AU2006303998A1 - Targeted biocides - Google Patents

Targeted biocides Download PDF

Info

Publication number
AU2006303998A1
AU2006303998A1 AU2006303998A AU2006303998A AU2006303998A1 AU 2006303998 A1 AU2006303998 A1 AU 2006303998A1 AU 2006303998 A AU2006303998 A AU 2006303998A AU 2006303998 A AU2006303998 A AU 2006303998A AU 2006303998 A1 AU2006303998 A1 AU 2006303998A1
Authority
AU
Australia
Prior art keywords
antibody
composition
epitope
pct
cell
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
AU2006303998A
Inventor
Michael Imboden
Michael W. Riggs
Deborah Schaefer
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.)
Arizona Board of Regents of University of Arizona
ioGenetics LLC
Original Assignee
Arizona Board of Regents of University of Arizona
ioGenetics LLC
University of Arizona
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 Arizona Board of Regents of University of Arizona, ioGenetics LLC, University of Arizona filed Critical Arizona Board of Regents of University of Arizona
Publication of AU2006303998A1 publication Critical patent/AU2006303998A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/14Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from fungi, algea or lichens
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/20Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans from protozoa
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/10Immunoglobulins specific features characterized by their source of isolation or production
    • C07K2317/12Immunoglobulins specific features characterized by their source of isolation or production isolated from milk
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/60Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments
    • C07K2317/62Immunoglobulins specific features characterized by non-natural combinations of immunoglobulin fragments comprising only variable region components
    • C07K2317/622Single chain antibody (scFv)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/30Non-immunoglobulin-derived peptide or protein having an immunoglobulin constant or Fc region, or a fragment thereof, attached thereto
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/55Fusion polypeptide containing a fusion with a toxin, e.g. diphteria toxin
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Medicinal Chemistry (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Immunology (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)
  • Peptides Or Proteins (AREA)
  • Micro-Organisms Or Cultivation Processes Thereof (AREA)

Description

WO 2007/047189 PCT/US2006/039361 TARGETED BIOCIDES This application is a continuation in part of U.S. Patent application serial number 11/254,500, filed 10/20/2005, which claims priority to provisional patent application serial 5 number 60/620,642, filed 10/20/04, and which is a continuation in part of copending patent application serial number 10/844,837, filed 5/13/04, which claims priority to provisional patent application serial number 60/470,841, filed 5/15/03. The present application further claims priority to provisional patent application serial number 60/725,180, filed 10/11/2005. Each of the aforementioned patent applications is herein incorporated by reference in its 10 entirety. FIELD OF THE INVENTION The present invention relates to retroviral constructs that encode novel monoclonal antibodies, novel fusion proteins, and chimeric monoclonal antibodies and to methods of 15 using and producing the same. In particular, the present invention relates to methods of producing a fusion protein comprising a microorganism targeting molecule (e.g., immunoglobulin or innate immune system receptor molecule) and a biocide (e.g., bactericidal enzyme) in transgenic animals (e.g., bovines) and in cell cultures. The present invention also relates to therapeutic and prophylactic methods of using a fusion protein 20 comprising a microorganism targeting molecule and a biocide in health care (e.g., human and veterinary), agriculture (e.g., animal and plant production), and food processing (e.g., beef carcass processing). The present invention also relates to methods of using a fusion protein comprising a microorganism targeting molecule and a biocide in various diagnostic applications in number of diverse fields such as agriculture, medicine, and national defense. 25 BACKGROUND OF THE INVENTION The majority of people in the industrialized world have access to an abundance of inexpensive processed food products. The safety, quality, and wholesomeness of these products are usually unquestioned. The availability of inexpensive food products is largely 30 a result of advances in farm mechanization and improved industries of scale in food processing and distribution operations. The mechanization of the family fanr has not come without certain drawbacks however. 1 WO 2007/047189 PCT/US2006/039361 One of the drawbacks of large-scale food processing operations, and of meat processing in particular, is the occasional contamination (e.g., bacterial, fungal, etc.) and subsequent distribution of large quantities of contaminated products sometimes with dire consequences. Food safety researchers have determined that the introduction of even a few contaminated 5 carcasses into the production lines of large scale food processing operations is often enough to contaminate entire batches of product. The meat packing industry is particularly susceptible to carcass contamination during dehiding, evisceration, splitting, chilling, and fabrication. Further contamination of previously uncontaminated meat products may occur during grinding, processing, and transport. This type of contamination has lead to several 10 major meat product recalls, including the recall of 24 million pounds of ground beef by the Hudson Beef Co. in 1997, and more recently, the recall of 19 million pounds of beef and related products by the ConAgra Beef Company in July 2002. (See, Recall Release, FSIS RC-055-2002). The economic impact of food safety and spoilage is very large. USDA ERS estimates that the leading six bacterial food borne pathogens cause $2.9-6.7 billion in 15 medical costs and lost productivity annually in the US (Buzby et al., Bacterial Foodborne Disease: Medical Costs and Productivity Losses. 1996. Food and Consumer Economics Division, Economic Research Service U.S. Department of Agriculture. Agricultural Economic Report 741) Many meat product recalls are the result of contamination by the bacterium 20 Escherichia coli 0157:H7. This bacterium is commonly isolated from the gastrointestinal tract and feces of cattle. Direct contact with cattle can be a source of human infection. However, the principal route of transmission to humans is through fecal contamination of carcasses at slaughter. (J. Tuttle et al., Epidemiol Infect., 122:185-192 [1999]). Every year in the United States the 01 57:H7 bacterium causes about 70,000 cases of hemorrhagic 25 diarrhea and renal disease. Children, the elderly, and the immunocompromised are most susceptible to foodborne illness caused by Escherichia coli 01 57:H7. Virulent strains of Escherichia coli are not the only foodborne pathogens of concern. Listeria inonocytogenes has emerged as another dangerous, but relatively uncommon foodborne pathogen. Despite being an uncommon source of illness, L. monocytogenes is 30 ubiquitous in agricultural and food processing environments and can cause serious human and animal infections. The infection caused by L. monocytogenes is commonly called Listeriosis. Listeriosis occurs in sporadic and epidemic forms throughout the world. (See 2 WO 2007/047189 PCT/US2006/039361 e.g., B. Lorber, Clin. Infect. Dis., 24(1):1-9 [1997]; J.M. Farber et al., Microbiol. Rev., 55:476-511 [1991]; and W.F. Schlech, Clin. Infect. Dis., 31:770-775 [2000]). A multistate outbreak of Listeriosis has been reported in the United States. (Morb. Mortal. Wkly. Report, 49(50):1129-1130 [2000] erratum in Morb. Mortal. Wkly. Report, 50(6):101 5 [2001]). Since May 2000, 29 illnesses caused by a strain of Listeria monocytogenes have been identified in 10 states: New York (15 cases); Georgia (3 cases); Connecticut, Ohio, and Michigan (2 cases each); and California, Pennsylvania, TennesSee, Utah, and Wisconsin (1 case each). Listeriosis, in its most severe form, is an invasive disease that affects 10 immunocompromised patients and has the highest case-fatality rate of any foodborne illnesses. (B.G. Gellin et al., Amer. J. Epidemiol., 133:392-401 [1991]; D.B. Louria et al., Ann. NY Acad. Sci., 174:545-551 [1970]; J. McLauchlin, Epidemiol. Infect., 104:191-201 [1990]; V. Goulet and P. Marchetti, Scand. J. Infect. Dis., 28:367-374 [1996]; and C.J. Bula et al., Clin. Infect Dis., 20:66-72 [1995]). In immunocompetent persons, it can also cause 15 severe disease as well as outbreaks of benign febrile gastroenteritis. (P. Aureli et al., New Engl. J. Med., 342:1236-41 [2000]). Another form of human disease is perinatal infection, which is associated with a high rate of fetal loss (including full-term stillbirths) and serious neonatal disease (J. McLauchlin, Epidemiol. Infect., 104:181-190 [1990]). Most, perhaps all, of listeriosis in humans occurs after consumption of contaminated 20. food (e.g., meat and cheese) products. (A. Schuchat et al., J. Amer. Med. Assoc., 267:2041 2045 [1992]). While uncommon, Listeriosis causes about half the foodborne disease fatalities in the US each year. Additionally, many mild cases of listeriosis and inapparent Listeria infections go unreported. For those susceptible to listeriosis, ingestion of even small doses of L. monocytogenes is often sufficient for infection. About 2,500 cases of 25 listeriosis are reported in the US each year, of these about 20% or 500 cases are fatal. In 1989, the USDA FSIS implemented a testing program for L. monocytogenes in cooked meat products and adopted a zero tolerance position for L. monocytogenes contamination in ready to eat products. Guidelines promulgated by the American Association of Meat Processors for current Good Manufacturing Practices for Ready to Eat 30 meat products address the need for environmental monitoring for Listeria as a component of HACCP programs. The ecology of L. monocytogenes and its increasing prevalence and/or detection in food preparation establishments has lead to major recalls of processed meat 3 WO 2007/047189 PCT/US2006/039361 products. In October 2002 the USDA issued a recall notice, which when further expanded, constituted the largest meat product recall on record for 28 million pounds of processed turkey products (See, USDA FSIS Recall Notification Report 090-2002 EXP Recall from Pilgrims Pride Corp dba Wampler Foods Inc. 11-4-2002). The recall was in response to 5 detection of L. monocytogenes at multiple points in the facilities and equipment used to process the recalled turkey. A number of approaches have been tried to increase the safety and wholesomeness of the nation's meat and agricultural products. For example, some approaches have focused on the exposing food products to one or more types of pathogen destroying processes, 10 including ionizing radiation or ultra high temperatures and pressures. (See e.g., US 5,891,490; 6,013,918; 6,086,936; and 6,165,526 etc.). US 6,165,526 is representative of these approaches. This patent describes an UV radiation and ultra high temperature method for sterilizing food products. A number of other approaches have focused on providing mixtures of chemicals 15 (e.g., acids, surfactants, emulsifying agents, and organic phosphates) that inactivate bacteria and bacterial spores in food products. (See e.g., US 5,550,145; and 5,618,840). US 5,618,840, for instance, describes an antibacterial oil-in-water emulsion for inhibiting the growth of Helicobacter pylori. The various compositions and methods previously described for food sterilization 20 have certain advantages and certain other disadvantages. One disadvantage is that the manufacture and additional or large quantities of artificial chemicals to food products can be costly and logistically difficult. Moreover, the current chemical food sterilization agents are indiscriminate and are thus inappropriate for addition into food products such as cheese and yogurt that require the beneficial action of certain bacteria for their production. The 25 addition of artificial chemical compounds to food products or subjecting the products to irradiation or temperature and pressure extremes can also produce unpleasant organoleptic qualities. Another disadvantage is the publics' generally negative perception of food irradiation and the addition of chemical additives. Still other efforts have been directed to producing food washes to remove residual 30 surface impurities such as waxes and pesticides sometimes acquired during food product production, processing, and transporting. For instance, US 6,367,488 describes a chemical wash for fruits and vegetables made from surfactants, such as oleate, and alcohol 4 WO 2007/047189 PCT/US2006/039361 ethoxylates, and neutralized phosphoric acid. While these washes are useful for removing surface contaminates and surface bacteria from solid food products, these compositions are inappropriate for sterilizing homogenized food products such as ground beef. While each of these above-mentioned compositions and methods has particular 5 advantages and disadvantages, the need still exists for compositions and methods that reduce the amount of pathogenic bacteria shed by feedlot animals (e.g., bovines, porcines, and the like), that induce immunity in feedlot animals to pathogens, and for edible compositions that safety destroy harmful foodborne pathogens. A further major economic problem confronting the food processing industry is that 10 of bacterial spoilage. In particular, dairy and processed meat products are susceptible to bacterial spoilage by organisms such as the Lactic acid bacteria (e.g., Lactobacillus etc.) (Kraft AA. Health hazards vs. food spoilage. Boca Raton, FL.: CRC Press, Inc., 1992). These organisms are widely distributed in nature, and can easily out-compete other bacteria under low oxygen tension and low pH conditions that are common in processed dairy and 15 meat foods (Stainer. Lactic acid bacteria. In: Defigueiredo MP and Splittstoesser DF eds. Westport, CT: AVI Publishing, 1976). Over 20% of the fruit and vegetable products harvested for human consumption are believed to be lost to post-harvest microbial spoilage (Jay, J. Modern Food Microbiology 4 th ed Van Norstand Reinhold New York, 1992). SUMMARY OF THE INVENTION 20 The present invention relates to retroviral constructs that encode novel monoclonal antibodies, novel fusion proteins, and chimeric monoclonal antibodies and to methods of using and producing the same. In particular, the present invention relates to methods of producing a fusion protein comprising a microorganism targeting molecule (e.g., immunoglobulin or innate immune system receptor molecule) and a biocide (e.g., 25 bactericidal enzyme) in transgenic animals (e.g., bovines) and in cell cultures. The present invention also relates to therapeutic and prophylactic methods of using a fusion protein comprising a microorganism targeting molecule and a biocide in health care (e.g., human and veterinary), agriculture (e.g., animal and plant production), and food processing (e.g., beef carcass processing). The present invention also relates to methods of using a fusion 30 protein comprising a microorganism targeting molecule and a biocide in various diagnostic applications in number of diverse fields such as agriculture, medicine, and national defense. 5 WO 2007/047189 PCT/US2006/039361 Accordingly, in some embodiments, the present invention provides compositions comprising recombinant monoclonal antibodies that bind to a surface epitope of Ciyptosporidiumparvum, vectors and transgenic animal including the antibodies, and methods of using the antibodies to treat subjects infected with Cryptosporidiuiparvum. 5 For example, in some embodiments, the present invention provides a vector comprising a gene encoding a recombinant monoclonal antibody that binds to a surface epitope of Cryptosporidium sp. (e.g., Cryptosporidiunparvum or Cryptosporidiuin hominis), for example a CSL epitope, a p 23 epitope, a GP25-200 epitope or a beta mannosylated glycolipid epitope. In some embodiments, the recombinant monoclonal 10 antibody comprises a heavy chain variable region selected from the group consisting of a 3E2 variable region, a 18.44 variable region, a 1E10 variable region, and a 4H9 variable region. In some embodiments, the 3E2 recombinant antibody is a IgM isotype. In other embodiments, the recombinant antibody has a hybrid isotype, wherein the light chain has an isotype selected from the group consisting of an IgM isotype and an IgGl isotype and the 15 heavy chain has an variable region selected from the group consisting of an IgM isotype and an IgGl isotype and a constant region selected from the group consisting of an murine IgG2b isotype and an human IgG3 isotype. In some embodiments, the gene encoding a recombinant monoclonal antibody encodes an antibody heavy chain and an antibody light chain, and wherein the gene encoding an antibody heavy chain is fused to a gene encoding 20 an antimicrobial polypeptide. In some embodiments, the antimicrobial polypeptide is a LL37 polypeptide (e.g., encoded by nucleotides 1474-1584 of SEQ ID NO:102). In other embodiments, the antimicrobial polypeptide is lactoferrin hydrolysate, lactoferrin b, indolicidin, beta-defensin-2, deta-defensin-1, phopholipase A2, or phospho-inositol specific phospholipase C. 25 The present invention further provides a composition comprising a recombinantly expressed monoclonal antibody that binds to a surface epitope of Cyptosporidium sp. (e.g., a CSL epitope, a p23 epitope, a GP25-200 epitope or a beta-mannosylated glycolipid epitope). In some embodiments, the recombinant monoclonal antibody comprises a heavy chain variable region selected from the group consisting of a 3E2 variable region, a 18.44 30 variable region, a 1E10 variable region, and a 4H9 variable region. In some embodiments, the 3E2 recombinant antibody is a IgM isotype (e.g., a hexamer or a pentamer). In other embodiments, the recombinant antibody has a hybrid isotype, wherein the light chain has an 6 WO 2007/047189 PCT/US2006/039361 isotype selected from the group consisting of an IgM isotype and an IgG1 isotype and the heavy chain has an variable region selected from the group consisting of an IgM isotype and an IgG1 isotype and a constant region selected from the group consisting of an murine IgG2b isotype and an human IgG3 isotype. In some embodiments, the recombinant 5 monoclonal antibody comprises a heavy chain and a light chain and wherein the heavy chain is fused to an antimicrobial polypeptide. In some embodiments, the antimicrobial polypeptide is a LL37 polypeptide (e.g., encoded by nucleotides 1474-1584 of SEQ ID NO: 102). In other embodiments, the antimicrobial polypeptide is lactoferrin hydrolysate, lactoferrin b, indolicidin, beta-defensin-2, deta-defensin-1, phopholipase A2, or phospho 10 inositol specific phospholipase C. In preferred embodiments, the composition neutralizes Cryptosporidiun sp. sporozoite infectivity. The present invention also provides a method of treating a subject comprising: providing a recombinantly expressed monoclonal antibody that binds to a surface epitope of Cyptosporidium sp. (e.g., a CSL epitope, a p23 epitope, a GP25-200 epitope or a beta 15 mannosylated glycolipid epitope); and a subject suspected of being contaminated or infected with Cryptosporidium sp.; applying the recombinantly expressed monoclonal antibody that binds to a surface epitope of Cryptosporidium sp. to the subject under conditions such that the recombinantly expressed monoclonal antibody that binds to a surface epitope of Cryptosporidiun sp. neutralizes the Cryptosporidium sp. suspected of contaminating the 20 subject. In some embodiments, the recombinant monoclonal antibody comprises a heavy chain variable region selected from the group consisting of a 3E2 variable region, a 18.44 variable region, a 1E10 variable region, and a 4H9 variable region. In some embodiments, the 3E2 recombinant antibody is a IgM isotype (e.g., a hexamer or a pentamer). In other embodiments, the recombinant antibody has a hybrid isotype, wherein the light chain has an 25 isotype selected from the group consisting of an IgM isotype and an IgGl isotype and the heavy chain has an variable region selected from the group consisting of an IgM isotype and an IgGl isotype and a constant region selected from the group consisting of an murine IgG2b isotype and an human IgG3 isotype. In some embodiments, the recombinant monoclonal antibody comprises a heavy chain and a light chain and wherein the heavy chain 30 is fused to an antimicrobial polypeptide. In some embodiments, the antimicrobial polypeptide is a LL37 polypeptide (e.g., encoded by nucleotides 1474-1584 of SEQ ID NO: 102). In other embodiments, the antimicrobial polypeptide is lactoferrin hydrolysate, 7 WO 2007/047189 PCT/US2006/039361 lactoferrin b, indolicidin, beta-defensin-2, deta-defensin- 1, phopholipase A2, or phospho inositol specific phospholipase C. In some embodiments, the subject is a mammal (e.g., a ruminant (e.g., bovine) or a human). In yet other embodiments, the present invention provides a method of treating a 5 subject comprising: providing a composition comprising a 18.44 monoclonal antibody, and wherein the heavy chain of said antibody is fused to an antimicrobial polypeptide; and a subject suspected of being contaminated or infected with an apicomplexan parasite (e.g., Cryptosporidiun sp); applying the composition to the subject under conditions such that composition neutralizes the microorganism suspected of contaminating the subject. In 10 preferred embodiments, the antimicrobial polypeptide is a LL37 polypeptide (e.g., encoded by nucleotides 1474-1584 of SEQ ID NO: 102). In other embodiments, the antimicrobial polypeptide is lactoferrin hydrolysate, lactoferrin b, indolicidin, beta-defensin-2, deta defensin-1, phopholipase A2, or phospho-inositol specific phospholipase C. In some embodiments, the subject is a mammal (e.g., a human or a ruminant (e.g. bovine) or an 15 avian species. In yet other embodiments, the present invention provides a transgenic organism (e.g., an animal, a plant or a microorganism) comprising a gene encoding a recombinant monoclonal antibody that binds to a surface epitope of Cryptosporidium sp. (e.g., a CSL epitope, a p23 epitope, a GP25-200 epitope or a beta-mannosylated glycolipid epitope). In 20 some embodiments, the recombinant monoclonal antibody comprises a heavy chain variable region selected from the group consisting of a 3E2 variable region, a 18.44 variable region, a 1E10 variable region, and a 4H9 variable region. The present invention also provides an antimicrobial polypeptide encoded by nucleotides 1474-1584 of SEQ ID NO:102. In preferred embodiments, the antimicrobial 25 polypeptide neutralizes Cryptosporidium sp. sporozoite infectivity. In some embodiments, the antimicrobial polypeptide is fused to a microorganism targeting molecule. DESCRIPTION OF THE FIGURES Fig. 1 shows one contemplated retrovector construct embodiment of the present 30 invention. Figures 2A-2D show various contemplated retrovector elements used for production in mammalian cell culture of certain biocide fusions. Figure 2A shows a full size antibody 8 WO 2007/047189 PCT/US2006/039361 with biocide linked to the N-terminus of the heavy chain. Figure 2B shows a full size antibody with biocide linked to the C-terminus of the heavy chain. Figure 2C shows a single chain antibody with biocide linked to the N-terminus of the light chain. Figure 2D shows a single chain antibody with biocide linked to the C-terminus of the heavy chain. In 5 Figures 2A-2D abbreviations used are as follows: LTR, long terminal repeat; EPR, extended packaging region; neo, neomycin selection marker; sCMV, simian cytomegalovirus; SP, signal peptide; X, biocide; L, (G4S)3-4 linker; HC, antibody heavy chain; IRES1, internal ribosome entry site from encephalomyocarditis virus; LC, antibody light chain; and RESE (RNA stabilization element). 10 Figure 3 shows PLA2 neutralization of C. parvum in one embodiment of the present invention. Figure 4A shows retrovector elements used for mammalian cell culture production of recombinant 3E2 IgM antibody as a hexamer. Figure 4B shows retrovector gene construct used for production of recombinant 3E2 IgM antibody as a pentamer with J-chain. Figure 15 4C shows C a retrovector construct used for transgenic production of recombinant 3E2 IgM antibody as a hexamer. Figure 4D, shows a retrovector gene construct used for transgenic production of recombinant 3E2 IgM antibody as a pentamer with J-chain Figures 5A-5D show retrovector elements used for mammalian cell culture production of biocide fusion proteins in certain embodiments of the present invention. 20 Figure 5A shows a full size antibody with biocide linked to the N-terminus of the heavy chain. Figure 5B shows a full size antibody with biocide linked to the C-terminus of the heavy chain. Figure 5C shows a single chain antibody with biocide linked to the N-tenninus of the light chain. Figure 5D shows a single chain antibody with biocide linked to the C terminus of the heavy chain. 25 Figure 6 shows an exemplary Human CD14-PLA2 construct of the present invention (SEQ ID NO:97). Figure 7 shows an exemplary Human LBP-PLA2 construct of the present invention (SEQ ID NO:98). Figure 8 shows an exemplary Human MBL-PLA2 construct of the present invention 30 (SEQ ID NO:99). Figure 9 shows an exemplary Human SP-D-PLA2 construct of the present invention (SEQ ID NO:100). 9 WO 2007/047189 PCT/US2006/039361 Figure 10 shows an exemplary Mouse IgM-PLA2 construct of the present invention (SEQ ID NO:101). Figure 11 shows the parasticidal activity of different biocides against C. parvum spores. 5 Figure 12 shows the P-values for the data of Figure 11 against a no-biocide control. Figure 13 shows the effect of biocides on C. parvum sporozoite infectivity for Caco 2 human intestinal epithelial cells. Figure 14 shows the P-values for the data in Figure 13. Figure 15 shows a schematic of the isolation of immunoglobulin heavy chain genes 10 using degenerate primers. Figure 16 shows elements used in the construction of the 3E2 recombinant hexamer (A) and pentamer (A+B) antibody. Figure 17 shows In vitro neutralization of C. parvum sporozoite infectivity by recombinant 3E2 Hexamer (r3E2-HEX), recombinant 3E2 Pentamer (r3E2-PENT), and 15 hybridoma-derived 3E2 (3E2 HYB) MAbs. Figure 18 shows the efficacy of orally administered rMAb 3E2 against intestinal infection in C. parvum oocyst challenged neonatal mice. Figure 19 shows the nucleic acid sequence of an exemplary IgG2b biocide fusion protein (SEQ ID NO:102). 20 Figure 20 shows elements used in the construction of the recombinant IgG2b fusion proteins. Figure 21 shows In vitro neutralization of C. parvum sporozoite infectivity by recombinant 4H9 IgGl (r4H9 GI), r4H9 G1-CAT fusion (r4H9 G1-CAT), recombinant 4H9 IgG2b-CAT fusion (r4H9 2b-CAT), CAT alone, and r4H9 G1 + CAT. 25 Figure 22 shows In vitro neutralization of C. parvun sporozoite infectivity by recombinant human PLA2 (r-PLA2) and bee venom PLA2. Figure 23 shows efficacy of orally administered r4H9 GI, r4H9 G1 + CAT, and r4H9 G2b-CAT (two different concentrations) against intestinal infection in C. parvum oocyst challenged neonatal mice. 30 Figure 24 shows the nucleic acid sequence the human IgG3 heavy chain (SEQ ID NO: 103). 10 WO 2007/047189 PCT/US2006/039361 Figure 25 shows the effect of PLA2-antibody fusions on reduction in infectivity of Cryptosporidium in a mouse model. Figure 26 shows dose-dependent efficacy of 4H9 fusions. Figure 27 shows the effect of 18.44 antibody fusions on infectivity in a mouse 5 model. DEFINITIONS To facilitate an understanding of the present invention, a number of terms and phrases are defined below: 10 As used herein, the terms "biocide" or "biocides" refer to at least a portion of a naturally occurring or synthetic molecule (e.g., peptides) that directly kills or promotes the death and/or attenuation of, or otherwise neutralizes infectivity without killing (e.g., prevents growth and/or replication) of biological targets (e.g., bacteria, parasites, yeast, viruses, fungi, protozoans and the like). Examples of biocides include, but are not limited 15 to, bactericides, viricides, fungicides, parasiticides, and the like. As used herein, the terms "protein biocide" and "protein biocides" refer to at least a portion of a naturally occurring or synthetic peptide molecule that directly kills or promotes the death and/or attenuation of, or otherwise neutralizes infectivity without killing (e.g., prevents growth and/or replication) of biological targets (e.g., bacteria, parasites, yeast, 20 viruses, fungi, protozoans and the like). Examples of biocides include, but are not limited to, bactericides, viricides, fungicides, parasiticides, and the like. As used herein, the term "neutralization," "pathogen neutralization," "and spoilage organism neutralization" refer to destruction or inactivation (e.g., loss of virulence) of a "pathogen" or "spoilage organism" (e.g., bacterium, parasite, virus, fungus, mold, prion, and 25 the like) thus preventing the pathogen's or spoilage organism's ability to initiate a disease state in a subject or cause degradation of a food product. As used herein, the term "spoilage organism" refers to microorganisms (e.g., bacteria or fungi), which cause degradation of the nutritional or organoleptic quality of food and reduces its economic value and shelf life. Exemplary food spoilage microorganisms 30 include, but are not limited to, Zygosaccharomyces bailii, Aspergillus niger, Saccharomyces cerivisiae, Lactobacillus plantarun, Streptococcus faecalis, and Leuconostoc inesenteroides. 11 WO 2007/047189 PCT/US2006/039361 As used herein, the term "microorganism targeting molecule" refers to any molecule (e.g., protein) that interacts with a microorganism. In preferred embodiments, the microorganism targeting molecule specifically interacts with microorganisms at the exclusion of non- microorganism host cells. Preferred microorganism targeting molecules 5 interact with broad classes of microorganism (e.g., all bacteria or all gram positive or negative bacteria). However, the present invention also contemplates microorganism targeting molecules that interact with a specific species or sub-species of microorganism. In some preferred embodiments, microorganism targeting molecules interact with "Pathogen Associated Molecular Patterns (PAMPS)". In some embodiments, microorganism targeting 10 molecules are recognition molecules that are known to interact with or bind to PAMPS (e.g., including, but not limited to, as CD14, lipopolysaccharide binding protein (LBP), surfactant protein D (SP-D), and Mannan binding lectin (MBL)). In other embodiments, microorganism targeting molecules are antibodies (e.g., monoclonal antibodies directed towards PAMPS or monoclonal antibodies directed to specific organisms or serotype 15 specific epitopes). As used herein the term "biofilm" refers to an aggregation of microorganisms (e.g., bacteria) surrounded by an extracellular matrix or slime adherent on a surface in vivo or ex vivo, wherein the microorganisms adopt altered metabolic states. As used herein, the term "host cell" refers to any eukaryotic cell (e.g., mammalian 20 cells, avian cells, amphibian cells, plant cells, fish cells, insect cells, yeast cells, and bacteria cells, and the like), whether located in vitro or in vivo (e.g., in a transgenic organism). As used herein, the term "cell culture" refers to any in vitro culture of cells. Included within this term are continuous cell lines (e.g., with an immortal phenotype), primary cell cultures, finite cell lines (e.g., non-transformed cells), and any other cell population 25 maintained in vitro, including oocytes and embryos. As used herein, the term "vector" refers to any genetic element, such as a plasmid, phage, transposon, cosmid, chromosome, retrovirus, virion, etc., which is capable of replication when associated with the proper control elements and which can transfer gene sequences between cells. Thus, the tenn includes cloning and expression vehicles, as well 30 as viral vectors. As used herein, the term "multiplicity of infection" or "MOI" refers to the ratio of integrating vectors: host cells used during transfection or infection of host cells. For 12 WO 2007/047189 PCT/US2006/039361 example, if 1,000,000 vectors are used to transfect 100,000 host cells, the multiplicity of infection is 10. The use of this term is not limited to events involving infection, but instead encompasses introduction of a vector into a host by methods such as lipofection, microinjection, calcium phosphate precipitation, and electroporation. 5 As used herein, the term "genome" refers to the genetic material (e.g., chromosomes) of an organism or a host cell. The term "nucleotide sequence of interest" refers to any nucleotide sequence (e.g., RNA or DNA), the manipulation of which may be deemed desirable for any reason (e.g., treat disease, confer improved qualities, etc.), by one of ordinary skill in the art. Such 10 nucleotide sequences include, but are not limited to, coding sequences, or portions thereof, of structural genes (e.g., reporter genes, selection marker genes, oncogenes, drug resistance genes, growth factors, etc.), and non-coding regulatory sequences that do not encode an mRNA or protein product (e.g., promoter sequence, polyadenylation sequence, termination sequence, enhancer sequence, etc.). 15 The term "gene" refers to a nucleic acid (e.g., DNA or RNA) sequence that comprises coding sequences necessary for the production of a polypeptide or precursor (e.g., proinsulin). The polypeptide can be encoded by a full length coding sequence or by any portion of the coding sequence so long as the desired activity or functional properties (e.g., enzymatic activity, ligand/binding, signal transduction, etc.) of the full-length or fragment 20 are retained. The term also encompasses the coding region of a structural gene and includes sequences located adjacent to the coding region on both the 5' and 3' ends for a distance of about 1 kb or more on either end such that the gene corresponds to the length of the full length mRNA. The sequences that are located 5' of the coding region and which are present on the mRNA are referred to as 5'untranslated sequences. The sequences that are located 3' 25 or downstream of the coding region and which are present on the mRNA are referred to as 3' untranslated 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 that are transcribed into nuclear RNA (hnRNA); introns may 30 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 13 WO 2007/047189 PCT/US2006/039361 (mRNA) transcript. The mRNA functions during translation to specify the sequence or order of amino acids in a nascent polypeptide. DNA molecules (e.g., genes) are said to have "5' ends" and "3' ends" because mononucleotides are reacted to make oligonucleotides in a manner such that the 5' 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 mononucleotide 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 10 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 the transcription of a linked gene are generally 15 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 "exogenous gene" refers to a gene that is not naturally present in a host organism or cell, or is artificially introduced into a host organism or cell. 20 As used herein, the tenn "transgene" means a nucleic acid sequence (e.g., encoding one or more fusion protein polypeptides), which is introduced into the genome of a transgenic organism. A transgene can include one or more transcriptional regulatory sequences and other nucleic acid, such as introns, that may be necessary for optimal expression and secretion of a nucleic acid encoding the fusion protein. A transgene can 25 include an enhancer sequence. A fusion protein sequence can be operatively linked to a tissue specific promoter, e.g., mammary gland specific promoter sequence that results in the secretion of the protein in the milk of a transgenic mammal, a urine specific promoter, or an egg specific promoter. As used herein, the tenn "transgenic cell" refers to a cell containing a transgene. 30 A "transgenic organism," as used herein, refers to a transgenic animal or plant. As used herein, a "transgenic animal" is a non-human animal in which one or more, and preferably essentially all, of the cells of the animal contain a transgene introduced by 14 WO 2007/047189 PCT/US2006/039361 way of human intervention, such as by transgenic techniques known in the art. The transgene can be introduced into the cell, directly or indirectly by introduction into a precursor of the cell, by way of deliberate genetic manipulation, such as by microinjection or by infection with a recombinant virus. 5 Mammals are defined herein as all animals which have mammary glands and produce milk. As used herein, a "dairy animal" refers to a milk producing non-human mammal that is larger than a laboratory rodent (e.g., a mouse). In preferred embodiments, the dairy animals produce large volumes of milk and have long lactating periods (e.g., cows or goats). 10 As used herein, the term "plant" refers to either a whole plant, a plant part, a plant cell, or a group of plant cells, including plants that are actively growing (e.g. in soil) and those that have been harvested. The class of plants used in methods of the invention is generally as broad as the class of higher plants amenable to transformation techniques, including both monocotyledonous and dicotyledonous plants. It includes plants of a variety 15 of ploidy levels, including polyploid, diploid and haploid. As used herein, a "transgenic plant" is a plant, preferably a multi-celled or higher plant, in which one or more, and preferably essentially all, of the cells of the plant contain a transgene introduced by way of human intervention, such as by transgenic techniques known in the art. 20 As used herein, the term "gene expression" refers to the process of converting genetic infonnation encoded in a gene into RNA (e.g., mRNA, rRNA, tRNA, or snRNA) through "transcription" of the gene (i.e., via the enzymatic action of an RNA polymerase), and for protein encoding genes, into protein through "translation" of mRNA. Gene expression can be regulated at many stages in the process. "Up-regulation" or "activation" 25 refers to regulation that increases the production of gene expression products (i.e., RNA or protein), while "down-regulation" or "repression" refers to regulation that decrease production. Molecules (e.g., transcription factors) that are involved in up-regulation or down-regulation are often called "activators" and "repressors," respectively. As used herein, the term "protein of interest" refers to a protein encoded by a nucleic 30 acid of interest. As used herein, the term "native" (or wild type) when used in reference to a protein refers to proteins encoded by partially homologous nucleic acids so that the amino acid 15 WO 2007/047189 PCT/US2006/039361 sequence of the proteins varies. As used herein, the term "variant" encompasses proteins encoded by homologous genes having both conservative and nonconservative amino acid substitutions that do not result in a change in protein function, as well as proteins encoded by homologous genes having amino acid substitutions that cause decreased (e.g., null 5 mutations) protein function or increased protein function. The term "isolated" when used in relation to a nucleic acid, as in "an isolated oligonucleotide" refers to a nucleic acid sequence that is identified and separated from at least one contaminant nucleic acid with which it is ordinarily associated in its natural source. Isolated nucleic acids are nucleic acids present in a form or setting that is different 10 from that in which they are found in nature. In contrast, non-isolated nucleic acids are nucleic acids such as DNA and RNA that are found in the state in which they exist in nature. As used herein, the terms "nucleic acid molecule encoding," "DNA sequence encoding," "DNA encoding," "RNA sequence encoding," and "RNA encoding" refer to the order or sequence of deoxyribonucleotides or ribonucleotides along a strand of 15 deoxyribonucleic acid or ribonucleic acid. The order of these deoxyribonucleotides or ribonucleotides determines the order of amino acids along the polypeptide (protein) chain translated from the mRNA. The DNA or RNA sequence thus codes for the amino acid sequence. As used herein, the terms "complementary" or "complementarity" are used in 20 reference to polynucleotides (i.e., a sequence of nucleotides) related by the base-pairing rules. For example, for the sequence "A-G-T," is complementary to the sequence "T-C-A." Complementarity may be "partial," in which only some of the nucleic acids' bases are matched according to the base pairing rules. Or, there may be "complete" or "total" complementarity between the nucleic acids. The degree of complementarity between 25 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 that depend upon binding between nucleic acids. The terms "homology" and "percent identity" when used in relation to nucleic acids refers to a degree of complementarity. There may be partial homology (i.e., partial identity) 30 or complete homology (i.e., complete identity). A partially complementary sequence is one that at least partially inhibits a completely complementary sequence from hybridizing to a target nucleic acid sequence and is referred to using the functional term "substantially 16 WO 2007/047189 PCT/US2006/039361 homologous." 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 (i.e., an oligonucleotide which is capable of hybridizing to 5 another oligonucleotide of interest) 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 10 specific binding may be tested by the use of a second target 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 art knows well that numerous equivalent conditions may be employed to comprise low stringency conditions; factors such as the length and nature (DNA, RNA, base 15 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) are considered and the hybridization solution may be varied to generate conditions of low stringency hybridization different from, but equivalent to, the above listed conditions. In 20 addition, the art knows conditions that 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.). When used in reference to a double-stranded nucleic acid sequence such as a cDNA or genomic clone, the term "substantially homologous" refers to any probe that can 25 hybridize to either or both strands of the double-stranded nucleic acid sequence under conditions of low stringency as described above. When used in reference to a single-stranded nucleic acid sequence, the term "substantially homologous" refers to any probe that can hybridize (i.e., it is the complement of) the single-stranded nucleic acid sequence under conditions of low stringency as 30 described above. As used herein, the tern "hybridization" is used in reference to the pairing of complementary nucleic acids. Hybridization and the strength of hybridization (i.e., the 17 WO 2007/047189 PCT/US2006/039361 strength of the association between the nucleic acids) is impacted by such factors as the degree of complementary between the nucleic acids, stringency of the conditions involved, the Tm of the formed hybrid, and the G:C ratio within the nucleic acids. A single molecule that contains pairing of complementary nucleic acids within its structure is said to be "self 5 hybridized." As used herein, the term "Tm" is used in reference to the "melting temperature" of a nucleic acid. The melting temperature is the temperature at which a population of double stranded nucleic acid molecules becomes half dissociated into single strands. The equation for calculating the Tm of nucleic acids is well known in the art. As indicated by standard 10 references, a simple estimate of the Tm value may be calculated by the equation: Tm= 81.5 + 0.41(% G + C), when a nucleic acid is in aqueous solution at 1 M NaCl (See e.g., Anderson and Young, Quantitative Filter Hybridization, in Nucleic Acid Hybridization [1985]). Other references include more sophisticated computations that take structural as well as sequence characteristics into account for the calculation of Tm. 15 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. With "high stringency" conditions, nucleic acid base pairing will occur only between nucleic acid fragments that have a high frequency of complementary base sequences. Thus, conditions of "weak" or "low" 20 stringency are often required with nucleic acids that are derived from organisms that are genetically diverse, as the frequency of complementary sequences is usually less. "High stringency conditions" when used in reference to nucleic acid hybridization comprise conditions equivalent to binding or hybridization at 42'C in a solution consisting of 5X SSPE (43.8 g/l NaCl, 6.9 g/l NaH 2
PO
4
H
2 0 and 1.85 g/l EDTA, pH adjusted to 7.4 25 with NaOH), 0.5% SDS, 5X Denhardt's reagent and 100 tg/ml denatured salmon sperm DNA followed by washing in a solution comprising 0. IX SSPE, 1.0% SDS at 42'C when a probe of about 500 nucleotides in length is employed. "Medium stringency conditions" when used in reference to nucleic acid hybridization comprise conditions equivalent to binding or hybridization at 42*C in a 30 solution consisting of 5X SSPE (43.8 g/l NaCl, 6.9 g/l NaH 2
PO
4
H
2 0 and 1.85 g/l EDTA, pH adjusted to 7.4 with NaOH), 0.5% SDS, 5X Denhardt's reagent and 100 pig/ml denatured 18 WO 2007/047189 PCT/US2006/039361 salmon sperm DNA followed by washing in a solution comprising 1.OX SSPE, 1.0% SDS at 42 0 C when a probe of about 500 nucleotides in length is employed. "Low stringency conditions" comprise conditions equivalent to binding or hybridization at 42'C in a solution consisting of 5X SSPE (43.8 g/l NaCl, 6.9 g/l NaH 2
PO
4 5 H 2 0 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, Pharamcia), 5 g BSA (Fraction V; Sigma)] and 100 ig/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. 10 A gene may produce multiple RNA species that are generated by differential splicing of the primary RNA transcript. cDNAs that are splice variants of the same gene will contain regions of sequence identity or complete homology (representing the presence of the same exon or portion of the same exon on both cDNAs) and regions of complete non-identity (for example, representing the presence of exon "A" on cDNA 1 wherein cDNA 2 contains exon 15 "B" instead). Because the two cDNAs contain regions of sequence identity they will both hybridize to a probe derived from the entire gene or portions of the gene containing sequences found on both cDNAs; the two splice variants are therefore substantially homologous to such a probe and to each other. The terms "in operable combination," "in operable order," and "operably linked" as 20 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. As used herein, the tenn "selectable marker" refers to a gene that encodes an 25 enzymatic activity that confers the ability to grow in medium lacking what would otherwise be an essential nutrient (e.g., the HIS3 gene in yeast cells); in addition, a selectable marker may confer resistance to an antibiotic or drug upon the cell in which the selectable marker is expressed. Selectable markers may be "dominant"; a dominant selectable marker encodes an enzymatic activity that can be detected in any eukaryotic cell line. Examples of dominant 30 selectable markers include, but are not limited to, the bacterial aminoglycoside 3' phosphotransferase gene (also referred to as the neo gene) that confers resistance to the drug G418 in mammalian cells, the bacterial hygromycin G phosphotransferase (h2yg) gene that 19 WO 2007/047189 PCT/US2006/039361 confers resistance to the antibiotic hygromycin and the bacterial xanthine-guanine phosphoribosyl transferase gene (also referred to as the gpt gene) that confers the ability to grow in the presence of mycophenolic acid. Other selectable markers are not dominant in that their use must be in conjunction with a cell line that lacks the relevant enzyme activity. 5 Examples of non-dominant selectable markers include the thymidine kinase (tk) gene that is used in conjunction with tk - cell lines, the CAD gene, which is used in conjunction with CAD-deficient cells and the mammalian hypoxanthine-guanine phosphoribosyl transferase (hprt) gene which is used in conjunction with hprt - cell lines. A review of the use of selectable markers in mammalian cell lines is provided in Sambrook, J. et al., Molecular 10 Cloning: A Laboratory Manual, 2nd ed., Cold Spring Harbor Laboratory Press, New York (1989) pp.
16
.
9
-
16
.
15 . As used herein, the term "reporter gene" refers to a gene encoding a protein that may be assayed. Examples of reporter genes include, but are not limited to, luciferase (See, e.g., deWet et al., Mol. Cell. Biol. 7:725 [1987] and US Pat Nos., 6,074,859; 5,976,796; 15 5,674,713; and 5,618,682; all of which are incorporated herein by reference), green fluorescent protein (e.g., GenBank Accession Number U43284; a number of GFP variants are commercially available from CLONTECH Laboratories, Palo Alto, CA), chloramphenicol acetyltransferase, p-galactosidase, alkaline phosphatase, and horseradish peroxidase. 20 As used herein, the term "regulatory element" refers to a genetic element that controls some aspect of the expression of nucleic acid sequences. For example, a promoter is a regulatory element that facilitates the initiation of transcription of an operably linked coding region. Other regulatory elements are splicing signals, polyadenylation signals, termination signals, RNA export elements, internal ribosome entry sites, etc. (defined infra). 25 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 et al., Science 236:1237 [1987]). Promoter and enhancer elements have been isolated from a variety of eukaryotic sources including genes in yeast, insect and mammalian cells, and viruses 30 (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. Some eukaryotic promoters and enhancers have a broad host range while 20 WO 2007/047189 PCT/US2006/039361 others are functional in a limited subset of cell types (for review See e.g., Voss et al., Trends Biochem. Sci., 11:287 [1986]; and Maniatis et al., supra). For example, the SV40 early gene enhancer is very active in a wide variety of cell types from many mammalian species and has been widely used for the expression of proteins in mammalian cells (Dijkema et al., 5 EMBO J. 4:761 [1985]). Two other examples of promoter/enhancer elements active in a broad range of mammalian cell types are those from the human elongation factor la gene (Uetsuki et al., J. Biol. Chem., 264:5791 [1989]; Kim et al., Gene 91:217 [1990]; and Mizushima and Nagata, Nuc. Acids. Res., 18:5322 [1990]) and the long terminal repeats of the Rous sarcoma virus (Gorman et al., Proc. Natl. Acad. Sci. USA 79:6777 [1982]) and the 10 human cytomegalovirus (Boshart et al., Cell 41:521 [1985]). In preferred embodiments, inducible retroviral promoters are utilized. As used herein, the term "promoter/enhancer" denotes a segment of DNA which contains sequences capable of providing both promoter and enhancer functions (i.e., the functions provided by a promoter element and an enhancer element, see above for a 15 discussion of these functions). For example, the long tenninal repeats of retroviruses contain both promoter and enhancer functions. The enhancer/promoter may be "endogenous" or "exogenous" or "heterologous." An "endogenous" enhancer/promoter is one that is naturally linked with a given gene in the genome. An "exogenous" or "heterologous" enhancer/promoter is one that is placed in juxtaposition to a gene by means 20 of genetic manipulation (i.e., molecular biological techniques such as cloning and recombination) such that transcription of that gene is directed by the linked enhancer/promoter. Regulatory elements may be tissue specific or cell specific. The term "tissue specific" as it applies to a regulatory element refers to a regulatory element that is capable of 25 directing selective expression of a nucleotide sequence of interest to a specific type of tissue (e.g., mammillary gland) in the relative absence of expression of the same nucleotide sequence(s) of interest in a different type of tissue (e.g., liver). Tissue specificity of a regulatory element may be evaluated by, for example, operably linking a reporter gene to a promoter sequence (which is not tissue-specific) and to 30 the regulatory element to generate a reporter construct, introducing the reporter construct into the genome of an animal such that the reporter construct is integrated into every tissue of the resulting transgenic animal, and detecting the expression of the reporter gene (e.g., 21 WO 2007/047189 PCT/US2006/039361 detecting mRNA, protein, or the activity of a protein encoded by the reporter gene) in different tissues of the transgenic animal. The detection of a greater level of expression of the reporter gene in one or more tissues relative to the level of expression of the reporter gene in other tissues shows that the regulatory element is "specific" for the tissues in which 5 greater levels of expression are detected. Thus, the term "tissue-specific" (e.g., liver specific) as used herein is a relative term that does not require absolute specificity of expression. In other words, the term "tissue-specific" does not require that one tissue have extremely high levels of expression and another tissue have no expression. It is sufficient that expression is greater in one tissue than another. By contrast, "strict" or "absolute" 10 tissue-specific expression is meant to indicate expression in a single tissue type (e.g., liver) with no detectable expression in other tissues. The term "cell type specific" as applied to a regulatory element refers to a regulatory element which is capable of directing selective expression of a nucleotide sequence of interest in a specific type of cell in the relative absence of expression of the same nucleotide 15 sequence of interest in a different type of cell within the same tissue (e.g., cells infected with retrovirus, and more particularly, cells infected with BLV or HTLV). The term "cell type specific" when applied to a regulatory element also means a regulatory element capable of promoting selective expression of a nucleotide sequence of interest in a region within a single tissue. 20 The cell type specificity of a regulatory element may be assessed using methods well known in the art (e.g., immunohistochemical staining and/or Northern blot analysis). Briefly, for immunohistochemical staining, tissue sections are embedded in paraffin, and paraffin sections are reacted with a primary antibody specific for the polypeptide product encoded by the nucleotide sequence of interest whose expression is regulated by the 25 regulatory element. A labeled (e.g., peroxidase conjugated) secondary antibody specific for the primary antibody is allowed to bind to the sectioned tissue and specific binding detected (e.g., with avidin/biotin) by microscopy. Briefly, for Northern blot analysis, RNA is isolated from cells and electrophoresed on agarose gels to fractionate the RNA according to size followed by transfer of the RNA from the gel to a solid support (e.g., nitrocellulose or a 30 nylon membrane). The immobilized RNA is then probed with a labeled oligo deoxyribonucleotide probe or DNA probe to detect RNA species complementary to the probe used. Northern blots are a standard tool of molecular biologists. 22 WO 2007/047189 PCT/US2006/039361 The term "promoter," "promoter element," or "promoter sequence" as used herein, refers to a DNA sequence which when ligated to a nucleotide sequence of interest is capable of controlling the transcription of the nucleotide sequence of interest into mRNA. A promoter is typically, though not necessarily, located 5' (i.e., upstream) of a nucleotide 5 sequence of interest whose transcription into mRNA it controls, and provides a site for specific binding by RNA polymerase and other transcription factors for initiation of transcription. Promoters may be constitutive or regulatable. The term "constitutive" when made in reference to a promoter means that the promoter is capable of directing transcription of an 10 operably linked nucleic acid sequence in the absence of a stimulus (e.g., heat shock, chemicals, etc.). In contrast, a "regulatable" promoter is one that is capable of directing a level of transcription of an operably linked nucleic acid sequence in the presence of a stimulus (e.g., heat shock, chemicals, etc.), which is different from the level of transcription of the operably linked nucleic acid sequence in the absence of the stimulus. 15 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 et al., 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 20 acceptor site is the splice junction from the 16S RNA of SV40. Efficient expression of recombinant DNA sequences in eukaryotic cells requires 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 "poly A site" 25 or "poly A sequence" as used herein denotes a DNA sequence that 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 30 naturally at the 3' end of the coding region of a given gene in the genome. A heterologous poly A signal is one that is isolated from one gene and placed 3' of another gene. A commonly used heterologous poly A signal is the SV40 poly A signal. The SV40 poly A 23 WO 2007/047189 PCT/US2006/039361 signal is contained on a 237 bp BamHI/BclI restriction fragment and directs both termination and polyadenylation (Sambrook, supra, at 16.6-16.7). Eukaryotic expression vectors may also contain "viral replicons "or "viral origins of replication." Viral replicons are viral DNA sequences that allow for the extrachromosomal 5 replication of a vector in a host cell expressing the appropriate replication factors. Vectors that contain either the SV40 or polyoma virus origin of replication replicate to high "copy number" (up to 104 copies/cell) in cells that express the appropriate viral T antigen. Vectors that contain the replicons from bovine papillomavirus or Epstein-Barr virus replicate extrachromosomally at "low copy number" (~100 copies/cell). However, it is not intended 10 that expression vectors be limited to any particular viral origin of replication. As used herein, the term "long terminal repeat" or "LTR" refers to transcriptional control elements located in or isolated from the U3 region 5' and 3' of a retroviral genome. As is known in the art, long terminal repeats may be used as control elements in retroviral vectors, or isolated from the retroviral genome and used to control expression from other 15 types of vectors. As used herein, the terms "RNA export element" or "Pre-mRNA Processing Enhancer (PPE)" refer to 3' and 5' cis-acting post-transcriptional regulatory elements that enhance export of RNA from the nucleus. "PPE" elements include, but are not limited to Mertz sequences (described in US 5,914,267 and 5,686,120, all of which is incorporated 20 herein by reference) and woodchuck mRNA processing enhancer (WPRE; WO 99/14310, incorporated herein by reference). As used herein, the term "polycistronic" refers to an mRNA encoding more than one polypeptide chain (See, e.g., WO 93/03143, WO 88/05486, and European Pat. No. 117058, each of which is incorporated herein by reference). Likewise, the term "arranged in 25 polycistronic sequence" refers to the arrangement of genes encoding two different polypeptide chains in a single mRNA. As used herein, the term "internal ribosome entry site" or "IRES" refers to a sequence located between polycistronic genes that permits the production of the expression product originating from the second gene by internal initiation of the translation of the 30 dicistronic mRNA. Examples of internal ribosome entry sites include, but are not limited to, those derived from foot and mouth disease virus (FDV), encephalomyocarditis virus, poliovirus and RDV (Scheper et al., Biochem. 76: 801-809 [1994]; Meyer et al., J. Virol. 24 WO 2007/047189 PCT/US2006/039361 69: 2819-2824 [1995]; Jang et al., 1988, J. Virol. 62: 2636-2643 [1998]; Haller et al., J. Virol. 66: 5075-5086 [1995]). Vectors incorporating IRESs maybe assembled as is known in the art. For example, a retroviral vector containing a polycistronic sequence may contain the following elements in operable association: nucleotide polylinker, gene of interest, an 5 internal ribosome entry site and a mannnalian selectable marker or another gene of interest. The polycistronic cassette is situated within the retroviral vector between the 5' LTR and the 3' LTR at a position such that transcription from the 5' LTR promoter transcribes the polycistronic message cassette. The transcription of the polycistronic message cassette may also be driven by an internal promoter (e.g., cytomegalovirus promoter) or an inducible 10 promoter (e.g., the inducible promoters of the present invention), which may be preferable depending on the use. The polycistronic message cassette can further comprise a cDNA or genomic DNA (gDNA) sequence operatively associated within the polylinker. Any mammalian selectable marker can be utilized as the polycistronic message cassette mammalian selectable marker. Such mammalian selectable markers are well known to 15 those of skill in the art and can include, but are not limited to, kanamycin/G41 8, hygromycin B or mycophenolic acid resistance markers. As used herein, the term "retrovirus" refers to a retroviral particle which is capable of entering a cell (i.e., the particle contains a membrane-associated protein such as an envelope protein or a viral G glycoprotein which can bind to the host cell surface and 20 facilitate entry of the viral particle into the cytoplasm of the host cell) and integrating the retroviral genome (as a double-stranded provirus) into the genome of the host cell. As used herein, the tenn "retroviral vector" refers to a retrovirus that has been modified to express a gene of interest. Retroviral vectors can be used to transfer genes efficiently into host cells by exploiting the viral infectious process. Foreign or heterologous 25 genes cloned (i.e., inserted using molecular biological techniques) into the retroviral genome can be delivered efficiently to host cells that are susceptible to infection by the retrovirus. Through well-known genetic manipulations, the replicative capacity of the retroviral genome can be destroyed. The resulting replication-defective vectors can be used to introduce new genetic material to a cell but they are unable to replicate. A helper virus or 30 packaging cell line can be used to permit vector particle assembly and egress from the cell. Such retroviral vectors comprise a replication-deficient retroviral genome containing a nucleic acid sequence encoding at least one gene of interest (i.e., a polycistronic nucleic acid 25 WO 2007/047189 PCT/US2006/039361 sequence can encode more than one gene of interest), a 5' retroviral long terminal repeat (5' LTR); and a 3' retroviral long terminal repeat (3' LTR). The term "pseudotyped retroviral vector" refers to a retroviral vector containing a heterologous membrane protein. The term "membrane-associated protein" refers to a 5 protein (e.g., a viral envelope glycoprotein or the G proteins of viruses in the Rhabdoviridae family such as VSV, Piry, Chandipura and Mokola), which is associated with the membrane surrounding a viral particle; these membrane-associated proteins mediate the entry of the viral particle into the host cell. The membrane associated protein may bind to specific cell surface protein receptors, as is the case for retroviral envelope proteins or the membrane 10 associated protein may interact with a phospholipid component of the plasma membrane of the host cell, as is the case for the G proteins derived from members of the Rhabdoviridae family. A "subject" is an animal such as vertebrate, preferably a mammal, more preferably a human or a bovine. Mammals, however, are understood to include, but are not limited to, 15 murines, simians, humans, bovines, cervids, equines, porcines, canines, felines etc.). An "effective amount" is an amount sufficient to effect beneficial or desired results. An effective amount can be administered in one or more administrations, "Co-administration" refers to administration of more than one agent or therapy to a subject. Co-administration may be concurrent or, alternatively, the chemical compounds 20 described herein may be administered in advance of or following the administration of the other agent(s). One skilled in the art can readily determine the appropriate dosage for co administration. When co-administered with another therapeutic agent, both the agents may be used at lower dosages. Thus, co-administration is especially desirable where the claimed compounds are used to lower the requisite dosage of known toxic agents. 25 As used herein, the term "toxic" refers to any detrimental or harmful effects on a cell or tissue. A "pharmaceutical composition" is intended to include the combination of an active agent with a carrier, inert or active, making the composition suitable for diagnostic or therapeutic use in vivo, in vivo or ex vivo. 30 As used herein, the term "pharmaceutically acceptable carrier" encompasses any of the standard phannaceutical carriers, such as a phosphate buffered saline solution, water, and an emulsion, such as an oil/water or water/oil emulsion, and various types of wetting 26 WO 2007/047189 PCT/US2006/039361 agents. The compositions also can include stabilizers and preservatives. For examples of carriers, stabilizers and adjuvants see Martin, Remington's Pharmaceutical Sciences, 15th Ed., Mack Publ. Co., Easton, PA (1975). "Pharmaceutically acceptable salt" as used herein, relates to any pharmaceutically 5 acceptable salt (acid or base) of a compound of the present invention, which, upon administration to a recipient, is capable of providing a compound of this invention or an active metabolite or residue thereof. As is known to those of skill in the art, "salts" of the compounds of the present invention may be derived from inorganic or organic acids and bases. Examples of acids include hydrochloric, hydrobromic, sulfuric, nitric, perchloric, 10 fumaric, maleic, phosphoric, glycolic, lactic, salicylic, succinic, toluene-p-sulfonic, tartaric, acetic, citric, methanesulfonic, ethanesulfonic, formic, benzoic, malonic, naphthalene-2 sulfonic and benzenesulfonic acid. Other acids, such as oxalic, while not in themselves pharmaceutically acceptable, may be employed in the preparation of salts useful as intennediates in obtaining the compounds of the invention and their pharmaceutically 15 acceptable acid. As used herein, the term "nutraceutical," refers to a food substance or part of a food, which includes a fusion protein. Nutraceuticals can provide medical or health benefits, including the prevention, treatment, or cure of a disorder. The transgenic protein will often be present in the nutraceutical at concentration of at least 100 pg/kg, more preferably at least 20 1 mg/kg, most preferably at least 10 mg/kg. A nutraceutical can include the milk of a transgenic animal. As used herein, the term "purified" or "to purify" refers to the removal of undesired components from a sample. As used herein, the term "substantially purified" refers to molecules, either nucleic or amino acid sequences, that are removed from their natural 25 environment, isolated or separated, and are at least 60% free, preferably 75% free, and most preferably 90% free from other components with which they are naturally associated. An "isolated polynucleotide" is therefore a substantially purified polynucleotide. The terms "bacteria" and "bacterium" refer to all prokaryotic organisms, including those within all of the phyla in the Kingdom Procaryotae. It is intended that the term 30 encompass all microorganisms considered to be bacteria including Mycoplasma, Chlamydia, Actinomyces, Streptomyces, and Rickettsia. All forns of bacteria are included within this definition including cocci, bacilli, spirochetes, spheroplasts, protoplasts, etc. Also included 27 WO 2007/047189 PCT/US2006/039361 within this term are prokaryotic organisms that are gram negative or gram positive. "Gram negative" and "gram positive" refer to staining patterns with the Gram-staining process that is well known in the art. (See e.g., Finegold and Martin, Diagnostic Microbiology, 6th Ed., CV Mosby St. Louis, pp. 13-15 [1982]). "Gram positive bacteria" are bacteria that retain 5 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 the counterstain. Thus, gram negative bacteria appear red. In some embodiments, the bacteria are those capable of causing disease (pathogens) and those that cause product degradation or spoilage. 10 The terms "specific binding" or "specifically binding" when used in reference to the interaction of an antibody and an antigen means that the interaction is dependent upon the presence of a particular structure (i.e., the antigenic determinant or epitope) on the antigen; in other words the antibody is recognizing and binding to a specific structure rather than to antigens in general. For example, if an antibody is specific for epitope "A," the presence of 15 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. As used herein, the terms "non-specific binding" and "background binding" when used in reference to the interaction of an antibody and an antigen refer to an interaction that is not dependent on the presence of a particular structure (i.e., the antibody is binding to 20 antigens in general rather that a particular structure such as an epitope). As used herein, the tenn "antigen binding protein" refers to proteins that bind to a specific antigen. "Antigen binding proteins" include, but are not limited to, immunoglobulins, including polyclonal, monoclonal, chimeric, single chain, and humanized antibodies, Fab fragments, F(ab')2 fragments, and Fab expression libraries. Various 25 procedures known in the art are used for the production of polyclonal antibodies. For the production of antibody, various host animals can be immunized by injection with the peptide corresponding to the desired epitope including but not limited to rabbits, mice, rats, sheep, goats, etc. In a preferred embodiment, the peptide is conjugated to an immunogenic carrier (e.g., diphtheria toxoid, bovine serum albumin (BSA), or keyhole limpet hemocyanin 30 (KLH)). Various adjuvants are used to increase the immunological response, depending on the host species, including but not limited to Freund's (complete and incomplete), mineral gels such as aluminum hydroxide, surface active substances such as lysolecithin, pluronic 28 WO 2007/047189 PCT/US2006/039361 polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanins, dinitrophenol, and potentially useful human adjuvants such as BCG (Bacille Calmette-Guerin) and Corynebacterium parvum. For preparation of monoclonal antibodies, any technique that provides for the 5 production of antibody molecules by continuous cell lines in culture may be used (See e.g., Harlow and Lane, Antibodies: A Laboratory Manual, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, NY). These include, but are not limited to, the hybridoma technique originally developed by K6hler and Milstein (K6hler and Milstein, Nature, 256:495-497 [1975]), as well as the trioma technique, the human B-cell hybridoma technique (See e.g., 10 Kozbor et al., Immunol. Today, 4:72 [1983]), and the EBV-hybridoma technique to produce human monoclonal antibodies (Cole et al., in Monoclonal Antibodies and Cancer Therapy, Alan R. Liss, Inc., pp. 77-96 [1985]). In other embodiments, suitable monoclonal antibodies, including recombinant chimeric monoclonal antibodies and chimeric monoclonal antibody fusion proteins are prepared as described herein. 15 According to the invention, techniques described for the production of single chain antibodies (US 4,946,778; herein incorporated by reference) can be adapted to produce specific single chain antibodies as desired. An additional embodiment of the invention utilizes the techniques known in the art for the construction of Fab expression libraries (Huse et al., Science, 246:1275-1281 [1989]) to allow rapid and easy identification of 20 monoclonal Fab fragments with the desired specificity. In some embodiments, monoclonal antibodies are generated using the ABL-MYC method (See e.g., U.S. Patent 5,705,150 and 5,244,656, each of which is herein incorporated by reference) (Neoclone, Madison, WI). ABL-MYC is a recombinant retrovirus that constitutively expresses v-abl and c-myc oncogenes. When used to infect antigen-activated 25 splenocytes, this retroviral system rapidly induces antigen-specific plasmacytomas. ABL MYC targets antigen-stimulated (Ag-stimulated) B-cells for transformation. Antibody fragments that contain the idiotype (antigen binding region) of the antibody molecule can be generated by known techniques. For example, such fragments include but are not limited to: the F(ab')2 fragment that can be produced by pepsin digestion 30 of an antibody molecule; the Fab' fragments that can be generated by reducing the disulfide bridges of an F(ab')2 fragment, and the Fab fragments that can be generated by treating an antibody molecule with papain and a reducing agent. 29 WO 2007/047189 PCT/US2006/039361 Genes encoding antigen-binding proteins can be isolated by methods known in the art. In the production of antibodies, screening for the desired antibody can be accomplished by techniques known in the art (e.g., radioimmunoassay, ELISA (enzyme-linked immunosorbant assay); "sandwich" immunoassays, immunoradiometric assays, gel diffusion 5 precipitin reactions, immunodiffusion assays, in situ immunoassays (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.) etc. 10 As used herein, the term "Cryptosporidium sp." refers to any species of Cryptosporidium. Examples include, but are not limited to, Cryptosporidiui parvum and Cyptosporidiuin hominis. As used herein, the term "a recombinant antibody that binds to a surface epitope of Cryptosporidium sp." refers to a recombinantly expressed monoclonal antibody that binds to 15 a specific epitope on the surface of Cryptosporidium sp. Preferred recombinant antibodies bind to epitopes on Cryptosporidium parvun but not to other microorganisms. Exemplary Cryptosporidium parvun epitopes include, but are not limited to, GP25-200, p23, CSL, or beta-mannosylated glycolipid. Exemplary recombinant monoclonal antibodies include, but are not limited to 3E2, which recognizes CSL, 1E10, which recognizes p23, 3H2, which 20 recognizes GP25-200, 4H9, which recognizes GP25-200, 18.44, which recognizes beta mannosylated glycolipid. As used herein the term "recombinant antibody having substantially the same properties as antibodies secreted by a hybridoma with accession number ATCC HB 12075" refers to an antibody that has substantially the same properties as those described for 25 hybridoma HB 12075 (e.g., including, but not limited to, binding to the CSL epitope and/or neutralizing Cryptosporidium parvum sporozoites). In some embodiments, the recombinant antibody is a "recombinant 3E2 antibody." As used herein, the tenn "recombinant 3E2 antibody" refers to a recombinant antibody isolated from hybridoma cell line deposited with the American Type Culture 30 Collection, Manassas, VA with accession number ATCC HB 12075 (See e.g., U.S. Patent 6,110,463; Riggs, Microbes Infect 2002; 4:1067 and Riggs et al., J Immunol 1997; 158:1787-95; each of which is herein incorporated by reference in its entirety). In some 30 WO 2007/047189 PCT/US2006/039361 embodiments, recombinant 3E2 antibodies have the IgM isotype. In other embodiments, recombinant 3E2 antibodies are engineered to have hybrid isotypes where the heavy chain has a IgM variable region and a murine IgG2b or human IgG3 isotype. As used herein, the term "recombinant 1 E10 antibody" refers to a recombinant monoclonal 5 antibody isolated from a hybridoma cell line (See e.g., U.S. Patent 6,323,020 and Schaefer et al., Infection and Immunity 68:2508 [2000], each of which herein incorporated by reference in its entirety). In some embodiments, recombinants 1E10 antibodies have the IgG1 isotype. In other embodiments, recombinant 1E10 antibodies are engineered to have hybrid isotypes where the heavy chain has a IgM variable region and a murine IgG2b or 10 human IgG3 isotype. As used herein, the tenn "recombinant 4H9 antibody" refers to a recombinant monoclonal antibody isolated from hybridoma cell line (See e.g., Schaefer et al., Infection and Immunity 68:2508 [2000] and Riggs, Microbes Infect 2002; 4:1067; each of which is herein incorporated by reference in its entirety). In some embodiments, recombinant 4H9 15 antibodies are IgG1 isotypes. In other embodiments, recombinant 4H9 antibodies are engineered to have hybrid isotypes where the heavy chain has a IgM variable region and a murine IgG2b or human IgG3 isotype. As used herein, the term "recombinant 18.44 antibody" refers to a recombinant antibody isolated from hybridoma cell line (See e.g., Priest et al., J Biol Chem 2003; 278:52212-22 20 and Riggs et al., Infect Immun 1999; 67:1317-22; each of which herein incorporated by reference in its entirety). In some embodiments, recombinant 18.44 antibodies have Murine IgG3 isotypes. In other embodiments, recombinant 18.44 antibodies are engineered to have hybrid isotypes where the heavy chain has a IgM variable region and a murine IgG2b or human IgG3 isotype. 25 GENERAL DESCRIPTION OF THE INVENTION One embodiment of the present invention provides compositions and methods for treating and/or preventing illnesses in animals caused by pathogens. More particularly, the present invention provides therapeutic and prophylactic compositions directed to combating 30 bacterial, parasitical, and fungal infections in humans and other animals (e.g., feedlot and domestic animals such as cows, chickens, turkeys, pigs, and sheep). 31 WO 2007/047189 PCT/US2006/039361 In preferred embodiments, the present invention provides fusion proteins comprising microorganism targeting molecules (e.g., including, but not limited to, monoclonal antibody and innate immune system receptors) directed against bacterial, parasitic, and fungal pathogens and methods of using and creating these molecules. In some of these 5 embodiments, the antibodies are chimeras (e.g., murine-bovine). The present invention is not limited however to providing fusion proteins or chimeras. In other embodiments, the present invention provides chimeric monoclonal antibodies directed against foodborne bacterial, protozoan and parasitic pathogens. However, the bacterial pathogens need not be foodborne (e.g., gastrointestinal). For 10 example, additional embodiments are directed to providing therapeutic compositions and methods to combat other bacterial infections via other possible routes of transmission (e.g., respiratory, salivary, fecal-oral, skin-to-skin, bloodborne, genital, urinary, eye-to-eye, zoonotic, etc.). Moreover, other aspects of the present invention provide chimeric monoclonal antibodies against viruses, prions, fungal, protozoan and other parasitic and 15 pathogenic sources of illness. In addition to the compositions and methods discussed above, the present invention - further provides chimeric recombinant monoclonal antibody fusion proteins. In some of these embodiments, the fusion proteins comprise one or more portions of an immunoglobulin and a portion of a biocide molecule, such as bactericides, viricides, 20 fungicides, parasiticides, and the like. In preferred embodiments, the present invention provides antibody biocide fusion proteins, wherein the biocide component comprises a bactericidal enzyme such as human lysozyme, phospholipase A2 (groups I, II, V, X, and XII), lactoferrin, lactoperoxidase, and bacterial permeability increasing protein. In additional embodiments, the present provides fusion proteins comprising immune system 25 complement proteins including cytokines such as the interferons (e.g., IFN-a, IFN-3, and IFN-y) and the tumor necrosis factors (e.g., TNF- a, and TNF- @) and defensins. In preferred embodiments, the antibody portion of these fusion proteins binds specifically to a foodborne bacterial pathogen (e.g., E. coli 0157:H7, Listeria monocytogenes, Canpylobacterjejuni, and the like). 30 The present invention also provides compositions comprising fusion proteins in an edible carrier such as whey protein. Preferred methods of using these compositions include, but are not limited to, food additives for human and animal (e.g., bovines) consumption, 32 WO 2007/047189 PCT/US2006/039361 carcass decontaminating compounds used during processing and finishing feedlot animal (e.g., bovine) carcasses and poultry, as well as pharmaceutical compositions for both human and veterinary medicine. The present invention is not limited to the uses specifically recited herein. 5 In some embodiments, suitable food additive formulations of the present compositions include, but are not limited to, compositions directly applied to food products such as processed meat slices and dairy products in the form of sprays, powders, injected solutions, coatings, gels, rinses, dips, films (e.g., bonded), extrusions, among other known formulations. 10 Likewise, the present invention further provides compositions (e.g., rinses, sprays, and the like) for sanitizing food-processing, medical, military or household equipment. For example, some preferred embodiments of the present invention provide compositions for disinfecting meat-processing equipment. In this regard, the present invention contemplates that a number of food (e.g., meat) processors will benefit from using the compositions and 15 methods of the present invention in their operations. In this regard, the present invention contemplates providing compositions to the entire range of meat processing operations from the largest commercial slaughterhouses to individual consumers. Those skilled in the art will appreciate that the compositions disclosed herein can be readily formulated to include additional compounds common in the pharmaceutical arts 20 such as, excipients, extenders, preservatives, and bulking agents depending on the intended use of a composition. Furthermore, ingestible formulations of these compositions may also comprise any material approved by the United States Department of Agriculture (USDA) for incorporation into food products such as substances that are generally recognized as safe (GRAS) including, food additives, flavorings, colorings, vitamins, minerals, and 25 phytonutrients. The term phytonutrients as used herein, refers to organic compounds isolated from plants having biological effects including, but not limited to, compounds from the following classes of molecules: isoflavonoids, oligomeric proanthcyanidins, indol-3-carbinol, sulforaphone, fibrous ligands, plant phytosterols, ferulic acid, anthocyanocides, triterpenes, omega 3/6 fatty acids, polyacetylene, quinones, terpenes, 30 cathechins, gallates, and quercitin. In still further embodiments, the fusion proteins of the present invention are purified from the lactations of transgenic non-human mammals such as, cows, pigs, sheep, and 33 WO 2007/047189 PCT/US2006/039361 goats. In particularly preferred embodiments, the transgenic animal is a cow. Consequently, the present invention further provides novel genetic constructs and methods of producing transgenic animals that express the compositions of the present invention in their lactation. The present invention also provides methods of inducing transgenic animals 5 (e.g., bovines) to lactate upon maturation. The present invention also provides methods of stably transfecting cell lines (e.g., mammalian, plant, insect, and amphibian) with encoding the fusion proteins disclosed herein. In preferred embodiments, the constructs of the present invention allow complex multicistronic gene constructs to be stably inserted into cells (e.g., mammalian, bacteria, 10 fungal cells, plant, etc). The production of fusion proteins in mammalian cell lines (or in transgenic mammals) allows for their proper assembly and processing. Another method suitable for use in some embodiments of the present invention is protein production in mammalian tissue culture bioreactors. Monoclonal antibodies are typically produced in mammalian cells to ensure correct 15 processing, however mammalian tissue culture bioreactors are often expensive to operate thus placing products beyond mass applications. The ability to manufacture monoclonals in the milk of transgenic animals (e.g., bovines) is contemplated to expand the scope of monoclonal antibodies typically from individual medicine to applications for large populations. Production of the disclosed compositions in the milk of transgenic mammals 20 (e.g., bovines) provides large quantities for economical distribution to food safety and processing operations. For instance, in preferred embodiments, the present invention contemplates that at reasonable expression levels of about one gram per liter of milk, a herd of 100 transgenic cows will produce about a metric ton of recombinant protein per year. This enables production of recombinant monoclonals at 100 fold less cost than in cell 25 culture bioreactors. Accordingly, in preferred embodiments the present invention provides methods of creating transgenic bovines that produce the compositions of the present invention in their lactation. The present invention also provides methods of isolating and purifying the compositions of the present invention from the lactation of milk producing herd animals (e.g., cows, sheep, and goats and the like). 30 In still further embodiments, the present invention provides fusion protein enriched colostrum, or colostrum like products, for use as milk substitutes and nutritional supplements for nursing mammals and in particular for nursing feedlot animals. In 34 WO 2007/047189 PCT/US2006/039361 preferred embodiments, these compositions comprise the microorganism targeting molecule fusion proteins of the present invention. The present invention also contemplates that introducing these compositions to nursing feedlot animals will reduce the colonization of the animal's gastrointestinal tract by pathogenic organisms such as E coli 0157:H7 and 5 Listeria monocytogenes and Cryptosporidium parvum. Furthermore, the compositions may be added to feeds to control diseases such as coccidiosis, which are common in both cattle and chicken feeding operations. In particular, providing a fusion protein enriched milk replacer or colostrum supplement reduces the load of E. coli 01 57:H7 in the gastrointestinal tract of the neonate and specifically places the targeted pathogenic organisms at a 10 competitive disadvantage in relation to normal gastrointestinal flora. The present invention further contemplates inducing a protective immune response in animals fed the preset fusion protein enriched colostrums and colostrum-like compositions. Accordingly, additional preferred embodiments of the present invention are directed to conferring passive immunity in animals fed the present compositions. 15 The present invention provides compositions and methods directed against foodborne pathogens such as, but not limited to, E. coli 01 57:H7, Listeria monocytogenes, Campylobacterjejuni, Clostridium botulinum, Clostridium peifringens, Staphylococcus aureus, Staphylococcus epidermidis, Staphylococcus pneumoniae, Staphylococcus saprophyticus, Staphylococcus mutans, Shigella dysenteriae, Salmonella typhi, Salmonella 20 paratyphi, Salmonella enteritidis, Cryptosporidium parvum, fungi, and the like. The present invention further provides composition and methods directed against food spoilage organisms such as, but not limited to, bacteria (e.g., Lactobacillus, Leuconostoc, Pediococcus, and Streptococcus and fungi (e.g., Monilia, Trichoderma, Crinipellis, Moniliophthora, Phytophthora, Botrytis, and Fusarium). 25 The present invention also provides compositions and methods directed against protozoans, particularly, apicomplexan protozoans including, but not limited to coccidian, cryptosporidian, toxoplasman, malarian and trypanosomatid protozoans. In preferred embodiments, the compositions of the present invention comprise a targeting molecule, for example an immunoglobulin subunit (or portion thereof), a biocide 30 molecule (or portion thereof) such as, a bactericidal enzyme, (e.g., lysozyme), and a linker that connects the targeting molecule and the biocide molecule. In other preferred 35 WO 2007/047189 PCT/US2006/039361 embodiments, the compositions further comprise a signaling molecule or sequence that predictably directs the composition to an intracellular or extracellular location. In certain embodiments, the present invention provides broad spectrum antimicrobials. Broad spectrum antimicrobials find use as a preventative tool where the 5 identity of possible food contaminants is unknown, and new organisms can emerge as serious threats. Broad spectrum antimicrobials are also well suited for use in medicine and biodefense in confronting an infection of unknown etiology. Broad spectrum antimicrobials take advantage of the innate immune system, which provides an important front line defense through receptors on specialized cells (e.g., 10 macrophages, neutrophils) that are capable of binding the vast majority of microbes to which these body surfaces are exposed. In some embodiments, recognition molecules such as CD 14, lipopolysaccharide binding protein (LBP), surfactant protein D (SP-D), Toll receptors, and Mannan binding lectin (MBL) that recognize and bind to Pathogen Associated Molecular Patterns (PAMPs) common to many organisms are used as the 15 targeting portion of fusion proteins of the present invention. In other embodiments, broadly reactive monoclonal antibodies that bind to PAMPS are use as the targeting portion of the fusion proteins of the present invention. In preferred embodiments, biocidal enzymes are delivered in high concentrations to the surface of bacteria by expressing the two components, a microorganism targeting molecule and a 20 biocidal payload as a fusion protein. In still further embodiments, IgM (e.g., for increased avidity of binding to repetitive PAMPs) and secretory IgA (e.g., for greater stability in harsh environments) are used and instead of attaching the biocide directly to the targeting molecule, it is attached to the J chain that is used to assemble both pentameric IgM and dimeric IgA. 25 By using components of the innate immune system the present invention provides antimicrobials that function effectively ex vivo (e.g., in food safety settings), as well as in vivo (e.g., in clinical medicine and veterinary medicine), which can confront a broad range of bacteria through the broad affinity of the innate recognition. Furthermore, because the recognition targets bacterial features that are essential to bacterial invasion and attachment, 30 resistance is very unlikely to occur. The present invention thus provides a novel class of antimicrobials that find use in a variety of settings. 36 WO 2007/047189 PCT/US2006/039361 I. Immunoglobulins Immunoglobulins (antibodies) are proteins generated by the immune system to provide a specific molecule capable of complexing with an invading molecule commonly referred to as an antigen. Natural antibodies have two identical antigen-binding sites, both 5 of which are specific to a particular antigen. The antibody molecule recognizes the antigen by complexing its antigen-binding sites with areas of the antigen termed epitopes. The epitopes fit into the conformational architecture of the antigen-binding sites of the antibody, enabling the antibody to bind to the antigen. The inmmunoglobulin molecule is composed of two identical heavy and two identical 10 light polypeptide chains, held together by interchain disulfide bonds. Each individual light and heavy chain folds into regions of about 110 amino acids, assuming a conserved three dimensional conformation. The light chain comprises one variable region (termed VL) and one constant region (CL), while the heavy chain comprises one variable region (VH) and three constant regions (CH1, CH 2 and CH3). Pairs of regions associate to form discrete 15 structures. In particular, the light and heavy chain variable regions, VL and VH, associate to form an "Fv " area that contains the antigen-binding site. The variable regions of both heavy and light chains show considerable variability in structure and amino acid composition from one antibody molecule to another, whereas the constant regions show little variability. Each antibody recognizes and binds an antigen 20 through the binding site defined by the association of the heavy and light chain, variable regions into an Fv area. The light-chain variable region VL and the heavy-chain variable region VH of a particular antibody molecule have specific amino acid sequences that allow the antigen-binding site to assume a confonnation that binds to the antigen epitope recognized by that particular antibody. 25 Within the variable regions are found regions in which the amino acid sequence is extremely variable from one antibody to another. Three of these so-called "hypervariable" regions or "complementarity-determining regions" (CDR's) are found in each of the light and heavy chains. The three CDRs from a light chain and the three CDRs from a corresponding heavy chain form the antigen-binding site. 30 Cleavage of naturally occurring antibody molecules with the proteolytic enzyme papain generates fragments that retain their antigen-binding site. These fragments, commonly known as Fab's (for Fragment, antigen binding site) are composed of the CL, VL, 37 WO 2007/047189 PCT/US2006/039361 CHI and VH regions of the antibody. In the Fab the light chain and the fragment of the heavy chain are covalently linked by a disulfide linkage. Monoclonal antibodies against target antigens (e.g., a cell surface protein, such as receptors) are produced by a variety of techniques including conventional monoclonal 5 antibody methodologies such as the somatic cell hybridization techniques of Kohler and Milstein, Nature, 256:495 (1975). Although in some embodiments, somatic cell hybridization procedures are preferred, other techniques for producing monoclonal antibodies are contemplated as well (e.g., viral or oncogenic transformation of B lymphocytes). 10 The preferred animal system for preparing hybridomas is the murine system. Hybridoma production in the mouse is a well-established procedure. Immunization protocols and techniques for isolation of immunized splenocytes for fusion are known in the art. Fusion partners (e.g., murine myeloma cells) and fusion procedures are also known. Human monoclonal antibodies (mAbs) directed against human proteins can be 15 generated using transgenic mice carrying the complete human immune system rather than the mouse system. Splenocytes from the transgenic mice are immunized with the antigen of interest, which are used to produce hybridomas that secrete human mAbs with specific affinities for epitopes from a human protein. (See e.g., Wood et al., WO 91/00906, Kucherlapati et al., WO 91/10741; Lonberg et al., WO 92/03918; Kay et al., WO 92/03917 20 [each of which is herein incorporated by reference in its entirety]; N. Lonberg et al., Nature, 368:856-859 [1994]; L.L. Green et al., Nature Genet., 7:13-21 [1994]; S.L. Morrison et al., Proc. Nat. Acad. Sci. USA, 81:6851-6855 [1994]; Bruggeman et al., Immunol., 7:33-40 [1993]; Tuaillon et al., Proc. Nat. Acad. Sci. USA, 90:3720-3724 [1993]; and Bruggernan et al. Eur. J. Immunol., 21:1323-1326 [1991]). 25 Monoclonal antibodies can also be generated by other methods known to those skilled in the art of recombinant DNA technology. An alternative method, referred to as the "combinatorial antibody display" method, has been developed to identify and isolate antibody fragments having a particular antigen specificity, and can be utilized to produce monoclonal antibodies. (See e.g., Sastry et al., Proc. Nat. Acad. Sci. USA, 86:5728 [1989]; 30 Huse et al., Science, 246:1275 [1989]; and Orlandi et al., Proc. Nat. Acad. Sci. USA, 86:3833 [1989]). After immunizing an animal with an immunogen as described above, the antibody repertoire of the resulting B-cell pool is cloned. Methods are generally known for 38 WO 2007/047189 PCT/US2006/039361 obtaining the DNA sequence of the variable regions of a diverse population of immunoglobulin molecules by using a mixture of oligomer primers and the PCR. For instance, mixed oligonucleotide primers corresponding to the 5' leader (signal peptide) sequences and/or framework 1 (FRI) sequences, as well as primer to a conserved 3' constant 5 region primer can be used for PCR amplification of the heavy and light chain variable regions from a number of murine antibodies. (See e.g., Larrick et al., Biotechniques, 11:152-156 [1991]). A similar strategy can also been used to amplify human heavy and light chain variable regions from human antibodies (See e.g., Larrick et al., Methods: Companion to Methods in Enzymology, 2:106-110 [1991]). 10 In one embodiment, RNA is isolated from B lymphocytes, for example, peripheral blood cells, bone marrow, or spleen preparations, using standard protocols (e.g.,US 4,683,292 [incorporated herein by reference in its entirety]; Orlandi, et al.,Proc. Nat. Acad. Sci. USA, 86:3833-3837 [1989]; Sastry et al., Proc. Nat. Acad. Sci. USA, 86:5728-5732 [1989]; and Huse et al., Science, 246:1275 [1989]). First strand cDNA is synthesized using 15 primers specific for the constant region of the heavy chain(s) and each of the x and X light chains, as well as primers for the signal sequence. Using variable region PCR primers, the variable regions of both heavy and light chains are amplified, each alqne or in combination, and ligated into appropriate vectors for further manipulation ingenerating the display packages. Oligonucleotide primers useful in amplification protocols may be unique or 20 degenerate or incorporate inosine at degenerate positions. Restriction endonuclease recognition sequences may also be incorporated into the primers to allow for the cloning of the amplified fragment into a vector in a predetermined reading frame for expression. The V-gene library cloned from the immunization-derived antibody repertoire can be expressed by a population of display packages, preferably derived from filamentous phage, 25 to fonn an antibody display library. Ideally, the display package comprises a system that allows the sampling of very large variegated antibody display libraries, rapid sorting after each affinity separation round, and easy isolation of the antibody gene from purified display packages. In addition to commercially available kits for generating phage display libraries, examples of methods and reagents particularly amenable for use in generating a variegated 30 antibody display library can be found in, for example, US 5,223,409; WO 92/18619; WO 91/17271; WO 92/20791; WO 92/15679; WO 93/01288; WO 92/01047; WO 92/09690; WO 90/02809 [each of which is herein incorporated by reference in its entirety]; Fuchs et al., 39 WO 2007/047189 PCT/US2006/039361 Biol. Technology, 9:1370-1372 [1991]; Hay et al., Hum. Antibod. Hybridomas, 3:81-85 [1992]; Huse et al., Science, 46:1275-1281 [1989]; Hawkins et al., J. Mol. Biol., 226:889 896 [1992]; Clackson et al., Nature, 352:624-628 [1991]; Gram et al., Proc. Nat. Acad. Sci. USA, 89:3576-3580 [1992]; Garrad et al., Bio/Technolog, 2:1373-1377 [1991]; 5 Hoogenboom et al., Nuc. Acid Res., 19:4133-4137 [1991]; and Barbas et al., Proc. Nat. Acad. Sci. USA, 88:7978 [1991]. In certain embodiments, the V region domains of heavy and light chains can be expressed on the same polypeptide, joined by a flexible linker to form a single-chain Fv fragment, and the scFV gene subsequently cloned into the desired expression vector or phage genome. 10 As generally described in McCafferty et al., Nature, 348:552-554 (1990), complete VH and VL domains of an antibody, joined by a flexible linker (e.g., (Gly 4 -Ser) 3 ) can be used to produce a single chain antibody which can render the display package separable based on antigen affinity. Isolated scFV antibodies immunoreactive with the antigen can subsequently be formulated into a pharmaceutical preparation for use in the subject method. 15 Once displayed on the surface of a display package (e.g., filamentous phage), the antibody library is screened with the target antigen, or peptide fragment thereof, to identify and isolate packages that express an antibody having specificity for the target antigen. Nucleic acid encoding the selected antibody can be recovered from the display package (e.g., from the phage genome) and subcloned into other expression vectors by standard 20 recombinant DNA techniques. Specific antibody molecules with high affinities for a surface protein can be made according to methods known to those in the art, e.g., methods involving screening of libraries US 5,233,409 and US 5,403,484 (both incorporated herein by reference in their entireties). Further, the methods of these libraries can be used in screens to obtain binding 25 determinants that are mimetics of the structural detenninants of antibodies. In particular, the Fv binding surface of a particular antibody molecule interacts with its target ligand according to principles of protein-protein interactions, hence sequence data for VH and VL (the latter of which may be of the ic or X chain type) is the basis for protein engineering techniques known to those with skill in the art. Details of the protein surface 30 that comprises the binding determinants can be obtained from antibody sequence in formation, by a modeling procedure using previously detennined three-dimensional structures from other antibodies obtained from NMR studies or crytallographic data. 40 WO 2007/047189 PCT/US2006/039361 In one embodiment, a variegated peptide library is expressed by a population of display packages to form a peptide display library. Ideally, the display package comprises a system that allows the sampling of very large variegated peptide display libraries, rapid sorting after each affinity separation round, and easy isolation of the peptide-encoding gene 5 from purified display packages. Peptide display libraries can be in, e.g., prokaryotic organisms and viruses, which can be amplified quickly, are relatively easy to manipulate, and which allows the creation of large number of clones. Preferred display packages include, for example, vegetative bacterial cells, bacterial spores, and most preferably, bacterial viruses (especially DNA viruses). However, the present invention also 10 contemplates the use of eukaryotic cells, including yeast and their spores, as potential display packages. Phage display libraries are known in the art. Other techniques include affinity chromatography with an appropriate "receptor," e.g., a target antigen, followed by identification of the isolated binding agents or ligands by conventional techniques (e.g., mass spectrometry and NMR). Preferably, the soluble 15 receptor is conjugated to a label (e.g., fluorophores, colorimetric enzymes, radioisotopes, or luminescent compounds) that can be detected to indicate ligand binding. Alternatively, immobilized compounds can be selectively released and allowed to diffuse through a membrane to interact with a receptor. Combinatorial libraries of compounds can also be synthesized with "tags" to encode 20 the identity of each member of the library. (See e.g., W.C. Still et al., WO 94/08051 incorporated herein by reference in its entirety). In general, this method features the use of inert but readily detectable tags that are attached to the solid support or to the compounds. When an active compound is detected, the identity of the compound is determined by identification of the unique accompanying tag. This tagging method permits the synthesis 25 of large libraries of compounds that can be identified at very low levels among to total set of all compounds in the library. The term modified antibody is also intended to include antibodies, such as monoclonal antibodies, chimeric antibodies, and humanized antibodies which have been modified by, for example, deleting, adding, or substituting portions of the antibody. For 30 example, an antibody can be modified by deleting the hinge region, thus generating a monovalent antibody. Any modification is within the scope of the invention so long as the antibody has at least one antigen binding region specific. 41 WO 2007/047189 PCT/US2006/039361 Chimeric mouse-human monoclonal antibodies can be produced by recombinant DNA techniques known in the art. For example, a gene encoding the Fc constant region of a murine (or other species) monoclonal antibody molecule is digested with restriction enzymes to remove the region encoding the murine Fc, and the equivalent portion of a gene 5 encoding a human Fc constant region is substituted. (See e.g., Robinson et al., PCT/US86/02269; European Patent Application 184,187; European Patent Application 171,496; European Patent Application 173,494; WO 86/01533; US 4,816,567; European Patent Application 125,023 [each of which is herein incorporated by reference in its entirety]; Better et al., Science, 240:1041-1043 [1988]; Liu et al., Proc. Nat. Acad. Sci. 10 USA, 84:3439-3443 [1987]; Liu et al., J. Immunol., 139:3521-3526 [1987]; Sun et al., Proc. Nat. Acad. Sci. USA, 84:214-218 [1987]; Nishimura et al., Canc. Res., 47:999-1005 [1987]; Wood et al., Nature, 314:446-449 [1985]; and Shaw et al., J. Natl. Cancer Inst., 80:1553 1559 [1988]). The chimeric antibody can be further humanized by replacing sequences of the Fv 15 variable region that are not directly involved in antigen binding with equivalent sequences from human Fv variable regions. General reviews of humanized chimeric antibodies are provided by S.L. Morrison, Science, 229:1202-1207 (1985) and by Oi et al., Bio. Techniques, 4:214 (1986). Those methods include isolating, manipulating, and expressing the nucleic acid sequences that encode all or part of immunoglobulin Fv variable regions 20 from at least one of a heavy or light chain. Sources of such nucleic acid are well known to those skilled in the art and, for example, may be obtained from 7E3, an anti-GPIIbIIIa antibody producing hybridoma. The recombinant DNA encoding the chimeric antibody, or fragment thereof, can then be cloned into an appropriate expression vector. Suitable humanized antibodies can alternatively be produced by CDR substitution 25 (e.g., US 5,225,539 (incorporated herein by reference in its entirety); Jones et al., Nature, 321:552-525 [1986]; Verhoeyan et al., Science, 239:1534 [1988]; and Beidler et al., J. Inimunol., 141:4053 [1988]). All of the CDRs of a particular human antibody may be replaced with at least a portion of a non-human CDR or only some of the CDRs may be replaced with non-human CDRs. It is only necessary to replace the number of CDRs 30 required for binding of the humanized antibody to the Fc receptor. An antibody can be humanized by any method that is capable of replacing at least a portion of a CDR of a human antibody with a CDR derived from a non-human antibody. 42 WO 2007/047189 PCT/US2006/039361 The human CDRs may be replaced with non-human CDRs; using oligonucleotide site directed mutagenesis. Also within the scope of the invention are chimeric and humanized antibodies in which specific amino acids have been substituted, deleted or added. In particular, preferred 5 humanized antibodies have amino acid substitutions in the framework region, such as to improve binding to the antigen. For example, in a humanized antibody having mouse CDRs, amino acids located in the human framework region can be replaced with the amino acids located at the corresponding positions in the mouse antibody. Such substitutions are known to improve binding of humanized antibodies to the antigen in some instances. 10 In preferred embodiments, the fusion proteins include a monoclonal antibody subunit (e.g., a human, murine, or bovine), or a fragment thereof, (e.g., an antigen binding fragment thereof). The monoclonal antibody subunit or antigen binding fragment thereof can be a single chain polypeptide, a dimer of a heavy chain and a light chain, a tetramer of two heavy and two light chains, or a pentamer (e.g., IgM). IgM is a pentamer of five 15 monomer units held together by disulfide bonds linking their carboxyl-terminal (Cp4/Cp4) domains and C 3/C3 domains. The pentameric structure of IgM provides 10 antigen binding sites, thus serum IgM has a higher valency than other types of antibody isotypes. With its high valency, pentameric IgM is more efficient than other antibody isotypes at binding multidimensional antigens (e.g., viral particles and red blood cells. However, due 20 to its large pentameric structure, IgM does not diffuse well and is usually found in low concentrations in intercellular tissue fluids. The J chain of IgM allows the molecule to bind to receptors on secretary cells, which transport the molecule across epithelial linings to the external secretions that bathe the mucosal surfaces. In some embodiments, of the present invention take advantage of the low diffusion rate of pentameric IgM to help concentrate the 25 fusion proteins of present invention at a site of interest. In preferred embodiments, monoclonal IgM, and fusion and chimeric proteins thereof, are directed to destroying Cyptosporidiun parvum and other types of parasitic pathogens. In some embodiments, an IgA is utilized to make a directed biocide. IgA's are preferably produced using either one, two or three constructs. IgA made by use of two or 30 three retrovector constructs. For example, a retroviral construct can be produced in which the J-chain expression is driven by the long terminal repeat (LTR) promoter, and expression of a heavy chain and light chain separated by an IRES sequence is driven by an internal 43 WO 2007/047189 PCT/US2006/039361 promoter. In another example, the heavy chain and light chain are provided in one vector and the J chain is provided in another vector. In. another example, a third construct expressing the secretory component truncated form from poly IgR is provided. In still other embodiments, secretion of a directed biocide is enhanced by 5 transfecting a cell producing a directed biocide with a vector (e.g., a retroviral vector) that expressed secretory component. See U.S. Pat. No. 6,300,104; Koteswarra and Morrison, Proc. Natl. Acad. Sci. USA 94:6364-68 (1997). In some preferred embodiments, the monoclonal antibody is a murine antibody or a fragment thereof. In other preferred embodiments, the monoclonal antibody is a bovine 10 antibody or a fragment thereof. For example, the murine antibody can be produced by a hybridoma that includes a B cell obtained from a transgenic mouse having a genome comprising a heavy chain transgene and a light chain transgene fused to an immortalized cell. The antibodies can be of various isotypes, including, but not limited to: IgG (e.g., IgGl, IgG2, IgG2a, IgG2b, IgG2c, IgG3, IgG4); IgM; IgAl; IgA2; IgAsco; IgD; and IgE. In 15 some preferred embodiments, the antibody is an IgG isotype. In other preferred embodiments, the antibody is an IgM isotype. The antibodies can be full-length (e.g., an IgG1, IgG2, IgG3, or IgG4 antibody) or can include only an antigen-binding portion (e.g., a Fab, F(ab') 2 , Fv or a single chain Fv fragment). In preferred embodiments, the immunoglobulin subunit of the fusion proteins is a 20 recombinant antibody (e.g., a chimeric or a humanized antibody), a subunit, or an antigen binding fragment thereof (e.g., has a variable region, or at least a complementarity detennining region (CDR)). In preferred embodiments, the immunoglobulin subunit of the fusion protein is monovalent (e.g., includes one pair of heavy and light chains, or antigen binding portions 25 thereof). In other embodiments, the immunoglobulin subunit of the fusion protein is a divalent (e.g., includes two pairs of heavy and light chains, or antigen binding portions thereof). In preferred embodiments, the transgenic fusion proteins include an inununoglobulin heavy chain or a fragment thereof (e.g., an antigen binding fragment thereof). 30 In still other embodiments, the fusion proteins and/or or recombinant antibodies comprise an immunoglobulin having only heavy chains such as the HCAbs found in certain Camelidae (e.g., camels, dromedaries, and llamas) species, spotted ratfish, and nurse shark. 44 WO 2007/047189 PCT/US2006/039361 While the present invention is not limited to any particular mechanisms, the present invention contemplates that there are differences between conventional antibodies and HCAbs in both the VH and CH regions. For instance, as reported by Muyldermans et al. and Nguyen et al., the sequences of HCAbs variable domains (VHH) differ significantly from 5 those of conventional antibodies (VH). (S. Muyldermans et al., Protein, Eng., 7:1129-1135 [1994]; V.K. Nguyen et al., J. Mol. Biol., 275:413-418 [1998]; and V.K. Nguyen et al., Immunogenetics DOI 10.1007/s00251-002-0433-0 [2002]). Additionally, HCAbs lack the first domain of the constant region (CH); the matured VHH-DJ is directly joined to the hinge region. Separate sets of V and C genes encode conventional antibodies and HCAbs, 10 however, conventional antibodies and HCAbs have some common D genes and appear to have identical JH regions. (V.K. Nguyen et al., EMBO J., 19:921-930 [2000]; and V.K. Nguyen et al., Adv. Immunol., 79:261-296 [2001]). In yet other embodiments, IgM is used as the microorganism targeting molecule. IgMs bind with multiple epitopes, effectively enhancing the avidity of the binding. The 15 genes for both SP-D and MBL of these molecules have been sequenced and both have been produced as recombinant molecules in full or truncated forms (Shrive et al., J Mol Biol 2003; 331:509-23; Arora et al., J Biol Chem 2001; 276:43087-94). In other embodiments, the microorganism targeting molecules are monoclonal antibodies that target PAMPs. In some embodiments, the monoclonal antibodies utilize the 20 multimeric structure of IgM and IgA. The repetitive structure of many PAMPs allows antibodies to bind to two identical epitopes on one molecule or on separate molecules (cross-linking) on the bacterial surface. This type of interaction results in an overall higher binding energy per antibody molecule than the engagement of one single arm of the immunoglobulin in the binding; for the antibody to detach, both binding sites would have to 25 be released at once. The avidity is proportionally higher for IgA, which is a dimer, bearing a total of 4 binding sites, or IgM which usually is a pentamer, having 10 binding sites. Packing plants are harsh environments; secretory IgA is adapted to function in the gastrointestinal tract and its dimeric configuration is supported by a portion of the secretory component that assists its membrane transport. Thus IgM and IgA may offer advantages as 30 targeting molecules over IgG. In certain embodiments, the fact that they are linked by a J chain, and in the case of IgA coexpressed with secretory component, allows for attaching the payload biocide to these auxiliary chains instead of to the Fc portion of the immunoglobulin 45 WO 2007/047189 PCT/US2006/039361 itself. Accordingly, in some embodiments, the immunoglobulin J-chain is used as a microorganism targeting molecule. In some embodiments, a system of hybridoma-like antibody preparation, developed by Neoclone (Madison, WI), is used in the production of monoclonal antibodies. 5 Splenocytes from immunized mice are immortalized using a retrovector-mediated introduction of the abl-myc genes. On reintroduction into recipient mice one dominant immortalized B cell clone (plasmacytoma) outgrows all others and produces a monoclonal antibody in the ascitic fluid. The B cell clone can be harvested with the ascitic fluid that contains high concentration of monoclonal antibody. This process can be completed in 8-10 10 weeks. In still further embodiments, murine IgG2b (nucleotides 1-1425 of SEQ ID NO:102) or human IgG3 (SEQ ID NO:103) biocide fusions are utilized as directed biocides. The murine immunoglobulin gamma heavy chain isotype IgG2b has a substantially longer hinge region than the other IgG isotypes, which makes the Fe portion more flexible when bound to 15 the antigen. Its human equivalent is the IgG3 isotype. The present invention is not limited to a particular mechanism. Indeed, an understanding of the mechanism of the present invention is not necessary to practice the present invention. Nonetheless, it is contemplated that a c-terminal fusion of a biocide to the IgG2b or human IgG3 heavy chain will extend its radius of activity and make it more likely that the biocide finds its target (substrate). This 20 flexibility allows a construct with a very short or no linker at all for attachment of the fusion to the c-terminus (e.g., a very short stiff linker such as glycine-proline-proline-glycine or a flexible linker). In other embodiments, it is contemplated that an extended length linker will enhance the activity radius. It is contemplated that this additional flexibility allows for better binding of the biocide to its substrate present on the target organism's surface and 25 therefore result in a greater efficacy of the antibody-biocide fusion. Experiments conducted during the course of development of the present invention resultedin the construction of several IgG2b fusions to different biocides and testing of them. The data show that there is a marked difference in reduction of infection of a gut epithelial cell line (Caco-2) with cryptosporidium sporozoites between the 4H9 GI -CAT 30 fusion protein and the 4H9 G2b-CAT fusion protein treatment. Both fusion proteins were used at the same concentration yet the IgG2b version of the fusion shows a much higher efficacy than the fusion to the IgG1 isotype. Further experiments tested the 4119 G2b-CAT 46 WO 2007/047189 PCT/US2006/039361 fusion in a neonatal mouse sytem and confirmed the cell culture results. The 4H9 G2b-CAT fusion was superior to all other treatments. The IgG2b constant region was combined with the variable region by overlap PCR and the resulting product was cloned and sequenced. Binding tests by IFA (indirect fluorescent assay) revealed that the engineered IgG2b version 5 of the 4H9 monoclonal antibody showed a similar binding pattern to its target epitope on the sporozoite. Accordingly, in some embodiments, proteins of interest (e.g., biocides) are fused to murine IgG2b or IgG3 sequences. The present invention is not limited to a particular protein of interest. In some embodiments, the protein of interest is a protein biocide (e.g. 10 those described herein). However, the protein of interest is not limited to protein biocides. In other embodiments the protein of interest is an effector molecule for murine IgG2b or human IgG3. Examples include, but are not limited to, cytokines (eig., INFy, IL-2, IL-12, etc), receptors for cytokines and growth factors, ligands for receptors, and label or marker proteins (e.g., including, but not limited to, HRP, fluorescent molecules, biotin). 15 II. Innate System Receptors In some embodiments of the present invention, innate immunity receptors are used as microorganism targeting molecules due to their high affinity of interaction with a multitude of microorganisms. These receptors are all highly conserved structures across 20 species, even across classes (Ezekowitz RAB and Hoffinann JA. Innate Immunity. Totowa, NJ: HUmana Press, 2003). Many of these evolutionarily ancient receptors are found in Invertebrae (Aderem A and Ulevitch RJ. Nature 2000; 406:782-7). Preferred microorganism targeting molecules are those that exist as soluble molecules in circulation. In some embodiments, the, microorganism targeting molecule is CD 14, found on 25 monocytes/macrophages and neutrophils (Haziot et al., J Immunol 1988; 141:547-52). This molecule exists as both a membrane-bound form, with a GPI anchor, and as a soluble form. Both versions of CD14 bind LPS with high affinity. The reported dissociation constant for LPS binding to CD14 is KD 3-7 x 10 8 M (Tobias et al., J Biol Chem 1995; 270:10482-8). Haziot and co-workers have produced recombinant soluble human CD14 and have used it to 30 study the binding to LPS (Haziot et al., J Immunol 1995; 154:6529-32), showing that soluble CD14 binds LPS under various conditions. LPS is one of the main components of the cell wall of Gram-negative bacteria. CD14 also binds to peptidoglycan structures with high affinity (Kd=25nM) (Dziarski, Cell Mol Life Sci 2003; 60:1793-804; Dziarski et al., 47 WO 2007/047189 PCT/US2006/039361 Chem Immunol 2000; 74:83-107; Dziarski et al., Infect Immun 2000; 68:5254-60.). Peptidoglycan (PGN) structures make up a substantial portion of the cell wall of Gram negative and Gram-positive bacteria. In addition, CD14 binds to lipoteichoic acid on Gram positive bacteria, lipoarabinomannan of mycobacteriae, lipoproteins from spirochetes and 5 mycobacteriae, and others (Dziarski, Cell Mol Life Sci 2003; 60:1793-804). CD14 is a highly versatile receptor that interacts with an impressive variety of different bacteria. In other embodiments, the microorganism targeting molecule are LPS-binding protein (LBP) (Tobias et al., J Exp Med 1986; 164:777-93). LBP is an acute phase protein that is released into circulation upon a bacterial infection with Gram-negative bacteria or 10 exposure to LPS. LBP can bind circulating or bound LPS, and through this interaction inhibit LPS-related septic shock (Lamping et al., J Clin Invest 1998; 101:2065-71). LBP can inhibit LPS-induced signaling by blocking LPS transfer from CD 14 to Toll like receptor 4 (Thompson et al., J Biol Chem 2003; 278:28367-71). In addition, LBP is capable of removing LPS from mCD14 with high efficiency, indicating that LBP binds LPS more 15 strongly than CD14. Separate studies aimed at comparing binding efficiencies between LPS, LBP, and CD14 have confirmed that LBP binds LPS with a roughly ten-fold higher affinity KD 3.5 x 10~9 M (Tobias et al., J Biol Chem 1995; 270:10482-8) than CD14. LBP has been cloned, sequenced and expressed by various groups (Schumann et al., Science 1990; 249:1429-31; Theofan et al., J Immunol 1994; 152:3623-9; Thompson et al., J Biol 20 Chem 2003; 278:28367-71). Other LBPs are described in U.S. patent 6,103,888, herein incorporated by reference in its entirety. In still further embodiments, the microorganism targeting molecule is a member of the collectins, also called defense collagens (Van De Wetering et al., Eur J Biochem 2004; 271:1229-49). Surfactant protein D (SP-D) has been shown to interact with rough and 25 smooth LPS on bacterial surfaces (Clark et al., Microbes Infect 2000; 2:273-8; Lawson and Reid, Immunol Rev 2000; 173:66-78) and therefore can target Gram-negative microorganisms. In other embodiments, mannan-binding lectin (MBL), from this family, which is known to target peptidoglycans (and hence Gram-positive bacteria) (Lu et al., Biochim Biophys Acta 2002; 1572:387-400), is utilized. Collectins assemble into 30 multimers, effectively multiplying the number of binding sites per complex available for interaction with the microorganism's repetitive surface. 48 WO 2007/047189 PCT/US2006/039361 In other embodiments the Toll receptor family are used as pathogen targeting molecules; this group of cell bound receptors functions singly or in concert with other innate immune system receptors (Ezekowitz and Hoffmann, Innate Immunity. Totowa, NJ: HUmana Press, 2003; Janeway and Medzhitov. Annu Rev Immunol 2002; 20:197-216; 5 Medzhitov and Janeway, Trends Microbiol 2000; 8:452-6. Toll like receptors (TLRs) comprise a family of cell surface receptors that are related to the Drosophila Toll protein, a molecule involved in defense against fungal infection in the fly (Aderem and Ulevitch, Nature, 406:785-787 [2000]). Ten mammalian TLRs have been identified (Aderem and Ulevitch, Supra). Two members of the family, TLR2 and TLR4, have been better 10 characterized and shown to mediate the response to multiple bacterial cell-wall components including lipopolysaccharide (LPS), lipopeptides, peptidoglycans (PGN) and lipoteichoic acid (LTA) (Yang et al,, Nature, 395:284-288 [1998]; Poltorak et al., Science, 282:2085 2088 [1998]; Aliprantis et al., Science, 285:736-739 [1999]; Chow et al., J. Biol. Chem., 274:10689-10692 [2000]; and Schwandner et al., J. Biol. Chem., 274: 17406-17409 15 [2000]). Mammalian TLRs have multiple leucine-rich repeats in the ectodomain and an intracellular Toll-ILl receptor (TIR) domain that mediates a signaling cascade to the nucleus (Aderem and Ulevitch, Supra). Stimulation of TLR2,and TLR4 leads to the recruitment of the adaptor molecule MyD88 and the serine kinase IL-1R-associated kinase (IRAK), two signaling components that together with TRAF-6 mediate activation of NF-KB (Aderem and 20 Ulevitch, Supra). IL. Linkers In preferred embodiments, the transgenic fusion proteins comprise a targeting molecule (e.g., immunoglobulin heavy chain (or fragment thereof) and a light chain or (a 25 fragment thereof)) connected to a biocide molecule by a linker. In preferred embodiments, the targeting molecule is linked via a peptide linker or is directly fused (e.g., covalently bonded) to the biocide molecule. In preferred embodiments, the transgenic fusion proteins assemble into dimeric, trimeric, tetrameric, pentameric, hexameric or higher polymeric complexes. 30 In preferred embodiments, the present invention provides retroviral constructs that encode in operable configuration an immunoglobulin (or portion thereof), a biocide molecule (or portion thereof), and a linker group that connects the immunoglobulin and the 49 WO 2007/047189 PCT/US2006/039361 biocide. In some of these embodiments, the linker group comprises one amino acid moiety (e.g., X,,; wherein X is any amino acid or amino acid derivative; and n = 1). In some of these embodiments, the linker group comprises at least one amino acid moiety (e.g., X"; wherein X is any amino acid or amino acid derivative; and n > 2). Similarly, in other 5 embodiments, the linker group comprises two or more repeating amino acids (e.g., X.Yz; wherein X and Y are any amino acid or amino acid derivative; and n > 1 and z > 1). In still further embodiments, the linker group comprises two or more repeating amino acids that form a repeating unit (e.g., (XnYz)r; wherein r > 1). The present invention is not intended to be limited, however, to the aforementioned linker groups. Those skilled in the art will 10 appreciate that a number of other linker group configurations and compositions find use in certain embodiments of the present invention. In particularly preferred embodiments, the linker group used has one or more of the following characteristics: 1) sufficient length and flexibility to allow for the rotation of the targeting molecule (e.g., immunoglobulin) and the biocide molecule (e.g., lysozyme) 15 relative to one another; 2) a flexible extended conformation; 3) a propensity for developing ordered secondary or tertiary structures that interact with functional components; 4) nonreactive with the functional components of the construct (e.g., minimal hydrophobic or charged character to react with the functional protein domains); 5) sufficient resistant to degradation (e.g., digestion by proteases); and 6) allows the fusion protein to form a 20 complex (e.g., a di-, tri-, tetra-, penta-, or higher multimeric complex) while retaining biological (e.g., biocidal) activity. The linker sequence should separate the target molecule and the biocide molecule of the fusion protein by a distance sufficient to ensure that each component properly folds into its secondary and tertiary structures. In preferred embodiments, the peptide linker is from about 2 to 500, more preferably 25 of from about 50 to 100, and even more preferably, from about 10 to 30 amino acids long. A polypeptide linker sequence of about 20 amino acids provides a suitable separation of functional protein domains, although longer or shorter linker sequences are contemplated. For example, in particularly preferred embodiments, the peptide linker is between 17 to 20 amino acids in length. 30 The present invention further contemplates peptide linkers comprised of the following amino acids: Gly, Ser, Asn, Thr or Ala. Typical surface amino acids in flexible protein regions include Gly, Ser, and Asn. The present invention contemplates that various 50 WO 2007/047189 PCT/US2006/039361 amino acid sequence permutations of Gly, Ser, and optionally Asn, provide suitable linker sequences. However, the present invention is not limited to peptide linkers comprised of the aforementioned amino acids. For example, in some embodiments, the peptide linkers comprise further uncharged polar amino acids (e.g., Gln, or Tyr) and/or nonpolar amino 5 acids (e.g., Val, Leu, Ileu, Pro, Phe, Met, Trp, Cys). In some preferred embodiments, the peptide linker comprises one (or more) Gly-Ser elements. Fore example, in some of these embodiments, the peptide linker has the formula (Sern-Glyx)y, wherein n and x > 1, and y > 1. In some preferred embodiments, the peptide linker has the formula (Ser-Gly 4 )y, wherein y = 1, 2, 3, 4, 5, 6, 7, 8 or more. In some other 10 preferred embodiments, the peptide linker includes a sequence having the formula (Ser Gly 4
)
3 . In still other preferred embodiments, the peptide linker comprises a sequence of the fonnula ((Ser-Gly 4
)
3 -Ser-Pro). Other peptide linker sequences are contemplated, including, but not limited to, Gly 4 SerGly 5 Ser, and ((Ser 4 -Gly) 3 -Ser-Pro). In still further embodiments, the target molecule and the biocidal molecule 15 comprising the fusion protein are fused directly without a linker sequence. In some embodiments, linker sequences are unnecessary where the fusion protein components have non-essential N-or C-terminal amino acid regions that separate functional domains and prevent steric interference. IV. Biocides 20 The present invention provides novel fusion proteins. In preferred embodiments, the recombinant fusion proteins comprise one or more biocide molecules (e.g., a bactericidal enzyme) attached to the antibody portion of the construct via a linking group. The specificity of the monoclonal antibody portion of the construct targets the biocide molecule to a pathogen such as, for example, E. coli 0157:H7, Listeria inonocytogenes, 25 Campylobacterjejuni, or Cryptosporidium parvum. One benefit of the specific targeting ability of the fusion protein construct is that it allows for relative accumulation of biocide at locations where the targeted pathogens are challenging the animal. Increasing the local concentration of biocide relative to the targeted pathogens enhances the biocidal activity of the fusion protein construct. In particular, the 30 present invention contemplates that directing the biocide (e.g., lysozyme, PLA2, and the like) to the immediate vicinity of the pathogen (e.g., a bacterium) via the antibody portion of the construct effectively increasing the biocide's local concentration, thus providing a 51 WO 2007/047189 PCT/US2006/039361 significantly greater biocidal (e.g., bactericidal) effect than administering biocide alone (parasiticidal compounds). For example in the case of lysozyme, the affinity constant (Kmn) of lysozyme for its substrate is approximately 10- M, while that of phospholipase A2 is approximately 10-4 M. However, the K of a monoclonal antibody is usually in the range of 5 10- 8 M to 101 M, thus antibodies have about 5 orders of magnitude higher affinity for their substrates than do biocidal molecules alone. Therefore, preferred embodiments of the present invention utilize monoclonal antibodies (or portions thereof) to specifically direct biocide molecules to a target by taking advantage of the antibody's very high affinity for target pathogens. Additionally, directing the fusion protein constructs to target pathogens 10 also reduces the possible deleterious effects to the animal caused by systemic administration of the biocidal molecules. In preferred embodiments, the directed biocidal approach described herein uses a monoclonal antibody to direct a naturally occurring bactericidal enzyme to the target pathogen. In some of these embodiments, the bactericidal enzyme(s) are components of the 15 innate immune system. One such preferred bactericidal enzyme is lysozyme. Lysozyme is naturally present in mammalian tissues and in secretions such as tears and mucus. Lysozyme is also found in many foods including, egg whites, cow milk, and human colostrum. The enzyme is widely reported to have antibacterial properties. Lysozyme is a glycosidase that targets the polysaccharides of many bacterial cell walls 20 rendering them more susceptible to osmotic lysis. Lysozyme is a 1,4-p-N-acetylmurmidase that cleaves the glycosidic bond between C-1 of N-acetylmuramic acid and C-4 of N acetylglucosamine of the peptidoglycan layer present in many bacterial cell walls (See e.g., M. Schindler et al., Biochemistry, 16(3):423-431 [1977]). While it is not clear whether this cleavage contributes to the bactericidal action of lysozyme (K. During et al., FEBS Lett., 25 449(2-3):93-100 [1999]; and H.R. Ibrahim et al., FEBS Lett., 506(l):27-32 [2001]), it is widely accepted that lysozyme plays an important role in defense against bacterial infection. Lysozyme has also been shown to bind to the lipid A portion of bacterial endotoxin. This interaction prevents the endotoxin from inducing the release of inflammatory components by lymphocytes and macrophages (See e.g., B. Reusens-Billen et al., Diabetes Res. Clin. 30 Pract., 23(2):85-94 [1994]; K. Takada et al., Infect. Immun., 62(4):1171-1175 [1994]; and K. Takada et al., Circ. Shock, 44(4):169-174 [1994]). 52 WO 2007/047189 PCT/US2006/039361 Other proteins that form part of the innate immune system, and especially those secreted by the intestinal Paneth cells, are contemplated for targeting the structural integrity of sporozoites. For example, phopholipase A2 (PLA2) is another naturally occurring bactericidal enzyme contemplated for use in certain embodiments of the present invention. 5 Secretory type II phospholipase A2 (sPLA(2)-IIA) is a 14 kD enzyme synthesized in a number of gland cells, including Paneth cells of intestinal mucosa, prostate gland cells, and lacrimal glands. It is present in cellular secretions on mucosal surfaces including intestinal mucus, seminal plasma, and tears (X.D. Qu and R.I. Lehrer, Infect. Immun., 66:2791-2797 [1998]; and X.D. Qu et al., Infect. Immun., 64:5161-5165 [1996]). Evidence suggests that 10 phopholipase A2 has an important antibacterial role in addition to its inflammatory mediating role (See e.g., A.G. Buckland and D.C. Wilton, Biochim. Biophys. Acta, 1488(1 2):71-82 [2000]). Elevated amounts of phospholipase A2 is found in patients with acute bacterial diseases (J.O. Gronoos et al., J. Infect. Dis., 185:1767-1772 [2002]). The enzyme appears to effective in controlling E coli. infections when expressed in transgenic mice (See 15 e.g., V.J. Laine et al., Infect. Immun., 68(l):87-92 [2000]). While the present invention is not limited to any mechanisms, PLA2 appears to hydrolyze membrane phospholipids, thus destroying the membranes of invading microbes. PLA2 serves as a critical component of the innate immune system, functioning in combination with lysozyme and the defensins to provide an effective barrier to invasion by a diverse range of organisms. 20 Mammalian cells are generally highly resistant to sPLA(2) IIA (R.S. Koduri et al., J. Biol. Chem., 273:32142-32153 [1998]). The substrate specificity of the different members of the PLA2 family may be related to the differences in interfacial binding characteristics to charge-neutral phosphotidyl choline (PC) versus anionic phospholipids. Indeed, sPLA(2) family members sPLA2-V and -X bind efficiently and hydrolyze PC vesicles in vitro 25 whereas the vesicles are a poor binding substrate for -IIA. Plasma membranes with a high PC content would therefore be stable in the presence of sPLA(2)-IIA. The composition of the phospholipids on the surface of the organism therefore contributes to the susceptibility of the organism to the action of sPLA2. Some parasitic eukaryotic organisms may evade the innate immune system by not stimulating the cells of the immune system to release biocidal 30 enzymes and defensins (e.g., G. lamblia and C. albicans appear not to stimulate Paneth cells). However, one recent report suggests that Plasinodium is susceptible to sPLA2 (Type III, from bee venom). Type III sPLA2 has an activity that is similar to the type IIA enzyme, 53 WO 2007/047189 PCT/US2006/039361 but is a slightly larger molecule having N- and C-terminal extensions. Systemically, sPLA(2)-IIA has a role in generalized inflammatory responses. In acute inflammation, the levels of the enzyme are elevated many hundreds of fold, however, it appears to have no adverse effect at epithelial surfaces. In vitro, sPLA(2) apparently has no deleterious effect 5 on various types of cultured mammalian cells. Healthy transgenic mice chronically over expressing sPLA(2)-IIA have been produced and exhibit an elevated resistance to infection by gram positive organisms (V.J. Laine et al., J. Immunol., 162:7402-7408 [1999]; and V.J. Laine et al., Infect, Immun., 68:87-92 [2000]). A number of inhibitors have been identified that have activity against C. parvum by 10 targeting the parasite's metabolic pathways. These include, but are not limited to, metalloprotease inhibitors (P.C. Okhuysen et al., Antimicrob. Agents Chemother., 40:2781 2784 [1996]) and serine protease antagonists (J.R. Forney et al., J. Parasitol., 82:638-640 [1996]). Other enzymes essential to C. parvum infectivity provide useful inhibitor targets. These include, for example, phosphoinositide 3-kinase (J.R. Forney et al., Infect. Irmmun., 15 67:844-852 [1999]) and cysteine proteinase (M.V. Nesterenko et al., Microbios., 83:77-88 [1995]). Other naturally occurring bactericidal molecules (e.g., enzymes) contemplated for use in certain embodiments of the present invention, include, but are not limited to, lactoferrin, lactoperoxidase, bacterial permeability increasing protein (BPI), and Aprotinin. 20 (See e.g., B.A. Mannion et al., J. Clin. Invest., 85(3):853-860 [1990]; A. Pellegrini et al., Biochem. Biophys. Res. Commun., 222(2):559-565 [1996]; and P. Prohinar et al., Mol. Microbiol., 43(6):1493-1504 [2002]). In some embodiments of the present invention, the biocide component of the fusion protein comprises an antimicrobial polypeptide (See e.g., Antimicrobial Peptide Protocols, 25 ed. W. M. Shafer, Humana Press, Totowa, NJ [1997]) or a pore fonning agent. In some embodiments, the antimicrobial peptide or pore forming agent is a compound or peptide selected from the following: magainin (e.g., magainin I, magainin II, xenopsin, xenopsin precursor fragment, caeralein precursor fragment), magainin I and II analogs (PGLa, magainin A, magainin G, pexiganin, Z-12, pexigainin acetate, D35, MSI-78A, MGO [K1OE, 30 K11 E, F12W-magainin 2], MG2+ [K1 OE, F12W-magainin-2], MG4+ [F12W-magainin 2], MG6+ [fl2W, E19Q-magainin 2 amide], MSI-238, reversed magainin II analogs [e.g., 53D, 87-ISM, and A87-ISM], Ala-magainin II amide, magainin II amide), cecropin P1, cecropin 54 WO 2007/047189 PCT/US2006/039361 A, cecropin B, indolicidin, nisin, ranalexin, lactoferricin B, poly-L-lysine, cecropin A (1-8) magainin 11 (1-12), cecropin A (1-8)-melittin (1-12), CA(1-13)-MA(1-13), CA(1-13)-ME(1 13), gramicidin, gramicidin A, gramicidin D, gramicidin S, alamethicin, protegrin, histatin, dermaseptin, lentivirus amphipathic peptide or analog, parasin I, lycotoxin I or II, 5 globomycin, gramicidin S, surfactin, ralinomycin, valinomycin, polymyxin B, PM2 [(±/-) 1-(4-aminobutyl)-6-benzylindane], PM2c [ (+/-) -6-benzyl-1-(3-carboxypropyl)indane], PM3 [(+/-)1-benzyl-6-(4-aminobutyl)indane], tachyplesin, buforin I or II, misgurin, melittin, PR-39, PR-26, 9-phenylnonylamine, (KLAKKLA)n, (KLAKLAK)n, where n = 1, 2, or 3, (KALKALK)3, KLGKKLG)n, and KAAKIKAA)n, wherein N = 1, 2, or 3, paradaxin, Bac 5, 10 Bac 7, ceratoxin, mdelin 1 and 5, bombin-like peptides, PGQ, cathelicidin, HD-5, Oabac5alpha, ChBac5, SMAP-29, Bac7.5, lactoferrin, granulysin, thionin, hevein and knottin-like peptides, MPG1, IbAMP, snakin, lipid transfer proteins, and plant defensins. In further embodiments, the antimicrobial polypeptide is a modified cathelicidin derived polypeptide (e.g., LL37, Ganz and Lehrer, 1997, Current Opinion in Hematology 4:53-58 15 and described by nucleotides 1474-1584 of SEQ ID NO: 102). Exemplary sequences for the above compounds are provided in Table 1. In some embodiments, the antimicrobial peptides are synthesized from L-amino acids, while in other embodiments, the peptides are synthesized from or comprise D-amino acids. Table 1 Antimicrobial Peptides SEQ ID Name Organism Sequence NO: 1 lingual antimicrobial Bos taurus MRLHHLLLALLFLVLSAGSGFTQGVR peptide precursor NSQSCRRNKGICVP (Magainin) IRCPGSMRQIGTCLGAQVKCCRRK 2 antimicrobial peptide Xenopus GVLSNVIGYLKIKLGTGALNAVLKQ PGQ laevis 3 Xenopsin Xenopus MYKGIFLCVLLAVICANSLATPSSDA laevis DEDNDEVERYVRGW 55 WO 2007/047189 PCT/US2006/039361 Table 1 Antimicrobial Peptides SEQ ID Name Organism Sequence NO: ASKIGQTLGKIAKVGLKIELIQPKREA MLRSAEAQGKRPWIL 4 magainin precursor Xenopus MFKGLFICSLIAVICANALPQPEASAD laevis EDMDEREVRGIGKFLHSAGKFGKAF VGEIMKSKRDAEAVGPEAFADEDLD EREVRGIGKFLHSAKKFGKAFVGEIM NSKRDAEAVGPEAFADEDLDEREVR GIGKFLHSAKKFGKAFVGEIMNSKRD AEAVGPEAFADEDLDEREVRGIGKFL HSAKKFGKAFVGEIMNSKRDAEAVG PEAFADEDFDEREVRGIGKFLHSAKK FGKAFVGEIMNSKRDAEAVGPEAFA DEDLDEREVRGIGKFLHSAKKFGK AFVGEIMNSKRDAEAVDDRRWVE 5 tachyplesin I Tachypleus KWCFRVCYRGICYRRCR gigas 6 tachyplesin II Tachypleus RWCFRVCYRGICYRKCR gigas 7 buforin I Bufo bufo MSGRGKQGGKVRAKAKTRSSRAGL gagarizans QFPVGRVHRLLRKGNYAQRVGAGA PVYLAAVLEYLTAEILELAGNAARD NKKTRIIPRHLQLAVRNDEELNKLLG GVTIAQGGVLPNIQAVLLPKT ESSKPAKSK 8 buforin II Bufo bufo TRSSRAGLQFPVGRVHRLLRK 56 WO 2007/047189 PCT/US2006/039361 Table 1 Antimicrobial Peptides SEQ ID Name Organism Sequence NO: gagarizans 9 cecropin A Bombyx mori MNFVRILSFVFALVLALGAVSAAPEP RWKLFKKIEKVGRNVRDGLIIAGPAI AVIGQAKSLGK 10 cecropin B Bombyx mori MNFAKILSFVFALVLALSMTSAAPEP RWKIFKKIEKMGRN IRDGIVKAGPAIEVLGSAKAIGK 11 cecropin C Drosophila MNFYKIFVFVALILAISIGQSEAGWLK melanogaste KLGKRIERIGQHT r RDATIQGLGIAQQAANVAATARG 12 cecropin P1 Sus scrofa SWLSKTAKILENSAKKRISEGIAIAIQ GGPR 13 indolicidin Bos taurus ILPWKWPWWPWRR 14 nisin Lactococcus ITSISLCTPGCKTGALMGCNMKTATC lactis HCSIHVSK 15 ranalexin Rana FLGGLIKIVPAMICAVTKKC catesbeiana 16 lactoferricin B Bos taurus FKCRRWQWRMKKLGAPSITCVRRAF 17 protegrin-1 Sus scrofa RGGRLCYCRRRFCVCVGRX 18 protegrin-2 Sus scrofa GGRLCYCRRRFCICVG 19 histatin precursor Hoino MKFFVFALILALMLSMTGADSHAKR sapiens HHGYKRKFHEKIHHSHRGYRSNYLY DN 57 WO 2007/047189 PCT/US2006/039361 Table 1 Antimicrobial Peptides SEQ ID Name Organism Sequence NO: 20 histatin 1 Macaca DSHEERHHGRHGHHKYGRKFHEKH fascicularis HSHRGYRSNYLYDN 21 dermaseptin Phyllomedus ALWKTMLKKLGTMALHAGKAALGA a sauvagei AADTISQTQ 22 dermaseptin 2 Phyllomedus ALWFTMLKKLGTMALHAGKAALGA a sauvagei AANTISQGTQ 23 dermaseptin 3 Phyllomedus ALWKNMLKGIGKLAGKAALGAVKK a sauvagei LVGAES 24 misgurin Misgurnus RQRVEELSKFSKKGAAARRRK anguillicaud atus 25 melittin Apis GIGAVLKVLTTGLPALISWISRKIRQ mellifera Q 26 pardaxin-1 Pardachirus GFFALIPKIISSPLFKTLLSAVGSALSSS pavoninus GEQE 27 pardaxin-2 Pardachirus GFFALIPKIJSSPIFKTLLSAVGSALSSS pavoninus GGQE 28 bactenecin 5 Bos taurus METQRASLSLGRCSLWLLLLGLVLPS precursor ASAQALSYREAVLR AVDQFNERSSEANLYRLLELDPTPND DLDPGTRKPVSFRV IETDCPRTSQQPLEQCDFIGENGLVK QCVGTVTLDPSNDQFDINCNELQSVR FRPPIRRPPIRPPFYPPFRPPIRPPIFPPIR 58 WO 2007/047189 PCT/US2006/039361 Table 1 Antimicrobial Peptides SEQ ID Name Organism Sequence NO: PPFRPPLGPFPGRR 29 bactenecin precursor Bos taurus METPRASLSLGRWSLWLLLLGLALPS ASAQALSYREAVLR AVDQLNEQSSEPNIYRLLELDQPPQD DEDPDSPKRVSFRVKETVCSRTTQQP PEQCDFKENGLLKRCEGTVTLDQVR GNFDITCNNHQSIRITKQPWAPPQAA RLCRIVVIRVCR 30 ceratotoxin A Ceratitis SIGSALKKALPVAKICGKIALPIAKAA capitata LP 31 ceratotoxin B Ceratitis SIGSAFKKALPVAKKIGjKALPAjCA capitata ALP 32 cathelicidin Homo MKTQRNGHSLGRWSLVLLLLGLVMP antimicrobial peptide sapiens LAIJAQVLSYKEAVL RAIDGINQRSSDANLYRLLDLDPRPT MDGDPDTPKPVSFT VKETVCPRTTQQSPEDCDFKKDGLV KRCMGTVTLNQARGSFDISCDKDNK RFALLGDFFRSIGKEIGKEFKRIVQRI KDFLRNLVPRTES 33 myeloid cathelicidin Equus METQRNTRCLGRWSPLLLLLGLVIPP 3 caballus ATTQALSYIEAVLRAVDGLNQRSSD ENLYRLLELDPLPKGDKDSDTPKPVS FMVIETVCPRIMKQTPEQCDFIGNG LVKQCVGTVILDPVKDYFDASCDEP QRVERFHSVGSLIQRHQQMIRDKSEA 59 WO 2007/047189 PCT/US2006/039361 Table 1 Antimicrobial Peptides SEQ ID Name Organism Sequence NO: TRHGIRIITRPKLLLAS 34 myeloid Bos taurus METQRASLSLGRWSLWLLLLGLALP antimicrobial peptide SASAQALSYREAVLR BMAP-28 AVDQLNEKSSEANLYRLLELDPPPIE DDENPNIPKPVSFRVIETVCPRTSQQ SPEQCDFIGNGLLKECVGTVTLDQV GSNFDITCAVPQSVGGLRSLGRKILR AWKKYGPIIVPIIRIG 35 myeloid cathelicidin Equus METQRNTRCLGRWSPLLLLLGLVIPP 1 caballus ATTQALSYIGAVLR AVDGLNQRSSDENLYRLLELDPLPKG DKDSDTPKPVSFMVIETVCPRIMKQ TPEQCDFKENGLVKQCVGTVILGPV KDHFDVSCGEPQRVERFGRLAKSFL RMRILLPRRKILLAS 36 SMAP 29 Ovis aries METQRASLSLGRCSLWLLLLGLALPS ASAQVLSYREAVLRAADQLNEKSSE ANLYRLLELDPPPKQDDENSNIKPV SFRVIETVCPRTSQQPAEQCDFIENG LLKECVGTVTLDQVRNNFDITCAEPQ SVRGLRRLGRKIAHGVKKYGPTVLRI IRIAG 37 BNP-1 Bos taurus RLCRIVVIRVCR 38 HNP-1 Homo ACYCRIPACIAGERRYGTCIYQGRLW sapiens AFCC 60 WO 2007/047189 PCT/US2006/039361 Table 1 Antimicrobial Peptides SEQ ID Name Organism Sequence NO: 39 HNP-2 Homo CYCRIPACIAGERRYGTCIYQGRLWA sapiens FCC 40 HNP-3 Homo DCYCRIPACIAGERRYGTCIYQGRLW sapiens AFCC 41 HNP-4 Homo VCSCRLVFCRRTELRVGNCLIGGVSF sapiens TYCCTRV 42 NP-1 Oryctolagus VVCACRRALCLPRERRAGFCRIRGRI cuniculus HPLCCRR 43 NP-2 Oryctolagus VVCACRRALCLPLERRAGFCRIRGRI cuniculus HPLCCRR 44 NP-3A Oryctolagus GICACRRRFCPNSERFSGYCRVNGAR cuniculus YVRCCSRR 45 NP-3B Oryctolagus GRCVCRKQLLCSYRERRIGDCKIRGV cuniculus RFPFCCPR 46 NP-4 Oryctolagus VSCTCRRFSCGFGERASGSCTVNGG cuniculus VRHTLCCRR 47 NP-5 Oryctolagus VFCTCRGFLCGSGERASGSCTINGVR cuniculus HTLCCRR 48 RatNP-1 Rattus VTCYCRRTRCGFRERLSGACGYRGRI norvegicus YRLCCR 49 Rat-NP-3 Rattus CSCRYSSCRFGERLLSGACRLNGRIY norvegicus RLCC 50 Rat-NP-4 Rattus ACTCRIGACVSGERLTGACGLNGRIY 61 WO 2007/047189 PCT/US2006/039361 Table 1 Antimicrobial Peptides SEQ ID Name Organism Sequence NO: norvegicus RLCCR 51 GPNP Guinea pig RRCICTTRTCRFPYRRLGTCIFQNRVY TFCC 52 beta defensin-3 Homo MRIHYLLFALLFLFLVPVPGHGGIINT sapiens LQKYYCRVRGGRC AVLSCLPKEEQIGKCSTRGRKCCRRK K 53 theta defensin-1 Macaca RCICTRGFCRCLCRRGVC mulatta 54 defensin CUA1 Helianthus MKSSMKMFAALLLVVMCLLANEMG annuus GPLVVEARTCESQSHKFKGTCLSDTN CANVCHSERFSGGKCRGFRRRCFCT THC 55 defensin SD2 Helianthus MKSSMKMFAALLLVVMCLLANEMG annuus GPLVVEARTCESQSHKFKGTCLSDTN CANVCHSERFSGGKCRGFRRRCFCT THC 56 neutrophil defensin 2 Macaca ACYCRIPACLAGERRYGTCFYMGRV mulatta WAFCC 57 4 KDA defensin Androctonus GFGCPFNQGACHRHCRSIRRRGGYC australis AGLFKQTCTCYR hector 58 defensin Mytilus GFGCPNNYQCHRHCKSIPGRCGGYC galloprovinci GGXHRLRCTCYRC 62 WO 2007/047189 PCT/US2006/039361 Table 1 Antimicrobial Peptides SEQ ID Name Organism Sequence NO: alis 59 defensin AMPI Heuchera DGVKLCDVPSGTWSGHCGSSSKCSQ sanguinea QCKDREHFAYGGACH YQFPSVKCFCKRQC 60 defensin AMP1 Clitoria NLCERASLTWTGNCGNTGHCDTQCR ternatea NWESAKHGACHKRGN WKCFCYFNC 61 cysteine-rich Mus MKKLVLLFALVLLAFQVQADSIQNT cryptdin-1 homolog musculus DEETKTEEQPGEKDQAVSVSFGDPQ GSALQDAALGWGRRCPQCPRCPSCP SCPRC PRCPRCKCNPK 62 beta-defensin-9 Bos taurus QGVRNFVTCRINRGFCVPIRCPGHRR QIGTCLGPQIKCCR 63 beta-defensin-7 Bos taurus QGVRNFVTCRlNRGFCVPIRCPGHRR QIGTCLGPRIKCCR 64 beta-defensin-6 Bos taurus QGVRNHVTCRIYGGFCVPIRCPGRTR QIGTCFGRPVKCCRRW 65 beta-defensin-5 Bos taurus QVVRNPQSCRWNMGVCIPISCPGNM RQIGTCFGPRVPCCR 66 beta-defensin-4 Bos taurus QRVRNPQSCRWNMGVCIPFLCRVG MRQIGTCFGPRVPCCRR 67 beta-defensin-3 Bos taurus QGVRNHVTCRINRGFCVPIRCPGRTR QIGTCFGPRI(CCRSW 63 WO 2007/047189 PCT/US2006/039361 Table 1 Antimicrobial Peptides SEQ ID Name Organism Sequence NO: 68 beta-defensin-10 Bos taurus QGVRSYLSCWGNRGICLLNRCPGRM RQIGTCLAPRVKCCR 69 beta-defensin-13 Bos taurus SGISGPLSCGRNGGVCIPIRCPVPMRQ IGTCFGRPVKCCRSW 70 beta-defensin-1 Bos taurus DFASCHTNGGICLPNRCPGHMIQIGIC FRPRVKCCRSW 71 coleoptericin Zophobas SLQGGAPNFPQPSQQNGGWQVSPDL atratus GRDDKGNTRGQIEIQNKGKDHDFNA GWGKVIRGPNKAKPTWHVGGTYRR 72 beta defensin-3 Homo MRIHYLLFALLFLFLVPVPGHGGIINT sapiens LQKYYCRVRGGRCAVLSCLPKEEQI GKCSTRGRKCCRRKK 73 defensin C Aedes ATCDLLSGFGVGDSACAAHCIARGN aegypti RGGYCNSKKVCVCRN 74 defensin B Mytilus GFGCPNDYPCHRHCKSIPGRYGGYC edulis GGXHRLRCTC 75 sapecin C Sarcophaga ATCDLLSGIGVQHSACALHCVFRGN peregrina RGGYCTGKGICVCRN 76 macrophage Oryctolagus MRTLALLAAILLVALQAQAEHVSVSI antibiotic peptide cuniculus DEVVDQQPPQAEDQDVAIYVKEHES MCP-1 SALEALGVKAGVVCACRRALCLPRE RRAG FCRIRGRIHPLCCRR 77 cryptdin-2 Mus MKIPLVLLSALVLLSFQVQADPIQNTD 64 WO 2007/047189 PCT/US2006/039361 Table 1 Antimicrobial Peptides SEQ ID Name Organism Sequence NO: musculus EETKTEEQSGEEDQAVSVSFGDREG ASLQEESLRDLVCYCRTRGCKRRER MNGT CRKGHLMYTLCC 78 cryptdin-5 Mus MKTFVLLSALVLLAFQVQADPIHKTD musculus EETNTEEQPGEEDQ AVSISFGGQEGSALHEELSKKLICYCR IRGCKRRERVFGT CRNLFLTFVFCCS 79 cryptdin 12 Mus LRDLVCYCRARGCKGRERMNGTCR musculus KGHLLYMLCCR 80 defensin Pyrrhocoris ATCDILSFQSQWVTPNHAGCALHCVI apterus KGYKGGQCKITVCHCRR 81 defensin R-5 Rattus VTCYCRSTRCGFRERLSGACGYRGRI norvegicus YRLCCR 82 defensin R-2 Rattus VTCSCRTSSCRFGERLSGACRLNGRI norvegicus YRLCC 83 defensin NP-6 Oiyctolagus GICACRRRFCLNFEQFSGYCRVNGAR cuniculus YVRCCSRR 84 beta-defensin-2 Pan MRVLYLLFSFLFIFLMPLPGVFGGISD troglodytes PVTCLKSGAICHP VFCPRRYKQIGTCGLPGTKCCKIKP 85 beta-defensin-2 Homo MRVLYLLFSFLFIFLMPLPGVFGGIGD sapiens PVTCLKSGAICHP VFCPRRYKQIGTCGLPGTKCCKKP 86 beta-defensin-1 Homo MRTSYLLLFTLCLLLSEMASGGNFLT 65 WO 2007/047189 PCT/US2006/039361 Table 1 Antimicrobial Peptides SEQ ID Name Organism Sequence NO: sapiens GLGHRSDHYNCVSS GGQCLYSACPIFTKIQGTCYRGKAKC CK 87 beta-defensin-1 Capra circus MRLHHLLLVLFFLVLSAGSGFTQGIR SRRSCHRNKGVCAL TRCPRNMRQIGTCFGPPVKCCRKK 88 beta defensin-2 Capra hircus MRLHHLLLALFFLVLSAGSGFTQGIN HRSCYRNKGVCAP ARCPRNMRQIGTCHGPPVKCCRCKC 89 defensin-3 Macaca MRTLVILAAILLVALQAQAEPLQART mulatta DEATAAQEQIPTDNPEVVVSLAWDE SLAPKDSVPGLRKNMACYClgIPACL AGER RYGTCFYRRRVWAFCC 90 defensin-1 Macaca MRTLVILAAILLVALQAQAEPLQART mulatta DEATAAQEQIPTDNPEVVVSLAWDE SLAPKDSVPGLRKNMACYCRIPACL AGER RYGTCFYLGRVWAFCC 91 neutrophil defensin 1 Mesocricetus VTCFCRRRGCASRERHIGYCRFGNTI auratus YRLCCRR 92 neutrophil defensin 1 Mesocricetus CFCKRPVCDSGETQIGYCRLGNTFYR auratus LCCRQ 93 Gallinacin 1-alpha Gallus gallus GRKSDCFRKNGFCAFLKCPYLTLISG KCSRFHLCCKRIW 94 defensin Allonyrina VTCDLLSFEAKGFAANHSLCAAHCL 66 WO 2007/047189 PCT/US2006/039361 Table1I Antimicrobial Peptides SEQ ID Name Organism Sequence NO: dichotoma AIGRRGGSCERGVCICRR 95 neutrophil cationic Cavia RRCICTTRTCRFPYRRLGTCIFQNRVY peptide 1 porcellus TFCC In some embodiments of the present invention, the antimicrobial polypeptide is a defensin. In preferred embodiments, the compositions of the present invention comprise one or more defensins. In some of these embodiments, the antimicrobial polypeptide 5 defensin is BNP1 (also known as bactanecin and bovine dodecapeptide). In certain embodiments, the defensin comprises the following consensus sequence: (SEQ ID NO:96 X 1
CN
1
CRN
2
CN
3
ERN
4
CN
5
GN
6
CCX
2 , wherein N and X represent conservatively or nonconservatively substituted amino acids and N 1 = 1, N 2 = 3 or 4, N 3 = 3 or 4, N 4 1, 2, or 3, N 6 = 5-9, Xi and X 2 may be present, absent, or equal from 1-2. The present invention is 10 not limited to any particular defensin. Representative defensins are provided in Tables 1 and 2. Table 2 Defensins SEQ ID NO Name Organism Sequence 38 HNP-1 Human ACYCRIPACIAGERRYGTCIYQGRLWA FCC 39 HNP-2 Human CYCRIPACIAGERRYGTCIYQGRLWAF CC 40 HNP-3 Human DCYCRIPACIAGERRYGTCIYQGRLWA FCC 67 WO 2007/047189 PCT/US2006/039361 Table 2 Defensins SEQ ID NO Name Organism Sequence 41 HNP-4 Human VCSRLVCRLVFCRRTELRVGNCLIGGVSFTY CCTRV 42 NP-1 Rabbit VVCACRRALCLPRERRAGFCRIRGRIHP LCCRR 43 NP-2 Rabbit VVCACRRALCLPLERRAGFCRIRGRIHP LCCRR 44 NP-3A Rabbit GICACRRRFCPNSERFSGYCRVNGARY VRCCSRR 45 NP-3B Rabbit GRCVCRKQLLCSYRERRIGDCKIRGVR FPFCCPR 46 NP-4 Rabbit VSCTCRRFSCGFGERASGSCTVNGVRH TLCCRR 47 NP-5 Rabbit VFCTCRGFLCGSGERASGSCTINGVRHT LCCRR 48 RatNP-1 Rat VTCYCRRTRCGFRERLSGACGYRGRIY RLCCR 49 Rat-NP-3 Rat CSCRYSSCRFGERLLSGACRLNGRIYRL cc 50 Rat-NP-4 Rat ACTCRIGACVSGERLTGACGLNGRIYR LCCR 51 GPNP Guinea pig RRCICTTRTCRFPYRRLGTCIFQNRVYTF cc 68 WO 2007/047189 PCT/US2006/039361 In general, defensins are a family of highly cross-linked, structurally homologous antimicrobial peptides found in the azurophil granules of polymorphonuclear leukocytes (PMN's) with homologous peptides being present in macrophages. (See e.g., Selsted et al., 5 Infect. Immun., 45:150-154 [1984]). Originally described as "Lysosomal Cationic Peptides" in rabbit and guinea pig PMN (Zeya et al., Science, 154:1049-1051 [1966]; Zeya et al., J. Exp. Med., 127:927-941 [1968]; Zeya et al., Lab. Invest., 24:229-236 [1971]; Selsted et al., [1984], supra.), this mixture was found to account for most-of the microbicidal activity of the crude rabbit PMN extract against various microorganisms (Zeya et al., [1966], supra; 10 Lehrer et al., J. Infect. Dis., 136:96-99 [1977]; Lehrer et al., Infect. Immun., 11:1226-1234 [1975]). Six rabbit neutrophil defensiis have been individually purified and are designated NP-1, NP-2, NP-3A, NP-3B, NP-4, and NP-5. Their amino acid sequences were determined, and their broad spectra of activity were demonstrated against a number of bacteria (Selsted et al., Infect. Immun., 45:150-154 [1984]), viruses (Lehrer et al., J. Virol. 15 54:467 [1985]), and fungi (Selsted et al., Infect. Immun., 49:202-206 [1985]; Segal et al., 151:890-894 [1985]). Defensins have also been shown to possess mitogenic activity (e.g., Murphy et al., J. Cell. Physiol., 155:408-13 [1993]). Four peptides of the defensin family have been isolated from human PMN's and are designated HNP-1, HNP-2, HNP-3, and HNP-4 (Ganz et al., J. Clin. Invest., 76:1427-1435 20 [1985]; Wilde et al., J. Biol. Chem., 264:11200-11203 [1989]). The amino acid sequences of HNP-1, HNP-2, and HNP-3 differ from each other only in their amino tenninal residues, while each of the human defensins are identical to the six rabbit peptides in 10 or 11 of their 29 to 30 residues. These are the same 10 or 11 residues that are shared by all six rabbit peptides. Human defensin peptides have been shown to share with the rabbit defensins a 25 broad spectrum of antimicrobial activity against bacteria, fungi, and enveloped viruses (Ganz et al., [1985], supra). Three defensins designated RatNP-1, RatNP-2, and RatNP-4, have been isolated from rat. (Eisenhauer et al., Infection and Immunity, 57:2021-2027 [1989]). A guinea pig defensin (GPNP) has also been isolated, purified, sequenced and its broad spectrum 30 antimicrobial properties verified (Selsted et al., Infect. Immun., 55:2281-2286 [1987]). Eight of its 31 residues were among those invariant in six rabbit and three human defensin peptides. The sequence of GPNP also included three nonconservative substitutions in 69 WO 2007/047189 PCT/US2006/039361 positions otherwise invariant in the human and rabbit peptides. Of the defensins tested in a quantitative assay HNP- 1, RatNP- 1, and rabbit NP-1 possess the most potent antimicrobial properties, while NP-5 possesses the least amount of antimicrobial activity when tested against a panel of organisms in stationary growth phase. (Selsted et al., Infect. Inimun., 5 45:150-154 [1984]; Ganz et al., J. Clin. Invest. 76:1427-1435 [1985]). Defensin peptides are further described in US 4,543,252; 4,659,692; and 4,705,777 (each of which is incorporated herein by reference). Accordingly, in some embodiments, the compositions of the present invention comprise one or more defensins selected from the group consisting of SEQ ID NOs: 37-95. 10 In preferred embodiments, suitable antimicrobial peptides comprise all or part of the amino acid sequence of a known peptide, more preferably incorporating at least some of the conserved regions identified in Table 2. In particularly preferred embodiments, the antimicrobial peptides incorporate at least one of the conserved regions, more usually incorporating two of the conserved regions, preferably conserving at least three of the 15 conserved regions, and more preferably conserving four or more of the conserved regions. In preferred embodiments, the antimicrobial peptides comprise fifty amino acids or fewer, although there may be advantages in increasing the size of the peptide above that of the natural peptides in certain instances. In certain embodiments, the peptides have a length in the range from about 10 to 50 amino acids, preferably being in the range from about 10 to 20 40 amino acids, and most preferably being in the range from about 30 to 35 amino acids which corresponds generally to the length of the natural defensin peptides. In some embodiments, the present invention provides antibodies (or portions thereof) fused to biocidal molecules (e.g., lysozyme) (or portions thereof) suitable for use with processed food products as a whey based coating applied to food packaging and/or as a 25 food additive. In still other embodiments, the compositions of the present invention are formulated for use as disinfectants for use in food processing facilities. Additional embodiments of the present invention provide human and animal therapeutics. V. Applications 30 The methods and compositions of the present invention find use in a variety of applications, including, but not limited to, those described below. 70 WO 2007/047189 PCT/US2006/039361 A. Exemplary Target Pathogens i) Escherichia coli 0157:H7 Preferred embodiments of the present invention provide effective therapeutic 5 treatments and prophylactic methods for combating the foodborne pathogen E. coli 0157:H7. E. coli 0157:H7 is a common component of the normal flora of the bovine gastrointestinal tract. Surveys of clinically normal cattle detect E. coli 0157:H7 shedding in the feces of about 1-25% of animals. (D.D. Hancock DD et al., Epidemiol. Infect., 113(2):199-207 [1994]; MMWR Morb. Mortal. Wkly. Rep., 48(36):803-805 [1999]; and 10 National Dairy Heifer Evaluation Project, USDA APHIS NAHMS [1994]). Human infection by E. coli 0157:H7 most often occurs through the fecal-oral route, either directly through handling of infected cattle, or more commonly as a result consuming contaminated meat products. Small-scale outbreaks of E. coli 0157:H7 disease have been associated with petting zoos and agricultural fairs. (See e.g., G.C. Pritchard et al., Vet. Rec., 147:259-264 15 [2000]). Larger outbreaks of E. coli 0157:H7 disease have been traced to the widespread dissemination and consumption of contaminated meat products such as ground beef. (J. Tuttle et al., Epidemiol. Infect., 122:185-192 [1999]). Contamination of meat products with fecal matter harboring E. coli 0157:H7 appears to occur primarily during slaughterhouse dehiding, evisceration, splitting, chilling, 20 and fabrication operations. Further dissemination of E. coli 0157:H7 to otherwise uncontaminated meat products also occurs during grinding, processing, and transportation of meat products. Because of the severity of E. coli 0157:H7 disease and the potential for the contamination of large quantities of otherwise wholesome meat by a relatively small amount of contaminated meat, the recalls issued for potentially contaminated meat products are 25 often very large. Recent E. coli 0157:H7 contaminated meat recalls, include the recall of 24 million pounds of ground beef by the Hudson Beef in 1997, and most recently the recall of 19 million pounds of ground beef by the ConAgra Beef Company in July 2002. Recalls of this magnitude are obviously very costly; not only for actual value of the beef being destroyed, 30 but also the logistical effort required to collect and dispose of the contaminated beef. More importantly, immeasurable costs arise from decreased consumer confidence in the meat packing industry and in the wholesomeness of food supply generally. 71 WO 2007/047189 PCT/US2006/039361 E. coli 0157:H7 is particularly pathogenic because it produces a multi-unit verotoxin (or a shiga-like toxin) protein that binds receptors in the kidney and gastrointestinal tract of man. This toxin produces a hemolytic uremic syndrome in children and the elderly, and a hemorrhagic colitis in adults. The Center for Disease Control 5 reported over 70,000 cases of E. coli 0157:H7 disease each year and 60 deaths annually. The symptoms of hemorrhagic colitis last an average of 8 days. This implies that over half a million work days are lost per year due to E. coli 0157:117 infection. E. coli 0157:H7 disease is a costly and frustrating zoonosis, in part because its epidemiology is well understood yet very difficulty to prevent. The recent massive 10 dissemination of the organism I contaminated meat products is a function of the industrialization processes that are essential to providing affordable food. Preferred embodiments of the present invention combat E. coli 0157:H7, by providing chimeric murine-bovine monoclonal antibodies to provide passive immunity in host animals (e.g., bovines), to induce specific immunity in host animals due to the bovine 15 portion of the antibody, to topically control E. co/i 0157:H7 post harvest. Additional preferred embodiments provide chimeric murine-bovine monoclonal antibody fusion proteins that directly reduce E. coli 0157:H7 in host animals by providing highly controlled and targeted bactericidal microenvironments that destroy the pathogens without affecting normal microbiological flora. 20 ii) Listeria monocytogenes Ingestion of the bacterium Listeria monocytogenes probably occurs quite often, as the bacteria has been isolated in food products worldwide. (B. Lorber, Clin. Infect. Dis., 24:1-11 [1997]; and J.M. Farber and P.I. Peterkin, Microbiol. Rev., 55:476-511 [1991]). 25 Development of Listeriosis, the invasive disease caused by L. monocytogenes ingestion, is determined primarily by the integrity of the host's immune system (predominantly cell mediated immune defects) and possibly also by inoculum size. (See e.g., P. Aureli et al., New Engl. J. Med., 342:1236-1241 [2000]). L. monocytogenes crosses the mucosal barrier of the intestines and invades the bloodstream. The bacterium may disseminate to any organ, 30 but has a clear predilection for the placenta and central nervous system (CNS), thus determining the main clinical syndromes caused by infection. Listeriosis is life-threatening zoonosis, especially in human fetuses and neonates, the elderly, and patients with certain 72 WO 2007/047189 PCT/US2006/039361 predisposing conditions. Many cases of Listeriosis probably go unreported especially in perinates and newborns. L. monocytogenes is a difficult bacterium to control because it thrives in vacuum packed food products and at temperatures typically used in food refrigeration. (S. Liu et al., 5 Appl. Environ. Microbiol., 68(4):1697-1705 [2002]). Thus, L. nonocytogenes is a particular problem in ready to eat foods that consumers expect are safe to eat with no further cooking. Typical food products contaminated with L. monocytogenes include unpasteurized or low acid dairy products and ready-to-eat (RTE) meat products such as luncheon meat and pates. 10 Many of the larger listeriosis outbreaks have been associated with fresh dairy products, especially Mexican soft cheeses and other non-aged or fermented cheese products. (M.J. Linnan et al., New Eng. J. Med., 319:823-828 [1988]). In specialty cheese production, L. monocytogenes has been found to accumulate in ripening rooms. (S.I. Pak et al., supra). Outbreaks have also been linked to processed and deli meat products, including 15 turkey hot dogs, pate, and jellied tongue. (See e.g., C. Jacquet et al., Appl. Env. Microbiol., 61:2242-2246 [1995]; J. McLaughlin et al., Brit Med. J., 303:773-775 [1991]; and Morbidity and Mortality Weekly Reports, 47:1085-1086 [1998]). L. monocytogenes is however, easily destroyed by cooking. As Listeria is heat labile, food products that cooked prior to eating and served hot have a lower risk profile. 20 The bacterium often contaminants food processing equipment, where it is typically found as a biofilm on steel and glass parts. L. monocytogenes tends to form biofilms on containers used to store food products. (A.C. Wong, J. Dairy Sci., 81(10):2765-2770 [1998]). It is also a common environmental contaminant of food storage facilities. (See e.g., M.S. Chae and H. Schraft, Int. J. Food. Microbiol., 62:103-111 [2000]). Thus, a 25 pathogen originally considered an environmentall" organism on farms has invaded industrial food preparation facilities. Strains of L. monocytogenes have emerged with progressive resistance to antimicrobial agents. (C. Arizcun et al., J. Food Prot., 61:731-734 [1988]). Contamination of food processing equipment and storage often seed small amounts of bacteria onto food products which multiply during refrigeration. The length of 30 time food is kept refrigerated, both at the retail outlet and in home, and the actual storage temperatures influence the risk that even low levels of initial L. nonocytogenes contamination will grow to become problematic. Outbreaks of listeriosis have been 73 WO 2007/047189 PCT/US2006/039361 associated with breakdowns in the environmental controls at food processing facilities (e.g., during plant renovation). Given the capacity of L. monocytogenes to grow under refrigeration, even very low levels of L. monocytogenes contamination in food products as they leave the processor can ultimately result in inoculums large enough to cause lethal 5 infections in the consumer. Consequently, the present invention contemplates controlling foodborne listeriosis requires focusing on post-processing food handling and storage. L. monocytogenes is recognized as an apparent and inapparent infection of livestock and is widespread in agricultural environments. (See e.g., L. Hassan et al., J. Dairy Sci., 83(11):2441-2447 [2000]). L. monocytogenes is widespread in the environment and shed in 10 the feces and milk of inapparently infected cattle. (L. Hassan et al., supra). Even low levels of L. monocytogenes contamination are enough to support the continued growth of the organism and are the primary source of human listeriosis. Listeriosis is a serious disease. In milder its forms, listeriosis is a febrile illness with gastrointestinal signs but often progresses to bacteremia and meningitis with nervous system 15 clinical manifestations including headache, loss of balance, and convulsions. Incubation periods can be several weeks making epidemiologic investigation more difficult. Listeriosis is particularly serious for pregnant women, infants, and individuals with compromised immune systems. (See e.g., A. Schuchat et al., Clinical Microbiol. Rev., 4:169-183 [1991]). Individuals with Acquired Immune Deficiency Syndrome (AIDS) are almost 300 times more 20 likely to contract listeriosis than people with normally functioning immune systems. (J.G. Morris and M. Potter, Emerging Infectious Diseases, 3:435-441 [1997]). Diabetics and those affected by cancer and kidney disease are also at greater risk of contracting Listeriosis and are more prone to serious nervous system manifestations of the disease. Listeriosis in pregnant women can result in premature birth, still births, or birth of a critically infected 25 child. Perinates and newborns are particularly at risk as the relative immaturity of their immune systems has been shown to contribute to the severity of disease (See e.g., L. Slutzker and A. Schuchat, Listeriosis in humans. In: Listeria, Listeriosis and Food Safety, E.T. Ryser and E.M. Marth eds. Marcel Dekker Inc., New York, New York pp. 75-95 [1999]. Human foodborne listeriosis tends to result in either sporadic cases or epidemics 30 depending on whether a common food source infects a group of people (H.R. Ibrahim et al., FEBS Lett., 506(l):27-32 [2001]). Contaminated meats, seafood, vegetables, fruits, and 74 WO 2007/047189 PCT/US2006/039361 dairy products have all been the source of sporadic cases of listeriosis (S.I. Pak et al., Prev. Vet. Med., 53(1-2):55-65 [2002]). iii) Cryptosporidium parvum 5 Cryptosporidium parvum is a zoonotic apicomplexan parasite recognized as the cause of large outbreaks of acute diarrheal disease. The disease caused by Cryptosporidium infection is called cryptosporidiosis. Cryptosporidiosis has emerged as an important opportunistic infection in patients infected with HIV. With the advent of more effective HIV therapies, the association between Cryptosporidium infection and HIV has lessened in 10 the US, however opportunistic Ciyptosporidiosis following infection by HIV continues to be a major problem in developing countries. Cryptosporidium is also recognized as a leading cause of traveler's diarrhea. An acutely debilitating diarrheal disease, accompanied by stomach cramps, cryptosporidiosis typically lasts 2-10 days. In the otherwise healthy host, cryptosporidiosis is rarely fatal, but 15 deaths occur among the immunocompromised including AIDS patients, chemotherapy patients, malnourished individuals, and the elderly, who may become chronically diarrheic and in whom the parasite may establish hard-to-eliminate hepatobiliary and pancreatic infections. C. parvum infects cattle and other, livestock usually within the first few hours or days 20 of life. Infected animals can become long-term shedders of C. parvum oocysts. C. parvum is an economically important cause of diarrheal disease and mortality among calves which provide a significant reservoir for human infection. In swine, clinical disease cryptosporidiosis is less common, but C. parvum has been recognized as a highly prevalent contaminant of swine manure holding facilities. 25 C. parvun oocysts can survive for extended periods of time in water and soil contaminated from human or animal fecal shedding. The oocysts are not inactivated by chlorination, nor removed by many water filtration systems. Drinking water, recreational water contact, and fecally contaminated foodstuffs are the principal sources of infection for humans. Cryptosporidiun genotypes are epidemiologically and genetically distinct, 30 although overlap occurs and heterogeneous infections can occur. C. honinis is transmitted from human to human, while C. parvunz is zoonotic and transmitted between cattle and other livestock, and humans. While genetic polymorphisms occur in both species, the 75 WO 2007/047189 PCT/US2006/039361 extent of epitope homology between the genotypes and cross-protection is not fully understood. Dual infections may occur, but with no reproductive mixing of the genotypes. Nevertheless, the present invention shows that three functionally defined antigens are conserved at the protein level between several C. parvuin and C. hominis isolates. These 5 antigens, CSL, P23 and GP25-200, were originally defined on C. parvum isolates and previously shown to be the targets of neutralizing antibodies. The apparent conservation of antigens indicates that the compositions of the present invention using monoclonals antibodies to these antigens as neutralizing agents, either alone or to target a biocide to the parasite surface, will have application to infections by both species. The present invention 10 further contemplates compositions comprising polyvalent antibody passive immunotherapies to treat epidemics of unknown origin. Large outbreaks of human cryptosporidiosis demonstrate the potential for Cryptosporidium to be used as a bioterrorism agent. A 1993 outbreak of cryptosporidiosis in Milwaukee resulted in over 400,000 cases of clinical disease and several dozen deaths, 15 following dissemination of C. parvum through the public water supply. The Milwaukee experience suggests that large waterborne or food borne outbreaks of cryptosporidiosis could be brought about by deliberate contamination. With the ratio of infective oocysts per gram of feces shed by an individual to the infective dose approximating one million to one (shedding 106 or 107 oocysts per gram, compared to an infective dose 10-100), the potential 20 for producing major urban outbreaks is real. C. parvun has several attributes that lend it for use as a potential bioterrorism agent: infectious oocysts are very hardy and easily transported; infective oocysts are shed in very large numbers but have a low infective dose; cryptosporidiosis is unlikely to be fatal to the terrorist handler; oocysts are readily available without access to reference collections or high 25 security laboratories and can be easily propagated in neonatal ruminants (up to ~1010 oocysts from a single calf); and widespread dissemination can be achieved in food or water. Given a high background incidence of C parvum infections, an acute epidemic would be harder to trace back to a point source. Nevertheless, clinical signs are dramatic enough to cause panic, and to allow terrorist claims of responsibility to ring true. C. parvun thus fits 30 the profile of an organism which might be deployed by a "low tech" terrorist group without access to a well-developed laboratory infrastructure. 76 WO 2007/047189 PCT/US2006/039361 The recent British detection of "ricin laboratories" indicate that low tech bioterrorism is today's reality. If the intent of bioterrorism is to produce mass hysteria rather than mass disease and fatalities, a few cases of cryptosporidiosis implicating contamination of a major food or water supplies could have a dramatic psychological effect. 5 Water distribution systems across the country are relatively difficult to secure. Security of water supply is also of concern in assuring a safe food supply. In an urban society dependent on complex food distribution chains, a point source contamination could affect people across a wide geographic area. For example, the recent nationwide recall of 28 million pounds of processed turkey due to Listeria contamination in a single processing 10 plant illustrates that point source food contamination can have a very wide ripple effect. With only a moderate increment in microbiologic expertise, the effects of food bioterrorism could be devastating. Dairy effluent is considered an important source of natural C. parvum infection, likewise, swine effluent is suspected as being reservoir for infection. Controlling infection 15 in animal populations would help reduce the environmental risk of natural infections. Accordingly, certain embodiments of the present invention provide compositions to control zoonotic pathogens in animal reservoirs and agricultural environments. Prior to the present invention, there were no effective parasite-specific drug therapies to control or curtail cryptosporidiosis in man or animals. Nitazoxanide was recently 20 approved by FDA as the first drug for use in treating human cryptosporidiosis, although its parasite specificity remains equivocal and its lack of efficacy in animal models and inimuncompromised humans raises concerns. Other existing treatments are palliative and directed to avoiding onset of dehydration. (S. Tzipori and H. Ward, Microbes Infect., 4:1047 [2002]). Naturally occurring cases of cryptosporidiosis in human and animal hosts 25 with normal immunological systems can be severe, but are typically self limiting. (C.L. Chappell et al., Amer. J. Trop. Med. Hyg., 60:157-164 [1999]). In certain embodiments, colostral antibodies fed to calves limit infection and prevent clinical disease. In some other embodiments, polyclonal hyperimmune antibodies raised against C. parvum effectively limit clinical cases of the disease while allowing some active immunity to develop. 30 The present invention provides antibody-based immunoprophylaxis and immunotherapy that effectively control acute C. parvun infections. The present invention contemplates that the efficacy of compositions and methods of passive immunotherapy 77 WO 2007/047189 PCT/US2006/039361 comprising administering antibodies specifically developed against neutralization-sensitive epitopes is distinguishable from the host-produced antibodies in protection against natural infection, which depends on competent cell mediated immune responses (M. Riggs, Microbes Infect., 4:1067 [2002]). 5 Preferred embodiments provide compositions and methods for administering passive immunotherapies against pathogens (e.g., C. parvun infections). Faced with a population exposed to deliberately contaminated food or water, or in a battle theater setting, rapidly deployable, rapidly effective, passive immunotherapies are strategically and clinically very valuable. In some of these embodiments, the present invention provides orally administered 10 monoclonal antibody compositions that specifically target pathogens (e.g., parasites) and either prevent infection, or reduce an existing infection to subclinical levels and abbreviate existing clinical effects. In some embodiments, the present invention provides monoclonal antibodies against defined apical complex and surface-exposed antigens to specifically neutralize infective 15 stages of C. parvum in vitro and in vivo. The present invention also provides previously unavailable recombinant antibodies to C. parvun. Prior to the present invention, high cost and inefficient production systems for recombinant and hybridoma monoclonals alike have generally removed widespread immunoprophylaxis and/or immunotherapies for cryptosporidiosis from serious clinical consideration. 20 Some preferred embodiments of the present invention make use of an extensive bank of hybridoma lines directed to cryptosporidial antigens. A large number of C. parvum antigens of distinct function have been identified and characterized. (M.W. Riggs, Microbes. Infect., 4:1067 [2002]). Several antigens in particular have shown potential for independent targeting to neutralize sporozoite and merozoite infectivity, including, but not 25 limited to, CSL, P23, and GP25-200. Briefly, CSL (-1300 kDa) is an apical complex derived glycoprotein expressed on the surface of sporozoite and merozoite infective stages. After antibody binding to CSL, sporozoites release the antigen in membranous antibody CSL complexes and are rendered non-infective. (M.W. Riggs et al., J. Immunol., 158:1787 1795 [1997]). Since CSL has been shown to contain a ligand for a surface receptor on 30 human intestinal epithelial cells (See, R.C. Langer and M.W. Riggs, Infect. Immun., 67:5282-5291 [1999]; and R.C. Langer et al., Infect. Immun., 69:1661-1670 [2001]), blocking of CSL is contemplated to account for the efficacy of anti-CSL antibodies in 78 WO 2007/047189 PCT/US2006/039361 inhibiting sporozoite attachment. P23 (~23 kDa) is a surface protein of sporozoites and merozoites believed to be involved in motility and invasion processes (See, L.E. Perryman et al., Vaccine, 17:2142-2149 [1999]). Monospecific antibodies to P23 have been shown to curtail disease in neonatal calves. (L.E. Perryman et al., supra). GP25-200 is a 5 glycoprotein complex of variable size, found in the apical complex and on the surface of sporozoites and merozoites. (M.W. Riggs et al., supra). Schaefer et al. demonstrated that when hybridoma derived monoclonal antibodies to CSL, P23, and GP25-200 were applied singly, or in combination, significant sporozoite neutralization could be obtained. (D.A. Schaefer et al., Infect. Immun., 68:2608-2616 [2000]). 10 In some embodiments, optimal protection against oocyst challenge in neonatal mice is achieved by combining three antibodies: 3E2 (IgM) to target CSL; 3H2 (IgM) to target GP25-200; and IE1O (IgG1) to target P23. When these three antibodies are orally administered, individually, in the neonatal mouse infection model, they are able to reduce intestinal infection by 50-60%. When these antibodies are administered as a polyvalent 15 "cocktail" the three monoclonal antibodies reduced intestinal infection by 86-93% (D.A. Schaefer et al. supra). In an adult immunodeficient SCID mouse model of persistent, disseminated infection, 3E2, alone or in combination with 1 E10 and/or 3H2, also significantly reduced an established gastrointestinal infection (Riggs et al, Antimicrob. Agents. Chemother. 46:275-282). Preferred embodiments of the present invention provide 20 recombinant analogues of 3E2, 3H2, and 1E1O antibodies. Additional preferred embodiments provide fusion proteins comprising cryptosporocidal enzymes and antibodies (e.g., IgG), or portions thereof,. including, but not limited to, 3E2, 3H2, 1E1O, 18.44 and 4H9. 4H9 is a second antibody directed to GP25-200. It is an IgG1 antibody that is though to recognize a different epitope on GP25-200 than does 3H2. In some embodiments, 4H9 is 25 able to reduce infection in neonatal mice by ~50% when administered orally. Thus, ion stil other embodiments, compositions comprising 1E1O and 4H9 provide vehicles for delivering biocides to two different neutralization-sensitive molecules on the surface of sporozoites and merozoites. In other embodiments, monoclonal antibody 18.44 is utilized. 18.44 recognizes a b 30 mannosylated glycolipid (CPS-500). This is a conserved neutralization-sensitive antigen of C. parvum sporozoites and merozoites (Priest et al., J Biol Chem 2003; 278:52212-22; Riggs et al. Infect Immun 1999; 67:1317-22). It is also contemplated that 18.44 has pan 79 WO 2007/047189 PCT/US2006/039361 apicomplexan parasite eitope binding properties (e.g., Eimeria sp.). Eimeria is the causative agent of coccidiosis, an economically important parasite in birds. The present invention contemplates that using a monoclonal antibody fusion protein to direct otherwise naturally occurring biocides to specific pathogenic organisms (e.g., E. 5 coli 0157:H7, L. monocytogenes, Cryptosporidiumparvum and the like) has wide applicability in human and animal health. The present invention further contemplates compositions to control other pathogenic feedlot organisms including, but not limited to, E. coli K99 in calves and E. coli K88 in piglets. In still further embodiments, the present invention provides embodiments to control 10 other pathogens responsible for foodborne illnesses and other emerging infectious diseases as a component of the Nation's food security and bioterrorism response. For example, some embodiments of the present invention are focused on controlling potential foodborne bioterrorism agents such as, Clostridium botulinum, Clostridium perfringens, Staphylococcus aureus, Staphylococcus epidermidis, Staphylococcus pneumoniae, 15 Staphylococcus saprophyticus, Shigella dysenteriae, Salmonella typhi, Salmonella paratyphi, Salmonella enteritidis, fungal agents and the like. ii). Other Exemplary Target Pathogens In some other embodiments, the present methods and compositions are directed to 20 specifically controlling (e.g., therapeutic treatments or prophylactic measures) diseases caused by the following pathogens: Bartonella henselae, Borrelia burgdorferi, Campylobacterjejuni, Campylobacter fetus, Chlamydia trachomatis, Chlamydia pneumoniae, Chylamydia psittaci, Simkania negevensis, Escherichia coli (e.g., 0157:H7 and K88), Ehrlichia chafeensis, Clostridium botulinum, Clostridium perfringens, 25 Clostridium tetani, Enterococcusfaecalis, Haemophilius influenzae, Haemophilius ducreyi, Coccidioides immitis, Bordetella pertussis, Coxiella burnetii, Ureaplasma urealyticum, Mycoplasma genitalium, Trichomatis vaginalis, Helicobacter pylori, Helicobacter hepaticus, Legionella pneunzophila, Mycobacterium tuberculosis, Mycobacterium bovis, Mycobacterium africanum, Mtycobacterium leprae, Mycobacterium asiaticum, 30 Mycobacterium avium, Mycobacterium celatum, Mycobacterium celonae, Mycobacterium fortuitum, Mycobacterium genavense, Mycobacterium haemophilum, Mycobacterium intracellulare, Mycobacterium kansasii, Mycobacterium malmoense, Mycobacterium 80 WO 2007/047189 PCT/US2006/039361 marinum, Mycobacterium scrofulaceum, Mycobacterium simiae, Mycobacterium szulgai, Mycobacterium ulcerans, Mycobacterium xenopi, Coiynebacterium diptheriae, Rhodococcus equi, Rickettsia aeschlimannii, Rickettsia africae, Rickettsia conorii, Arcanobacterium haemolyticum, Bacillus anthracis, Bacillus cereus, Lysteria 5 monocytogenes, Yersinia pestis, Yersinia enterocolitica, Shigella dysenteriae, Neisseria meningitides, Neisseria gonorrhoeae, Streptococcus bovis, Streptococcus hemolyticus, Streptococcus mutans, Streptococcus pyogenes, Streptococcus pneumoniae, Staphylococcus aureus, Staphylococcus epidermidis, Staphylococcus pneumoniae, Staphylococcus saprophyticus, Vibrio cholerae, Vibrio parahaemolyticus, Salmonella typhi, Salmonella 10 paratyphi, Salmonella enteritidis, Treponema pallidum, Human rhinovirus, Human coronavirus, Dengue virus, Filoviruses (e.g., Marburg and Ebola viruses), Hantavirus, Rift Valley virus, Hepatitis B, C, and E, Human Immunodeficiency Virus (e.g., HIV-1, HIV-2), HHV-8, Human papillomavirus, Herpes virus (e.g., HV-I and HV-II), Human T-cell lymphotrophic viruses (e.g., HTLV-I and HTLV-II), Bovine leukemia virus, Influenza virus, 15 Guanarito virus, Lassa virus, Measles virus, Rubella virus, Mumps virus, Chickenpox (Varicella virus), Monkey pox, Epstein Bahr virus, Norwalk (and Norwalk-like) viruses, Rotavirus, Parvovirus B 19, Hantaan virus, Sin Nombre virus, Venezuelan equine encephalitis, Sabia virus, West Nile virus, Yellow Fever virus, causative agents of transmissible spongiform encephalopathies, Creutzfeldt-Jakob disease agent, variant 20 Creutzfeldt-Jakob disease agent, Candida, Cryptcooccus, Cryptosporidium, Giardia lamblia, Microsporidia, Plasmodium vivax, Pneumocystis carinii, Toxoplasma gondii, Trichophyton mentagrophytes, Enterocytozoon bieneusi, Cyclospora cayetanensis, Encephalitozoon hellem, Encephalitozoon cuniculi, among other viruses, bacteria, archaea, protozoa, fungi, and the like). 25 The present invention is not limited to the exemplary microorganisms described herein. One skilled in the art understands that the methods and compositions of the present invention are suitable for the targeting of any microorganism or group or class of microorganism. 30 B. Biofilms In some embodiments, the methods and compositions of the present invention target bacteria present as a biofilm. Listeria monocytogenes can fonn biofilms on a variety of 81 WO 2007/047189 PCT/US2006/039361 materials used in food processing equipment and other food and non-food contact surfaces (Blackman, J Food Prot 1996; 59:827-31; Frank, J Food Prot 1990; 53:550-4; Krysinski, J Food Prot 1992; 55:246-51; Ronner, J Food Prot 1993; 56:750-8). Biofilms can be broadly defined as microbial cells attached to a surface, and which are embedded in a matrix of 5 extracellular polymeric substances produced by the microorganisms. Biofilms are known to occur in many environments and frequently lead to a wide diversity of undesirable effects. For example, biofilms cause fouling of industrial equipment such as heat exchangers, pipelines, and ship hulls, resulting in reduced heat transfer, energy loss, increased fluid frictional resistance, and accelerated corrosion. Biofilm accumulation on teeth and gums, 10 urinary and intestinal tracts, and implanted medical devices such as catheters and prostheses frequently lead to infections (Characklis WG. Biofilm processes. In: Characklis WG and Marshall KC eds. New York: John Wiley & Sons, 1990:195-231; Costerton et al., Annu Rev Microbiol 1995; 49:711-45). Biofilm formation is a serious concern in the food processing industry because of the 15 potential for contamination of food products, leading to decreased food product quality and safety (Kumar CG and Anand SK, Int J Food Microbiol 1998; 42:9-27; Wong, J Dairy Sci 1998; 81:2765-70; Zottola and Sasahara, Int J Food Microbiol 1994; 23:125-48). The surfaces of equipment used for food handling or processing are recognized as major sources of microbial contamination. (Dunsmore et al., J Food Prot 1981; 44:220-40; Flint et al., 20 Biofouling 1997; 11:81-97; Grau, In: Smulders FJM ed. Amsterdam: Elsevier, 1987:221 234; Thomas et al., In: Smulders FJM ed. Amsterdam: Elsevier, 1987:163-180). Biofilm bacteria are generally hardier than their planktonic (free-living) counterparts, and exhibit increased resistance to antimicrobial agents such as antibiotics and disinfectants. It has been shown that even with routine cleaning and sanitizing procedures consistent with good 25 manufacturing practices, bacteria can remain on equipment, food and non-food contact surfaces and can develop into biofilms. In addition, L. monocytogenes attached to surfaces such as stainless steel and rubber, materials commonly used in food processing environments, can survive for prolonged periods (Helke and Wong, J Food Prot 1994; 57:963-8). This would partially explain their ability to persist in the processing plant. 30 Common sources of L. monocytogenes in processing facilities include equipment, conveyors, product contact surfaces, hand tools, cleaning utensils, floors, drains, walls, and 82 WO 2007/047189 PCT/US2006/039361 condensate (Tomkin et al., Dairy, Food Environ Sanit 1999; 19:551-62; Welbourn and Williams, Dairy, Food Environ Sanit 1999; 19:399-401). C. Plant Pathogens 5 In still further embodiments, the present invention provides methods and compositions targeted towards plant pathogens. Plant fungi have caused major epidemics with huge societal impacts. The Irish potato famine, with its consequent economic disaster and human population displacement, was the result of the sudden introduction of the fungus Phytophthora infestans. 10 South American cocoa crops are under threat of two major fungal diseases: witches broom caused by Crinipellis perniciosa and frosty pod (Moniliophthora roreri), which together threaten the viability of the chocolate production industry in the western hemisphere. Phytophthora blight, caused by the oomycete Phytophthora capsici, has become one 15 of the most serious threats to production of cucurbits (cucumbers, squash, pumpkins) and peppers, both in the United States and worldwide (Erwin, D. C., and Ribeiro, 0. K. 1996. Phytophthora Diseases Worldwide. American Phytopathological Society, St. Paul, MN). Banana crops worldwide are affected by Black Sigatoka is caused by the ascomycete, Mycosphaerellafijiensis. Fusarium scab affects small grain crops (wheat and 20 barley). Ganoderma spp fungi have produced deaths of ornamental palms, as do several species of Phytophthora (Elliott and Broschat, T. K. 2001. Palms 45:62-72; Nagata and Aragaki, M. 1989. Plant Dis. 73:661-663). Plants are also affected by bacteria and viruses. Burkholderia cepacia is a bacterium which produces economic losses to onion crops (Burkholder 1950. Phytopathology 40:115 25 118). Numerous plant viruses cause significant crop losses worldwide. Exemplary of such plant viruses are soybean mosaic virus, bean pod mottle virus, tobacco ring spot virus, barley yellow dwarf virus, wheat spindle streak virus, soil born mosaic virus, wheat streak virus in maize, maize dwarf mosaic virus, maize chlorotic dwarf virus, cucumber mosaic virus, tobacco mosaic virus, alfalfa mosaic virus, potato virus X, potato virus Y, potato leaf 30 roll virus and tomato golden mosaic virus. 83 WO 2007/047189 PCT/US2006/039361 D. Sanitation In yet other embodiments, the methods and compositions of the present invention find use in the sanitation of household and other areas. Listeria spp are common contaminants of the domestic environment. As many as 47% of households sampled were 5 contaminated, with dishcloths, and drain areas being common sites of contamination. (Beumer et al., Epidemiol Infect. 1996 Dec; 117(3):437-42). Pseudomonas aeruginosa is frequently isolated from showers and baths and hot tubs (Zichichi et al., Int J Dermatol. 2000 Apr;39(4):270-3; Silverman and Nieland, J Am Acad Dermatol. 1983 Feb;8(2):153-6). 10 Where a family member has an infectious disease, transmission may occur through contamination of domestic objects (Barker et al., J Appl Microbiol. 2000 Jul;89(l):137-44). Domestic food preparation and storage areas are also a source of bacterial food contaminants. The public is showing increasing awareness of the need to control domestic microbial contamination (Mattick et al., it J Food Microbiol. 2003 Aug 25;85(3):213-26; 15 Kusumaningrum et al., Int J Food Microbiol. 2003 Aug 25;85(3):227-36). E. Building structures In other embodiments, the present invention provides methods of targeting building structures. There has been increasing public attention to the potential health risks of mold 20 exposure, particularly in wet buildings. A variety of molds have been isolated from both damaged homes and businesses, including agents that secrete toxigenic materials. Stachybotrys chartarum is a fungus that has become notorious as a mycotoxin producer that can cause animal and human mycotoxicosis. Indeed, over the past 15 years in North America, evidence has accumulated implicating this fungus as a serious problem in homes 25 and buildings and one of the causes of the "sick building syndrome." (Mahmoudi et al., J Asthma. 2000 Apr;37(2):191-8). Legionella spp.bacteria replicate in manmade water containing structures, especially when these are heated, such as industrial cooling towers, heating and air conditioning systems. Legionnaires disease pneumonia is contracted by susceptible individuals that 30 breathe water droplets from such sources. Preventive and remedial treatment of water containing structures is needed to eliminate the source of infection to building inhabitants (Shelton et al., AIHAJ. 2000 Sep-Oct;61(5):738-42). 84 WO 2007/047189 PCT/US2006/039361 F. Military and Bioterrorism Applications The methods and compositions of the present invention further find use in military and bioterrorism applications. For example, in some embodiments, the methods and 5 compositions of the present invention are used in the decontamination of surfaces exposed to unknown bacteria and potentially other microorganisms (e.g., military equipment and personal protective gear). In yet other embodiments, microorganisms engineered for use in combatting bioterror agents (e.g., B. Anthracis, smallpox, etc.) are targeted by the methods and 10 compositions of the present invention. The methods and compositions of the present invention further find use in combating unknown and drug resistant organisms. The prevalence of bacteria that resist standard antibiotic therapy is increasing rapidly. Furthermore, the ability to engineer organisms with multiple drug resistance to standard antibiotics creates a significant threat in 15 bioweapons development. Because the broad spectrum antimicrobials of the present invention are suitable for use against broad classes of pathogens, they can respond to unknown bacteria and their bactericidal effect is independent of antibiotic resistance mechanisms. 20 G. Medical Applications The methods and compositions of the present invention additionally find use in the' treatment of subjects (e.g., humans) infected with a microorganism (e.g., food borne pathogens). The methods and compositions of the present invention are particularly well suited for use against antibiotic resistant organisms are targeted by the methods and 25 compositions of the present invention. VI. Cryptosporidium Biocides The present invention is not limited to the use of fusion protein biocides. As described above, Cryptosporidium parvum is a common cause of gastrointestinal infection 30 for which there is presently no parasite-specific curative therapy. In some embodiments, the present invention provides isolated biocides for use against Cryptosporidium parvum (e.g., 85 WO 2007/047189 PCT/US2006/039361 sporozoites). Experiments conducted during the course of development of the present invention demonstrated that several biocides had Cryptosporidiumparvum sporozoite killing activity, as well as the ability to reduce Cryptosporidium parvum infectivity. The biocides evaluated have the further advantage of showing minimal toxicity against human 5 cells. The biocides of the present invention thus find use in the treatment of objects (e.g., swimming pools) and food products (e.g., drinking water) contaminated with Cryptosporidiun. The present invention is not limited to a particular biocide. Any biocide that is effective against Cryptosporidium parvum (e.g., killing of spores or reduction in infectivity) 10 may be utilized. Preferred biocides are those that are non-toxic to animal (e.g., human) cells. Exemplary biocides include, but are not limited to, lactoferrin hydrolysate, lactoferrin b, cathelicidin, indolicidin, beta-defensin-2, deta-defensin-1, phopholipase A2, and phospho-inositol specific phospholipase C. 15 DETAILED DESCRIPTION OF THE INVENTION The present invention relates to constructs that encode novel microorganism targeting molecules (e.g., innate immune receptors ligands or monoclonal antibodies), novel fusion proteins, and chimeric monoclonal antibodies and to methods of using and producing the same. In particular, the present invention relates to methods of producing novel 20 monoclonal antibody biocide (e.g., bactericidal enzymes) fusion proteins in transgenic animals (e.g., bovines) and in cell cultures. The present invention also relates to therapeutic and prophylactic methods of using monoclonal antibody biocide fusion proteins in health care (e.g., human and veterinary), agriculture (e.g., animal and plant production), and food processing (e.g., beef carcass processing). The present invention also relates to methods of 25 using monoclonal antibody biocide fusion proteins in various diagnostic applications in number of diverse fields such as agriculture, medicine, and national defense. Certain embodiments of the present invention relate to the production of novel monoclonal antibodies and chimeric monoclonal antibody fusion proteins in host cells containing multiple integrated copies of an integrating vector. Preferred embodiments of 30 the present invention utilize integrating vectors (i.e., vectors that integrate via an integrase or transposase or have the capability to code for these enzymes) to create cell lines containing a high copy number of a nucleic acid encoding a gene of interest. The 86 WO 2007/047189 PCT/US2006/039361 transfected genomes of the high copy number cells are stable through repeated passages (e.g., at least 10 passages, preferably at least 50 passages, and most preferably at least 100 passages). Furthermore, in preferred embodiments, the host cells of the present invention are capable of producing high levels of protein (e.g., more than 1 pg/cell/day, preferably 5 more than 10 pg/cell/day, more preferably more than 50 pg/cell/day, and most preferably more than 100 pg/cell/day). Additional embodiments provide methods for the production of transgenic non human animals that express novel proteins in their tissues (e.g., mammary glands). In preferred embodiments, the transgenic animals are non-human ruminant (Ruminantia) 10 mammals. In other preferred embodiments, the transgenic animals are ungulates. In particularly preferred embodiments, the mammals are female ruminants (e.g., bovines) that preferentially express the novel proteins in their mammary glands. In some additional embodiments, the novel protein compositions produced in the transgenic animals are collected, purified, and subsequently incorporated into a variety of additional compositions 15 (e.g., food additives, pharmaceuticals, disinfecting agents, etc.) and/or used in a variety of therapeutic or prophylactic methods. In some embodiments, the proteins of interest (the novel fusion proteins disclosed herein) are mixed with colostrum (or colostrum substitute(s)) and subsequently feed to nursing feedlot animals (e.g., beef calves, piglets, lambs, kids, and the like). In other embodiments, the proteins of interest are formulated 20 with one or more carriers (e.g., whey) and used in the meat processing industry either a topical disinfecting agent applied to animal carcasses or as an edible supplement mixed into finished meat products. In still further embodiments, the proteins of interest (e.g., novel monoclonal antibodies and chimeric monoclonal antibody fusion proteins disclosed herein) are purified from the lactation of transgenic non-human animals and subsequently processed 25 and formulated for administration to subjects (e.g., humans and non-human animals) as therapeutic or prophylactic medicaments. Further embodiments of the present invention provide methods for producing transgenic non-human animals by the introduction of exogenous DNA into pre-maturation oocytes and mature, unfertilized oocytes (i.e., pre-fertilization oocytes) using retroviral 30 vectors that transduce dividing cells (e.g., vectors derived from murine leukemia virus [MLV], Moloney murine leukemia virus [MMLV], and the like). In addition, the present invention provides methods and compositions for cytomegalovirus promoter-driven, as well 87 WO 2007/047189 PCT/US2006/039361 as, mouse mammary tumor LTR expression of various recombinant proteins. The present invention is not limited however to the aforementioned constructs, promoters, and other genetic elements. Indeed, the present invention provides numerous examples of contemplated genetic constructs (e.g., retroviral vectors) and methods of producing stably 5 transfected cell lines (e.g., mammalian, amphibian, insect, and plant) and transgenic non human animals (e.g., bovines). In some preferred embodiments, retrovector transgenic technologies (described in greater detail herein) are used to overcome problems inherent in earlier methods for creating transgenic mammals. In preferred embodiments, unlike earlier transgenic methods, genes of 10 interest (e.g., genes encoding at least a portion of a recombinant antibody) are introduced into unfertilized oocytes (e.g., bovine oocytes). After entry of the retroviral RNA into the cell, and reverse transcription into DNA, the integration of the DNA provirus into the host cell genome is mediated by the retroviral integrase and specific nucleotide sequences at the ends of the retroviral genome. By introducing genes to the oocyte (e.g., a bovine oocyte) at 15 metaphase II arrest, the vector has access to the oocyte DNA when there is no nuclear membrane in place. The present invention contemplates this technology negates the need for dividing cells for retroviral integration to occur. Depending on the conditions, such integrations can occur at one or several independent sites in the genome and are transmitted in standard Mendelian patterns upon subsequent animal (e.g., bovine) breeding. The 20 integrated gene is transcribed like other indigenous cell genes, and the proteins it encodes are expressed at high levels. In still other preferred embodiments, the retrovector backbone used lacks the genes essential for viral structure and enzyme functions, therefore the retroviral constructs are replication defective. In yet other embodiments, the present invention uses constructs that preclude the need for a selectable marker. Importantly, 25 preferred embodiments contemplate that the removal of selectable markers (e.g., antibiotic dependent selection markers) provides a significant advantage, especially upon consideration of regulatory requirements for transgenic livestock. 88 WO 2007/047189 PCT/US2006/039361 In some of preferred embodiments directed to producing transgenic animals, the contemplated approach has major advantages. For example, the efficiency of transgenic live births using the contemplated transgenic methods is high e.g., from about 25% -75% of animals (cattle) born when genes without a selection marker are used. Additionally, in 5 preferred embodiments, retroviral genes insert as single copies, thus decreasing the risk of genetic instability upon subsequent cell replication, which tends to splice out tandem repeats of genes typical in pronuclear injection and nuclear transfer technologies. The present invention further contemplates in some preferred embodiments, where transgenes are inserted prior to fertilization the risk of producing mosaic animals, which are only 10 transgenic in some tissues but not all, is greatly reduced. Exemplary compositions and methods of the present invention are described in more detail in the following sections: I. Production of recombinant antibodies; II. Production of recombinant chimeric antibodies; III. Production of pathogen specific monoclonal antibodies in a multigenic expression system; IV. Comparison of murine and 15 chimeric murine-bovine antibodies; and V. Transgenic animal technologies; VI. Considerations for combating Cryptosporidium and other parasites; VII. Transgenic plant technologies; and VIII. Pharmaceutical compositions. I. Production of Recombinant Antibodies 20 The present invention contemplates obtaining hybridoma cell lines that produce monoclonal antibodies against particular pathogens of interest (e.g., E. coli strain 0157:H7) from one or more sources (e.g., ATCC). The cell lines are subsequently used to isolate the heavy and light chain genes that encode for pathogen (e.g., E. coli 0157:H7 and Listeria monocytogenes) specific monoclonal antibodies according to standard molecular biology 25 methods. For example, in one embodiment, hybridoma cell line ATCC HB 10452, which makes monoclonal antibody 4E8C12 specific for E. coli 0157:H7 and 026:Hl 1, are grown according to the depositors instructions. The cells are maintained in cRPMI at 37 'C and 5% CO 2 atmosphere and split biweekly at a 1:10 ratio. In another example, monoclonal 30 antibody hybridomas to the pathogen L. monocytogenes from the cell line ATCC 4689-4708 are likewise grown according to the depositors instructions. In some embodiments, candidate monoclonal antibodies are chosen based upon their binding affinity to the 89 WO 2007/047189 PCT/US2006/039361 pathogen of interest (e.g., L. monocytogenes) as well as their binding specificity that in certain instances includes as many different pathogen serotypes as possible. In some other embodiments, candidate monoclonals preferably show no or only weak cross-reaction with other species of bacteria and mammalian cells. 5 These cultures are expanded and grown in roller bottles under the described conditions to allow production of approximately milligram amounts of purified monoclonal antibodies. In preferred embodiments, the antibodies are purified using any suitable protocol such as ammonium sulfate precipitation. In some embodiments, the purified monoclonal antibodies are used to perform various in vitro functionality tests. For example, 10 the present invention contemplates using purified monoclonal antibodies to perfonn affinity and specificity tests in order to select for the antibodies that have the best binding properties to the surface of the pathogen of interest (e.g., L. monocytogenes) and/or that include binding to a broad range of serotypes. Contemplated functionality tests include, but are not limited to, enzyme-linked immunosorbent assays (ELISA) and competitive ELISA assays. 15 In one embodiment, varying concentrations of different monoclonal antibodies are allowed to bind to immobilized heat killed pathogens (e.g., L. monocytogenes). In another embodiment using a competitive assay, various concentrations of competing antigen are added to the wells of test plate and the binding of the monoclonal antibodies is measured. In yet another embodiment, quantitative immunofluorescence assays are used to allow the 20 determination of binding affinity based on fluorescence intensity per cell. The present invention contemplates that by determining the affinity of the monoclonal antibodies based on their binding capacity to the pathogen of interest (e.g., L. monocytogenes), the present invention allows the selection of the one monoclonal antibody that is best for topical applications against viable pathogens. 25 Cells from the highest affinity hybridoma clone will be used to extract total RNA with the purpose of isolating the monoclonal antibody-specific heavy and light chain gene transcripts. Upon total RNA extraction, the RNA is reverse transcribed using standard molecular biology kits and protocols, such as the RIBOCLONE cDNA synthesis system from Promega (Promega Corp., Madison, WI). Preferably, the procedures used create 30 double stranded cDNA of all RNA transcripts in a cell, including the transcripts from the murine heavy and light chain genes. The total cDNA is used as a template to specifically amplify the mouse IgG2a heavy chain and the Igk light chain. Site-directed mutagenesis 90 WO 2007/047189 PCT/US2006/039361 primers are used to amplify these sequences. The present invention contemplates that the use of these primers adds short sequences of DNA, and introduces suitable restriction sites thus allowing direct cloning of the product into the retrovector backbone. In preferred embodiments, once the genes for the murine heavy and light chain have 5 been isolated, they are separated by an IRES element and inserted into the retrovector expression system under the control of the simian cytomegalovirus and the bovine alpha lactalbumin promoter. In particularly preferred embodiments, the genes for the murine heavy and light antibody chains are cloned into a retrovector expression system (e.g., under the control of the simian cytomegalovirus (sCMV)) or other suitable multigenic gene 10 expression systems. This process allows for the production of cell lines that secrete high levels of the monoclonal antibodies. In particular, the heavy chain followed by an internal ribosome entry site (IRES) element are cloned into the retrovector backbone at the same site. Similarly, the light chain is then cloned into the retrovector backbone. Once the retroviral construct is complete, 15 quality control sequencing will confirm that all the elements are present. The present invention contemplates that the use of the IRES element in between heavy and light chain genes yields fully functional antibodies expressed and secreted into the medium at exceptionally high levels (e.g., >1 00pg/cell/day in CHO cells). In some preferred embodiments, after the retroviral constructs are complete, quality control sequencing is used 20 to confirm that all the elements are present. The retrovector construct are then used to transform host cells along with the plasmid that encodes the vesicular stomatitis virus glycoprotein (VSV-G) used for pseudotyping the retrovirus. This procedure creates intermediate level viral titer that is used to infect production cell lines (e.g., 293H or CHO cells). The population of transduced cells are subjected to clonal selection based on the 25 antibody levels present in the medium supernatant. The clone with the highest level of antibodies secreted into the supernatant is selected to produce milligram amounts of murine monoclonal antibody 4E8C12. In preferred embodiments, the recombinant antibodies are purified from cell supernatants using standard techniques well known to those in the art. Figure 1 shows one contemplated retroviral construct for expression of murine and 30 chimeric bovine murine antibodies with lysozyme. In some cell culture expression embodiments, the alpha-lactalbumin promoter is replaced with simian cytomegalovirus promoter. 91 WO 2007/047189 PCT/US2006/039361 II. Production of Recombinant Chimeric Antibodies In some embodiments, the bovine IgG1 and IgG2 heavy chain genes are used to modify the constructs made above to produce constructs encoding chimeric bovine-murine 5 antibodies. For example, in one contemplated embodiment, the constant portion of the murine heavy chain gene is replaced with the constant portion of the bovine heavy chain gene to create a chimeric bovine-murine monoclonal antibodies. A suitable bovine heavy chain IgGl sequence may be selected from, but is not limited to, the following GenBank Accession Numbers: BD105809; S82409; U32264; U32263; U32262; U32261; U32260; 10 U32259; U32258; U32257; U32256; U32255; U32254; U32253; U32252; U32251; U32250; U32249; U34749; U34748; U32852; U32851; U32850; U36824; U36823; S82407; X62917; X62916; and X16701. Likewise, a suitable bovine heavy chain IgG2 sequence may be selected from, but is not limited to, the following GenBank Accession Numbers: S82409; S82407; Z37506; and X16702. In preferred embodiments, GenBank 15 Accession No. S282409 (SEQ ID NO:l) provides bovine IgG1/IgG2 sequences. (See, I. Kacskovics and J.E. Butler, Mol. Immunol., 33(2):189-195 [1996]). Preferably, the murine IgG2a heavy chain gene will be replaced by the bovine sequence for IgG1 or IgG2a. Thus, modified with bovine IgG1/IgG2 sequences, the vectors described above are used in subsequent cloning steps. 20 In preferred embodiments, following sequence analysis of the construct, the constructs are used to create vectors for the transduction of production cell lines (e.g., 293H) and packaging cell lines (e.g., 293gp). Standard clonal analysis techniques are used to select for clones that produce high levels of the bovine-murine chimeric antibody. Once a top clone has been selected, enough chimeric antibody will be produced from this clone to 25 conduct functionality tests with the derived chimeric monoclonal antibody. In preferred embodiments, production cell lines that secrete high levels of the monoclonal antibodies are made from the above-mentioned constructs. The retroviral construct containing the chimeric murine-bovine monoclonal antibody genes are used to transduce at least one production cell line (e.g., the 293H production cell line). Upon 30 transduction and expansion, the cell pool is subjected to limited dilution cloning to select for clones that produce high levels of the chimeric monoclonal antibody as determined by standard assay techniques (e.g., ELISA assays). One of the top clones is used to produce 92 WO 2007/047189 PCT/US2006/039361 chimeric murine-bovine monoclonal antibodies in milligram amounts that are subsequently used in the functionality tests described below. The present invention further contemplates the production of retrovector packaging cell lines that produce high titers of retrovector containing the gene for the monoclonal 5 antibodies in preparation for making transgenic animals, such as bovines. For example, the retrovector construct containing the chimeric murine-bovine monoclonal antibody genes are used to transduce a packaging cell line (e.g., 293gp packaging cell line). The transduced packaging cell pool is then subjected to limiting dilution cloning and clones that produce the highest infectious viral titers are used for virus production. After a thorough quality control 10 of the top virus titer producing clone, which ensures that the construct is complete, an appropriate amount of pseudotyped virus are purified and cryopreserved for use in oocyte injections. III. Production of Pathogen Specific Monoclonal Antibodies in a Multigenic 15 Expression System In certain preferred embodiments, the production of L. monocytogenes-specific monoclonal antibody is conducted in a retrovector gene product expression system. In an initial step, the transduced production cell pool is subjected to clonal analysis to select the top antibody producing clones. Preferably, the retrovector construct will be used to 20 transform host cells along with the plasmid that encodes the vesicular stomatitis virus glycoprotein (VSV-G) used for pseudotyping the retrovirus. This procedure creates intermediate level viral titer used to infect production cell lines (e.g., 293H and CHO cells among others). The population of transduced cells is then subjected to a clonal selection, based on antibody levels present in the medium supernatant. 25 In additional embodiments, the selected clones are then expanded and used to produce sufficient quantities of monoclonal L. monocytogenes-specific antibodies to perform one or more functionality studies similar to those mentioned above. The clone with the highest level of antibody secreted into the supernatant is then chosen to produce milligram amounts of recombinant murine monoclonal antibody against 30 L. monocytogenes. Additional experiments with the purified monoclonal antibodies, similar to those mentioned above are contemplated. The objective of these experiments is to detennine whether the production of the selected high-affinity monoclonal antibody affects 93 WO 2007/047189 PCT/US2006/039361 the binding capacity when compared to the original hybridoma-derived antibody. Since the present invention contemplates using a mammalian expression system, no changes in affinity of the retrovector produced monoclonal antibody are expected. 5 IV. Comparison of Murine and Chimeric Murine-bovine Antibodies In preferred embodiments, the present invention contemplates additional functionality testing of the purified marine monoclonal antibody as compared to the hybridoma-derived product. For example, in one embodiment, a number of tests are conducted to demonstrate that the 4E8C12 monoclonal is highly specific for E. coli strain 10 0157:H7 and strain 026:H11 and no other related strains or species. In some of these embodiments, the assays contemplated for determining the specific bactericidal activity are divided into two phases. First, the bactericidal activity of the monoclonal antibody and fusion proteins are tested in vitro for inactivation of the pathogenic strain (e.g., E. coli 0157:H7). Second, the monoclonal antibody and fusion proteins are evaluated by adding to 15 formulations in turkey slurries. In particular, to assess in vitro inactivation, E. coli 0157:H7 (five food and outbreak isolates) are grown in trypticase soy broth (TSB) until late log phase (-24 h). The cells are harvested by centrifugation, washed in 67 mM sodium phosphate buffer, pH 6.6 (PB), and strains mixed in approximately equal concentrations. The E. coli mixture is then added to a 20 level of 105 per ml to PB. The monoclonal conjugates are added starting at concentrations that correspond to the bactericidal concentration of lysozyme and phospholipas A2 alone and down at least 3 logs. The suspensions are incubated at 40 and 100 C and cell viability determined at 0, 1, 4, 8 and 24 h by direct plating on TSB and MacConkey sorbitol agars. The cell suspensions are examined microscopically for clumping. If clumping is observed, 25 further experimental techniques are used to separate the cells (e.g., addition of surfactants, such as Tween 80, changing pH, and mild sonication). Controls without added monoclonal conjugates are also contemplated for testing. All conditions are tested in triplicate and standard deviations of viability are determined. In some embodiments, cooked, uncured, and unsmoked turkey breast is obtained 30 from a manufacturer. Slurries of this meat product are prepared as described by Schlyter et al., Int. J. Food Microbiol., 19(4):271-281 (1993) adjusted to the appropriate brine content, and pasteurized to 68'C. Two levels of filter-sterilized monoclonal conjugates, depending 94 WO 2007/047189 PCT/US2006/039361 on in vitro results, are added to the slurries after pasteurization Flasks are cooled to 4 0 C and subsequently inoculated with E. coli 0157:H7 (five strain mixture of food and outbreak isolates) to yield about a 105 cfu/ml slurry, and dispensed 3 ml per sterile polystyrene tube for incubation at 4 and 10 C for up to 4 weeks. In preferred embodiments, triplicate 5 samples per variable are assayed weekly for changes in E. coli 0157:H7 populations using direct plating on MacConkey sorbitol agar techniques. In addition to the bactericidal tests, the present invention further contemplates additional experiments to determine whether the chimeric bovine-murine antibodies contemplated are more effective than their murine counterparts in mediating pathogen 10 ingestion by phagocytes. While there is a substantial amount of data available on the efficacy of humanizing therapeutic murine antibodies in order to improve beneficial reactions between immune cells and target cells (for example ADCC, phagocytosis, antigen presentation) in humans, however, the efficacy of a chimeric bovine-murine antibodies in mediating ingestion and killing of a pathogen in cattle has yet to be determined. 15 Accordingly, the present invention provides functional assays of bovine monocyte/macrophage to measure killing/ingestion of E. coli 01 57:H7 in the presence of the murine monoclonal antibody, or the chimeric antibody, or no antibody. It is expected that the chimeric bovine-murine antibody of the present invention are superior in mediating phagocytosis compared to murine only versions. 20 In yet other embodiments, the present invention contemplates the purification of sufficient quantities of retrovectors containing genes for the chimeric monoclonal antibodies to conduct further functional assays and additional tests. In still other embodiments, based upon the results obtained in the above-mentioned assays and tests, further clonal analysis of packaging cell lines that express the chimeric antibody 25 are contemplated. Briefly, a high viral titer producing clone is chosen and expanded. The expanded culture are subsequently induced to produce infective viral particles and viral preparations to enrich viral particles to a titer of approximately 1-5x108 cfu/ml. Such titers have proven effective in producing transgenic animals when used for oocyte injection in transgametic systems. 30 V. Transgenic Animal Technologies 95 WO 2007/047189 PCT/US2006/039361 The methods and compositions used in certain embodiments of the present invention for creating transgenic animals (e.g., bovines and other ungulates) for expression of the biocidal fusion proteins are described in greater detail below. 5 A. Retroviruses and Retroviral Vectors Retroviruses (family Retroviridae) are divided into three groups: the spumaviruses (e.g., human foamy virus); the lentiviruses (e.g., human immunodeficiency virus and sheep visna virus) and the oncoviruses (e.g., MLV, Rous sarcoma virus). Retroviruses are enveloped (i.e., surrounded by a host cell-derived lipid bilayer 10 membrane) single-stranded RNA viruses that infect animal cells. When a retrovirus infects a cell, its RNA genome is converted into a double-stranded linear DNA form (i.e., it is reverse transcribed). The DNA form of the virus is then integrated into the host cell genome as a provirus. The provirus serves as a template for the production of additional viral genomes and viral mRNAs. Mature viral particles containing two copies of genomic RNA 15 bud from the surface of the infected cell. The viral particle comprises the genomic RNA, reverse transcriptase and other pol gene products inside the viral capsid (which contains the viral gag gene products), which is surrounded by a lipid bilayer membrane derived from the host cell containing the viral envelope glycoproteins (also referred to as membrane associated proteins). 20 The organization of the genomes of numerous retroviruses is well known in the art and this has allowed the adaptation of the retroviral genome to produce retroviral vectors. The production of a recombinant retroviral vector carrying a gene of interest is typically achieved in two stages. First, the gene of interest is inserted into a retroviral vector which contains the sequences necessary for the efficient expression of the gene of interest 25 (including promoter and/or enhancer elements which may be provided by the viral long terminal repeats [LTRs] or by an internal promoter/enhancer and relevant splicing signals), sequences required for the efficient packaging of the viral RNA into infectious virions (e.g., the packaging signal [Psi], the tRNA primer binding site [-PBS], the 3' regulatory sequences required for reverse transcription [+PBS] and the viral LTRs). The LTRs contain sequences 30 required for the association of viral genomic RNA, reverse transcriptase and integrase functions, and sequences involved in directing the expression of the genomic RNA to be packaged in viral particles. For safety reasons, many recombinant retroviral vectors lack 96 WO 2007/047189 PCT/US2006/039361 functional copies of the genes that are essential for viral replication (these essential genes are either deleted or disabled); the resulting virus is said to be replication defective. Second, following the construction of the recombinant vector, the vector DNA is introduced into a packaging cell line. Packaging cell lines provide viral proteins required in 5 trans for the packaging of the viral genomic RNA into viral particles having the desired host range (i.e., the viral-encoded gag, pol and env proteins). The host range is controlled, in part, by the type of envelope gene product expressed on the surface of the viral particle. Packaging cell lines may express ecotrophic, amphotropic or xenotropic envelope gene products. Alternatively, the packaging cell line may lack sequences encoding a viral 10 envelope (env) protein. In this case the packaging cell line will package the viral genome into particles that lack a membrane-associated protein (e.g., an env protein). In order to produce viral particles containing a membrane associated protein that will permit entry of the virus into a cell, the packaging cell line containing the retroviral sequences is transfected with sequences encoding a membrane-associated protein (e.g., the G protein of vesicular 15 stomatitis virus [VSV]). The transfected packaging cell will then produce viral particles that contain the membrane-associated protein expressed by the transfected packaging cell line; these viral particles, which contain viral genomic RNA derived from one virus encapsidated by the envelope proteins of another virus are said to be pseudotyped virus particles. 20 Viral vectors, including recombinant retroviral vectors, provide a more efficient means of transferring genes into cells as compared to other techniques such as calcium phosphate-DNA co-precipitation or DEAE-dextran-mediated transfection, electroporation or microinjection of nucleic acids. It is believed that the efficiency of viral transfer is due in part to the fact that the transfer of nucleic acid is a receptor-mediated process (i.e., the virus 25 binds to a specific receptor protein on the surface of the cell to be infected). In addition, the virally transferred nucleic acid once inside a cell integrates in controlled manner in contrast to the integration of nucleic acids which are not virally transferred; nucleic acids transferred by other means such as calcium phosphate-DNA co-precipitation are subject to rearrangement and degradation. 30 The most commonly used recombinant retroviral vectors are derived from the amphotropic Moloney murine leukemia virus (MoMLV) (Miller and Baltimore, Mol. Cell. Biol., 6:2895 [1986]). The MoMLV system has several advantages: 1) this specific 97 WO 2007/047189 PCT/US2006/039361 retrovirus can infect many different cell types, 2) established packaging cell lines are available for the production of recombinant MoMLV viral particles and 3) the transferred genes are permanently integrated into the target cell chromosome. The established MoMLV vector systems comprise a DNA vector containing a small portion of the retroviral sequence 5 (the viral long terminal repeat or "LTR" and the packaging or "psi" signal) and a packaging cell line. The gene to be transferred is inserted into the DNA vector. The viral sequences present on the DNA vector provide the signals necessary for the insertion or packaging of the vector RNA into the viral particle and for the expression of the inserted gene. The packaging cell line provides the viral proteins required for particle assembly (Markowitz et 10 al., J. Virol., 62:1120 [1988]). Despite these advantages, existing retroviral vectors based upon MoMLV are limited by several intrinsic problems: 1) they do not infect non-dividing cells (Miller et al., Mol. Cell. Biol., 10:4239 [1992]), 2) they produce low titers of the recombinant virus (Miller and Rosman, BioTechn., 7: 980 [1989]; and Miller, Nature 357: 455 [19921) and 3) they infect 15 certain cell types (e.g., human lymphocytes) with low efficiency (Adams et al., Proc. Natl. Acad. Sci. USA 89:8981 [1992]). The low titers associated with MoMLV-based vectors has been attributed, at least in part, to the instability of the virus-encoded envelope protein. Concentration of retrovirus stocks by physical means (e.g., ultracentrifugation and ultrafiltration) leads to a severe loss of infectious virus. 20 The low titer and inefficient infection of certain cell types by MoMLV-based vectors has been overcome by the use of pseudotyped retroviral vectors which contain the G protein of VSV as the membrane associated protein. Unlike retroviral envelope proteins which bind to a specific cell surface protein receptor to gain entry into a cell, the VSV G protein interacts with a phospholipid component of the plasma membrane (Mastromarino et al., J. 25 Gen. Virol., 68:2359 [1977]). Because entry of VSV into a cell is not dependent upon the presence of specific protein receptors, VSV has an extremely broad host range. Pseudotyped retroviral vectors bearing the VSV G protein have an altered host range characteristic of VSV (i.e., they can infect almost all species of vertebrate, invertebrate and insect cells). Importantly, VSV G-pseudotyped retroviral vectors can be concentrated 2000 30 fold or more by ultracentrifugation without significant loss of infectivity (Bums et al., Proc. Natl. Acad. Sci. USA, 90:8033 [1993]). 98 WO 2007/047189 PCT/US2006/039361 The VSV G protein has also been used to pseudotype retroviral vectors based upon the human immunodeficiency virus (HIV) (Naldini et al., Science 272:263 [1996]). Thus, the VSV G protein may be used to generate a variety of pseudotyped retroviral vectors and is not limited to vectors based on MoMLV. 5 The present invention is not limited to the use of the VSV G protein when a viral G protein is employed as the heterologous membrane-associated protein within a viral particle. The G proteins of viruses in the Vesiculovirus genera other than VSV, such as the Piry and Chandipura viruses, that are highly homologous to the VSV G protein and, like the VSV G protein, contain covalently linked palmitic acid (Brun et al., Intervirol., 38:274 [1995]; and 10 Masters et al., Virol., 171:285 [1990]). Thus, the G protein of the Piry and Chandipura viruses can be used in place of the VSV G protein for the pseudotyping of viral particles. In addition, the VSV G proteins of viruses within the Lyssa virus genera such as Rabies and Mokola viruses show a high degree of conservation (amino acid sequence as well as functional conservation) with the VSV G proteins. For example, the Mokola virus G 15 protein has been shown to function in a manner similar to the VSV G protein (i.e., to mediate membrane fusion) and therefore may be used in place of the VSV G protein for the pseudotyping of viral particles (Mebatsion et al., J. Virol., 69:1444 [1995]). Viral particles may be pseudotyped using either the Piry, Chandipura or Mokola G protein as described in the art with the exception that a plasmid containing sequences encoding either the Piry, 20 Chandipura or Mokola G protein under the transcriptional control of a suitable promoter element (e.g., the CMV intermediate-early promoter; numerous expression vectors containing the CMV IE promoter are available, such as the pcDNA3.1 vectors [Invitrogen]) is used in place of pHCMV-G. Sequences encoding other G proteins derived from other members of the Rhabdoviridae family may be used; sequences encoding numerous 25 rhabdoviral G proteins are available from the GenBank database. B. Integration of Retroviral DNA The majority of retroviruses can transfer or integrate a double-stranded linear fonn of the virus (the provirus) into the genome of the recipient cell only if the recipient cell is 30 cycling (i.e., dividing) at the time of infection. Retroviruses that have been shown to infect dividing cells exclusively, or more efficiently, include MLV, spleen necrosis virus, Rous 99 WO 2007/047189 PCT/US2006/039361 sarcoma virus and human immunodeficiency virus (HIV; while HIV infects dividing cells more efficiently, HIV can infect non-dividing cells). It has been shown that the integration of MLV virus DNA depends upon the host cell's progression through mitosis and it has been postulated that the dependence upon 5 mitosis reflects a requirement for the breakdown of the nuclear envelope in order for the viral integration complex to gain entry into the nucleus (Roe et al., EMBO J., 12:2099 [1993]). However, as integration does not occur in cells arrested in metaphase, the breakdown of the nuclear envelope alone may not be sufficient to permit viral integration; there may be additional requirements such as the state of condensation of the genomic DNA 10 (Roe et al., supra). C. Introduction of Retroviral Vectors into Gametes Before the Last Meiotic Division The nuclear envelope of a cell breaks down during meiosis as well as during mitosis. 15 Meiosis occurs only during the final stages of gametogenesis. The methods of the present invention exploit the breakdown of the nuclear envelope during meiosis to permit the integration of recombinant retroviral DNA and pennit for the first time the use of unfertilized oocytes (i.e., pre-fertilization and pre-maturation oocytes) as the recipient cell for retroviral gene transfer for the production of transgenic animals. Because infection of 20 unfertilized oocytes permits the integration of the recombinant provirus prior to the division of the one cell embryo, all cells in the embryo will contain the proviral sequences. Oocytes which have not undergone the final stages of gametogenesis are infected with the retroviral vector. The injected oocytes are then permitted to complete maturation with the accompanying meiotic divisions. The breakdown of the nuclear envelope during 25 meiosis permits the integration of the proviral form of the retrovirus vector into the genome of the oocyte. When pre-maturation oocytes are used, the injected oocytes are then cultured in vitro under conditions that permit maturation of the oocyte prior to fertilization in vitro. Conditions for the maturation of oocytes from a number of mammalian species (e.g., bovine, ovine, porcine, murine, caprine) are well known to the art. In general, the base 30 medium used herein for the in vitro maturation of bovine oocytes, TC-M199 medium, may be used for the in vitro maturation of other mammalian oocytes. TC-M199 medium is supplemented with hormones (e.g., luteinizing honnone and estradiol) from the appropriate 100 WO 2007/047189 PCT/US2006/039361 mammalian species. The amount of time a pre-maturation oocyte must be exposed to maturation medium to pennit maturation varies between mammalian species as is known to the art. For example, an exposure of about 24 hours is sufficient to permit maturation of bovine oocytes while porcine oocytes require about 44-48 hours. 5 Oocytes may be matured in vivo and employed in place of oocytes matured in vitro in the practice of the present invention. For example, when porcine oocytes are to be employed in the methods of the present invention, matured pre-fertilization oocytes may be harvested directly from pigs that are induced to superovulate as is known to the art. Briefly, on day 15 or 16 of estrus the female pig(s) is injected with about 1000 units of pregnant 10 mare's serum (PMS; available from Sigma and Calbiochem). Approximately 48 hours later, the pig(s) is injected with about 1000 units of human chorionic gonadotropin) (hCG; Sigma) and 24-48 hours later matured oocytes are collected from oviduct. These in vivo matured pre-fertilization oocytes are then injected with the desired retroviral preparation as described herein. Methods for the superovulation and collection of in vivo matured (i.e., oocytes at the 15 metaphase 2 stage) oocytes are known for a variety of mammals (e.g., for superovulation of mice, see Hogan et al., supra at pp. 130-133 [1994]; for superovulation of pigs and in vitro fertilization of pig oocytes see Cheng, Doctoral Dissertation, Cambridge University, Cambridge, United Kingdom [1995]). Retroviral vectors capable of infecting the desired species of non-human animal, 20 which can be grown and concentrated to very high titers (e.g., $ 1 x 108 Cfu/ml) are preferentially employed. The use of high titer virus stocks allows the introduction of a defined number of viral particles into the perivitelline space of each injected oocyte. The perivitelline space of most mammalian oocytes can accommodate about 10 picoliters of injected fluid (those in the art know that the volume that can be injected into the 25 perivitelline space of a mammalian oocyte or zygote varies somewhat between species as the volume of an oocyte is smaller than that of a zygote and thus, oocytes can accommodate somewhat less than can zygotes). The vector used may contain one or more genes encoding a protein of interest; alternatively, the vector may contain sequences that produce anti-sense RNA sequences or 30 ribozymes. The infectious virus is microinjected into the perivitelline space of oocytes (including pre-maturation oocytes) or one cell stage zygotes. Microinjection into the perivitelline space is much less invasive than the microinjection of nucleic acid into the 101 WO 2007/047189 PCT/US2006/039361 pronucleus of an embryo. Pronuclear injection requires the mechanical puncture of the plasma membrane of the embryo and results in lower embryo viability. In addition, a higher level of operator skill is required to perform pronuclear injection as compared to perivitelline injection. Visualization of the pronucleus is not required when the virus is 5 injected into the perivitelline space (in contrast to injection into the pronucleus); therefore injection into the perivitelline space obviates the difficulties associated with visualization of pronuclei in species such as cattle, sheep and pigs. The virus stock may be titered and diluted prior to microinjection into the perivitelline space so that the number of proviruses integrated in the resulting transgenic 10 animal is controlled. The use of a viral stock (or dilution thereof) having a titer of 1 x 108 cfu/ml allows the delivery of a single viral particle per oocyte. The use of pre-maturation oocytes or mature fertilized oocytes as the recipient of the virus minimizes the production of animals which are mosaic for the provirus as the virus integrates into the genome of the oocyte prior to the occurrence of cell cleavage. 15 In order to deliver, on average, a single infectious particle per oocyte, the micropipets used for the injection are calibrated as follows. Small volumes (e.g., about 5-10 pl) of the undiluted high titer viral stock (e.g., a titer of about 1 x 108 cfu/ml) are delivered to the wells of a microtiter plate by pulsing the micromanipulator. The titer of virus delivered per a given number of pulses is determined by diluting the viral stock in each well 20 and determining the titer using a suitable cell line (e.g., the 208F cell line) as described in the art. The number of pulses which deliver, on average, a volume of virus stock containing one infectious viral particle (i.e., gives a MOI of 1 when titered on 208F cells) are used for injection of the viral stock into the oocytes. Prior to microinjection of the titered and diluted (if required) virus stock, the 25 cumulus cell layer is opened to provide access to the perivitelline space. The cumulus cell layer need not be completely removed from the oocyte and indeed for certain species of animals (e.g., cows, sheep, pigs, mice) a portion of the cumulus cell layer must remain in contact with the oocyte to permit proper development and fertilization post-injection. Injection of viral particles into the perivitelline space allows the vector RNA (i.e., the viral 30 genome) to enter the cell through the plasma membrane thereby allowing proper reverse transcription of the viral RNA. 102 WO 2007/047189 PCT/US2006/039361 D. Detection of the Retrovirus Following Injection into Oocytes or Embryos The presence of the retroviral genome in cells (e.g., oocytes or embryos) infected with pseudotyped retrovirus may be detected using a variety of means. The expression of the gene product(s) encoded by the retrovirus may be detected by detection of mRNA 5 corresponding to the vector-encoded gene products using techniques well known to the art (e.g., Northern blot, dot blot, in situ hybridization and RT-PCR analysis). Direct detection of the vector-encoded gene product(s) is employed when the gene product is a protein which either has an enzymatic activity (e.g., a-galactosidase) or when an antibody capable of reacting with the vector-encoded protein is available. 10 Alternatively, the presence of the integrated viral genome may be detected using Southern blot or PCR analysis. For example, the presence of the LZRNL or LSRNL genomes may be detected following infection of oocytes or embryos using PCR as follows. Genomic DNA is extracted from the infected oocytes or embryos (the DNA may be extracted from the whole embryo or alternatively various tissues of the embryo may be 15 examined) using techniques well known to the art. The LZRNL and LSRNL viruses contain the neo gene and the following primer pair can be used to amplify a 349-bp segment of the neo gene: upstream primer: 5'-GCATTGCATCAGCCATGATG-3' (SEQ ID NO:103) and downstream primer: 5'-GATGGATTGCACGCAGGTTC-3' (SEQ ID NO:104). The PCR is carried out using well known techniques (e.g., using a GeneAmp kit according to the 20 manufacturer's instructions [Perkin-Elmer]). The DNA present in the reaction is denatured by incubation at 94EC for 3 min followed by 40 cycles of 94EC for 1 min, 60EC for 40 sec and 72EC for 40 sec followed by a final extension at 72EC for 5 min. The PCR products may be analyzed by electrophoresis of 10 to 20% of the total reaction on a 2% agarose gel; the 349-bp product may be visualized by staining of the gel with ethidium bromide and 25 exposure of the stained gel to UV light. If the expected PCR product cannot be detected visually, the DNA can be transferred to a solid support (e.g., a nylon membrane) and hybridized with a 32 P-labeled neo probe. Southern blot analysis of genomic DNA extracted from infected oocytes and/or the resulting embryos, offspring and tissues derived therefrom is employed when infonnation 30 concerning the integration of the viral DNA into the host genome is desired. To examine the number of integration sites present in the host genome, the extracted genomic DNA is typically digested with a restriction enzyme, which cuts at least once within the vector 103 WO 2007/047189 PCT/US2006/039361 sequences. If the enzyme chosen cuts twice within the vector sequences, a band of known (i.e., predictable) size is generated in addition to two fragments of novel length which can be detected using appropriate probes. 5 E. Detection of Foreign Protein Expression in Transgenic Animals The present invention also provides transgenic animals that are capable of expressing foreign proteins in their milk, urine and blood. The transgene is stable, as and shown to be passed from a transgenic bull to his offspring. In addition, the transgenic animals produced according to the present invention express foreign proteins in their body 10 fluids (e.g., milk, blood, and urine). Thus, the present invention further demonstrates the utility of using the MoMLV LTR as a promoter for driving the constitutive production of foreign proteins in transgenic cattle. It is also contemplated that such a promoter could be used to control expression of proteins that would prevent disease and/or infection in the transgenic animals and their offspring, or be of use in the production of a consistent level of 15 protein expression in a number of different tissues and body fluids. For example, it is contemplated that the MoMLV LTR of the present invention will find use in driving expression of antibody to pathogenic organisms, thereby preventing infection and/or disease in transgenic animals created using the methods of the present invention. For example, it is contemplated that antibodies directed against organisms such 20 as E. coli, Sahnonella ssp., Streptococcus ssp., Staphylococcus spp., Mycobacterium spp., produced by transgenic animals will find use preventing mastitis, scours, and other diseases that are common problems in young animals. It is also contemplated that proteins expressed by transgenic animals produced according to the present invention will find use as bacteriostatic, bactericidal, fungistatic, fungicidal, viricidal, and/or anti-parasitic 25 compositions. Thus, it is contemplated that transgenic animals produced according to the present invention will be resistant to various pathogenic organisms. Furthermore, the milk produced by female transgenic animals would contain substantial antibody levels. The present invention contemplates that these antibodies are useful in the protection of other animals (e.g., through passive immunization methods). 30 V. Considerations for Combating Cryptosporidium and Other Parasites 104 WO 2007/047189 PCT/US2006/039361 A. Production of Transgenic Expression System for Monoclonal 3E2 Antibodies Against C. parvun In certain embodiments, the present invention uses an established hybridoma line (as described herein) as a source for the 3E2 genes for insertion into a replication defective 5 retrovector. While the present invention is not limited to any mechanism, it is contemplated that 3E2 has especially potent neutralizing capabilities against sporozoites because it is of the IgM isotype. It is thought that through binding to repetitive epitopes of the CSL antigen the circumsporozoite precipitate (CSP)-like reaction is induced (M.W. Riggs et al., J. Inmunol., 158:1787-1795 [1997]) that renders the sporozoite non-infective. IgM antibodies 10 exist in several forms, one, in unstimulated B-lymphocytes they are membrane-bound and, two, upon stimulation of the B-lymphocyte, IgM is secreted as a pentamer joined by the J chain. J-chain expression plays an important role in inducing the pentamerization process of IgM. In studies done by Niles et al., high expression of the J-chain resulted in a high percentage of pentameric IgM. (M.J. Niles et al., Proc. Natl. Acad. Sci. USA, 92:2884-2888 15 [1995]). A third possible configuration for IgM was shown to be a hexamer. (A. Cattaneo and M.S. Neuberger, EMBO J., 6:2753-2758; and T.D. Randall et al., Eur. J. Immunol., 20:1971-1979 [1990]). In one embodiment, the present invention specifically provides a cloning strategy that addresses the pentamer and hexamer configurations. In some embodiments, the hexamer configuration of IgM is contemplated to provide better efficacy 20 against Cryptosporidium sporozoites than IgG. In some embodiments, an IgM isotype control (of irrelevant specificity) is constructed in parallel following the cloning strategy described herein. Briefly, the retrovectors are pseudotyped with VSVg to give pantropic infectivity and used to achieve gene transfer to bovine oocytes and to CHO cells (component C). For transgenic expression 25 in mammals (e.g., bovines), as opposed to expression in cell culture, the construct is designed to remove antibiotic-based selection markers (i.e., undesirable in an animal population), and to insert a promoter that links expression closely to lactation thus restricting expression to the mammary cells. In some embodiments, an alphalactalbumin promoter is used for this purpose. To assure high probability of infection and transgene 30 integration into the oocyte genome, very high retrovector titer is needed for injection into the very small perivitelline space. It is contemplated that using pseudotyped VSVG vector envelope stabilizes the vector and increases the ability to concentrate vector sufficiently for 105 WO 2007/047189 PCT/US2006/039361 injection in picoliter amounts. Preferably, transgenic embryos are produced by injection of unfertilized oocytes, in vitro fertilization, and transfer to recipient animals (e.g., surrogate bovine mothers). After transgenic offspring have been verified as transgenic and grown to 6-8 months, a hormone regimen is used to initiate lactation. 5 A consideration in using retrovectors is the need to provide assurances that no reversion, recombination, or mutation of replication defective retrovectors to viral competence has occurred. Thus, in preferred embodiments a testing protocol is followed for testing packaging cell lines and transgenic offspring. In some embodiments, two different IRES elements are used to reduce the likelihood 10 of recombination events that can be triggered by different identical sequences in a vector. The use of the IRES element in between heavy and light chain genes has been tested extensively and proven to yield fully functional antibodies, expressed and secreted into the medium at high levels (up to 100 pg/cell/day in CHO cells in serum free medium). 15 B. Selection and Testing of Biocidess, and Preparation of Vector for Cryptosporidium Neutralizing Monoclonal Antibodies and Fusion Proteins In some additional embodiments, additional antibodies are selected from a large previously reported test panel. (See, D.A. Schaefer et al., Infect. Immun., 68:2608-2616 20 [2000]). For example, lE10 is an IgGl isotype, that targets the P23 antigen; 3H2 is an IgM, that targets the GP25-200 antigen. Because, in some embodiments, IgG may be preferred for biocide fusion proteins, the present invention also expresses the 4H9 antibody. 4H9 is an IgG that targets GP25-200, but a different epitope than 3H2 (Schaefer et al., Infect. Immun., 68:2608-2616 [2000]). In one embodiment, 4 different antibody-biocide fusion 25 types (Figure 2) from each IgG antibody are constructed. These molecules are expressed in retrovector based cell culture systems and tested for their efficacy against sporozoites in vitro and in vivo. The considerations pertaining to production of tricistronic constructs for IgM, discussed above with respect to 3E2, are also relevant to 3H2. To select appropriate biocides, the present invention contemplates expanding the 30 preliminary testing of sporozoite neutralization by potential biocides to include additinoal candidates, and comparison of human PLA2 to bee venom PLA2. 106 WO 2007/047189 PCT/US2006/039361 In some preferred embodiments, molecular modeling is used to guide the structural assembly of the fusion molecules. The relative geometry of a monoclonal antibody molecule with a molecule of biocidal activity attached to the C-terminus is similar to that of complement binding to the Fc region of the MAb HC when bound to a pathogen, which 5 results in destruction of the membrane. Thus, the present invention contemplates using the C. parvum binding site affinity of the MAb molecule to bring the biocidal activity into close apposition to its substrate by attachment of the biocide to the C-terminus of the monoclonal heavy chain. Secretory PLA2 is a relatively small molecule (-14 kDa) and is comparable in size 10 to one of the CHl or CH2 domains of an antibody molecule. As an alternative, an N terminal extension linker on the PLA2 portion of the molecule is created to move the phospholipase domains a short distance from the MAb molecule. One linker contemplated for use for constructing single chain monoclonal-cytokine fusion proteins is a -(Gly4-Ser)3 extension (~16-20 angstrom extension). (See e.g., C.R. Robinson and R.T. Sauer, Proc. 15 Natl. Acad. Sci. USA, 95:5929-5934 [1998]). This is a relatively neutral sequence that is flexible and does not have a strong structure-forming propensity. In another exemplary embodiment, the present invention inserts a proline into the middle of the extension arm to provide a "kink", with freedom to rotate in the extension chain and thus allow different geometrical relationships between the biocide and the antibody molecule. 20 C. Expression of Monoclonal Antibodies and Monoclonal Antibody-Biocide Fusions in Cell Culture and Animal Models In some preferred embodiments, the present invention provides animal based expression systems for producing large quantities of present compositions, while, in other 25 preferred embodiments, the present invention provides high yielding cell lines, prepared. In some embodiments, cell based production is more expensive and on a smaller scale than production in transgenic animals (e.g., bovines), significant quantities of antibodies and fusion products are rapidly obtainable as compared to the proposed transgenic-derived products. 30 After expression and testing in vitro the present invention, contemplates scale up production using roller bottles to make sufficient recombinant product to test in mice. Then the most promising compounds, based on their efficacy in mice, are tested in an animal 107 WO 2007/047189 PCT/US2006/039361 model where clinical disease is observed. The neonatal mouse model provides an essential, cost-effective means for the initial in vivo evaluation of product efficacy in reducing intestinal infection levels and is widely accepted for this purpose. However, C. parvum infection in neonatal mice does not cause diarrhea or other signs of disease, hence the need 5 for subsequent evaluation in a clinical model for compositions having demonstrated anti cryptosporidial activity in mice. In some embodiments, piglets are selected as the clinical model of choice because of their small size, availability in adequate numbers to permit comparative studies and statistical analysis, and development of intestinal lesions resulting in acute watery diarrhea, 10 dehydration, malabsorption, and weight loss when infected with C. parvum (C.W. Kim, Cryptosporidiosis in Pigs and Horses. In: J.P. Dubey, C.A. Speer, and R. Fayer eds. Boca Raton, FL: CRC Press, pp. 105-111 [1990]). Importantly, as monogastrics, the pathogenesis and control of cryptosporidiosis in piglets is thought to closely model that of human infections and response to treatment. (S. Tzipori, Adv. Parasitol., 40:187-221 [1998]). 15 Criteria for determining efficacy in piglets include, but are not limited to, clinical signs, weight loss, fecal volume and dry matter, and fecal oocyst quantitation and duration of shedding. Following euthanasia at 10 days post infection, extensive histopathological examination completes the data set. 20 VII. Transgenic Plant Technologies In some embodiments, the fusion proteins of the present invention are expressed in transgenic organisms such as transgenic plants having a transgene inserted into its nuclear or plastidic genome. Techniques of plant transformation are known as the art. (See e.g., Wu and Grossman, Methods in Enzymology, Vol. 153, Recombinant DNA Part D, Academic 25 Press [1987], and EP 693554 (incorporated herein by reference in its entirety). Foreign nucleic acids can be introduced into plant cells or protoplasts by several methods. For example, nucleic acid can be mechanically transferred by microinjection directly into plant cells by use of micropipettes. In some embodiments, foreign nucleic acid can also be transferred into plant cells by using polyethylene glycol to forn a precipitation complex 30 with the genetic material that is taken up by the cell. (See e.g., Paszkowski et al., J. EMBO, 3:2712-2722 [1984]). In other embodiments, foreign nucleic acid are introduced into plant cells by electroporation. (See e.g., Fromm et al., Proc. Nat. Acad. Sci. USA, 82:5824 108 WO 2007/047189 PCT/US2006/039361 [1985]). Briefly, plant protoplasts are electroporated in the presence of plasmids or nucleic acids containing the relevant genetic construct. Electrical impulses of high field strength reversibly permeabilize the plant cell's biomembranes thus allowing the introduction of the plasmids. Electroporated plant protoplasts reform the cell wall, divide, and form a plant 5 callus. Preferably, selection of the transformed plant cells with the transformed gene is accomplished using phenotypic markers. In certain other embodiments, the cauliflower mosaic virus (CaMV) is used as a vector to introduce foreign nucleic acids into plant cells. (See e.g., Hohn et al., "Molecular Biology of Plant Tumors," Academic Press, New York, pp. 549-560 [1982]; and US 10 4,407,956 (incorporated by reference herein in its entirity). CaMV viral DNA genome is inserted into a parent bacterial plasmid creating a recombinant DNA molecule that can be propagated in bacteria. The recombinant plasmid can be further modified by introduction of the desired DNA sequence. The modified viral portion of the recombinant plasmid is then excised from the parent bacterial plasmid, and used to inoculate the plant cells or plants. 15 High velocity ballistic penetration by small particles can be used to introduce foreign nucleic acid into plant cells. Nucleic acid is disposed within the matrix of small beads or particles, or on the surface. (See e.g., Klein et al., Nature, 327:70-73 [1987]). Although typically only a single introduction of a new nucleic acid segment is required, this method also provides for multiple introductions. 20 A nucleic acid can be introduced into a plant cell by infection of a plant cell, an explant, an ineristem or a seed with Agrobacterium tumefaciens transformed with the nucleic acid. Under appropriate conditions, the transformed plant cells are grown to form shoots, roots, and develop further into plants. The nucleic acids can be introduced into plant cells, for example, by means of the Ti plasmid of Agrobacterium tumefaciens. The Ti 25 plasmid is transmitted to plant cells upon infection by Agrobacterium tumefaciens, and is stably integrated into the plant genome. (See e.g., Horsch et al., Science, 233:496-498 [1984]; and Fraley et al., Proc. Nat. Acad. Sci. USA, 80:4803 [1983]). Plants from which protoplasts can be isolated and cultured to give whole regenerated plants can be transfonned so that whole plants are recovered which contain the transferred 30 foreign gene. All plants that can be produced by regeneration from protoplasts can also be transfected using the process according to the invention (e.g., cultivated plants of the genera Fragaria, Lotus, Medicago, Onobiychis, Trifolium, Trigonella, Vigna, Citrus, Geranium, 109 WO 2007/047189 PCT/US2006/039361 Manihot, Daucus, Arabidopsis, Brassica, Raphanus, Sinapis, Atropa, Capsicum, Hyoscyarnus, Lycopersicon, Nicotiana, Solarium, Petunia, Digitalis, Majorana, Ciohorium, Helianthus, Lactuca, Bromus, Asparagus, Antirrhinum, Hererocallis, Nemesia, Pelargoniwn, Panicum, Pennisetum, Ranunculus, Senecio, Salpiglossis, Cucumis, 5 Browaalia, Glycine, Lolium, Zea, Triticum, Sorghum, Datura, Solanum, Beta, Pisum, Phaseolus, Allium, Avena, Hordeum, Oryzae, Setaria, Secale, Sorghum, Triticum, Musa, Cocos, Cydonia, Pyrus, Malus, Phoenix, Elaeis, Rubus, Fragaria, Prunus, Arachis, Saccharum, Coffea, Camellia, Ananas, or Vitis). In general, protoplasts are produced in accordance with conventional methods. (See e.g., US 4,743,548; 4,677,066, 5,149,645; and 10 5,508,184 all of which are incorporated herein by reference). Plant tissue may be dispersed in an appropriate medium having an appropriate osmotic potential (e.g., 3 to 8 wt. % of a sugar polyol) and one or more polysaccharide hydrolases (e.g., pectinase, cellulase, etc.), and the cell wall degradation allowed to proceed for a sufficient time to provide protoplasts. After filtration the protoplasts may be isolated by centrifugation and may then be 15 resuspended for subsequent treatment or use. Plant regeneration from cultured protoplasts is described in Evans et al., "Protoplasts Isolation and Culture," Handbook of Plant Cell Cultures 1:124-176 (MacMillan Publishing Co. New York 1983); M.R. Davey, "Recent Developments in the Culture and Regeneration of Plant Protoplasts," Protoplasts (1983)-Lecture Proceedings, pp. 12-29, (Birkhauser, Basal 20 1983); P.J. Dale, "Protoplast Culture and Plant Regeneration of Cereals and Other Recalcitrant Crops," Protoplasts (1983)-Lecture Proceedings, pp. 31-41, (Birkhauser, Basel 1983); and H. Binding, "Regeneration of Plants," Plant Protoplasts, pp. 21-73, (CRC Press, Boca Raton 1985). Regeneration from protoplasts varies from species to species of plants, but generally 25 a suspension of transformed protoplasts containing copies of the exogenous sequence is first generated. In certain species, embryo formation can then be induced from the protoplast suspension, to the stage of ripening and germination as natural embryos. The culture media can contain various amino acids and hormones, such as auxins and cytokinins. It can also be advantageous to add glutamic acid and proline to the medium, especially for such species 30 as corn and alfalfa. Shoots and roots normally develop simultaneously. Efficient regeneration will depend on the medium, on the genotype, and on the history of the culture. If these three variables are controlled, then regeneration is fully reproducible and repeatable. 110 WO 2007/047189 PCT/US2006/039361 In vegetatively propagated crops, the mature transgenic plants can be propagated by the taking of cuttings or by tissue culture techniques to produce multiple identical plants for trailing, such as testing for production characteristics. Selection of a desirable transgenic plant is made and new varieties are obtained thereby, and propagated vegetatively for 5 commercial sale. In seed propagated crops, the mature transgenic plants can be self crossed to produce a homozygous inbred plant. The inbred plant produces seed containing the gene for the newly introduced foreign gene activity level. These seeds can be grown to produce plants that have the selected phenotype. The inbreds according to this invention can be used to develop new hybrids. In this method, a selected inbred line is crossed with another inbred 10 line to produce the hybrid. Parts obtained from a transgenic plant, such as flowers, seeds, leaves, branches, fruit, and the like are covered by the invention, provided that these parts include cells which have been so transformed. Progeny and variants, and mutants of the regenerated plants are also included within the scope of this invention, provided that these parts comprise the 15 introduced DNA sequences. Progeny and variants, and mutants of the regenerated plants are also included within the scope of this invention. Selection of transgenic plants or plant cells can be based upon a visual assay, such as observing color changes (e.g., a white flower, variable pigment production, and uniform color pattern on flowers or irregular patterns), but can also involve biochemical assays of 20 either enzyme activity or product quantitation. Transgenic plants or plant cells are grown into plants bearing the plant part of interest and the gene activities are monitored, such as by visual appearance (for flavonoid genes) or biochemical assays (Northern blots); Western blots; enzyme assays and flavonoid compound assays, including spectroscopy. (See e.g., Harborne et al., (Eds.) "The Flavonoids, Vols: 1 and 2, Acad. Press 1975). Appropriate 25 plants are selected and further evaluated. Methods for generation of genetically engineered plants are further described in US 5,283,184; 5,482,852, and EPO Application EP 693,554 (each of which is herein incorporated by reference in its entirety). VIII. Pharmaceutical Compositions 30 The present invention provides novel methods and compositions for treating diseases characterized by pathogenic infection comprising administering subjects (e.g., bovines, humans, and other mammals) a pharmaceutical and/or nutraceutical composition comprising 111 WO 2007/047189 PCT/US2006/039361 chimeric recombinant antibodies either in food based (e.g., whey protein) carriers, or common pharmaceutical carriers, including any sterile, biocompatible pharmaceutical carrier (e.g., saline, buffered saline, dextrose, water, and the like) to subjects. In some embodiments, the methods of the present invention comprise administering 5 the compositions of the present invention in suitable pharmaceutical carriers. In some embodiments, these pharmaceutical compositions contain a mixture of at least two types of antibody-biocide compositions co-administered to a subject. In still further embodiments, the pharmaceutical compositions comprise a plurality of antibody-biocide compositions administered to a subject under one or more of the following conditions: at different 10 periodicities, different durations, different concentrations, different administration routes, etc. In some preferred embodiments, the compositions and methods of the present invention find use in treating diseases or altered physiological states characterized by pathogenic infection. However, the present invention is not limited to ameliorating (e.g., 15 treating) only these types of conditions in a subject. Indeed, various embodiments of the present invention are directed to treating a range of physiological symptoms and disease etiologies in subjects generally characterized by infection with a pathogen (e.g., bacteria, archeae, viruses, mycoplasma, fungi, etc.). Depending on the condition being treated, these pharmaceutical compositions are 20 formulated and administered systemically or locally. Techniques for fonnulation and administration are found in the latest edition of "Remington's Pharmaceutical Sciences" (Mack Publishing Co, Easton Pa.). Accordingly, the present invention contemplates administering pharmaceutical compositions in accordance with acceptable pharmaceutical delivery methods and preparation techniques. For example, some compounds of the present 25 invention are administered to a subject intravenously in a phannaceutically acceptable carrier such as physiological saline. For injection, the pharmaceutical compositions of the invention are formulated in aqueous solutions, preferably in physiologically compatible buffers (e.g., Hanks' solution, Ringer's solution, or physiologically buffered saline). For tissue or cellular administration, penetrants appropriate to the particular barrier to be 30 penneated are preferably used in the formulations. Such penetrants are generally known in the art. Standard methods for intracellular delivery of pharmaceutical agents are used in 112 WO 2007/047189 PCT/US2006/039361 other embodiments (e.g., delivery via liposomes). Such methods are well known to those skilled in the art. In some embodiments, present compositions are formulated for parenteral administration, including intravenous, subcutaneous, intramuscular, and intraperitoneal. In 5 some embodiments, these compositions optionally include aqueous solutions (i.e., water-soluble forms). Additionally, suspensions of the active compounds may also be prepared as oily injection suspensions as appropriate. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain 10 substances that increase the viscosity of the suspension, such as sodium carboxymethyl cellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents that increase the solubility of the compounds to allow for the preparation of highly concentrated solutions. Therapeutic co-administration of some contemplated compositions is also be 15 accomplished using gene therapy techniques described herein and commonly known in the art. In other embodiments, the present compositions are formulated using pharmaceutically acceptable carriers and in suitable dosages for oral administration. Such carriers enable the compositions to be formulated as tablets, pills, capsules, dragees, liquids, 20 gels, syrups, slurries, suspensions and the like, for oral or nasal ingestion by a patient to be treated. Phannaceutical preparations for oral use can be obtained by combining the active compounds (e.g., chimeric antibody biocide fusion proteins) with a solid excipient, optionally grinding the resulting mixture, and processing the mixture of granules, after 25 adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients are carbohydrate or protein fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; starch from corn, wheat, rice, potato, etc.; cellulose such as methyl cellulose, hydroxypropylmethyl-cellulose, or sodium carboxymethylcellulose; and gums including arabic and tragacanth; and proteins such as gelatin and collagen. If desired disintegrating or 30 solubilizing agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, alginic acid or a salt thereof such as sodium alginate. 113 WO 2007/047189 PCT/US2006/039361 Ingestible formulations of the present compositions may further include any material approved by the United States Department of Agriculture for inclusion in foodstuffs and substances that are generally recognized as safe (GRAS), such as, food additives, flavorings, colorings, vitamins, minerals, and phytonutrients. The term "phytonutrients" as used herein, 5 refers to organic compounds isolated from plants that have a biological effect, and includes, but is not limited to, compounds of the following classes: isoflavonoids, oligomeric proanthcyanidins, indol-3-carbinol, sulforaphone, fibrous ligands, plant phytosterols, ferulic acid, anthocyanocides, triterpenes, omega 3/6 fatty acids, polyacetylene, quinones, terpenes, cathechins, gallates, and quercitin. 10 Dragee cores are provided with suitable coatings such as concentrated sugar solutions, which may also contain gum arabic, talc, polyvinylpyrrolidone, carbopol gel, polyethylene glycol, and/or titanium dioxide, lacquer solutions, and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or dragee coatings for product identification or to characterize the quantity of active compound, (i.e., 15 dosage). Compositions of the present invention that can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a coating such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients mixed with fillers or binders such as lactose or starches, lubricants such as talc or magnesium stearate, 20 and, optionally, stabilizers. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycol with or without stabilizers. In some embodiments of the present invention, therapeutic agents are administered to a patient alone, or in combination with one or more other drugs or therapies (e.g., 25 antibiotics and antiviral agents etc.) or in pharmaceutical compositions where it is mixed with excipient(s) or other pharmaceutically acceptable carriers. In one embodiment of the present invention, the phannaceutically acceptable carrier is pharmaceutically inert. Phannaceutical compositions suitable for use in the present invention include compositions wherein the active ingredients are contained in an effective amount to achieve 30 the intended purpose. For example, an effective amount of therapeutic compound(s) maybe that amount that destroys or disables pathogens as compared to control pathogens. 114 WO 2007/047189 PCT/US2006/039361 In addition to the active ingredients, preferred pharmaceutical compositions optionally comprise pharmaceutically acceptable carriers, such as, excipients and auxiliaries that facilitate processing of the active compounds into preparations that can be used pharmaceutically. 5 In some embodiments, the pharmaceutical compositions used in the methods of the present invention are manufactured according to well-known and standard pharmaceutical manufacturing techniques (e.g., by means of conventional mixing, dissolving, granulating, dragee-making, levigating, emulsifying, encapsulating, entrapping or lyophilizing processes). 10 Dosing is dependent on severity and responsiveness of the disease state to be treated, with the course of treatment lasting from several days to several months, or until a cure is effected or a diminution of the disease state is achieved. Optimal dosing schedules are calculated from measurements of composition accumulation in the subject's body. The administering physician can easily determine optimum dosages, dosing methodologies and 15 repetition rates. Optimum dosages may vary depending on the relative potency of compositions agents, and can generally be estimated based on the EC 5 os found to be effective in in vitro and in vivo animal models. Additional factors that may be taken into account, include the severity of the disease state; the age, weight, and gender of the subject; the subject's diet; the time and frequency of administration; composition combinationss; 20 possible subject reaction sensitivities; and the subject's tolerance/response to treatments. In general, dosage is from 0.001 pg to 100 g per kg of body weight, and may be given once or more daily, weekly, monthly or yearly. The treating physician can estimate repetition rates for dosing based on measured residence times and concentrations of the drug in bodily fluids or tissues. Following successful treatment, it may be desirable to have the subject 25 undergo maintenance therapy to prevent the recurrence of the disease state, wherein the therapeutic agent is administered in maintenance doses, ranging from 0.001 ptg to 100 g per kg of body weight, once or more daily, weekly, or other period. For any compound used in the methods of the invention, the therapeutically effective dose can be estimated initially from cell culture assays. Then, preferably, dosage can be 30 formulated in animal models (particularly urine or rat models) to achieve a desirable circulating concentration range that results in increased PKA activity in cells/tissues characterized by undesirable cell migration, angiogenesis, cell migration, cell adhesion, 115 WO 2007/047189 PCT/US2006/039361 and/or cell survival. A therapeutically effective dose refers to that amount of compound(s) that ameliorate symptoms of the disease state (e.g., pathogenic infection). Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD 5 0 (the dose 5 lethal to 50% of the population) and the ED 5 o (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index, and it can be expressed as the ratio LD 5 o/ EDso. Compounds that exhibit large therapeutic indices are preferred. The data obtained from cell culture assays and additional animal studies can be used in formulating a range of dosage, for example, mammalian use 10 (e.g., humans). The dosage of such compounds lies preferably, however the present invention is not limited to this range, within a range of circulating concentrations that include the ED 50 with little or no toxicity. Guidance as to particular dosages and methods of delivery is provided in the literature (See, US 4,657,760; 5,206,344; or 5,225,212, all of which are herein incorporated 15 by reference in their entireties). Administration of some agents to a patient's bone marrow may necessitate delivery in a manner different from intravenous injections. EXAMPLES The present invention provides the following non-limiting examples to further 20 describe certain contemplated embodiments of the present invention. EXAMPLE 1 Effects of PLA2 on Sporozoite Infectivity This experiment describes the effects of PLA2 on sporozoite infectivity. Briefly, 25 sporozoites were incubated in an isotonic saline solution (37 'C, 30 min) with a range of concentrations of PLA2 isolated from honey bee venom (Sigma-Aldrich Corp., St. Louis, MO, 1.5 U/tg protein). Control sporozoites were identically incubated in buffer containing concentration-matched BSA but no PLA2. Sporozoites were then washed in medium and inoculated onto replicate Caco-2 human intestinal epithelial cell monolayers. 24 h later, 30 infection was quantified in test and control monolayers by immunofluorescence assay as described herein and the mean percent reduction of infection calculated. The results indicate that PLA2 achieved a highly significant reduction of sporozoite infectivity at a 116 WO 2007/047189 PCT/US2006/039361 concentration as low as 0.014 U/ml (Figure 3, P < 0.0005). Percent viability of Caco-2 cells following exposure to PLA2-treated sporozoites (86%) was similar to that of uninoculated control cells (91%) at 24 hrs, as determined by trypan blue dye exclusion. This finding suggests little if any toxic effect of residual PLA2 on host cells. The data suggests that 5 PLA2 is a viable candidate for antibody fusion. Through fusion of PLA2 to a monoclonal antibody with a high affinity for sporozoites, such as E10 or 4H9, lethal concentrations of biocide are deliverable to the sporozoite surface with relatively low amounts of biocide being used. 10 EXAMPLE 2 Target Antigens CSL, P23, and GP25-200 are Conserved in both Type 1 and Type 2 C. parvum Isolates Western blotting of type 1 and type 2 C. parvum was performed to evaluate expression of the antigens and epitopes defined by the monoclonal antibodies proposed 15 herein. For these studies, human C. parvun isolates were obtained from Peruvian patients and genotyped by nested PCR primers designed to amplify a region within the 18S rRNA gene, followed by RFLP analysis of the amplicons to differentiate Type 1 from Type 2 according to G.D. Sturbaum et al., Appl. Environ. Microbiol., 67:2665-2668 [2001]). Two human isolates determined to be of the Type 1 genotype were evaluated by Western blot for 20 recognition by monoclonal antibodies 3E2 (anti-CSL), 1El0 (anti-P23), and 3H2 (anti GP25-200) using previously described methods. (M.W. Riggs et al., Infect. Immun., 62:1927-1939 [1994]). The Iowa Type 2 isolate (J. Heine et al., J. Infect. Dis., 150:768-775 [1984]) was examined in parallel. Each monoclonal bound to both Type 1 isolates. In addition, the molecular weights and immunoreactivities of the Type 1 antigens recognized 25 by each monoclonal were indistinguishable from those recognized in the Type 2 isolate in blots of antigen resolved by either 2-12% or 4-20% reducing SDS-PAGE. Importantly, these findings suggest conservation of the antigens and epitopes defined by 3E2, 1El0, and 3H2 between Type 1 and Type 2 C. parvum, and are consistent with the functional role ascribed to each antigen. 30 EXAMPLE 3 Various Genetic Engineering Techniques 117 WO 2007/047189 PCT/US2006/039361 The present example describes the isolation of the genes for the heavy, light, and J chains from the 3E2 murine hybridoma cell line, cloning into the retrovector backbone in two configurations (for cell culture expression and for transgenic production), and clonal analysis of the vector producing packaging lines to identify high titer lines maintaining the 5 fidelity of the protein. In one embodiment, cells from the 3E2 hybridoma are used to extract total RNA with the purpose of isolating the monoclonal antibody-specific heavy, light and J-chain transcripts. Upon total RNA extraction, the RNA is reverse transcribed to create cDNA using standard molecular biology protocols. The total cDNA is then used as a template to 10 specifically amplify the mouse IgM-heavy and light chains as well as the J chain. Site directed mutagenesis primers are used to amplify the sequences. The use of these primers adds short sequences of DNA that introduce suitable restriction sites that allow direct cloning of the product into the retrovector backbone. As mentioned herein, in some embodiments, two retroviral constructs are made 15 containing the hybridoma-derived antibody genes. Preferably, one is a bicistronic construct, aimed at producing hexameric IgM. This construct bears the genes for IgM heavy and light chain and upon expression in a cell, spontaneous hexamer formation of IgM takes place. The elements of this and other contemplated constructs are shown in Figures 4A-4D grant. The construct is Figure 4A provides a bicistronic antibody. The construct provided in 20 Figure 4B is tricistronic and contains, in addition to the IgM heavy and light chain, the J chain that for pentamer formation of IgM. (Figure 4B). To create the tricistronic vector the following cloning steps are performed. First, the heavy chain (HC) is cloned into the multiple cloning site (MCS) of the disclosed retrovector backbone. Second, the genes for genes for the encephalomyocarditis virus (EMCV) IRES 25 (internal ribosome entry site) element, the signal peptide (SP) and the 1gM light chain (LC) are combined. Preferably, the IRES is engineered to optimize the secondary initiation of protein synthesis, thus allowing consistent perfonnance in obtaining equimolar expression of heavy and light chains. The genes for the foot-and-mouth disease virus (FMDV) IRES element, the SP and the J-chain gene are combined in parallel. (See e.g., M. Harries et al., J. 30 Gene Med., 2:243-249 [2000]; and X.Y. Wen et al., Cancer Gene Ther., 8:361-370 [2001]). Third, the IRES-SP-LC element is cloned into the backbone after the HC. Fourth, the third element of the construct, the J-chain, preceded by the second IRES is cloned into the 118 WO 2007/047189 PCT/US2006/039361 backbone. The present invention contemplates that using two different IRES elements reduces the likelihood of recombination events that are usually triggered by different identical sequences in a vector. The use of the IRES element in between heavy and light chain genes has been extensively tested and has proven to yield fully functional antibodies, 5 expressed and secreted into the medium at exceptionally high levels (up to 100 pg/cell/day in CHO cells in serum free medium). Once the retroviral constructs are complete, quality control sequencing is used to confirm that all the elements are present. The retrovector construct are then used to transfect production cell lines. Following production, purified product (either pentameric or 10 hexameric IgM 3E2) is tested using standard in vitro inhibition tests described herein and/or of known technologies. In some embodiments, for the r3E2 monoclonal antibody to be expressed in the milk of cows, lactation specific promoter based on the bovine alpha-lactalbumin promoter is used (G.T. Bleck and R.D. Bremel, Gene, 126:213-218 [1993]), and the neo-selectable marker is 15 removed from the construct. (See, Figures 4C and 4D). In a standard cloning step, the sCMV promoter used in the retrovector system is replaced with the alpha-lactalbumin promoter. Clonal analysis perfonned on the packaging cell lines to identify the antibody producing clones that give the highest expression. In additional embodiments, an IgM isotype control (of irrelevant specificity) is constructed in parallel following the same 20 cloning strategy. EXAMPLE 4 Production of Vector and Injection into Bovine Oocytes to Make Transgenic Embryos for Transfer to Recipient Animals (Cattle) 25 After quality assurance, the alphalactalbumin- bearing construct is used to transfect the 293 gp packaging cell line along with the plasmid encoding for the VSVg surface glycoprotein for pseudotyping the viral particles. (See, A.W. Chan et al., Proc. Natl. Acad. Sci. USA, 95:14028-14033 [1998]). 293gp packaging cells used in this process are derived from working stocks supplied by Gala Designs, Inc. (Middleton, WI) for cGMP production. 30 The resulting viral supernatant is used to infect packaging cells at a low virus to cell ratio so as to achieve single insertions of the virus. The packaging cell pool are then subjected to a clonal analysis and supernatants of single clones are monitored for high titer viral particle 119 WO 2007/047189 PCT/US2006/039361 production. One clone is chosen based on viral titer and quality assurance for use in the TRANSGAMETIC system (Gala Design, Inc., Middleton, WI). Bovine oocytes are harvested and grown in culture to metaphase 2 arrest (16 hrs). At this stage, which is prolonged in the bovine oocyte, the nuclear membrane has dispersed, allowing vector to 5 gain access to the nucleus. Pseudotyped vector injected into the perivitelline space infects the oocyte and provirus is integrated into the oocyte's haploid DNA. Upon viral particle injection, the oocytes are fertilized using sexed semen, which allows for almost 100 % female calves to be born. The transgenic frequency is expected to be between 25-75%. Preferably, embryos are biopsied and screened by PCR for presence of the transgene prior to 10 transfer into recipients, to optimize the transfer of transgene positive embryos. In vitro tests are done to confirm that the tricistronic expression of the 3E2 IgM antibody has no influence on specificity and affinity of this antibody. Following positive in vitro tests of the recombinant 3E2 monoclonal antibody and achievement of high numbers of transgenic embryos, the embryos are transferred into surrogate mothers. Accordingly, 15 groups of young mature female cattle (heifers) are hormonally synchronized to receive embryos at seven days post fertilization. Cattle are observed throughout pregnancy and ultrasounds are conducted to confirm pregnancy and sex of the embryo at 70 days. After the 280 day gestation period, calves are delivered by cesarian section (a routine surgery performed under epidural anesthesia in a standing surviving cow) and tested for the 20 transgene. EXAMPLE 5 Confirmation of Transgene Presence Following birth of the offspring they are tested for presence of the transgene and 25 raised to near puberty. Lactation is then honnonally induced to identify the best protein expression and to provide product for evaluation. At the age of approximately 8 months, lactation is induced in transgenic heifers, using a hormonal regimen, and milk analyzed for expression of the r3E2 product. Product is collected, purified from whey, and quantified for efficacy studies in mice and piglets. 30 A progestin implant is used to simulate a short pseudopregnancy and then initiate milking in peripubertal (6-8 months old) heifers. Heifers should yield up to 250-1000 ml per day of milk, increasing rapidly to approximate a first lactation heifer yield of 15-20 liters 120 WO 2007/047189 PCT/US2006/039361 a day. Subsequent fertility is not impaired. Milk product is tested for the presence of murine antibody using established Western blot and ELISA procedures. The animals are milked until enough product is obtained to conduct efficacy testing in mice and the neonatal pig model using assays described herein. 5 For quantification to carry out efficacy testing in pigs, monoclonal antibody is purified after fat removal from milk by continuous flow centrifuge while the milk is at animal body temperature. A skim milk product is used for further processing. In some embodiments, size exclusion chromatography and tangential-flow ultrafiltration allow purification of sufficient amounts. MAbs are recovered from the milk serum with affinity chromatography 10 or size exclusion chromatography with a similar efficiency as from cell culture fluids. EXAMPLE 6 Evaluation of Efficacy of Milk Production In some embodiments, efficacy studies of milk production are preformed in neonatal 15 mouse and piglet models respectively. In vivo efficacy assays for C. parvum neutralizing r3E2 are preformed in mice. Studies of the effect of milk expressed 3E2 on the infectivity of C. parvum sporozoites in mice are performed as described herein. In vivo efficacy assays for C. parvum neutralizing r3E2 are conducted in piglets. These studies are performed following the same protocol as described herein. Three groups of 8 20 piglets are assigned to treatment (milk derived r3E2), isotype rIgM control, and placebo control groups. Dosages, experimental regimens, and blinded evaluations are conducted as described herein. EXAMPLE 7 25 Founder Animals Lines of founder animals are identified for propagation to develop production herds. Suitable high expressing transgenic founder animals (e.g., cattle) are identified and superovulated for propagation of a herd of production animals for large scale production of r3E2. Yields of r3E2 in milk are compared between founder animals and the best animal(s) 30 selected for super ovulation and insemination. Embryos are harvested and stored in liquid nitrogen for future herd expansion. 121 WO 2007/047189 PCT/US2006/039361 EXAMPLE 8 Identification of Candidate for Expression as Recombinant Antibody Biocide Fusion Proteins In some embodiments, various biocides are evaluated for potential neutralizing 5 activity against C. parvum sporozoites using the in vitro assay described in herein. Candidate biocides include, but are not limited to: PLA2, both from human and bee venom; protease inhibitors such as leupeptin, aprotinin, antipain, amastatin, and soybean trypsin inhibitor; lysozyme; and phosphatidylinositol-specific phospholipase C. The preceding protease inhibitor candidates were selected based on their reported activity against C. 10 parvun. (See e.g., J.R. Forney et al., J. Parasitol., 82:638-640 [1996]; J.R. Forney et al., J. Parasitol., 83:771-774 [1997]; and P.C. Okhuysen et al., Antimicrob. Agents Chemother., 40:2781-2784 [1996]). For this assay, isolated sporozoites are incubated (15 min, 37C) with an individual biocide in isotonic buffer over a range of concentrations that would theoretically be 15 achievable at the sporozoite surface by targeted delivery as a MAb-biocide fusion protein. PLA2 concentrations are based in part on preliminary data which showed that < 0.02 units/ml was effective in neutralization. In parallel, viability of control sporozoites after incubation with the selected biocide concentrations is determined by fluorescein diacetate assay. (See, M.W. Riggs et al., Infect. Immun., 62:1927-1939 [1994]). Following 20 incubation with biocide, sporozoites are washed, and then inoculated onto individual Caco-2 human intestinal epithelial cell monolayers grown in microscopy grade 96-well plates (10 replicates per treatment). For comparison, control monolayers are inoculated with sporozoites identically incubated with: 1) MEM; 2) murine hybridoma-derived neutralizing MAb 3E2 as a positive control; or 3) non-toxic control proteins such as BSA, each 25 concentration-matched to the biocide being tested. Samples are then processed and evaluated as described herein. The mean numbers of intracellular parasite stages per host cell in test and control cultures is examined for significant differences using ANOVA. Each experiment is performed three times. In parallel experiments, to monitor potential host cell toxicity of residual biocide, control monolayers are inoculated with the final wash medium 30 from biocide incubation tubes to which no sporozoites were added, but which otherwise have been processed identically to test samples. Cell viability in control and sporozoite inoculated monolayers is determined 24 hrs post-inoculation using an acridine orange 122 WO 2007/047189 PCT/US2006/039361 ethidium bromide viability assay and epifluorescence microscopy. (See, R.C. Duke and J.J. Cohen, Morphological and biochemical assays of apoptosis. John Wiley & Sons, New York, NY, [2002]). 5 EXAMPLE 9 Isolation of Genes for Antibody Heavy, Light chains, and J chains In some embodiments, the genes for antibody heavy, light chains, and J chains where applicable, are isolated from the 1E10, 3H2, and 4H9 hybridoma cell lines. The genes are cloned into a retrovector as standalone antibody constructs for each antibody, and for the 10 IgGls lE10 and 4H9, as fusions to a biocide gene. In some embodiments, 4 structurally different antibody-biocide fusion variants are considered for lE10 and 4H9. At the same time, vectors with a promoter suitable for transgenic expression are prepared. The following hybridoma cell lines are used for antibody gene extraction: 3H2, which expresses an IgM against GP25-200; 4H9, which expresses an IgG1 against GP25 15 200; and IE10, which expresses an IgG1 against P23. An isotype control for IgG is constructed and prepared in parallel using a hybridoma of irrelevant specificity. Total RNA is extracted from cells with the purpose of isolating the monoclonal antibody-specific heavy and light chain genes as described herein. The immunoglobulin genes are be cloned into the retrovector backbone as bicistronic constructs; in the case of 3H2, the present invention 20 contemplates apply a cloning strategy identical to the one applied for the 3E2 constructs described herein. (See, Figures 5A-5D). Standalone recombinant constructs of each antibody are produced. In addition, two IgG isotypes are engineered to contain a biocide attached to either the N-terminus or the C-terminus of the antibody. The cDNA for the biocide found to be most effective in neutralizing C. parvum sporozoites in vitro, and least 25 toxic to host cells is acquired through either the NIH Mammalian Gene Collection (human PLA2) or synthesized (Blue Heron Biotechnology, Seattle). The PLA2, or other biocide, cDNA is expanded through standard amplification in E. coli laboratory strains. Plasmid are extracted and sequenced for quality control purposes. The biocide genes are then cloned into 4 different antibody fusion constructs using glycine-serine (G4S)3-4 linkers. When 30 expressed, these constructs produce: a full size antibody with a biocide fusion to either the N-tenninus (Figure 5A) or to the C-tenninus (Figure 5B) of the heavy chain; or a single chain antibody with a biocide fusion to the N-terminus of the light chain (Figure 5C) or to 123 WO 2007/047189 PCT/US2006/039361 the C-terminus of the heavy chain (Figure 5D). The antibody-biocide fusions are tested for their efficacy in mediating neutralization and killing of sporozoites in vitro and reducing infection in vitro and in vivo. Constructs are also prepared for the production of transgenic embryo monoclonal 5 antibody to be expressed in the milk of cows, using a lactation specific promoter based on the bovine alpha-lactalbumin promoter (G.T. Bleck and R.D. Bremel, Gene, 126:213-218 [1993]), and the neo-selectable marker is removed from the construct (Figure 4). In a standard cloning step, the sCMV promoter used in some retrovector systems is replaced with the alpha-lactalbumin promoter. 10 Retrovector constructs are used to transduce host cells and produce pseudotyped replication deficient retrovector. Pool populations of transduced cells are subjected to a clonal selection, based on antibody levels present in the medium supernatant determined by C. parvum ELISA. Clones with the highest level of antibody secreted into the supernatant are chosen to produce milligram amounts of recombinant murine monoclonal antibody and 15 monoclonal antibody-biocide fusions against C. parvum. Constructs needed for transgenic cattle production are also prepared. Constructs contain lactation specific promoter based on the bovine alpha-lactalbumin promoter (G.T. Bleck and R.D. Bremel, infra), and no neo-selectable marker (Figure 4). In a standard cloning step, the sCMV promoter used in some retrovector systems is replaced with the 20 alpha-lactalbumin promoter. EXAMPLE 10 Cloning of Vector Constructs 25 In some embodiments, the above vector construct, and those for antibody 3E2 are clonally selected and expressed in a retrovector cell culture system to obtain adequate quantities of assembled antibody or antibody-biocide fusion protein for testing in vitro and in vivo. Briefly, the retrovector constructs prepared above are used to transform host cells 30 along with the plasmid that encodes the vesicular stomatitis virus glycoprotein (VSV-G) used for pseudotyping the retrovirus. This procedure creates intermediate level viral titer that is used to infect production cell lines (CHO cells). CHO cells used in this process are 124 WO 2007/047189 PCT/US2006/039361 derived from a working stock used to established a cGMP production. The population of transduced cells is subjected to a clonal selection, based on antibody levels present in the medium supernatant. Antibody levels are determined by standard ELISA methods using sporozoite lysate antigen prepared as described in Schaefer et al. (D.A. Schaefer et al., 5 Infect. Immun., 68:2608-2616 [2000]). The clones with the highest level of antibody secreted into the supernatant are chosen to produce milligram amounts of recombinant monoclonal antibody. Using the cell culture in a roller bottle system, gram scale quantities of rMAbs 3E2, 3H2, lE10, 4H9, and the rMAb-fusion parasiticides are expressed. Based on a 30 pg/cell/day 10 average, one roller bottle produces approximately 20 mg product per week. In some embodiments, complete product purification is unnecessary to formulate oral immunotherapies, especially when milk derived. However, in some embodiments, for the purposes of standardization of tests, purification of the monoclonals from tissue culture medium follows protocols established for other monoclonals. Briefly, harvested media is 15 filtered through a 0.45 micron sterile filter to remove cells and the immunoglobulins (IgG1, IgG2, and IgG4) and are captured using a protein A affinity column, or in case of IgM, using HiTrap IgM Purification columns (Amersham Biosciences, Piscataway, NJ) or for the purification of single chain antibodies Thiophilic Resin columns (BD Biosciences Clontech, Palo Alto, CA). After washing, the immunoglobulins are eluted by low pH and the pooled 20 eluate fractions are neutralized to pH 7.5. In some embodiments, a second chromatography step is employed to remove contaminants, host cell DNA and to act as a viral clearance step. This typically utilizes anion exchange chromatography (e.g., Q-Sepharose). The final polishing step utilizes size exclusion chromatography (e.g., Sephadex 200), to separate aggregates from monomers. Antibody are further concentrated or formulated as required. 25 EXAMPLE 11 Recombinant Monoclonal Antibodies and Monoclonal Antibody Biocide Fusion Products Efficacy in Neutralizing Sporozoites In vitro In some embodiments, recombinant monoclonal antibodies and monoclonal antibody 30 biocide fusion products expressed herein are tested for their efficacy in neutralizing sporozoites in vitro. 125 WO 2007/047189 PCT/US2006/039361 Prior to testing in neutralization assays, the monoclonals are evaluated for retention of sporozoite and merozoite reactivity by IFA, and for antigen specificity by Western immunoblot. (See e.g., M.W. Riggs et al., Infect. Immun., 62:1927-1939 [1994]; M.W. Riggs et al., J. Immunol., 158:1787-1795 [1997]). 5 In vitro neutralization assay for C. parvum To quantify specific neutralizing activity of each of the four MABs and the fusion biocides against the infective sporozoite stage, an in vitro neutralization assay is used. (See, R.C. Langer et al., Infect. Immun., 67:5282-5291 [1999]). The antibody-biocide fusions 10 based on the r1E10 and r4H9 antibodies in the four configurations depicted in Figure 5, and full size versions of r1El0, r4H9, and r3H2 are each tested individually. For this assay, isolated sporozoites are incubated with the selected MAB (10 pg/ml final concentration), then inoculated onto individual Caco-2 human intestinal epithelial cell monolayers (ATCC HTB37) (M. Pinto et al., Biol. Cell, 47:323-330 [2002]). Prior to inoculation, monolayers 15 of Caco-2 cells are grown to ~90% confluency in microscopy grade 96-well tissue culture plates. For comparison, control monolayers are inoculated with sporozoites which have been identically incubated with: 1) tissue culture medium (MEM); 2) murine hybridoma derived neutralizing monoclonal; or 3) isotype- and concentration- matched recombinant control MAb of irrelevant specificity. Ten replicates are performed for each treatment. 20 After incubation, inoculation medium is aspirated from monolayers and replaced with MEM. At 24 hrs post-inoculation, monolayers are washed, fixed, blocked, and processed for automated immunofluorescence assay (IFA) using MAb 4B10 and AlexaFluor488 affinity-purified goat anti-mouse IgM to detect intracellular stages. MAb 4B10, prepared against C. parvum as previously described (M.W. Riggs et al., J. Immunol. 158:1787-1795 25 [1997]), recognizes all parasite stages in Caco-2 cells through 72 hrs post-inoculation. (R.C. Langer and M.W. Riggs, Infect. Immun., 67:5282-5291 [1999]). Intestinal epithelial cell nuclei are counterstained with 300 nM 4,6-diamidino-2-phenylindole. Using an Olympus-IMT2 inverted microscope equipped for automated digital image capture, 50 standardized visual fields per well are read and stored on the program computer. 30 Intracellular parasite stages and epithelial cell nuclei are then quantified using Compix SimplePCI software (Compix, Inc., Cranberry Township, PA). Mean numbers of intracellular parasite stages per host cell in test and control cultures are examined for 126 WO 2007/047189 PCT/US2006/039361 sig can differences using ANOVA. Each experiment is performed three times. BSL-2 precautions are observed to prevent accidental infection of project personnel with C. parvum. 5 Cryptosporidium parvum propagation for use in the proposed studies The Iowa C. parvuin isolate (J. Heine et al., J. Infect. Dis., 150:768-775 [1984]) (genotype 2, bovine origin) has been maintained since 1988 by propagation in newborn Cryptosporidium-free calves (M.W. Riggs et al., J. Inununol., 143:1340-1345 [1989]; and M.W. Riggs and L.E. Perryman, Infect. Immun., 55:2081-2087 [1987]). This well 10 characterized isolate is infectious for humans and animal models, including neonatal mice and pigs. (See e.g., J. Heine et al., J. Infect. Dis., 150:768-775 [1984]; R.C. Langer and M.W. Riggs, Infect. Immun., 67:5282-5291 [1999]; H.W. Moon and W.J. Bemrick, Vet. Pathol., 18:248-255 [1981]; and S. Tzipori H. and Ward, Microbes. Infect., 4:1047 [2002]). Parasites are obtained by propagation in newborn calves as previously described. (M.W. 15 Riggs and L.E. Perryman, supra). Oocysts are isolated from the feces of experimentally infected calves as previously described, and stored in 2.5% KCr 2 0 7 (4 'C) (M.J. Arrowood K. and Donaldson et al., J. Eukaryot. Microbiol., 43:895 [1996]; and M.W. Riggs and L.E. Perryman, supra). To obtain isolated sporozoites, oocysts are hypochlorite-treated prior to excystation, then passed through a sterile polycarbonate filter. For mouse and piglet 20 experiments, oocysts are used within 30 days of isolation and disinfected with 1% peracetic acid prior to administration. EXAMPLE 12 In vivo Neutralizing Activity Assays 25 Each of the monoclonal antibody-biocide fusions based on the rlE10 and r4H9 antibodies and monoclonal antibodies r3E2, r1E10, r4H9, and r3H2 determined to have significant in vitro sporozoite neutralizing activity, is individually tested to quantify in vivo efficacy against infection. The neonatal mouse model is used. (See, M.W. Riggs MW and L.E. Perryman, Infect. Imnun., 55:2081-2087 [1987]; and D.A. Schaefer et al., Infect. 30 Inmun., 68:2608-2616 [2000]). Groups of 15 six-day-old specific pathogen free ICR mice (Harlan Sprague Dawley) are administered 5 x 104 oocysts (50 X mouse IDso) by gastric intubation. After 48 hrs, culture-derived r3E2 (4 mg MAb/ml, 75 p) are given by 127 WO 2007/047189 PCT/US2006/039361 incubationn. Every 12 hrs thereafter, mice are administered additional r3E2 (4 mg MAb/ml, 100 pl), for a total of eight treatments. Cimetidine (10 mg/kg) are included with all treatments. For comparison, groups of 15 six-day-old control mice are infected and treated identically with: 1) murine hybridoma-derived neutralizing 3E2, or 2) isotype- and 5 concentration- matched recombinant control MAb of irrelevant specificity. After euthanasia at 140-142 hrs post-inoculation, the jejunum, ileum, cecum, and colon are collected from each mouse and processed for histopathology. Sections are coded and examined by the same investigator, without knowledge of treatment group, for C. parvum stages in mucosal epithelium. Scores are assigned to longitudinal sections representing the entire length of: i) 10 terminal jejunum; ii) ileum; iii) cecum; and (iv) colon, then summed to an infection score for each mouse. (See, M.W. Riggs MW and L.E. Perryman, Infect. Immun., 55:2081-2087 [1987]; and D.A. Schaefer et al., Infect. Immun., 68:2608-2616 [2000]). Each experiment is performed twice. Mean infection scores within each experiment are analyzed by Student's one-tailed t test. Mean infection scores between experiments are analyzed by ANOVA. 15 Additionally, all intestinal sections and sections of stomach, liver, and kidney from mice treated with antibody-biocide fusions are examined by an ACVP Board-Certified Veterinary Pathologist to determine if any lesions suggestive of biocide-host toxicity are present. EXAMPLE 13 20 In vivo Efficacy Assays of rMAbs and rMAb-Fusion Parasiticides in a Neonatal Piglet Model This example provides in vivo efficacy assays for C. parvum neutralizing rMAb. Newborn male piglets for the proposed studies are obtained by project personnel at the time of parturition from sows in which the perineum has been thoroughly cleaned using standard 25 methods equivalent to pre-surgical preparation. Piglets, collected as born and colostrum deprived, are immediately placed in disinfected isolation crates for transport to BSL-2 isolation facilities. Precautions are taken to prevent animal exposure to an exogenous source of C. parvum and other potential diarrheal agents. (See e.g., L.E. Perryman et al., Mol. Biochem. Parasitol., 80:137-147 [1996]; L.E. Perryman et al., Vaccine, 17:2142-2149 30 [1999]; and M.W. Riggs and L.E. Perryman, Infect. Immun., 55:2081-2087 [1987]). Following arrival at BSL-2 isolation facilities, piglets are assigned to either treatment (8 piglets) or control groups (8 piglets) by blind code. Group assignments and coding are 128 WO 2007/047189 PCT/US2006/039361 made by an independent third party not be involved in conducting the experiments, data collection, or interpretation of results. All personnel involved with the experiments have no knowledge of piglet group assignments. Codes are revealed only at completion of the study. Testing of rMAbs and rMAb-biocide fusion proteins, individually and in combination to be 5 selected, proceeds as follows. Testing of individual rMAbs To allow accurate comparisons between activities of the six rMAb constructs being evaluated, the concentration of each is standardized on an equimolar basis. Using the 10 experimental design for rMAb 3E2 as an example, each construct is evaluated, individually, as follows. One group of 8 piglets is administered 107 oocysts by gastric intubation at 24 hrs of age. Forty-eight hours later, each piglet receives 250 mg culture-derived rMAb 3E2 by intubation. At 12 hrs and every 12 hrs thereafter, each piglet is administered 50 mg additional rMAb 3E2 for a total of 10 treatments (750 mg MAb r3E2 total/piglet). 15 Omeprazole (PRILOSEC, Astra-Merck) [lmg/kg] is administered 6-8 hrs prior to each rMAb treatment to block production of gastric acid according to a regimen previously shown to elevate gastric pH in pigs to -7 (D.L. Foss and M.P. Murtaugh, Vaccine, 17:788 801 [1999]). As an additional precaution against gastric degradation, rMAb is formulated in NaHCO 3 buffer prior to administration. For comparison, a group of 8 control piglets is 20 identically infected with 107 oocysts and administered recombinant isotype control MAb construct according to the same treatment regimen as the principals. Piglets are confined, individually, in elevated metabolic isolation cages equipped with fecal collection pans, and maintained on ESBILAC (PetAg, Inc., Hampshire, IL) for the duration of the experiment. To prevent urine from contaminating feces for subsequent analyses, a diversion device is 25 attached and sealed around the prepucial orifice of each piglet to divert urine into a drainage outlet. Piglets are examined twice daily by a veterinarian, without knowledge of treatment group, and assigned numerical scores based on clinical assessment for symptoms of depression, anorexia, and dehydration. Piglet weights at the time of infection and at the end of the experiment are also recorded. The total volume of feces excreted and percent dry 30 matter for successive 24 hrs fecal collections is detennined to provide an objective, quantitative index of diarrhea for each piglet. Fecal samples are examined for oocysts prior to challenge and daily thereafter by IFA using oocyst-specific MAb 4D3 to determine pre 129 WO 2007/047189 PCT/US2006/039361 patent and patent periods as previously described. (See, M.W. Riggs et al., Antimicrob. Agents Chemother., 46:275-282 [2002]). Total oocyst counts (number oocysts per ml of feces X total ml feces) for each piglet is determined from samples of well-mixed feces collected over successive 12 hrs periods (M.W. Riggs et al., supra). Feces from each piglet 5 is examined for possible bacterial and viral enteropathogens by standard methods. Piglets are euthanized 10 days post-infection, or before if clinically indicated. Sections of duodenum, jejunum, ileum, cecum, and colon from identically sampled sites in each piglet are collected for histopathology. Sections are coded and examined histologically without knowledge of treatment group by an ACVP board-certified veterinary pathologist. Villus 10 length to crypt depth ratios and the density of organisms per unit length of mucosa is determined as previously described (See, M.W. Riggs et al., Infect. hlnmun., 62:1927-1939 [1994]; M.W. Riggs. and L.E. Perryman, Infect. Immun., 55:2081-2087 [1987]). Infection scores of 0, 1, 2 or 3 (0, no infection; 1, < 33% of mucosa infected; 2, 33 to 66% of mucosa infected; and 3, > 66% of mucosa infected) are assigned to longitudinal sections from the (i) 15 terminal jejunum, (ii) ileum, (iii) cecum, and (iv) proximal colon, then summed to obtain an infection score (0 to 12) for each piglet. (M.W. Riggs. and L.E. Perryman, supra). Additionally, all intestinal sections, and sections of stomach, liver, and kidney from piglets treated with rMAb-biocide constructs are examined by an ACVP Board-Certified Veterinary Pathologist to detennine if any lesions suggestive of biocide-host toxicity are present. 20 Clinical, parasitologic, and histologic data is analyzed statistically by ANOVA using the General Linear Models Program of SAS. Testing of combined rMAbs Following evaluation of the individual rMAbs above, the necessary data is available 25 to decide which rMAbs are the best candidates for testing in combination for additive efficacy. Based on previous findings in mice, an optimal combination comprises up to three MAbs, one against each of the three target antigens (CSL, P23, GP25-200) (L.E. Perryman et al., Mol. Biochem. Parasitol., 80:137-147 [1996]). Because the neutralizing activity of anti-CSL MAb 3E2 is profoundly greater than that of all other Mabs against C. parvum, in 30 some embodiments, this MAb is an important component in the selected combination. Either rMAb 1E10 or rMAb 1E1O-biocide fusion, whichever demonstrates greater efficacy in the above experiments, is included in the combination to target P23. In other 130 WO 2007/047189 PCT/US2006/039361 embodiments, to target GP25-200, rMAb 3H2, rMAb 4H9, or rMAb 4H9-biocide fusion, whichever demonstrates the greatest efficacy in the above experiments, is included. Thus, in one embodiment, the combination to be evaluated contains rMAbs 3E2 + 1E10 (standalone or biocide fusion) + 3H2 or 4H9 (standalone or biocide fusion). 5 Previous studies on MAb combinations show that hybridoma-derived MAbs 3E2, 1E10, 3H2, and 4H9 recognize distinct epitopes and do not inhibit binding of each other to C. parvuin. Nevertheless, it is useful to repeat binding inhibition experiments with the recombinant candidates selected for combination testing to confirm that they do not inhibit binding of each other due to steric hindrance or other influences introduced by recombinant 10 expression. In brief, this is evaluated by ELISA using biotin-labeled (Sulfo-NHS-biotin, Pierce) and unlabeled rMAb candidates as previously described. (See, L.E. Perryman et al., supra). Inmulon-4 96-well ELISA plates are coated with solubilized sporozoite antigen, washed, and blocked. Plates are incubated with an individual unlabeled rMAb, then biotinylated competitor rMAb, washed, and developed with peroxidase-labeled Streptavidin 15 and substrate. Mean ODs of replicate wells for each treatment and control group are analyzed for significant differences. After determining that the three rMAbs selected for combination testing in piglets do not significantly inhibit binding of each other, they are combined and the concentration of each standardized on an equimolar basis to match that previously evaluated individually. 20 Efficacy testing of the combined rMAbs in piglets then proceeds as described for individual rMAbs above. In brief, one group of 8 piglets are infected with 107 oocysts at 24 hrs of age and receive the combined rMAbs in NaHCO3 buffer 48 hrs later by intubation. At 12 hrs and every 12 hras thereafter, piglets receive additional combined rMAbs for a total of 10 treatments. For comparison, a group of 8 control piglets are identically infected and 25 administered an appropriate isotype control rMAb combination according to the same treatment regimen. Clinical assessment scores, piglet weights, total volume of feces excreted and percent dry matter for successive 24 hr collections, pre-patent and patent periods, and total oocyst counts are determined as above. Ten days post-infection, sections of duodenum, jejunum, ileum, cecum, and colon from each piglet are collected, and 30 examined histologically to assess lesions and assign infection scores. Tissues are also examined to determine if any lesions suggestive of biocide-host toxicity are present. Clinical, parasitologic, and histologic data is analyzed statistically as described above. Data 131 WO 2007/047189 PCT/US2006/039361 from testing of the individual rMAbs is compared with data from testing of the rMAb combination by one-way ANOVA stratified by treatment group. Example 14 5 Anti-Cryptosporidium Biocides This Example describes biocides directed towards Cryptosporidium. A. Materials and Methods 10 Oocyst and sporozoite isolation. The Iowa C. parvum isolate (Heine, 1984) used in all experiments was propagated in newborn Cryptosporidium-free Holstein bull calves to obtain parasite material for study (Riggs, 1987). Oocysts were isolated by sucrose density gradient centrifugation and stored in 2.5% (wt/vol) K2Cr2O7 (44C) prior to use (Arrowood, 1987). Immediately prior to 15 excystation, oocysts were treated with hypochlorite (Riggs, 1987). Sporozoites were isolated from excysted oocyst preparations by passage through a polycarbonate filter (2.0 mm, pore size; Poretics, Livermore, Calif.). Biocides and host cell toxicity assay 20 Lactoferrin (LF; Sigma), lactoferrin hydrolysate (LFH)(Murdock, 2002), lactoferricin B (LFB; Sigma), cathelicidin (CAT; LL-37, Phoenix), indolicidin (IND; Sigma), b-defensin 1 (BD1; Peptides international), b-defensin 2 (BD2; Peptides international), lysozyme (LYZ; Sigma), bee-venom phospholipase A2 (PLA2; Sigma), phospho-inositol specific phospholipase C (PI-PLC; Sigma). 25 Host cell toxicity assay was performed by measuring the release of lactate dehydrogenase (LDH) in the medium using a CYTOTOX 96 Non-Radioactive Cytotoxicity Assay kit (Promega, Madison, WI). Toxicity was therefore classified as non toxic 0-5% LDH release and mild toxicity 5-10% LDH release (ref). 30 Sporozoites viability Sporozoites viability was assessed using an adapted fluorescein diacetate (FDA) and propidium iodide (PI) vital dye technique (Arrowood, 1991 AAC). In brief, freshly 132 WO 2007/047189 PCT/US2006/039361 excysted sporozoites were incubated for 15 min at 37 0 C in the presence of the biocides or in control medium. Heat killed sporozoites (20 see at 1 00C) were used as positive controls. Aliquots were supplemented with FDA and PI (8 mg/ml and 3mg/ml final concentration, respectively), incubated for 5 min at room temperature and stored at 4*C until examined. 5 Viability was determined by counting the relative number of green-fluorescing (viable) and red-fluorescing (dead) sporozoites at a magnification of 200X on an epifluorescence microscope (a minimum of 100 sporozoites were counted for each condition, all experiments were performed in triplicates). 10 Infection of Caco-2 monolayers and automated counting system Caco-2 cells were grown to 90% confluency in complete MEM (MEM containing 10% fetal bovine serum, 1% nonessential amino acids, 100 U of penicillin per ml, and 100 mg of streptomycin per ml) on glass coverslips or in black plastic bottom 96-well plates for the automated counting system and infected as described before (Langer, 1999). Briefly, 15 purified sporozoites in 50 ml of minimum essential medium [MEM]) were incubated (15 min, 37'C, 10% C02) with MAb 3E2 or isotype-matched control MAb or with the biocide and then inoculated onto the monolayer (three replicates per treatment for manual counting and five replicates for the automated counting). At 24 h postinoculation, cultures were washed with PBS, methanol fixed (4 min, -20'C), blocked (PBS containing 3.2% [wt/vol] 20 fish gelatin and 1% [wt/vol] bovine serum albumin [BSA]), and processed for IFA by using MAb 3E2 and affinity-purified fluoresceinated goat anti-mouse IgG-IgM-IgA (Kirkegaard & Perry, Gaithersburg, Md.) to detect intracellular stages. MAb 3E2 were prepared against immunoaffinity chromatography-isolated GP25-200 as previously described (Riggs, 1997). Results were evaluated manually or using an automated system. Manually: Each 25 coverslip was then systematically examined by the same investigator by epifluorescence microscopy to directly quantitate the number of intracellular stages per monolayer. Automated : Each well was examined using an epifluorescence microscope with automated stage coupled to a computer using Simple PCI software, 24 images of each well were recorded and analyzed using breakpoints for fluorescence intensity, size (area and diameter), 30 and roundness in order to quantify the number of intracellular stages per monolayer. Non infected monolayers were also counted in order to measure background. The mean numbers 133 WO 2007/047189 PCT/US2006/039361 of intracellular stages in test and control cultures were examined for significant differences by using Student's two-tailed t test. B. Results 5 The results are shown in Table 3 and Figures 11-14. Table 3 shows the toxicity of biocides to host cells. Figure 11 shows the parasticidal activity of different biocides against C. parvum spores. Figure 12 shows the P-values for the data of Figure 11 against a no biocide control. Figure 13 shows the effect of biocides on C. parvum sporozoite infectivity for Caco-2 human intestinal epithelial cells. Figure 14 shows the P-values for the data in 10 Figure 13. The results indicate that several biocides have activity against C. parvum spores and infectivity without significant toxicity against human cells. Table 3 Non-Specific Toxicity of Biocides to Caco-2 Host Cells Measured by LDH Release 15 Compound % LDH release 10 [g/ml 100 ptg/ml Mab 3E2 0.7 ±1.1 nd LF 5.9 ±1.2 * 5.8 ±3.1 LFH 2.6 ± 3.5 0.7 ±2.1 LFB 3.7 ± 2.3 2.6 ±2.0 CAT 1.6 ± 0.3 * 5.6 ± 0.9 * 1ND 0.9 ± 0.7 8.3 3.5 * PD1 0.8 ± 1.0 2.3 2.1 pD2 -0.9 ± 0.5 5.3 i 1.5 * LYZ Nd -1.9 1.3 PLA2 Nd 0.7 0.7 PI-PLC Nd - 1.4 0.9 nd not determined * : P<0.05 to control (medium alone for 2h) 20 Example 15 3E2 Biocides 134 WO 2007/047189 PCT/US2006/039361 This example describes the isolation and characterization of 3E2 derived monoclonal antibody based biocides A. Isolation of the genes for the heavy, light, and J-chains from the 3E2 murine 5 hybridoma cell line. RNA was extracted from snap-frozen hybridoma cells of clone 3E2. Total RNA was reverse transcribed using oligo dT primer to generate cDNA from mRNA transcripts. This cDNA was used for extraction of the 3E2 immunoglobulin genes. The Ig-Primer Set kit from Novagen (EMD Biosciences, San Diego, CA) was used. Figure 15 illustrates a typical 10 immunoglobulin heavy chain isolation procedure by PCR. Specifically, the Novagen degenerate upper primers that are designed to anneal to the signal peptide region of the variable region were utilized and combined with a lower primer that was designed to anneal to the C3 region of the heavy chain. In a second step the constant region was isolated using two primers specific to the C2 region and to the 3'UTR of IgM. 15 PCR products were combined in an overlap-extension PCR (oePCR) reaction, which results in a single product made up from overlapping template sequences and two flanking primers. The sequence was confirmed as the entire IgM heavy chain. The variable light chain was obtained likewise, but in one step using a degenerate upper primer and a 3'UTR lower primer. The J-chain was obtained by PCR using primers to upstream and downstream 20 untranslated regions of the gene and the 3E2 cDNA as a template and was confirmed by sequencing. The PCR products were cloned into a retrovector backbone, followed by transduction of a CHO production cell line, first to produce hexamer IgM, and subsequently superinfection with the J chain to make pentamer antibody. Clonal analysis was performed 25 to identify the highest expressing clones. The IgM molecule exists naturally in two different configurations (1,12): i) the pentamer, in which the J-chain links five immunoglobulins together, and ii) the hexamer, a ring of six immunoglobulins, which forms spontaneously in the absence of J-chain. To test both versions for efficacy, a bicistronic construct was generated. This construct is designed 30 for antibiotic-based selection of transduced cells. Resistant cells are clonally selected by quantification of product expression. The clones expressed IgM at low levels (0.3 ug/ml), as determined by ELISA using anti-heavy and anti-light chain antibodies. A fluorescent 135 WO 2007/047189 PCT/US2006/039361 marker-based selection that included a microtiter plate-capable fluorescence activated cell sorter (FACS) was next utilized. It is contemplated that a good insertion site in the host cell genome results in a high expression of both the LTR-inherent promoter-driven EGFP and the sCMV promoter-driven IgM. High immunoglobulin-expressing clones were isolated 5 from cells that were selected based on high green fluorescence by FACS. Cells that were selected from a lower EGFP expressing population produced less product. Top clones from a first round of infection using fluorescence-based selection resulted in a top clone that produced 7 ug/ml in a T150 culture flask on day 4. This represented a >20-fold increase in production due to improved clonal analysis criteria. Initial testing of hexamer antibody 10 derived from the top clone from this infection (clone 285) for binding to Cryptosporidium showed a high level of reactivity with the epitope on the sporozoite surface defined by hybridoma MAb 3E2 (Figure 17). Clone 285 was reinfected with vectors containing additional bicistronic construct for IgM hexamer and with a separate construct for J-chain (co-infection) to make pentamer. The J-chain construct was equipped with a different 15 fluorescent marker (red) allowing a double selection for green and red in the cell sort. Equal amounts of retrovirus encoding either the 3E2 IgM heavy and light chain (Fig. 16A) or the J-chain (Fig. 16B) were used to transduce a clone that already contained one or more copies of 3E2. A dual color sort was done. The entire population of this pool is already bright green whereas the transduction with the J-chain construct resulted in a mixed population of 20 low level to high level red signal. The gate was chosen to include the highest level of positive cells for both colors. The clones resulting from this experiment were analyzed for the expression of IgM using an established ELISA assay. The presence of the J-chain in these clones was confirmed in two ways: First by quantitative PCR detection of J-chain transcripts (Figure 19) and second by molecular weight determination via Western Blot. 25 The quantitative PCR analysis was performed using total RNA extracted from the top 5 clones. Clone 116, which had a J-chain to heavy chain ratio closest to the hybridoma (3E2) ratio, was also the best producer of pentameric IgM. The data demonstrate that the high level of heavy chain transcripts detected for clone 127 is consistent with its superior productivity of hexameric IgM antibody. 30 B. Efficacy of the recombinant 3E2 monoclonal antibody (r3E2) in neutralizing sporozoites in vitro. 136 WO 2007/047189 PCT/US2006/039361 The activity of r3E2 against C. parvum sporozoite infectivity was determined in an in vitro assay. In brief, sporozoites were incubated (37'C, 15 min) with culture supernatant from CHO cells expressing r3E2 hexamer or pentamer (10, 1, or 0.1 pg/ml, final concentrations). Control sporozoites were incubated identically with culture supernatant 5 from 3E2 hybridoma cells or non-recombinant CHO cells. Following incubation, the sporozoite preparations were inoculated onto replicate Caco-2 human intestinal epithelial cell monolayers, incubated an additional 2 h at 3 7'C, after which monolayers were washed and medium replaced. At 24 h post-inoculation, the number of intracellular stages in test and control monolayers was quantified by automated immunofluorescence microscopy and 10 the mean % reduction of infection calculated. Data were analyzed by Student's one-tailed t test. All MAbs neutralized sporozoite infectivity and did so in a concentration-dependent manner. The neutralizing activity of r3E2 pentamer was comparable to that of hybridoma derived 3E2 pentamer and the activity of r3E2 hexamer was superior to that of either. 15 Because neutralization by hybridoma-derived 3E2 is mediated by binding to the CSL ligand and occurrence of the circumsporozoite precipitate (CSP) reaction, after which sporozoites are unable to attach to human intestinal epithelial cells, the ability of r3E2 to mediate this reaction was evaluated. Both hexameric and pentameric r3E2 elicited the CSP reaction; however, the rapidity and extent of the reaction following exposure of sporozoites to the 20 hexamer were greater than that observed with the pentamer. This observation parallels the greater anti-cryptosporidial activity of hexameric r3E2. C. Ability of r3E2 to reduce infection in a neonatal mouse model To quantify the in vivo efficacy of r3E2 against infection, a neonatal mouse model 25 was used. In brief, groups of 10-12 eight-day-old SPF ICR mice were infected with 5 x 104 oocysts (50 X mouse ID50) and treated twice daily for a total of nine treatments with either r3E2 hexamer or pentamer (1.5 mg rMAb/ml in CHO cell culture supernatant, 0.1 ml) by gastric intubation. For comparison, groups of 10-12 eight-day-old control mice were infected and treated identically with 1) hybridoma-derived 3E2, or 2) non-recombinant CHO 30 cell culture supernatant. Mice were euthanized at 92-94 hours post-infection. Intestinal sections were collected, processed for histopathology, examined without knowledge of treatment group for C. parvum stages in mucosal epithelium, and assigned infection scores. 137 WO 2007/047189 PCT/US2006/039361 Scores for treated and control mice were used to calculate the mean % reduction of infection. Data were analyzed by Student's one-tailed t test. All MAbs significantly reduced intestinal infection levels (Fig. 18). The therapeutic activity of r3E2 hexamer in vivo was greater than that of r3E2 pentamer. The results 5 indicate that recombinant-expressed 3E2 retains specific activity characteristic of the native hybridoma and is capable of mediating neutralization in the gastrointestinal lumen during the brief period that zoites are extracellular. Example 16 10 Antibody-Biocide Fusions This Example describes the generation and activity of antibody-biocide fusion proteins. A. Isolation of the genes for antibody heavy and light chains, from the 1E10, 18.44, 15 and 4H9 hybridoma cell lines. Cloning of antibody genes into a retrovector as standalone antibody constructs for each antibody, and asfusions to a biocide gene. The antibody heavy and light chain genes from 4H9, 1E10 and 18.44 were isolated. Degenerate upper primers were used along with lower primers specific to IgG1 constant region. The same constant region was used to 20 generate 4H9, 1E10 and 18.44 full size heavy chain genes. For the two light chains, different primer sets had to be used since the 1 E10 antibody contains a lambda light chain, whereas the 4H9 and 18.44 light chain is of the kappa isotype. A problem was encountered in the in-frame cloning step of the light chain behind the IRES element. Persistent deletions of one or multiple bases at the junction site occurred resulting in a faulty reading frame. A 25 successful alternative cloning strategy using overlap PCR ligation with a specially designed primer was used. In addition to using the IgG1 heavy chain for 4H9 and 18.44, a constant heavy chain of the murine IgG2b isotype was included. Murine IgG2b has a substantially longer hinge region, which makes the Fc portion more flexible when bound to the antigen. The present invention is not limited to a particular mechanism. Indeed, an understanding of 30 the mechanism is not necessary to practice the present invention. Nonetheless, it is contemplated that this additional flexibility allows for better binding of the biocide to the sporozoite membrane target. The IgG2b constant region was obtained from a mouse 138 WO 2007/047189 PCT/US2006/039361 monoclonal antibody library. The IgG2b constant region was combined with the variable region by overlap PCR and the resulting product was cloned and sequenced. Binding tests by IFA (indirect fluorescent assay) revealed that both the IgG1 and the engineered IgG2b version of 4H9 showed similar binding patterns on sporozoites. Based on the results of 5 efficacy tests in B 1 in which a number of biocides neutralized C. parvum sporozoites by either parasiticidal or non parasiticidal mechanisms in vitro, human LL-37 was selected as a candidate for fusion. LL-37 is a cathelicidin-derived peptide with very potent anti-bacterial properties against many bacteria and against several parasites. A new DNA sequence was generated by exchanging 5 single nucleotides to codons that were of similar usage frequency 10 and encoded the same amino acid (nucleotides 1474-1584 of SEQ ID NO:102). The nucleotide changes were introduced by primer mutagenesis overlap extension PCR using a set of 7 different primers in a single PCR reaction. The new sequence was designated cat5 and its product CAT. The gene for human phospholipase A2, group IIA was obtained from ATCC (MGC14516) in the form of a plasmid. All the antibody fusion proteins were made 15 using a Glycine-Serine linker attached to the C-terminus of the heavy chain (Figs. 19 and 20). This construct was designed to conserve the binding affinity of the antibody and to keep the fusion biocide away from the binding portion of the molecule. In order to confirm the production of the IgGl-cat5 and IgG2b-cat5 heavy chain fusions a Western Blot was performed using an anti-mouse IgG antibody. The presence of 4H9 heavy and light chain, 20 4H9-cat5 heavy chain and 4H9b-cat5 heavy chain was confirmed. The differences in size observed correspond to the expected theoretical amino acid weight differences. B. Clonal selection and expression of each construct All the in vitro and in vivo neutralization assays were performed with concentrated 25 and dialysed antibody samples including controls with cell supernatant devoid of antibody. The samples that were used for in vitro testing originated from T1 50 tissue culture flasks. Cell supernatant was harvested by centrifugation at 400g followed by a concentration step using molecular size exclusion centrifugation tubes of 50K or 100K exclusion limits. These concentrations typically resulted in a 20-30 fold concentration of the cell supernatant. For in 30 vitro tests, the concentrated samples were diluted in assay buffer. Batch production for the in vivo assays was perfonned in MantaRay disposable 800ml culture flasks (Wheaton, Alcan Packaging, Millville, NJ). Typically 400 ml of culture medium was inoculated with 139 WO 2007/047189 PCT/US2006/039361 2x1 0 7 cells; two days later another 400 ml of medium was added to reach the capacity of the vessel. The MantaRay vessels were then stirred and supplied with 5% CO 2 inside a standard tissue culture incubator. After 5-7 days, the MantaRays were harvested by spinning at 6000 g for 10 min to remove cells and debris. The supernatant was then 5 concentrated using a tangential flow filtration unit from NCSRT Inc. (Apex, NC) and either a 100k filter (for the IgM products), a 30k filter for IgG products, or a 5k filter for PLA2. This unit was also used to diafilter the product against PBS at the end of the concentration cycle. Using this procedure the supernatant was concentrated to obtain about 1.5 mg/ml end concentration. Following diafiltration the product was aliquoted and frozen immediately. 10 Aliquots were set aside for testing in ELISA to determine MAb concentration, including an assay for detection of human PLA2 (Cayman Chemicals, Ann Arbor, MI) where indicated. The PLA2 fusion was easily detected via the PLA2 specific test indicating that intact PLA2 was present in the fusion protein. 15 C. Activity of Antibodies Against C. parvum in vitro The activity of r4H9 IgG1, r4H9 IgG1-CAT, and r4H9 IgG2b-CAT products against C. parvum sporozoite infectivity in vitro was determined as described above. Products were tested at final concentrations of 20, 2, or 0.2 jLg rMAb/ml. , For comparison, sporozoites were incubated identically with CAT alone or r4H9 GI culture supernatant to which CAT 20 was added. To allow accurate comparisons between treatment groups, all products were matched on an equimolar basis for MAb and CAT. All products significantly neutralized sporozoite infectivity in vitro (Fig. 17). The neutralizing activity of 4H9 expressed as an IgG2b-CAT fusion was significantly greater than that of 4H9 expressed as an IgG1-CAT fusion at all concentrations evaluated. 25 The activity of recombinant human PLA2 and bee venom PLA2 against C. parvum sporozoite infectivity in vitro was determined as described in the above examples. Products were tested at final concentrations of 1, 0.1, or 0.01 mg/ml. Both phospholipases significantly neutralized sporozoite infectivity at all concentrations tested (Fig 22). Bee venom PLA2 had similar neutralizing activity at a 1000-fold lower concentration (0.01 30 pg/ml) than that evaluated in studies reported above (10 pg/ml; Table 3). In addition, no decrease in neutralizing activity of either human or bee venom PLA2 was observed at a 100 fold lower concentration (0.01 pLg/ml) than that observed with the highest concentration 140 WO 2007/047189 PCT/US2006/039361 evaluated in the present study (1 pg/ml). Collectively, these observations suggest that similar neutralizing activity by PLA2 would likely be obtained at concentrations far lower than 0.01 tg/ml, particularly-when the enzyme is delivered to the sporozoite surface via fusion to a MAb. These results indicate that human PLA2 is as active as bee venom PLA2 5 against C. parvum sporozoites. D. Activity of antibodies in a neonatal mouse model. To quantify the in vivo efficacy of r4H9 products against infection, the neonatal mouse model was used as described above. Individual groups of infected mice were treated 10 with r4H9 G1 (1.4 mg/ml) or r4H9 G2b-CAT fusion (1.2 mg/ml). For comparison, additional groups of mice were treated with a 10-fold lower dose of r4H9 G2b-CAT fusion (0.12 mg/ml), r4H9 G1 (1.4 mg/ml) to which CAT (42 pg/ml) (LL-37, Phoenix Pharmaceuticals, Bellmont CA) had been added, or hybridoma-derived 3E2 (3 mg/ml). Intestinal infection scores for mice in each of treatment group were compared with those for 15 control mice given non-recombinant CHO supernatant to calculate the mean % reduction of infection for each product. All treatments significantly reduced intestinal infection levels (Fig. 23). As observed in vitro, the in vivo therapeutic activity of 4H9 expressed as an IgG2b-CAT fusion and administered at 1.2 mg/ml was significantly greater than that of r4H9 expressed as an 20 IgG1-CAT fusion. This activity was concentration-dependent (Fig 23, IgG2b-CAT fusion at 1.2 mg/ml vs 0.12 mg/ml) and significantly greater at 1.2 mg/ml than a combination of r4H9 G1 and CAT or r4H9 G1 alone (Fig. 25). Thus, targeted delivery of a biocide to the sporozoite surface using a MAb-biocide fusion, in which the affinity of the antibody provides targeting, confers greater neutralizing activity than the MAb or biocide, alone or in 25 combination. r4H9 G2b-CAT (1.2 mg/ml) had significantly greater in vivo therapeutic efficacy than MAb 3E2, administered at twice the concentration. In additional mouse trials, the treatment dose for the 4H9 expressed as an IgG2b CAT or as an IgGl-CAT fusion was substantially reduced and tested for efficacy (Fig 25). Individual groups of infected mice were treated with r4H9-G1 -PLA2 fusion (one group at 30 88 gg/ml, one group at 9ptmg/ml), r4H9-Gl-CAT (63 ptg/ml) and r4H9 G2b-CAT fusion (52 pg/ml). 141 WO 2007/047189 PCT/US2006/039361 For comparison, additional groups of mice were treated with r4H9 GI (67 tg/ml) to which CAT (1.98 pg/ml) (LL-37, Phoenix Pharmaceuticals, Bellmont CA) had been added, r4H9-Gl to which rPLA2 (8.3 tg/ml) had been added, or hybridoma-derived 3E2 (one group at 3mg/ml and one group at 300 pg/ml). Intestinal infection scores for mice in each of the 5 treatment groups were compared with those for control mice given non-recombinant CHO cell supernatant to calculate the mean % reduction of infection for each product or product combination. All treatments significantly reduced intestinal infection levels (Fig. 25). The in vivo therapeutic effect of r4H9-G1-PLA2 administered as a fusion ((88 ptg/ml) was significantly 10 greater than that of a combination of r4H9-G1 and rPLA2 given as separate molecules. The in vivo therapeutic effect of r4H9-G1-CAT (63 ig/ml) and r4H9 G2b-CAT (52 pg/ml) administered as fusions were each significantly more effective than the combination of r4H9 and CAT alone. Each of the fusion products tested r4H9 G2b-CAT (52 pg/ml), r4H9-G1-CAT (63 15 pg/ml) and r4H9-PLA2 (88 pg/ml) had significantly greater in vivo therapeutic efficacy than MAb 3E2, when the MAb 3E2 was administered at 300 [tg/ml, approximately a 3-5 fold greater concentration. In yet another in vivo mouse trial, a dose titration for the treatment of two different 4H9 antibody fusions (4H9-G1 -CAT and 4H9-G2b-CAT) was perfonned to determine the 20 lowest efficacious dose. 4H9-G1-CAT or 4H9-G2b-CAT fusions were given to three different groups of mice at 50 tg/ml, 25 tg/ml and 10 pg/ml respectively. Fig. 26 shows dose-dependent efficacy of both 4H9 fusions tested, indicating a good responsiveness of the antibody fusions to dosing. Even at the 10 ptg/ml treatment dose both 4H9 fusions induced a significant (p<0.0001) better 25 In order to develop antibody-biocide fusions with a specificity against cryptosporidium sporozoites other than 4H9, two additional fusion proteins were developed based on the monoclonal antibody 18.44 which recognizes an antigen that is different from the antigen that is recognized by the 4H9 antibody. Fig 27 shows the results of the first in vivo mouse trial that was conducted with two different 18.44 fusions, 18.44-G1-CAT and 30 18.44-G1-PLA2. Of the two new fusion proteins, the 18.44-Gl-PLA2 demonstrated a reduction of infection (74 %). The data demonstrates that highly effective antibody fusions can be created against a target organism by using different specificities for the antibody 142 WO 2007/047189 PCT/US2006/039361 (target antigen) and different biocides as effector portions of the fusion indicating the potential to target many other organism by choosing optimal combinations of targeting portions and effector portions for the creation of antibody biocide fusions. Thus, this Example demonstrates that targeted delivery of a biocide to the sporozoite 5 surface using a MAb-biocide fusion, in which the affinity of the antibody provides targeting, confers greater neutralizing activity than the IAb or biocide, alone or in combination. All publications and patents mentioned in the above specification are herein incorporated by reference. Various modifications and variations of the described method 10 and system of the invention will be apparent to those skilled in the art without departing from the scope and spirit of the invention. Although the invention has been described in connection with specific preferred embodiments, it should be understood that the invention as claimed should not be unduly limited to such specific embodiments. Indeed, various modifications of the described modes for carrying out the invention that are obvious to those 15 skilled in the relevant fields are intended to be within the scope of the following claims. 143

Claims (26)

1. A composition comprising a recombinantly expressed monoclonal antibody 5 that binds to a surface epitope of Cryptosporidium sp.
2. The composition of claim 1, wherein said surface epitope of Cryptosporidiun sp. is selected from the group consisting of a CSL epitope, a p23 epitope, a GP25-200 epitope and a beta-mannosylated glycolipid epitope. 10
3. The composition of claim 1, wherein said recombinant monoclonal antibody comprises a heavy chain variable region selected from the group consisting of a 3E2 variable region, a 18.44 variable region, a 1E10 variable region, and a 4H9 variable region. 15
4. The composition of claim 3, wherein said 3E2 recombinant antibody is an IgM isotype.
5. The composition of claim 4, wherein said 3E2 is present as a hexamer. 20
6. The composition of claim 4, wherein said 3E2 is present as a pentamer.
7. The composition of claim 1, wherein said recombinant antibody has a hybrid isotype, wherein said light chain has an isotype selected from the group consisting of an IgM isotype and an IgG1 isotype and said heavy chain has an variable region selected from the 25 group consisting of an IgM isotype and an IgG1 isotype and a constant region selected from the group consisting of an murine IgG2b isotype and an human IgG3 isotype.
8. The composition of claim 1, wherein said recombinant monoclonal antibody comprises a heavy chain and a light chain and wherein said heavy chain is fused to an 30 antimicrobial polypeptide. 144 WO 2007/047189 PCT/US2006/039361
9. The composition of claim 8, wherein said antimicrobial polypeptide is a LL37 polypeptide.
10. The composition of claim 9, wherein said LL37 polypeptide is encoded by 5 nucleotides 1474-1584 of SEQ ID NO:102.
11. The composition of claim 8, wherein said antimicrobial polypeptide is selected from the group consisting of lactoferrin hydrolysate, lactoferrin b, indolicidin, beta defensin-2, deta-defensin-1, phopholipase A2, and phospho-inositol specific phospholipase 10 C.
12. The composition of claim 11, wherein said composition neutralizes Cryptosporidium parvum sporozoite infectivity. 15
13. The composition of claim 11, wherein said Cryptosporidium sp. is selected from the group consisting of Cryptosporidium parvum and Cryptosporidium hominis.
14. A vector encoding the polypeptide of claim 1. 20
15. A method of treating a subject comprising: a) providing i) a recombinantly expressed monoclonal antibody that binds to a surface epitope of Cryptosporidium sp.; and ii) a subject suspected of being contaminated or infected with 25 Cryptosporidium sp.; b) applying said recombinantly expressed monoclonal antibody that binds to a surface epitope of Cryptosporidiun sp. to said subject under conditions such that said recombinantly expressed monoclonal antibody that binds to a surface epitope of Cryptosporidiun sp. neutralizes said Cryptosporidiui sp. suspected of 30 contaminating said subject. 145 WO 2007/047189 PCT/US2006/039361
16. The method of claim 15, wherein said surface epitope of Cryptosporidium sp. is selected from the group consisting of a CSL epitope, a p 23 epitope, a GP25-200 epitope and a beta-mannosylated glycolipid epitope. 5
17. The method of claim 14, wherein said subject is a mammal.
18. The method of claim 17, wherein said mammal is a ruminant.
19. The method of claim 18, wherein said ruminant is a bovine. 10
20. The method of claim 14, wherein said mammal is a human.
21. A method of treating a subject comprising: a) providing 15 i) a composition comprising a 18.44 recombinant antibody, wherein a heavy chain of said 18.44 recombinant antibody is fused to an antimicrobial polypeptide; and ii) a subject suspected of being contaminated or infected with an apicomplexan parasite; 20 b) applying said composition to said subject under conditions such that composition neutralizes said apicomplexan parasite suspected of contaminating said subject.
22. The method of claim 21, wherein said apicomplexan parasite is 25 Cyyptosporidium sp or Eimeria sp.
23. The method of Claim 21, wherein said subject is an avian species.
24. An antimicrobial polypeptide encoded by nucleotides 1474-1584 of SEQ ID 30 NO:102. 146 WO 2007/047189 PCT/US2006/039361
25. The antimicrobial polypeptide of claim 24, wherein said antimicrobial polypeptide neutralizes Cryptosporidium sp. sporozoite infectivity.
26. A composition comprising a vector comprising a gene encoding an 5 immunoglobulin fused to a gene encoding a protein of interest, wherein said immunoglobulin comprises a murine IgG2b constant region. 10 147
AU2006303998A 2005-10-11 2006-10-10 Targeted biocides Abandoned AU2006303998A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US72518005P 2005-10-11 2005-10-11
US60/725,180 2005-10-11
PCT/US2006/039361 WO2007047189A2 (en) 2005-10-11 2006-10-10 Targeted biocides

Publications (1)

Publication Number Publication Date
AU2006303998A1 true AU2006303998A1 (en) 2007-04-26

Family

ID=37963026

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2006303998A Abandoned AU2006303998A1 (en) 2005-10-11 2006-10-10 Targeted biocides

Country Status (4)

Country Link
EP (1) EP1933874A4 (en)
AU (1) AU2006303998A1 (en)
CA (1) CA2625580A1 (en)
WO (1) WO2007047189A2 (en)

Families Citing this family (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8394379B2 (en) 2003-05-15 2013-03-12 Iogenetics, Llc Targeted cryptosporidium biocides
US8703134B2 (en) 2003-05-15 2014-04-22 Iogenetics, Llc Targeted cryptosporidium biocides
SG10201600531TA (en) 2011-01-24 2016-02-26 Univ Singapore Pathogenic mycobacteria-derived mannose-capped lipoarabinomannan antigen binding proteins
CN109942693B (en) * 2019-04-03 2020-12-25 周口师范学院 CTL epitope polypeptide of cryptosporidium parvum, application thereof and vaccine
CN113455394B (en) * 2021-07-23 2023-06-20 中国热带农业科学院橡胶研究所 Standardized production method for large-scale breeding of rubber tree body embryo seedlings

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6015882A (en) * 1992-05-29 2000-01-18 The Regents Of The University Of California Vaccines, antibodies, proteins, glycoproteins, DNAs and RNAs for prophylaxis and treatment of Cryptosporidium parvum infections
US6323020B1 (en) * 1996-08-23 2001-11-27 North Carolina State University Neutralization-sensitive epitopes of Cryptosporidium parvum
CA2869088A1 (en) * 2002-02-07 2003-08-14 Massachusetts Institute Of Technology Anti-pathogen treatments
AU2003298770A1 (en) * 2002-12-02 2004-06-23 Biosynexus Inc Wall teichoic acid as a target for anti-staphylococcal therapies and vaccines
US20050014932A1 (en) * 2003-05-15 2005-01-20 Iogenetics, Llc Targeted biocides
CN1878789A (en) * 2003-10-21 2006-12-13 加利福尼亚大学董事会 Human cathelicidin antimicrobial peptides
AU2005332655A1 (en) * 2004-10-20 2006-12-14 Iogenetics, Llc Biocides

Also Published As

Publication number Publication date
WO2007047189A3 (en) 2007-11-15
WO2007047189A2 (en) 2007-04-26
CA2625580A1 (en) 2007-04-26
EP1933874A4 (en) 2010-03-03
EP1933874A2 (en) 2008-06-25

Similar Documents

Publication Publication Date Title
AU2008203383B2 (en) Targeted biocides
US7566447B2 (en) Biocides
US20100034824A1 (en) Targeted biocides
US8394379B2 (en) Targeted cryptosporidium biocides
US20080274095A1 (en) Biocides
US20220152139A1 (en) Plant messenger packs encapsulating polypeptides and uses thereof
JP2006506053A (en) Modified transferrin-antibody fusion protein
AU2006303998A1 (en) Targeted biocides
US9518252B2 (en) Listeria bacteriophage P825 and uses thereof
US8703134B2 (en) Targeted cryptosporidium biocides
AU2003224073A1 (en) Antibodies, recombinant antibodies, recombinant antibody fragments and fusions mediated plant disease resistance against fungi
AU2010204809B2 (en) Targeted cryptosporidium biocides
CN101277970B (en) Improved chimeric toxin receptor proteins and chimeric toxin receptor proteins for treatment and prevention of anthrax
US20070031440A1 (en) Modified transferin-antibody fusion proteins
EP3155006B1 (en) Human cytolytic fusion proteins

Legal Events

Date Code Title Description
MK5 Application lapsed section 142(2)(e) - patent request and compl. specification not accepted