pellets resuspended in PBS before being stored at -80 C. The suspension was later thawed in an ambient-temperature water bath for 30 min and the cells lysed using a French press. Insoluble cell debris was removed by centrifugation at 28,000 x g for 20 min, followed by filtration through a 0.45-mm membrane. Recombinant exosporium protein was then initially purified using metal-chelating chromatography. Bacterial lysates were applied to a 5-ml Ni2-charged HiTrap chelating column (Amersham Pharmacia Biotech) and bound protein eluted with a ml linear gradient of 0-200 mM imidazole in 4 mM Tris-HC1, 100 mM NaCl, pH
7.9, at a flow rate of 5 ml/min. Fractions corresponding to recombinant exosporium protein, as determined by SDS-PAGE, were pooled and dialyzed against 25 mM Tris-HCI, pH
8.0, before further purification by ion-exchange chromatography. Dialyzed protein was applied to a 5-ml HiTrap Q column (Amersham Pharmacia Biotech) and bound protein eluted with a 200-m1 linear gradient of 0-0.5 M NaCl in 25 mM Tris-IICI, pit 8.0, at a flow rate of 5 ml/min. Fractions containing purified exosporium protein were identified by SDS-PAGE and estimated to be 90% pure.

Example XIII
Exosporium Protein Adherence Assays Binding of exosporium proteins to human skin protiens and/or EpiDerm differentiated human keratinocytes was performed essentially as described in Example X
and XI above with the exception that FITC conjugated spores were replaced with the exosporium protein. Detection of bound protein was accomplished using fluorescein conjugated Streptavidin (Rockland Immunochemicals). The total fluorescence (F(0ta1) per well was measured after a 1-h incubation using a Fluoroskan II fluorescence reader (Labsystems, Beverly, MA), with X < = 495 nm and X,,, = 528 run.

Claims (79)

Claims We claim:
1. An infection control composition capable of blocking and/or inhibiting the binding of a microbial spore to a substrate surface.
2. The composition of Claim 1, wherein said composition comprises a small organic molecule.
3. The composition of Claim 1, wherein said composition comprises a protein.
4. The composition of Claim 3, wherien said protein comprises a spore surface peptide.
5. The composition of Claim 4, wherein said peptide is derived from a Clostridia spore surface protein.
6. The composition of Claim 5, wherein said Clostridia surface protein comprises a Clostridia difficile spore surface protein.
7. The composition of Claim 4, wherein said peptide is derived from a Bacillus spore surface protein.
8. The composition of Claim 7, wherein said Bacillus surface protein comprises a Bacillus anthracis spore surface protein.
9. The composition of Claim 6, wherein said Clostridium difficile spore surface peptide comprises SEQ ID NO: 1 or a homologue of SEQ ID NO: 1.
10. The composition of Claim 6, said Clostridium difficile spore surface peptide is selected from the group comprising SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, and SEQ ID NO: 15.
11. The composition of Claim 1, wherein said composition comprises an antibody.
12. The composition of Claim 11, wherein said antibody is a polyclonal antibody.
13. The composition of Claim 11, wherein said antibody is a monoclonal antibody.
14. The composition of Claim 1, wherein said composition comprises a synthetic peptide.
15. The composition of Claim 1, wherein said composition comprises an inorganic ion complex.
16. The composition of Claim 1, wherein said composition comprises a protein mimetic.
17. The composition of Claim 1, wherein said substrate surface comprises an inanimate surface.
18. The composition of Claim 1, wherein said substrate surface comprises an animate surface.
19. The composition of Claim 18, wherein said animate surface comprises a body tissue surface.
20. The composition of Claim 19, wherein said body tissue surface comprises an epithelial surface.
21. The composition of Claim 20, wherein said epithelial surface comprises a mammalian skin surface.
22. The composition of Claim 17, wherein said inanimate surface comprises stainless steel.
23. The composition of Claim 17, wherein said inanimate surface comprises ceramic.
24. The composition of Claim 17, wherein said inanimate surface comprises vinyl.
25. The composition of Claim 17, wherein said inanimate surface comprises tile.
26. The composition of Claim 1, wherein said composition further comprises at least one antimicrobial drug.
27. The composition of Claim 1, wherein said composition is suitable for human administration.
28. The composition of Claim 1, wherein said composition further comprises an antimicrobial skin cleaner.
29. The composition of Claim 28, wherein said skin cleaner comprises a handwash cleaner.
30. The composition of Claim 1, wherein said composition further comprises an antimicrobial surface cleaner.
31. A method, comprising: a) providing: i) a subject at risk for exposure to a microbe, wherein the exposure is likely to result in a microbial infection and/or colonization; and ii) an infection control composition; b) administering the composition to the subject before the microbial exposure, under conditions such that a microbial infection barrier is formed.
32. The method of Claim 31, wherein said microbial infection barrier reduces said microbial infection.
33. The method of Claim 31, wherein said microbial infection barrier prevents said microbial infection.
34. The method of Claim 31, wherein said microbe comprises a bacteria.
35. The method of Claim 34, wherein said bacteria is selected from the group comprising Clostridia, Bacillus, Desulfotomaculum, Sporolactobacillus, Sporosarcina, or Thermoactinomyces.
36. The method of Claim 34, wherein said bacteria comprises a bacterial spore.
37. The method of Claim 36, wherein said bacterial spore comprises at least one spore surface protein.
38. The method of Claim 37, wherein said Clostridium spore surface protein is a C.
difficile spore surface protein.
39. The method of Claim 37, the Bacillus spore surface protein is a B.
anthracis spore surface protein.
40. The method of Claim 31, wherein said infection control composition comprises an amino acid sequence.
41. The method of Claim 40, wherein said amino acid sequence is derived from a microbial spore surface protein.
42. The method of Claim 40, wherein said amino acid sequence has substantial homology to a microbial spore surface protein.
43. The method of Claim 31, wherein said infection control composition comprises is capable of inhibiting, antagonizing, disrupting, displacing, and/or blocking the binding of a microbial spore to a substrate surface.
44. The method of Claim 31, wherein said infection control composition further comprises an antagonistic compound, wherein said compound has affinity for said microbial spore surface protein amino acid sequence.
45. The method of Claim 31, wherein said composition further comprises a carrier.
46. The method of Claim 31, wherein said infection control composition further comprises at least one antimicrobial drug.
47. The method of Claim 31, wherein said subject at risk is a healthcare worker.
48. The method of Claim 31, wherein said subject at risk is a medical first responder.
49. The method of Claim 31, wherein said subject at risk is a medical patient.
50. The method of Claim 49, wherein said medical patient is in a healthcare setting.
51. The method of Claim 49, wherein said medical patient has been administered at least one antibiotic.
52. The method of Claim 31, wherein said subject at risk is a fireman.
53. The method of Claim 31, wherein said subject at risk is a policeman.
54. The method of Claim 31, wherein said subject at risk is a military personnel.
55. The method of Claim 31, wherein said subject at risk is a food handler.
56. The method of Claim 31, wherein said subject at risk is an inanimate surface and/or object.
57. The method of Claim 31, wherein said administering is selected from the group consisting of topical, oral, parenteral, pulmonary, anal, vaginal, ocular, and intranasal.
58. A method, comprising: a) providing: i) a subject having been exposed to a microbe, wherein said exposure resulted in a microbial infection; ii) an infection control composition; b) administering said composition to said subject after said microbial exposure, under conditions such that said microbial infection is reduced.
59. The method of Claim 58, wherein said microbe comprises a bacteria.
60. The method of Claim 59, wherein said bacteria comprises a bacterial spore.
61. The method of Claim 60, wherein said bacterial spore comprises at least one spore surface protein.
62. The method Claim 61, wherein said bacteiral spore surface protein is a C.
difficile spore surface protein
63. The method of Claim 61, wherein said bacterial spore surface protein is a B.
anthracis spore surface protein.
64. The method of Claim 58, wherein said infection control composition comprises is capable of inhibiting, antagonizing, disrupting, displacing, and/or blocking the binding of a microbial spore to a substrate surface.
65. The method of Claim 58, wherein said infection control composition further comprises an antagonistic compound, wherein said compound has affinity for said microbial spore surface protein amino acid sequence.
66. The method of Claim 58, wherein said composition further comprises a carrier.
67. The method of Claim 58, wherein said infection control composition further comprises at least one antimicrobial drug.
68. The method of Claim 58, wherein said subject at risk is a healthcare worker.
69. The method of Claim 58, wherein said subject at risk is a medical first responder.
70. A screening method comprising: a) providing; i) an isolated peptide, wherein the peptide is derived from a microbial spore surface protein; and ii) an infection control test compound suspected of having an interaction with the peptide; b) contacting the peptide with the infection control test compound; and c) detecting the interaction of the peptide with the infection control test compound.
71. The method of Claim 70, wherein said method further coinprises a substrate surface capable of attaching the isolated peptide.
72. The method of Claim 71, wlierein said substrate surface comprises pigskin.
73. The method of Claim 70, wherein said peptide is derived from a Clostridia spore surface.
74. The method of Claim 73, wherein said Clostridia spore surface comprises a Clostridia difficile spore surface.
75. The method of Claim 70, wherein said peptide is derived from a Bacillus spore surface.
76. The method of Claim 75, wherein said Bacillus spore surface comprises a Bacillus anthracis spore surface.
77. The method of Claim 70, wherein said peptide comprises a spore surface peptide.
76. The method of Claim 77, wherein said spore surface peptide comprises SEQ
ID
NO: 1 or a homologue of SEQ ID NO: 1.
78. The method of Claim 77, wherein said spore surface peptide is selected from the group comprising SEQ ID NO: 3, SEQ ID NO: 5, SEQ ID NO: 7, and SEQ ID NO: 15.
79. The method of Claim 70, wherein said peptide comprises an antibody.
CA2756760A 2009-03-27 2010-03-26 Compositions and methods for screening and using compounds antagonizing spore-surface interactions Abandoned CA2756760A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US16415409P 2009-03-27 2009-03-27
US61/164,154 2009-03-27
PCT/US2010/028902 WO2010126670A2 (en) 2009-03-27 2010-03-26 Compositions and methods for screening and using compounds antagonizing spore-surface interactions

Publications (1)

Publication Number Publication Date
CA2756760A1 true CA2756760A1 (en) 2010-11-04

Family

ID=43032739

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2756760A Abandoned CA2756760A1 (en) 2009-03-27 2010-03-26 Compositions and methods for screening and using compounds antagonizing spore-surface interactions

Country Status (13)

Country Link
US (1) US20100291100A1 (en)
EP (1) EP2411410B1 (en)
JP (1) JP5911421B2 (en)
KR (1) KR20110140128A (en)
CN (1) CN102388064A (en)
AU (1) AU2010241947B2 (en)
BR (1) BRPI1014121A2 (en)
CA (1) CA2756760A1 (en)
DK (1) DK2411410T3 (en)
ES (1) ES2544435T3 (en)
PT (1) PT2411410E (en)
TW (1) TW201039841A (en)
WO (1) WO2010126670A2 (en)

Families Citing this family (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9133251B2 (en) * 2008-02-22 2015-09-15 The Curators Of The University Of Missouri Bacillus based delivery system and methods of use
US20130243794A1 (en) * 2010-12-03 2013-09-19 Beth Israel Deaconess Medical Center, Inc. Methods for predicting and treating infection-induced illnesses and predicting the severity of infection-induced illnesses
WO2012092469A2 (en) * 2010-12-29 2012-07-05 Cangene Corporation Clostridium difficile antigens
GB2491117A (en) 2011-05-20 2012-11-28 Royal Holloway & Bedford New College Coat proteins from Clostridium and Bacillus species
US9573980B2 (en) 2013-03-15 2017-02-21 Spogen Biotech Inc. Fusion proteins and methods for stimulating plant growth, protecting plants from pathogens, and immobilizing Bacillus spores on plant roots
WO2014193237A2 (en) * 2013-05-30 2014-12-04 Academisch Ziekenhuis Leiden H.O.D.N. Lumc C diff protease
EP3060246A4 (en) * 2013-10-23 2017-08-16 The Rockefeller University Compositions and methods for prophylaxis and therapy of clostridium difficile infection
GB2525177A (en) * 2014-04-14 2015-10-21 New Royal Holloway & Bedford Vaccine
BR122023020863A2 (en) 2014-09-17 2024-01-30 Spogen Biotech Inc METHOD FOR STIMULATING PLANT GROWTH
BR112018070380A2 (en) * 2016-04-05 2019-02-05 Nch Corp bacterial spore germination method; and nutrient germination composition.
EP3565533A4 (en) 2017-01-05 2021-02-17 The University of Chicago Inhibition of enteric infection through the modulation of microbiota
CN109706101B (en) * 2019-02-01 2022-10-11 东华大学 Sporosarcina psychrophilum and application thereof
WO2020247755A1 (en) * 2019-06-05 2020-12-10 Biovector, Inc. Aptamers against clostridium difficile
CN110879209A (en) * 2019-12-09 2020-03-13 济南大学 Rapid detection method of gamma-aminobutyric acid
CN111909945B (en) * 2020-08-31 2022-01-25 南京工业大学 Method for improving protein expression efficiency in clostridium
US11898146B2 (en) * 2020-12-03 2024-02-13 Liv Process, Inc. Aptamers against Clostridium difficile, compositions comprising aptamers against Clostridium difficile and methods of using the same
KR20230162685A (en) * 2021-03-30 2023-11-28 아사히 가세이 가부시키가이샤 Recombinant polypeptide with acyl CoA compound reducing activity
CN113247946B (en) * 2021-04-22 2022-07-05 哈尔滨工业大学 Self-assembled nano biocatalyst, preparation method thereof and application thereof in butanol production
CN113278578B (en) * 2021-06-15 2022-09-16 广西大学 Transplanting method of rice blast bacterium conidia
CN113308431B (en) * 2021-06-15 2022-09-16 广西大学 Transplanting and dispersing method for botrytis cinerea conidia

Family Cites Families (58)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NL7018669A (en) * 1969-12-23 1971-06-25
USRE28778E (en) 1969-12-23 1976-04-20 West Laboratories, Inc. Phenolic synthetic detergent-disinfectant
NL154598B (en) * 1970-11-10 1977-09-15 Organon Nv PROCEDURE FOR DETERMINING AND DETERMINING LOW MOLECULAR COMPOUNDS AND PROTEINS THAT CAN SPECIFICALLY BIND THESE COMPOUNDS AND TEST PACKAGING.
US3817837A (en) 1971-05-14 1974-06-18 Syva Corp Enzyme amplification assay
US3939350A (en) 1974-04-29 1976-02-17 Board Of Trustees Of The Leland Stanford Junior University Fluorescent immunoassay employing total reflection for activation
US3996345A (en) * 1974-08-12 1976-12-07 Syva Company Fluorescence quenching with immunological pairs in immunoassays
US4275149A (en) * 1978-11-24 1981-06-23 Syva Company Macromolecular environment control in specific receptor assays
US4277437A (en) 1978-04-05 1981-07-07 Syva Company Kit for carrying out chemically induced fluorescence immunoassay
US4458066A (en) 1980-02-29 1984-07-03 University Patents, Inc. Process for preparing polynucleotides
US4376110A (en) 1980-08-04 1983-03-08 Hybritech, Incorporated Immunometric assays using monoclonal antibodies
US4366241A (en) 1980-08-07 1982-12-28 Syva Company Concentrating zone method in heterogeneous immunoassays
US4517288A (en) 1981-01-23 1985-05-14 American Hospital Supply Corp. Solid phase system for ligand assay
US4632772A (en) 1982-02-22 1986-12-30 Dexide, Inc. Mild antimicrobial detergent composition
US5051448A (en) 1984-07-24 1991-09-24 The Mclean Hospital Corporation GABA esters and GABA analog esters
US5506337A (en) 1985-03-15 1996-04-09 Antivirals Inc. Morpholino-subunit combinatorial library and method
US4683195A (en) * 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US5288486A (en) * 1985-10-28 1994-02-22 Calgon Corporation Alcohol-based antimicrobial compositions
CA1291031C (en) * 1985-12-23 1991-10-22 Nikolaas C.J. De Jaeger Method for the detection of specific binding agents and their correspondingbindable substances
US5010175A (en) 1988-05-02 1991-04-23 The Regents Of The University Of California General method for producing and selecting peptides with specific properties
US4981785A (en) 1988-06-06 1991-01-01 Ventrex Laboratories, Inc. Apparatus and method for performing immunoassays
US5169862A (en) 1989-07-07 1992-12-08 Peptide Technologies Corporation Analogs of viscosin and their uses
IE66205B1 (en) 1990-06-14 1995-12-13 Paul A Bartlett Polypeptide analogs
US5650489A (en) 1990-07-02 1997-07-22 The Arizona Board Of Regents Random bio-oligomer library, a method of synthesis thereof, and a method of use thereof
US5192746A (en) 1990-07-09 1993-03-09 Tanabe Seiyaku Co., Ltd. Cyclic cell adhesion modulation compounds
US5114978A (en) 1990-10-15 1992-05-19 Rhone-Poulenc Surfactants And Specialties, L.P. Anhydrous blends of p-chloro-m-xylenol in surfactant mixtures
US5376313A (en) 1992-03-27 1994-12-27 Abbott Laboratories Injection molding a plastic assay cuvette having low birefringence
US5573905A (en) 1992-03-30 1996-11-12 The Scripps Research Institute Encoded combinatorial chemical libraries
US5541061A (en) 1992-04-29 1996-07-30 Affymax Technologies N.V. Methods for screening factorial chemical libraries
US5288514A (en) * 1992-09-14 1994-02-22 The Regents Of The University Of California Solid phase and combinatorial synthesis of benzodiazepine compounds on a solid support
EP0604289B1 (en) 1992-12-17 1997-09-24 Valeo Systemes D'essuyage Method and device for supplying power to an electric motor for driving a windscreen wiper of a automotive vehicle
US5576220A (en) * 1993-02-19 1996-11-19 Arris Pharmaceutical Corporation Thin film HPMP matrix systems and methods for constructing and displaying ligands
US5559103A (en) 1993-07-21 1996-09-24 Cytel Corporation Bivalent sialyl X saccharides
US5539085A (en) 1993-08-20 1996-07-23 Onyx Pharmaceuticals, Inc. Bcl-2 and R-ras complex
US5599677A (en) 1993-12-29 1997-02-04 Abbott Laboratories Immunoassays for prostate specific antigen
US5519134A (en) * 1994-01-11 1996-05-21 Isis Pharmaceuticals, Inc. Pyrrolidine-containing monomers and oligomers
US5593853A (en) * 1994-02-09 1997-01-14 Martek Corporation Generation and screening of synthetic drug libraries
US5539083A (en) 1994-02-23 1996-07-23 Isis Pharmaceuticals, Inc. Peptide nucleic acid combinatorial libraries and improved methods of synthesis
US5525735A (en) * 1994-06-22 1996-06-11 Affymax Technologies Nv Methods for synthesizing diverse collections of pyrrolidine compounds
US5549974A (en) * 1994-06-23 1996-08-27 Affymax Technologies Nv Methods for the solid phase synthesis of thiazolidinones, metathiazanones, and derivatives thereof
US5635462A (en) 1994-07-08 1997-06-03 Gojo Industries, Inc. Antimicrobial cleansing compositions
US5576423A (en) 1994-12-02 1996-11-19 Schering Corporation Antibodies to the slam protein expressed on activated T cells
US5585639A (en) * 1995-07-27 1996-12-17 Hewlett-Packard Company Optical scanning apparatus
US5569588A (en) 1995-08-09 1996-10-29 The Regents Of The University Of California Methods for drug screening
CA2239957A1 (en) 1995-12-22 1997-07-03 Abbott Laboratories Fluorescence polarization immunoassay diagnostic method
US5885529A (en) 1996-06-28 1999-03-23 Dpc Cirrus, Inc. Automated immunoassay analyzer
US7655252B2 (en) * 1999-04-28 2010-02-02 The Regents Of The University Of Michigan Antimicrobial nanoemulsion compositions and methods
US20050271689A1 (en) * 2003-07-11 2005-12-08 Chun-Ming Huang Novel targets and compositions for use in decontamination, immunoprophylaxis, and post-exposure therapy against anthrax
CA2397075A1 (en) * 2000-02-01 2001-08-09 Arthur D. Little, Inc. Chemical and/or biological decontamination system
US6656919B1 (en) * 2002-01-11 2003-12-02 Clarence L. Baugh Method and a product for the rapid decontamination and sterilization of bacterial endospores
US7192601B2 (en) * 2002-01-18 2007-03-20 Walker Edward B Antimicrobial and sporicidal composition
SG149043A1 (en) * 2003-08-18 2009-01-29 Novacal Pharmaceuticals Inc N,n-dihalogenated amino acids and derivatives
GB0410958D0 (en) * 2004-05-15 2004-06-16 Haptogen Ltd Methods for reducing biofilm formation in infectious bacteria
US7658706B2 (en) * 2005-12-05 2010-02-09 Rti Biologics, Inc. Vascular graft sterilization and decellularization
GB0612456D0 (en) * 2006-06-23 2007-03-28 Secr Defence Antibody for anthrax
CN101190946A (en) * 2006-12-01 2008-06-04 中国人民解放军第二军医大学 Recombination plasmodium falciparum circumsporozoite surface protein and its preparation method and use
EP2271366A4 (en) * 2008-02-28 2012-06-20 3M Innovative Properties Co Antibodies to clostridium difficile spores and uses thereof
TWI552751B (en) 2011-06-20 2016-10-11 H 朗德貝克公司 Method of administration of 4-((1r,3s)-6-chloro-3-phenyl-indan-1-yl)-1,2,2-trimethyl-piperazine and the salts thereof in the treatment of schizophrenia

Also Published As

Publication number Publication date
US20100291100A1 (en) 2010-11-18
PT2411410E (en) 2015-10-09
TW201039841A (en) 2010-11-16
KR20110140128A (en) 2011-12-30
CN102388064A (en) 2012-03-21
WO2010126670A3 (en) 2011-03-31
DK2411410T3 (en) 2015-09-07
ES2544435T3 (en) 2015-08-31
AU2010241947A1 (en) 2011-11-17
WO2010126670A9 (en) 2011-06-30
EP2411410A2 (en) 2012-02-01
EP2411410B1 (en) 2015-07-08
AU2010241947B2 (en) 2014-01-16
JP2012522010A (en) 2012-09-20
WO2010126670A2 (en) 2010-11-04
BRPI1014121A2 (en) 2016-11-08
JP5911421B2 (en) 2016-05-18

Similar Documents

Publication Publication Date Title
AU2010241947B2 (en) Compositions and methods for screening and using compounds antagonizing spore-surface interactions
Coburn et al. Lyme disease pathogenesis
Tucker et al. Discovery of next-generation antimicrobials through bacterial self-screening of surface-displayed peptide libraries
Gut et al. Mechanism of inhibition of Bacillus anthracis spore outgrowth by the lantibiotic nisin
Murray The molecular basis of leptospiral pathogenesis
Saar-Dover et al. D-alanylation of lipoteichoic acids confers resistance to cationic peptides in group B streptococcus by increasing the cell wall density
Somboonwiwat et al. Recombinant expression and anti-microbial activity of anti-lipopolysaccharide factor (ALF) from the black tiger shrimp Penaeus monodon
Saito et al. Incorporation of exogenous fatty acids protects Enterococcus faecalis from membrane-damaging agents
Siroy et al. Global comparison of the membrane subproteomes between a multidrug-resistant Acinetobacter baumannii strain and a reference strain
Lenz et al. Identification of a second Listeria secA gene associated with protein secretion and the rough phenotype
Saka et al. Vibrio cholerae cytolysin is essential for high enterotoxicity and apoptosis induction produced by a cholera toxin gene-negative V. cholerae non-O1, non-O139 strain
KR20100126760A (en) Antibodies to clostridium difficile spores and uses thereof
Reder-Christ et al. Model membrane studies for characterization of different antibiotic activities of lipopeptides from Pseudomonas
Belcher et al. Pathogenicity and virulence of Bordetella pertussis and its adaptation to its strictly human host
Zhang et al. A novel autolysin AtlASS mediates bacterial cell separation during cell division and contributes to full virulence in Streptococcus suis
Bednarz-Misa et al. Application of zwitterionic detergent to the solubilization of Klebsiella pneumoniae outer membrane proteins for two-dimensional gel electrophoresis
KR20190028746A (en) Lantibiotics variants and uses thereof
CA2526753A1 (en) Sdr proteins from staphylococcus capitis and their use in preventing and treating infections
US6255080B1 (en) Bacteriocins produced by ruminal bacteria
US20160199475A1 (en) Live-attenuated vaccine against plague
Garcia et al. Lyme Disease Pathogenesis
Reimer Search for novel antimicrobials against\(Neisseria\)\(gonorrhoeae\) and\(Chlamydia\)\(trachomatis\)
Afonina Characterizing the relationship between Sortase A-dependent substrates and the consequences of Srta deletion in Enterococcus faecalis
Kumru Surface localization determinants of Borrelia burgdorferi lipoproteins
JP2001233899A (en) Antibacterial and antifungal peptide

Legal Events

Date Code Title Description
EEER Examination request

Effective date: 20150302

FZDE Discontinued

Effective date: 20180816