AU758820B2 - Soluble recombinant botulinum toxin proteins - Google Patents

Soluble recombinant botulinum toxin proteins Download PDF

Info

Publication number
AU758820B2
AU758820B2 AU63043/99A AU6304399A AU758820B2 AU 758820 B2 AU758820 B2 AU 758820B2 AU 63043/99 A AU63043/99 A AU 63043/99A AU 6304399 A AU6304399 A AU 6304399A AU 758820 B2 AU758820 B2 AU 758820B2
Authority
AU
Australia
Prior art keywords
toxin
protein
difficile
recombinant
igy
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.)
Ceased
Application number
AU63043/99A
Other versions
AU6304399A (en
Inventor
Joseph R Firca
John A Kink
Nisha V Padhye
Douglas C Stafford
Bruce S Thalley
James A. Williams
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.)
Allergan Inc
Allergan Botox Ltd
Original Assignee
Allergan Inc
Allergan Botox Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority claimed from AU39683/95A external-priority patent/AU709586B2/en
Application filed by Allergan Inc, Allergan Botox Ltd filed Critical Allergan Inc
Priority to AU63043/99A priority Critical patent/AU758820B2/en
Publication of AU6304399A publication Critical patent/AU6304399A/en
Assigned to ALLERGAN BOTOX LIMITED, ALLERGAN, INC. reassignment ALLERGAN BOTOX LIMITED Alteration of Name(s) of Applicant(s) under S113 Assignors: OPHIDIAN PHARMACEUTICALS, INC.
Priority to AU2002317523A priority patent/AU2002317523B2/en
Application granted granted Critical
Publication of AU758820B2 publication Critical patent/AU758820B2/en
Anticipated expiration legal-status Critical
Ceased legal-status Critical Current

Links

Classifications

    • 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

  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Preparation Of Compounds By Using Micro-Organisms (AREA)

Description

Method of Treatment and Prevention of C. botulinum Disease FIELD OF THE INVENTION The present invention relates to clostridial antitoxin and vaccine therapy for humans and other animals. Antitoxins which neutralize the pathologic effects of clostridial toxins are provided. Vaccines which prevent the morbidity and mortality associated with clostridial diseases are provided.
BACKGROUND OF THE INVENTION The genus Clostridium is comprised of gram-positive, anaerobic. spore-forming bacilli.
The natural habitat of these organisms is the environment and the intestinal tracts of humans and other animals. Indeed, clostridia are ubiquitous: they are commonly found in soil, dust.
sewage, marine sediments. decaying vegetation, and mud. [See P.H.A. Sneath et al..
"Clostridium." Bergey's Manual® of Systematic Bacteriology, Vol. 2. pp. 1141-1200, Williams Wilkins (1986).] Despite the identification of approximately 100 species of Clostridium. only a small number have been recognized as etiologic agents of medical and veterinary importance. Nonetheless, these species are associated with very serious diseases.
including botulism. tetanus, anaerobic cellulitis, gas gangrene. bacteremia, pseudomembranous 20 colitis, and clostridial gastroenteritis. Table I lists some of the species of medical and veterinary importance and the diseases with which they are associated. As virtually all of i these species have been isolated from fecal samples of apparently healthy persons, some of these isolates may be transient rather than permanent residents of the colonic flora.
TABLE 1 Clostridium Species of Medical and Veterinary Importance* Species Disease C. aminovalericum Bacteriuria (pregnant women) Infected wounds; Bacteremia: Botulism; Infections of C. argentinense amniotic fluid Infected war wounds: Peritonitis: Infectious processes of C. baratii the eye. ear and prostate C. bei/erinckikii Infected wounds C. bifermentans Infected wounds: Abscesses: Gas Gangrene: Bacteremia TABLE 1 Clostridium Species of Medical and Veterinary Importance* St ecies Snecies Disease o o o 0 0 0 0 0 o C. botulinum Food poisoning: Botulism (wound. food. infant) Urinary tract. lower respiratory tract pleural cavity, and C. butvricum abdominal infections: Infected wounds: Abscesses; Bacteremia C. cadaveris Abscesses; Infected wounds C carnis Soft tissue infections: Bacteremia C. chauvoei Blackleg Abdominal. cervical, scrotal. pleural. and other infections: C. clostridioforme Septicemia: Peritonitis: Appendicitis Isolated from human disease processes. but role in disease C cochlearium unknown.
Antimicrobial-associated diarrhea: Pseudomembranous C. difficile enterocolitis: Bacteremia: Pyogenic infections C. fallax Soft tissue infections C ghnoii Soft tissue infections C. glycolicum Wound infections: Abscesses: Peritonitis C hastiforme Infected war wounds: Bacteremia: Abscesses Infected war wounds: Gas gangrene: Gingival plaque C. histolyticum isolate C. indolis Gastrointestinal tract infections C. innocuum Gastrointestinal tract infections: Empyema C irreulare C leptum C. limosum C. malenominatum *1 Penile lesions Isolated from human disease processes. but role in disease unknown.
Bacteremia: Peritonitis; Pulmonary infections Various infectious processes Infected wounds: Gas gangrene: Blackleg. Big head (ovine); Redwater disease (bovine) Urinary tract infections: Rectal abscesses Bacteremia: Peritonitis: Infected wounds: Appendicitis C. novyi C. oroticum C. paraputrificumn -2- TABLE 1 Clostridium Species of Medical and Veterinary Importance* Species Disease Gas gangrene: Anaerobic cellulitis: Intra-abdominal abscesses: Soft tissue infections: Food poisoning; C perfringens Necrotizing pneumonia: Empyema: Meningitis: Bacteremia: Uterine Infections; Enteritis necrotans: Lamb dysentery: Struck: Ovine Enterotoxemia: C. putrefaciens Bacteriuria (Pregnant women with bacteremia) C. putrificum Abscesses: Infected wounds: Bacteremia Infections of the abdominal cavity, genital tract, lung, and C. ramosum biliary tract: Bacteremia Isolated from human disease processes. but role in disease C sarnagforme unknown.
Gas gangrene: Bacteremia; Suppurative infections; C. septicum Necrotizing enterocolitis: Braxy Gas gangrene: Wound infections: Penile lesions; C. sordellii Bacteremia: Abscesses: Abdominal and vaginal infections Appendicitis; Bacteremia: Bone and soft tissue infections; C. sphenoides Intraperitoneal infections: Infected war wounds: Visceral gas gangrene: Renal abscesses Gas gangrene: Bacteremia: Endocarditis: central nervous C. sporogenes system and pleuropulmonary infections: Penile lesions: Infected war wounds: Other pyogenic infections Bacteremia: Empyema: Biliary tract. soft tissue and bone C. subterminale infections Liver abscesses: Bacteremia: Infections resulting due to C. svmbiosum bowel flora Gas gangrene: Appendicitis: Brain abscesses; Intestinal C. tertium tract and soft tissue infections: Infected war wounds; Periodontitis: Bacteremia Tetanus: Infected gums and teeth; Corneal ulcerations; Mastoid and middle ear infections: Intraperitoneal infections; Tetanus neonatorum: Postpartum uterine infections; Soft tissue infections, especially related to trauma (including abrasions and lacerations); Infections related to use of contaminated needles C. tetani
I
C. thermosaccharolyticum Isolated from human disease processes. but role in disease unknown.
-r Compiled from P.G. Engelkirk et al. "Classification". Principles and Practice of Clinical Anaerobic Bacteriology, pp. 22-23. Star Publishing Co., Belmont. CA (1992); J. Stephen and R.A. Petrowski. "Toxins Which Traverse Membranes and Deregulate Cells." in Bacterial Toxins. 2d ed., pp. 66-67, American Society for Microbiology (1986); R. Berkow and A.J. Fletcher "Bacterial Diseases." Merck Manual of Diagnosis and Therapy, 16th ed., pp. 116-126, Merck Research Laboratories, Rahway, N.J. (1992); and O.H. Sigmund and C.M. Fraser "Clostridial Infections." Merck Veterinary Manual. 5th ed., pp. 396-409. Merck Co.. Rahway. N.J. (1979).
In most cases. the pathogenicity of these organisms is related to the release of powerful exotoxins or highly destructive enzymes. Indeed. several species of the genus Clostridium produce toxins and other enzymes of great medical and veterinary significance.
Hatheway. Clin. Microbiol. Rev. 3:66-98 (1990).] Perhaps because of their significance for human and veterinary medicine, much 15 research has been conducted on these toxins, in particular those of C. botulinum and C.
difficile.
C. botulinum Several strains of Clostridium botulinum produce toxins of significance to human and 20 animal health. Hatheway, Clin. Microbiol. Rev. 3:66-98 (1990).] The effects of these toxins range from diarrheal diseases that can cause destruction of the colon, to paralytic effects that can cause death. Particularly at risk for developing clostridial diseases are neonates and humans and animals in poor health those suffering from diseases associated with old age or immunodeficiency diseases).
25 Clostridium hotulinum produces the most poisonous biological toxin known. The lethal human dose is a mere 10" mg/kg bodyweight for toxin in the bloodstream. Botulinal toxin blocks nerve transmission to the muscles, resulting in flaccid paralysis. When the toxin reaches airway and respiratory muscles, it results in respiratory failure that can cause death.
Arnon, J. Infect. Dis. 154:201-206 (1986).] C. botulinum spores are carried by dust and are found on vegetables taken from the soil. on fresh fruits, and on agricultural products such as honey. Under conditions favorable to the organism, the spores germinate to vegetative cells which produces toxin. Arnon.
Ann. Rev. Med. 31:541 (1980).] -4- Botulism disease may be grouped into four types. based on the method of introduction of toxin into the bloodstream. Food-borne botulism results from ingesting improperly preserved and inadequately heated food that contains botulinal toxin. There were 355 cases of food-borne botulism in the United States between 1976 and 1984. MacDonald et al., Am. J. Epidemiol. 124:794 (1986).] The death rate due to botulinal toxin is 12% and can be higher in particular risk groups. Tacket et al. Am. J. Med. 76:794 (1984).] Woundinduced botulism results from C. botulinum penetrating traumatized tissue and producing toxin that is absorbed into the bloodstream. Since 1950. thirty cases of wound botulism have been reported. Swartz. "Anaerobic Spore-Forming Bacilli: The Clostridia." pp. 633-646, in B.D. Davis et Microbiology, 4th edition. J.B_ Lippincott Co. (1990).] Inhalation botulism results when the toxin is inhaled. Inhalation botulism has been reported as the result of accidental exposure in the laboratory Holzer. Med. Klin. 41:1735 (1962)] and could arise if the toxin is used as an agent of biological warfare Franz et al.. in Botulinum and Tetanus Neurotoxins. B.R. DasGupta. ed.. Plenum Press. New York (1993). pp. 473-476].
15 Infectious infant botulism results from C. botulinum colonization of the infant intestine with production of toxin and its absorption into the bloodstream. It is likely that the bacterium gains entry when spores are ingested and subsequently germinate. Amon. J. Infect. Dis.
154:201 (1986).] There have been 500 cases reported since it was first recognized in 1976.
Swartz. supra.] 20 Infant botulism strikes infants who are three weeks to eleven months old (greater than of the cases are infants less than six months). Arnon. J. Infect. Dis. 154:201 (1986).] It is believed that infants are susceptible. due. in large part. to the absence of the full adult complement of intestinal microflora. The benign microflora present in the adult intestine provide an acidic environment that is not favorable to colonization by C botulinum. Infants begin life with a sterile intestine which is gradually colonized by microflora. Because of the limited microflora present in early infancy, the intestinal environment is not as acidic, allowing for C botulinum spore germination, growth, and toxin production. In this regard, some adults who have undergone antibiotic therapy which alters intestinal microflora become more susceptible to botulism.
An additional factor accounting for infant susceptibility to infectious botulism is the immaturity of the infant immune system. The mature immune system is sensitized to bacterial antigens and produces protective antibodies. Secretory IgA produced in the adult intestine has the ability to agglutinate vegetative cells of C. botulinum. Amon. J. Infect.
Dis. 154:201 (1986).] Secretory IgA may also act by preventing intestinal bacteria and their products from crossing the cells of the intestine. Arnon. Epidemiol. Rev. 3:45 (1981).] The infant immune system is not primed to do this.
Clinical symptoms of infant botulism range from mild paralysis, to moderate and severe paralysis requiring hospitalization, to fulminant paralysis. leading to sudden death. [S.
Amon. Epidemiol. Rev. 3:45 (1981).] The chief therapy for severe infant botulism is ventilatory assistance using a mechanical respirator and concurrent elimination of toxin and bacteria using cathartics.
enemas. and gastric lavage. There were 68 hospitalizations in California for infant botulism in a single year with a total cost of over $4 million for treatment. Frankovich and S.
Anon. West. J. Med. 154:103 (1991).] Different strains of Clostridium botulinum each produce antigenically distinct toxin designated by the letters A-G. Serotype A toxin has been implicated in 26% of the cases of food botulism: types B. E and F have also been implicated in a smaller percentage of the food botulism cases Sugiyama. Microbiol. Rev. 44:419 (1980)]. Wound botulism has been "i 20 reportedly caused by only types A or B toxins Sugiyama. supra]. Nearly all cases of infant botulism have been caused by bacteria producing either type A or type B toxin.
(Exceptionally. one New Mexico case was caused by Clostridium hotulinum producing type F toxin and another by Clostridium botulinum producing a type B-type F hybrid.) Anon.
Epidemiol. Rev. 3:45 (1981).] Type C toxin affects waterfowl. cattle. horses and mink. Type D toxin affects cattle. and type E toxin affects both humans and birds.
A trivalent antitoxin derived from horse plasma is commercially available from Connaught Industries Ltd. as a therapy for toxin types A. B, and E. However, the antitoxin has several disadvantages. First. extremely large dosages must be injected intravenously and/or intramuscularly. Second. the antitoxin has serious side effects such as acute anaphylaxis which can lead to death, and serum sickness. Finally. the efficacy of the antitoxin is uncertain and the treatment is costly. Tacket et al.. Am. J. Med. 76:794 (1984).] A heptavalent equine botulinal antitoxin which uses only the F(ab')2 portion of the antibody molecule has been tested by the United States Military. Balady, USAMRDC Newsletter, p. 6 (1991).] This was raised against impure toxoids in those large animals and is not a high titer preparation.
A pentavalent human antitoxin has been collected from immunized human subjects for use as a treatment for infant botulism. The supply of this antitoxin is limited and cannot be expected to meet the needs of all individuals stricken with botulism disease. In addition, collection of human sera must involve screening out HIV and other potentially serious human pathogens. Schwarz and S.S. Amon. Western J. Med. 156:197 (1992).] "0 Infant botulism has been implicated as the cause of mortality in some cases of Sudden Infant Death Syndrome (SIDS. also known as crib death). SIDS is officially recognized as 'infant death that is sudden and unexpected and that remained unexplained despite complete post-mortem examination. The link of SIDS to infant botulism came when fecal or blood specimens taken at autopsy from SIDS infants were found to contain C botulinum organisms and/or toxin in 3-4% of cases analyzed. Peterson et al., Rev. Infect. Dis. 1:630 (1979).] In contrast. only I of 160 healthy infants had C. botulinum organisms in the feces and no botulinal toxin. Anon et al.. Lancet, pp. 1273-76. June 17. 1978.) 0: 20 In developed countries. SIDS is the number one cause of death in children between one month and one year old. Anon et al.. Lancet, pp. 1273-77. June 17. 1978.) More children die from SIDS in the first year than from any other single cause of death in the first fourteen years of life. In the United States, there are 8.000-10,000 SIDS victims annually.
Id.
What is needed is an effective therapy against infant botulism that is free of dangerous side effects, is available in large supply at a reasonable price, and can be safely and gently delivered so that prophylactic application to infants is feasible.
Immunization of subjects with toxin preparations has been done in an attempt to induce immunity against botulinal toxins. A botulinum vaccine comprising chemically inactivated formaldehyde-treated) type A. B.C. D and E toxin is commercially available for human usage. However, this vaccine preparation has several disadvantages. First, the efficacy of this vaccine is variable (in particular. only 78% of recipients produce protective levels of anti-type B antibodies following administration of the primary series). Second, immunization is painful (deep subcutaneous inoculation is required for administration), with adverse reactions being common (moderate to severe local reactions occur in approximately 6% of recipients upon initial injection: this number rises to approximately 11% of individuals who receive booster injections) [Informational Brochure for the Pentavalent (ABCDE) Botulinum Toxoid. Centers for Disease Control]. Third, preparation of the vaccine is dangerous as active toxin must be handled by laboratory workers.
What is needed are safe and effective vaccine preparations for administration to those at risk of exposure to C botulinum toxins.
C. difficile C difficile. an organism which gained its name due to difficulties encountered in its isolation. has recently been proven to be an etiologic agent of diarrheal disease. (Sneath et al.. p. 1165.). C difficile is present in the gastrointestinal tract of approximately 3% of healthy adults. and 10-30% of neonates without adverse effect (Swartz. at p. 644); by other estimates. C difficile is a part of the normal gastrointestinal flora of 2-10% of humans. [G.F.
Brooks et al.. (eds.) "Infections Caused by Anaerobic Bacteria." Jawetz. Melnick. 20 Adelberg s Medical Microbiology, 19th ed.. pp. 257-262. Appleton Lange. San Mateo. CA (1991).] As these organisms are relatively resistant to most commonly used antimicrobials.
when a patient is treated with antibiotics. the other members of the normal gastrointestinal flora are suppressed and C. difficile flourishes, producing cytopathic toxins and enterotoxins.
It has been found in 25% of cases of moderate diarrhea resulting from treatment with antibiotics. especially the cephalosporins. clindamycin. and ampicillin. Swartz at 644.] Importantly, C difficile is commonly associated with nosocomial infections. The organism is often present in the hospital and nursing home environments and may be carried on the hands and clothing of hospital personnel who care for debilitated and immunocompromised patients. As many of these patients are being treated with 8 antimicrobials or other chemotherapeutic agents. such transmission of C. difficile represents a significant risk factor for disease. (Engelkirk et al., pp. 64-67.) C difficile is associated with a range of diarrhetic illness, ranging from diarrhea alone to marked diarrhea and necrosis of the gastrointestinal mucosa with the accumulation of inflammatory cells and fibrin. which forms a pseudomembrane in the affected area. (Brooks et al.) It has been found in over 95% of pseudomembranous enterocolitis cases. (Swartz. at p. 644.) This occasionally fatal disease is characterized by diarrhea, multiple small colonic plaques, and toxic megacolon. (Swartz. at p. 644.) Although stool cultures are sometimes used for diagnosis, diagnosis is best made by detection of the heat labile toxins present in fecal filtrates from patients with enterocolitis due to C difficile. (Swartz. at p. 644-645; and S' Brooks et al.. at p. 260.) C. difficile toxins are cytotoxic for tissue/cell cultures and cause enterocolitis when injected intracecally into hamsters. (Swartz. at p. 644.) The enterotoxicity of C. difficile is primarily due to the action of two toxins, designated A and B. each of approximately 300.000 in molecular weight. Both are potent cytotoxins. with toxin A possessing direct enterocytotoxic activity. [Lyerly et al.. Infect.
Immun. 60:4633 (1992).] Unlike toxin A of C. perfringens. an organism rarely associated with antimicrobial-associated diarrhea. the toxin of C difficile is not a spore coat constituent and is not produced during sporulation. (Swartz. at p. 644.) C difficile toxin A causes :i hemorrhage. fluid accumulation and mucosal damage in rabbit ileal loops and appears to increase the uptake of toxin B by the intestinal mucosa. Toxin B does not cause intestinal fluid accumulation, but it is 1000 times more toxic than toxin A to tissue culture cells and i causes membrane damage. Although both toxins induce similar cellular effects such as actin disaggregation. differences in cell specificity occurs.
Both toxins are important in disease. [Borriello et al.. Rev. Infect. Dis.. 12(suppl.
2):S185 (1990); Lyerly et al.. Infect. Immun.. 47:349 (1985): and Rolfe. Infect. Immun..
59:1223 (1990).] Toxin A is inought to act first by binding to brush border receptors, destroying the outer mucosal layer, then allowing toxin B to gain access to the underlying tissue. These steps in pathogenesis would indicate that the production of neutralizing antibodies against toxin A may be sufficient in the prophylactic therapy of CDAD. However.
antibodies against toxin B may be a necessary additional component for an effective -9therapeutic against later stage colonic disease. Indeed. it has been reported that animals require antibodies to both toxin A and toxin B to be completely protected against the disease.
[Kim and Rolfe, Abstr. Ann. Meet. Am. Soc. Microbiol., 69:62 (1987).] C difficile has also been reported to produce other toxins such as an enterotoxin different from toxins A and B [Banno et al.. Rev. Infect. Dis.. 6(Suppl. 1:S 1-S20 (1984)], a low molecular weight toxin [Rihn et al.. Biochem. Biophys. Res. Comm.. 124:690-695 (1984)], a motility aTtering factor [Justus et al.. Gastroenterol., 83:836-843 (1982)], and perhaps other toxins. Regardless. C. difficile gastrointestinal disease is of primary concern.
It is significant that due to its resistance to most commonly used antimicrobials, C.
S 10 difficile is associated with antimicrobial therapy with virtually all antimicrobial agents (although most commonly ampicillin. clindamycin and cephalosporins). It is also associated with disease in patients undergoing chemotherapy with such compounds as methotrexate. fluorouracil. cyclophosphamide. and doxorubicin. Finegold et al.. Clinical Guide to Anaerobic Infections. pp. 88-89. Star Publishing Co.. Belmont. CA (1992).] 15 Treatment of C difficile disease is problematic, given the high resistance of the organism. Oral metronidazole. bacitracin and vancomycin have been reported to be effective.
(Finegold et al.. p. 89.) However there are problems associated with treatment utilizing these compounds. Vancomycin is very expensive, some patients are unable to take oral medication.
and the relapse rate is high although it may not occur for several weeks. Id C. difficile disease would be prevented or treated by neutralizing the effects of these toxins in the gastrointestinal tract. Thus. what is needed is an effective therapy against C.
difficile toxin that is free of dangerous side effects, is available in large supply at a reasonable price, and can be safely delivered so that prophylactic application to patients at risk of developing pseudomembranous enterocolitis can be effectively treated.
DESCRIPTION OF THE DRAWINGS Figure 1 shows the reactivity of anti-C. botulinum IgY by Western blot.
Figure 2 shows the IgY antibody titer to C botulinum type A toxoid in eggs, measured by ELISA.
Figure 3 shows the results of C. difficile toxin A neutralization assays.
Figure 4 shows the results of C. difficile toxin B neutralization assays.
Figure 5 shows the results of C difficile toxin B neutralization assays.
Figure 6 is a restriction map of C difficile toxin A gene, showing sequences of primers 1-4 (SEQ ID NOS:1-4).
Figure 7 is a Western blot of C. difficile toxin A reactive protein.
Figure 8 shows C. difficile toxin A expression constructs.
Figure 9 shows C difficile toxin A expression constructs.
Figure 10 shows the purification of recombinant C. difficile toxin A.
Figure 11 shows the results of C. difficile toxin A neutralization assays with antibodies S" 10 reactive to recombinant toxin A.
Figure 12 shows the results for.a C difficile toxin A neutralization plate.
Figure 13 shows the results for a C difficile toxin A neutralization plate.
Figure 14 shows the results of recombinant C difficile toxin A neutralization assays.
Figure 15 shows C. difficile toxin A expression constructs.
15 Figure 16 shows a chromatograph plotting absorbance at 280 nm against retention time for a pMA1870-680 IgY PEG preparation.
Figure 17 shows two recombinant C. difficile toxin B expression constructs.
Figure 18 shows C difficile toxin B expression constructs.
"i Figure 19 shows C difficile toxin B expression constructs.
Figure 20 shows C. difficile toxin B expression constructs.
Figure 21 is an SDS-PAGE gel showing the purification of recombinant C difficile toxin B fusion protein.
Figure 22 is an SDS-PAGE gel showing the purification of two histidine-tagged recombinant C. difficile toxin B proteins.
Figure 23 shows C. difficile toxin B expression constructs.
Figure 24 is a Western blot of C. difficile toxin B reactive protein.
Figure 25 shows C. botulinum type A toxin expression constructs: constructs used to provide C. botulinum or C. difficile sequences are also shown.
Figure 26 is an SDS-PAGE gel stained with Coomassie blue showing the purification of recombinant C botulinum type A toxin fusion proteins.
11 Figure 27 shows C boiulinum type A toxin expression constructs: constructs used to provide C bolulinum sequences are also shown.
Figure 28 is an SDS-PAGE gel stained with Coomassie blue showing the purification of pHisBot protein using the Ni-NTA resin.
Figure 29 is an SDS-PAGE gel stained with Coomassie blue showing the expression of pHisBot protein in BL21(DE3) and BL21(DE3)pLysS host cells.
Figure 30 is an SDS-PAGE gel stained with Coomassie blue showing the purification of pHisBot protein using a batch absorption procedure.
Figure 31 shows C. difficile toxin A expression constructs.
10 Figure 32 shows an SDS-PAGE gel stained with Coomassie blue and a Western blot showing the expression of the pUC1960-2680 in E. coli host cells.
Figure 33 shows an SDS-PAGE gel stained with Coomassie blue and a Western blot showing the expression of the several recombinant C. difficile toxin A fusion proteins in E.
coli host cells.
Figure 34 is an SDS-PAGE gel stained with Coomassie blue showing the purification of recombinant C difficile toxin A and B fusion proteins.
Figure 35 shows the results of a prophylactic treatment study in hamsters.
Figure 36 shows the results of a therapeutic treatment study in hamsters.
Figure 37 shows the results of a therapeutic treatment study in hamsters.
Figure 38 shows the results of a therapeutic treatment study in hamsters.
Figure 39 shows the results of administration of vancomycin to hamsters having an established C. difficile infection.
Figure 40 shows the results of an ELISA analysis of IgY isolated from hens immunized with the recombinant C. difficile toxin A protein pMA1870-2680 and four different adjuvants.
Figure 41 shows the results of an ELISA analysis of IgY isolated from hens immunized with the recombinant C. difficile toxin A protein pPAI870-2680(N/C) and four different adjuvants.
Figure 42 shows dissolution profiles for Aquateric-coated IgY.
Figure 43 shows dissolution profiles for Eugragit®-coated IgY.
12 Figure 44 shows the results of an ELISA analysis of IgY isolated from hamsters vaccinated with recombinant C. difficile toxin A proteins.
Figure 45 shows the results of an ELISA analysis of IgY isolated from hamsters vaccinated with recombinant C difficile toxin A and B proteins: reactivity to recombinant C.
difficile toxin A is shown.
Figure 46 shows the results of an ELISA analysis of IgY isolated from hamsters vaccinated with recombinant C. difficile toxin A and B proteins: reactivity to recombinant C.
difficile toxin B is shown.
Figure 47 shows results of a therapeutic treatment study in hamsters.
10 Figure 48 shows results of a therapeutic treatment study in diarrhetic hamsters.
Figure 49 shows results of a therapeutic treatment study in hamsters.
Figure 50 shows a Western blot showing C difficile toxin A levels in culture S* supernatant. column flow through and column eluate from an affinity purification column.
Figure 51 shows a Western blot showing C difficile toxin A levels in culture 15 supernatant. column flow through and column eluate from an affinity purification column.
Figure 52 is a native PAGE gel stained with Coomassie blue showing C. difficile toxin B levels in liquid culture supematant.
Figure 53 is a native PAGE gel stained with Coomassie blue and a Western blot showing C dificile toxin B levels in dialysis bag cultures.
Figure 54 is a native PAGE gel stained with Coomassie blue and a Western blot showing C difficile toxin B levels in a commercial toxin B preparation and column flow through and column eluate from an affinity purification column.
Figure 55 shows the dissolution profiles of IgY tablets overcoated with a pH-sensitive enteric film.
Figure 56 shows the stability of the C difficile toxin A IgY reactivity after the tableting and enteric overcoating process.
Figure 57 shows mortality of hamsters prophylactically treated with IgY and infected with C. difficile toxin A.
Figure 58 shows mortality of hamsters therapeutically treated with IgY after infection with C difficile toxin A.
13
DEFINITIONS
To facilitate understanding of the invention, a number of terms are defined below.
As used herein, the term "neutralizing" is used in reference to antitoxins, particularly antitoxins comprising antibodies. which have the ability to prevent the pathological actions of the toxin against which the antitoxin is directed.
As used herein, the term "overproducing" is used in reference to the production of clostridial toxin polypeptides in a host cell and indicates that the host cell is producing more of the clostridial toxin by virtue of the introduction of nucleic acid sequences encoding said clostridial toxin polypeptide than would be expressed by said host cell absent the introduction S 10 of said nucleic acid sequences. To allow ease of purification of toxin polypeptides produced in a host cell it is preferred that the host cell express or overproduce said toxin polypeptide at a level greater than 1 me/liter of host cell culture.
As used herein, the term "fusion protein" refers to a chimeric protein containing the protein of interest C difficile toxin A or B and fragments thereof) joined to an o• 15 exogenous protein fragment (the fusion partner which consists of a non-toxin protein). The fusion partner may enhance solubility of the C difficile protein as expressed in a host cell.
may provide an affinity tag to allow purification of the recombinant fusion protein from the host cell or culture supernatant. or both. If desired. the fusion protein may be removed from the protein of interest toxin protein or fragments thereof) prior to immunization by a variety of enzymatic or chemical means known to the art.
As used herein the term "non-toxin protein" or "non-toxin protein sequence" refers to that portion of a fusion protein which comprises a protein or protein sequence which is not derived from a bacterial toxin protein.
The term "protein of interest" as used herein refers to the protein whose expression is desired within the fusion protein. In a fusion protein the protein of interest will be joined or fused with another protein or prctein domain, the fusion partner. to allow for enhanced stability of the protein of interest and/or ease of purification of the fusion protein.
As used herein, the term "maltose binding protein" refers to the maltose binding protein of E. coli. A portion of the maltose binding protein may be added to a protein of interest to generate a fusion protein: a portion of the maltose binding protein may merely 14 enhance the solubility of the resulting fusion protein when expressed in a bacterial host. On the other hand. a portion of the maltose binding protein may allow affinity purification of the fusion protein on an amylose resin.
As used herein, the term "poly-histidine tract" when used in reference to a fusion protein refers to the presence of two to ten histidine residues at either the amino- or carboxyterminus or both termini of a protein of interest or a fusion partner. A poly-histidine tract of six to ten residues is preferred. The poly-histidine tract is also defined functionally as being a number of consecutive histidine residues added to the protein of interest which allows the affinity purification of the resulting fusion protein on a nickel-chelate column.
The term "thioredoxin protein" when used in reference to a fusion protein refers to a the thioredoxin protein of E. coli It is noted that the invention is not limited by the source of the thioredoxin protein, while the E. coli thioredoxin protein is particularly preferred, thioredoxin proteins may be obtained from several sources. A portion of the thioredoxin protein may be added to a protein of interest to generate a fusion protein: a portion of the 15 thioredoxin protein may enhance the solubility of the resulting fusion protein when expressed in a bacterial host.
As used herein, the term "purified" or "to purify" refers to the removal of contaminants from a sample. For example. antitoxins are purified by removal of i contaminating non-immunoglobulin proteins: they are also purified by the removal of immunoglobulin that does not bind toxin. The removal of non-immunoglobulin proteins and/or the removal of immunoglobulins that do not bind toxin results in an increase in the percent of toxin-reactive immunoglobulins in the sample. The purification of antitoxin may be accomplished by a variety of means including the extraction and precipitation of avian antitoxin from eggs using polyethylene glycol. Purification of anticlostridal antitoxin may also be accomplished by affinity chromatography on a resin comprising a portion of a clostridial toxin protein. In another example, recombinant toxin polypeptides are expressed in bacterial host cells and the toxin polypeptides are purified by the removal of host cell proteins: the percent of recombinant toxin polypeptides is thereby increased in the sample.
Additionally. the recombinant toxin polypeptides are purified by the removal of host cell components such as lipopolysaccharide endotoxin).
The term "recombinant DNA molecule" as used herein refers to a DNA molecule which is comprised of segments of DNA joined together by means of molecular biological techniques.
The term "recombinant protein" or "recombinant polypeptide" as used herein refers to a protein molecule which is expressed from a recombinant DNA molecule.
The term "native protein" as used herein refers to a protein which is isolated from a natural source as opposed to the production of a protein by recombinant means.
As used herein the term "portion" when in reference to a protein (as in "a portion of a given protein") refers to fragments of that protein. The fragments may range in size from four amino acid residues to the entire amino acid sequence minus one amino acid.
As used herein "soluble" when in reference to a protein produced by recombinant DNA technology in a host cell is a protein which exists in solution in the cytoplasm of the host cell: if the protein contains a signal sequence the soluble protein is exported to the periplasmic space in bacteria hosts and is secreted into the culture medium in eucaryotic cells 15 capable of secretion or by bacterial host possessing the appropriate genes the kil gene).
In contrast. an insoluble protein is one which exists in denatured form inside cytoplasmic granules (called an inclusion bodies) in the host cell. High level expression greater than 10-20 mg recombinant protein/liter of bacterial culture) of recombinant proteins often results i in the expressed protein being found in inclusion bodies in the bacterial host cells. A soluble protein is a protein which is not found in an inclusion body inside the host cell or is found both in the cytoplasm and in inclusion bodies and in this case the protein may be present at high or low levels in the cytoplasm.
A distinction is drawn between a soluble protein a protein which when expressed in a host cell is produced in a soluble form) and a "solubilized" protein. An insoluble recombinant protein found inside an inclusion body may be solubilized rendered into a soluble form) by treating purified inclusion bodies with denaturants such as guanidine hydrochloride, urea or sodium dodecyl sulfate (SDS). These denaturants must then be removed from the solubilized protein preparation to allow the recovered protein to renature (refold). Not all proteins will refold into an active conformation after solubilization in a denaturant and removal of the denaturant. Many proteins precipitate upon removal of the 16 denaturant. SDS may be used to solubilize inclusion bodies and will maintain the proteins in solution at low concentration. However, dialysis will not always remove all of the SDS (SDS can form micelles which do not dialyze out); therefore, SDS-solubilized inclusion body protein is soluble but not refolded.
A distinction is drawn between proteins which are soluble dissolved) in a solution devoid of significant amounts of ionic detergents SDS) or denaturants urea. guanidine hydrochloride) and proteins which exist as a suspension of insoluble protein molecules dispersed within the solution. A soluble protein will not be removed from a solution containing the protein by centrifugation using conditions sufficient to remove bacteria 10 present in a liquid medium centrifugation at 5.000 x g for 4-5 minutes). For example, to test whether two proteins, protein A and protein B. are soluble in solution, the two proteins 0* are placed into a solution selected from the group consisting of PBS-NaCI (PBS containing S. 0.5 M NaCI). PBS-NaCI containing 0.2% Tween 20. PBS. PBS containing 0.2% Tween PBS-C (PBS containing 2 mM CaCI). PBS-C containing either 0.1 or 0.5 Tween 20. PBS- 15 C containing either 0.1 or 0.5% NP-40, PBS-C containing either 0.1 or 0.5% Triton X-100, PBS-C containing 0.1% sodium deoxycholate. The mixture containing proteins A and B is then centrifuged at 5000 x g for 5 minutes. The supernatant and pellet formed by centrifuation are then assayed for the presence of protein A and B. If protein A is found in the supernatant and not in the pellet [except for minor amounts less than 10%) as a result of trapping.] protein is said to be soluble in the solution tested. If the majority of protein B is found in the pellet greater than then protein B is said to exist as a suspension in the solution tested.
As used herein, the term "therapeutic amount" refers to that amount of antitoxin required to neutralize the pathologic effects of one or more clostridial toxins in a subject.
The term "therapeutic mixture" when used in reference to a mixture of antitoxins refers to that amount of antitoxin required neutralize the pathologic effects of one or more clostridial toxins in a subject.
The term "therapeutic vaccine" when used in reference to a vaccine comprising one or more recombinant clostridial toxin fusion proteins means that the vaccine contains an immunologically-effective amount of the fusion proteins the immunogens).
17 As used herein the term "immunogenically-effective amount" refers to that amount of an immunogen required to invoke the production of protective levels of antibodies in a host a subject) upon vaccination.
The term "pyrogen" as used herein refers to a fever-producing substance. Pyrogens may be endogenous to the host prostaglandins) or may be exogenous compounds bacterial endo- and exotoxins. nonbacterial compounds such as antigens and certain steroid compounds, etc.). The presence of pyrogen in a pharmaceutical solution may be detected using the U.S. Pharmacopeia (USP) rabbit fever test (United States Pharmacopeia. Vol. XXII (1990) United States Pharmacopeial Convention. Rockville. MD, p. 151).
10 The term "endotoxin" as used herein refers to the high molecular weight complexes associated with the outer membrane of gram-negative bacteria. Unpurified endotoxin contains lipids, proteins and carbohydrates. Highly purified endotoxin does not contain protein and is referred to as lipopolysaccharide (LPS). Because unpurified endotoxin is of concern in the production of pharmaceutical compounds proteins produced in E. coli using recombinant 15 DNA technology), the term endotoxin as used herein refers to unpurified endotoxin. Bacterial endotoxin is a well known pyrogen.
As used herein, the term "endotoxin-free" when used in reference to a composition to be administered parenterally (with the exception of intrathecal administration) to a host means that the dose to be delivered contains less than 5 EU/kg body weight [FDA Guidelines for Parenteral Drugs (December 1987)]. Assuming a weight of 70 kg for an adult human, the dose must contain less than 350 EU to meet FDA Guidelines for parenteral administration.
Endotoxin levels are measured herein using the Limulus Amebocyte Lysate (LAL) test (Limulus Amebocyte Lysate Pyrochrome
T
Associates of Cape Cod. Inc. Woods Hole. MA).
To measure endotoxin levels in preparations of recombinant proteins. 0.5 ml of a solution comprising 0.5 mg of purified recombinant protein in 50 mM NaPO,, pH 7.0. 0.3M NaCI and glycerol is used in the LAL assay according to the manufacturer's instructions for the endpoint chromogenic without diazo-coupling method. Compositions containing less than or equal to 450 endotoxin units (EU)/mg of purified recombinant protein are herein defined as "substantially endotoxin-free." Typically. administration of bacterial toxins or toxoids to adult humans for the purpose of vaccination involves doses of about 10-500 ig protein/dose.
18 Therefore. administration of 10-500 pg of a purified recombinant protein to a 70 kg human.
wherein said purified recombinant protein preparation contains 450 EU/mg protein, results in the introduction of only 4.5 to 225 EU 1.3 to 64.5% of the maximum allowable endotoxin burden per parenteral dose).
The LAL test is accepted by the U.S. FDA as a means of detecting bacterial endotoxins (21 C.F.R. 660.100 -105). Studies have shown that the LAL test is equivalent or superior to the USP rabbit pyrogcn test for the detection of endotoxin and thus the LAL test can be used as a surrogate for pyrogenicity studies in animals Perason. Pyrogens: endotoxins. LAL testing and depyrogenation. Marcel Dekker. New York (1985), pp.150-15 5 The FDA Bureau of Biologics accepts the LAL assay in place of the USP rabbit pyrogen test so long as the LAL assay utilized is shown to be as sensitive as. or more sensitive as the rabbit test [Fed. Reg.. 38. 26130 (1980)].
The term "monovalent" when used in reference to a clostridial vaccine refers to a vaccine which is capable of provoking an immune response in a host a subject) animal S 15 directed against a single type of clostridial toxin. For example, if immunization of a host with C. difficile type A toxin vaccine induces antibodies in the immunized host which protect against a challenge with type A toxin but not against challenge with type B toxin, then the type A vaccine is said to be monovalent. In contrast. a "multivalent" vaccine provokes an immune response in a host animal directed against several more than one) clostridial toxins. For example. if immunization of a host with a vaccine comprising C. difficile type A and B toxins induces the production of antibodies which protect the host against a challenge with both type A and B toxin, the vaccine is said to be multivalent (in particular. this hypothetical vaccine is bivalent).
As used herein, the terms "aggregate" and "aggregation" refer to the production of clumps, groupings. or masses of materials. It is not intended that the terms be limited to a particular type of clumping. Rather. it is intended that the term be used in its broadest sense to encompass any situation where multiple items are brought together into close contact.
Thus, the term encompasses agglutination of any type (including, but not limited to latex agglutination. hemagglutination. or any other method in which an immunological reaction is used to produce agglutination). The terms also apply to non-immunological methods, and 19also encompass non-specific relationships between multiple components: all that is required is that the individual components be clumped together.
The term "subject" when used in reference to administration of compositions comprising antitoxins or vaccines refers to the recipient animal to whom said antitoxins or vaccines are administered. The subject may be any animal. including mammals and more particularly. humans. in which it is desirable to administer said compositions. The subject may have'been previously exposed to one or more C d fficile toxins prior to administration of said compositions (this constitutes therapeutic administration to the subject). Alternatively.
the subject may not have been previously exposed to C difficile toxins prior to administration of said compositions (this constitutes prophylactic administration to the subject).
The term "sample" in the present specification and claims is used in its broadest sense.
On the one hand it is meant to include a specimen or culture microbiological cultures).
On the other hand. it is meant to include both biological and environmental samples.
Biological samples may be animal. including human. fluid. solid stool) or tissue.
as well as liquid and solid food and feed products and ingredients such as dairy items, vegetables. meat and meat by-products. and waste. Biological samples may be obtained from all of the various families of domestic animals. as well as eral or wild animals. including, but not limited to. such animals as ungulates. bear. fish. lagamorphs. rodents. etc.
Environmental samples include environmental material such as surface matter, soil, water and industriaI samples. as well as samples obtained from food and dairy processing instruments. apparatus. equipment. utensi Is. disposable and non-disposable items. These examples are not to be construed as limiting the sample types applicable to the present invention.
As used herein. the term "culture" is used in reference to the in vivo or in vitro growthof organisms. including. but not limited to bacteria. It is intended that the term encompass any form of microbial culture. It is intended that the term encompass the propagation of micoorganisms or other living cells in media and in an environment that is conducive to their growth. Such cultures may be grown in any forinat. including but not limited to agar plates.
broths. and semi-solid media. and may be grown in any environment suitable for the oreanisms cultured aerobic. anaerobic. microaerophilic. etc.).
20 As used herein, the term "supernatant" is used in reference to any liquid or fluid solution. This liquid or fluid may or may not contain soluble particles such as proteins antibodies or toxin molecules). The term encompasses any liquid lying above precipitated insoluble material, as well as liquids such as liquid culture media collected from a microbial or cell culture. It also encompasses the liquid portion of a sample which has been centrifuged to separate insoluble particles which are incapable of remaining in solution during centrifugation. from particles which are capable of remaining in solution during centrifugation. However. it is not intended that the term be limited to the situation in which centrifugation is utilized.
The term "protective level", when used in reference to the level of antibodies induced upon immunization of the host with an immunogen which comprises a bacterial toxin, means a level of circulating antibodies sufficient to protect the host from challenge with a lethal dose of the toxin.
As used herein the terms "protein" and "polypeptide" refer to compounds comprising 15 amino acids joined via peptide bonds and are used interchangeably.
The term "toxin" when used in reference to toxins produced by members species and strains) of the genus Clostridium refers to the proteins which are poisonous to tissue(s).
For example, the toxins produced by C. difficile are poisonous to intestinal tissues: the toxins *produced by C. botulinum are poisonous to nerve tissue.
The terms "encapsulation" or "encapsulating" refers to the covering of a solid Ivophilized) form of antitoxin. The covering may comprise an enteric coating or a capsule.
The terms "enteric coating" or "enteric film" are used interchangeably and refer to a material or compound which is resistant to acid pH an acid-resistant compound). such as that found in the stomach. An enteric coating when applied to a solid inhibits the dissolution of the solid in the stomach.
.i Standard techniques are known to the art for the encapsulation of solid compositions.
oThese techniques include microencapsulation of a solid composition wherein an enteric coating is applied to the solid composition. The coated material may be delivered orally to a subject by suspending the microencapsulated particles in pharmaceutical suspension solutions known to the art.
21 When a solid antitoxin is to be encapsulated using an enteric coating, the enteric coating may be applied using a one step coating process in which the enteric film is directly applied to the solid antitoxin: the coated antitoxin is said to be overcoated with the enteric film. Alternatively, a two step coating process may be employed wherein the solid antitoxin is first used to overcoat a non-pariel a sugar particle of about 40-60 mesh size) and then the antitoxin-coated non-pariel is overcoated with the enteric film. Desirable enteric coatings for the delivery of antitoxins include polymethacrylates such as Eudragit® L30D (Rahm Tech, Inc.) Solid antitoxin may formulated for oral delivery by insertion of the desired qunatity of antitoxin into a capsule: the capsule would preferable have the characteristic of being resistant to dissolution in the stomach and being capable of dissolving in the intestines. Numerous suitable capsule formulations are available to the art: in addition standard techniques are available for the filling of capsules including the use of inert filler materials to provide sufficient bulk of the filling of a capsule with a therapeutic composition in a solid form. In 15 addition to the use of microencapsulated antitoxin and antitoxin contained within a capsule.
the solid antitoxin may be delivered orally in tablet or pill form. The solid antitoxin may be combined with inert materials to provide sufficient bulk for the pressing of the tablet or pill.
Once formed, the tablet or pill may then be coated with an enteric film to prevent dissolution in the stomach and to enhance dissolution in the intestines.
The term "oral administration" refers to the delivery of a composition. such as a composition comprising antitoxin, via the mouth.
The term "parenteral administration" refers to the delivery of a composition. such as a composition comprising an antitoxin or vaccine, by a route other than through the Sgastrointestinal tract oral delivery) or the lungs. In particular. parenteral administration i may be via intravenous, subcutaneous. intramuscular or intramedullary intrathecal) injection.
6 The terms "symptoms" and "symptoms of intoxication" when used in reference to a subject exposed to or at risk of exposure to C difficile toxins refers to presence of any of the following phenomenon: diarrhea. enterocolitis. pseudomembranous colitis, hemorrhage, 22 ulceration and/or inflammation of the intestinal mucosa. cecitis inflammation of the cecum).
As used herein, the term "ceases to exhibit symptoms" refers to the situation in which a subject has stopped exhibiting the signs and/or symptoms associated with C. difficile disease and/or infection.
The term "substantial elimination" of the symptoms of intoxication with C. difficile disease means that in subject exposed to and suffering from the symptoms of intoxication, the symptoms are abated. attenuated or eliminated. For example. if an intoxicated subject presents with severe diarrhea voluminous, watery diarrhea). a return to an at least loosely formed stool would constitute a substantial elimination of this symptom.
The term "beyond the treatment period" when used in reference to a method of treating a subject exposed to a C difficile toxin means a period of time following the cessation of administration of a therapeutic compound antitoxin) to the subject for at least 7 days and more preferably at least 14 days. A therapeutic compound which results in 15 the substantial elimination of the symptoms of intoxication beyond the treatment period will i "prevent the reappearance (when symptoms are eliminated) or the increase in severity (when symptoms are abated) of these symptoms for at least 7 days following the withdrawal of S.administration of the therapeutic compound. In other words, no relapse reappearance or increase in severity) of the symptoms is seen in the majority a statistically significant 20 number of subjects for a period of at least 7 days following the cessation of therapy.
In contrast to the antitoxins of the present invention, existing therapeutic compounds for established C. difficile infections antibiotics such as vancomycin or metronidazole or bovine lgG concentrate from cows immunized with C. difficile toxoids A and B [Lyerly et al.
(1991) Infect. Immun.59:2 2 1 5 do not prevent relapse in a significant number of treated subjects. For example, about 25% of humans and up to 100% of hamsters suffering from C difficile associated disease treated with either vancomycin or metronidazole relapse symptoms of intoxication reappear).
Hamsters administered bovine IgG concentrate (BIC) from cows immunized with C.
difficile toxoids A and B prior to infection with C. difficile prophylactic treatment) 23 24 invariably relapse diarrheas returns) and die when the BIC is withdrawn [Lyerly et al, (1991), supra]. No therapeutic effect is observed when hamsters having established C.
difficile infections are treated with the BIC the administration of the BIC does not eliminate the diarrhea or prevent death) [Lyerly et al, (1991), supra].
In contrast, the antitoxins of the present invention, when used to treat established C.difficile infection (therapeutic regimen), substantially eliminate the symptoms of intoxication, including diarrhea and prevent death. The majority of animals treated with the anti-C. difficile toxin proteins do not relapse and remain healthy following cessation of antitoxin therapy for a period of at least 14 days [the animals remain healthy for long periods of time about 5 months)].
Summary of the Invention According to a first embodiment of the present invention there is provided a recombinant Clostridium botulinum toxin derived from the cleavage of a soluble, recombinant Clostridium botulinum toxin fusion protein.
S 15 According to a second embodiment of the present invention there is provided a recombinant Clostridium botulinum toxin protein derived from the cleavage of a soluble, recombinant Clostridium botulinum toxin protein or portion thereof, capable of eliciting an immune response, fused to a non-toxin protein sequence.
According to a third embodiment of the present invention there is provided a soluble fusion protein comprising: a portion of a Clostridium botulinum toxin capable of eliciting an immune response, and; a non-toxin protein.
According to a fourth embodiment of the present invention there is provided a 25 soluble fusion protein comprising: a Clostridium botulinum toxin, and; a non-toxin protein.
According to a fifth embodiment of the present invention there is provided a recombinant Clostridium botulinum toxin derived from the cleavage of a soluble, recombinant Clostridium botulinum toxin fusion protein as claimed in any one of the first to the fourth embodiments.
[I:\DayLib\LIBVV]02862spcci.doc:sxc 24a In a preferred embodiment, the toxin polypeptides comprise Clostridium botulinum neurotoxin. The invention contemplates the use of polypeptides derived from C.
botulinum toxin as immunogens for the production of vaccines and antitoxins. The C. botulinum vaccines and antitoxins find use in humans and other animals. In one embodiment, the present invention contemplates a fusion protein comprising a non-toxin protein sequence and a portion of the Clostridium botulinum type A toxin. In a preferred embodiment, the C. botulinum type A toxin sequences comprise a portion of the sequence of SEQ ID NO:28. In yet another preferred embodiment, the C botulinum type A toxin sequences comprise a portion of the sequence of SEQ ID NO:23. It is not intended that the present invention be limited by the nature of the fusion protein. For example, the fusion protein may comprise the Clostridium botulinum type A toxin sequence as set forth in SEQ ID NO:23 along with a poly-histidine tract.
e *g*o *#go *oo ooo *ooo [1:\DayLib\LIBVV]02862speci.doc:sxc The invention also contemplates a host cell containing a recombinant expression vector, wherein the vector encodes a fusion protein comprising a non-toxin protein sequence and a portion of the Clostridium botulinum type A toxin sequence of SEQ ID NO:28. In this embodiment, the host cell is capable of expressing the encoded Clostridium botulinum type A toxin protein as a soluble protein at a level greater than or equal to 0.25% to 10% of the total cellular protein and preferably at a level greater than or equal to 0.75% of the total cellular protein. It is not intended that the present invention be limited by the nature of the fusion protein expressed by the recombinant vector in the host cell. For example, the fusion protein may comprise the Clostridium botulinum type A toxin sequence as set forth in SEQ ID NO:23. along with a poly-histidine tract.
The present invention also contemplates a host cell containing a recombinant expression vector, wherein the vector encodes a protein derived from the Clostridium botulinum type A toxin sequence of SEQ ID NO:28. In this embodiment. the host cell is capable of expressing the encoded Clostridium botulinum type A toxin protein at a level greater than or equal to 10% to 40% of the total cellular protein and preferably at a level greater than or equal to 20% of the total cellular protein. It is not intended that the present invention be limited by the nature of the fusion protein expressed by the recombinant vector in the host cell. For example, the fusion protein may comprise the Clostridium botulinum type A toxin sequence as set forth in SEQ ID NO:23. along with a poly-histidine tract.
20 In one embodiment, the present invention contemplates a method of generating neutralizing antitoxin directed against Clostridium botulinum type A toxin comprising (in any order), a purified soluble fusion protein comprising a non-toxin protein sequence and a portion of the Clostridium botulinum type A toxin sequence of SEQ ID NO:28. as well as host is immunized with the purified fusion protein so as to generate antibodies capable of neutralizing native Clostridium botulinum type A toxin. By way of illustration only, the fusion protein may comprise a portion of the Clostridium'botulinum type A toxin sequence as set forth in SEQ ID NO:23. and a poly-histidine tract. The method may further comprise the additional step of collecting antibodies from the host. It is also contemplated that the collected antibodies be purified. The present invention contemplates the antibody, as a composition of matter. raised according to the above-described methods.
25 The present invention further contemplates a method of purifying a recombinant fusion protein derived from a Clostridium botulinum type A toxin. In this embodiment. the recombinant fusion protein comprises a poly-histidine tract. comprising (in any order) a solution comprising a fusion protein comprising a poly-histidine tract and a portion of the Clostridium botulinum type A toxin sequence of SEQ ID NO:28. and a chromatography resin comprising a divalent cation covalently linked to a solid support. In this embodiment, the solution is added to the chromatography resin to allow binding of the fusion protein to the chromatography resin. It is also contemplated that this embodiment further comprises the step of washing the chromatography resin containing said bound fusion protein to remove nonfusion protein from the chromatography resin, and eluting the bound fusion protein from the washed chromatography resin.
In a preferred embodiment. the chromatography resin comprises nickel ions immobilized on a solid support. Examples of commercially available nickel ion columns include the His-Bind® Resin (Novagen) and the Ni-NTA Agarose resin (Qiagen). Because S 15 the Ni-NTA Agarose resin has a very high affinity for binding proteins containing a polyhistidine tract, it is a preferred chromatography resin.
The invention is not intended to be limited by the nature of the solution comprising a fusion protein comprising a poly-histidine tract and a portion of the Clostridium botulinum type A toxin sequence of SEQ ID NO:28. In one embodiment, this solution comprises a 20 soluble extract derived from a cell pellet comprising host cells containing a recombinant fusion protein. In yet another embodiment. the soluble extract is generated from the cell pellet by suspension of the cell pellet in a binding buffer and disrupting the suspension to S. cause the disruption of the membranes of the host cell to generate a mixture comprising soluble proteins and insoluble cellular debris. In another embodiment, the method of purifying a recombinant fusion protein derived from a Clostridium bolulinum type A toxin.
wherein the recombinant fusion protein comprises a poly-histidine tract, further includes the additional step of removing the insoluble cellular debris from the disrupted cell suspension to generate a clarified soluble lysate. In yet a further embodiment, the method of purifying the recombinant fusion protein employs the addition of a non-ionic detergent to the clarified soluble lysate. A preferred non-ionic detergent is Nonidet P-40. In still another preferred -26embodiment of the method of purifying the recombinant fusion protein comprises, the additional step of incubating the clarified soluble lysate containing said non-ionic detergent with the chromatography resin for greater than one hour at four degrees Centigrade to allow the fusion protein to bind to the chromatography resin. Incubation steps of about 3 hours are particularly preferred.
The present invention relates to clostridial antitoxin therapy for humans and other animals. Antitoxins which neutralize the pathologic effects of clostridial toxins are generated by immunization of avian hosts with recombinant toxin fragments. In one embodiment, the present invention contemplates a fusion protein comprising a non-toxin protein sequence and a portion of the Clostridium difficile toxin B sequence of SEQ ID NO:10. It is not intended that the present invention be limited by the nature of the fusion protein. For example, the fusion protein may comprise the portion of the Clostridium difficile toxin B sequence as set forth in SEQ ID NO:20 and the maltose binding protein (or portion thereof). On the other hand. the fusion protein may comprise the Clostridium difficile toxin B sequence as set forth 15 in SEQ ID NO:21 along with a poly-histidine tract.
In one embodiment, the present invention contemplates a method of generating a neutralizing antitoxin directed against Clostridium difficile toxin B comprising: a) providing in /any order: i) a purified fusion protein comprising a non-toxin protein sequence and a portion 00' of the Clostridium difficile toxin B sequence of SEQ ID NO:10, and ii) an avian host; and b) 20 immunizing said host with said purified fusion protein so as to generate an antitoxin capable of neutralizing native Clostridium difficile toxin B. Again by way of illustration only. the fusion protein may comprise the portion of the Clostridium difficile toxin B sequence as set forth in SEQ ID NO:20 and the maltose binding protein (or portion thereof). On the other hand, the fusion protein may comprise the Clostridium difficile toxin B sequence as set forth in SEQ ID NO:21 along with a poly-histidine tract. The method may further comprise a step c) collecting said antitoxin from said host and. even further, a step d) purifying said antitoxin.
The present invention contemplates the antibody. as a composition of matter.. raised according to the above-described methods.
The present invention further contemplates a method of treatment comprising: a) providing: i) a subject and ii) at least one neutralizing antitoxin directed against a fusion 27 protein comprising a non-toxin protein sequence and a portion of Clostridium difficile toxin B sequence of SEQ ID NO:10: and b) administering said antitoxin to said subject. In one embodiment, the present invention contemplates administering by oral administration. The present invention further contemplates that the subject treated may or may not have been exposed to Clostridium difficile and its toxins. That is to say. in one instance, exposure to at Clostridium difficile toxin B may have occurred prior to administration of the antitoxin. In another instance, the subject has not been exposed to at Clostridium difficile toxin B prior to administration of the antitoxin.
The present invention also contemplates fusion proteins containing toxin A fragments.
In one embodiment, the present invention contemplates a fusion protein comprising a nontoxin protein sequence and a portion of the Clostridium difficile toxin A sequence consisting of SEQ ID NO:7. In still another embodiment. the present invention contemplates a fusion protein comprising a non-toxin protein sequence and a portion of the Clostridium difficile toxin A sequence comprising the amino acid sequence of SEQ ID NO:8. In the above 15 embodiments. the non-toxin part of the fusion protein may be selected from a variety of nontoxin protein sequence types. In a preferred embodiment, the non-toxin sequence is the Smaltose binding protein sequence (or a portion thereof).
The present invention contemplates a method of generating a neutralizing antitoxin directed against Clostridium difficile toxin A comprising: a) providing in any order: i) a 20 purified fusion protein comprising a non-toxin protein sequence (for example, the maltose binding protein sequence or portion thereof) and a portion of the Clostridium difficile toxin A sequence (for example, the toxin A sequence as set forth in SEQ ID NO:7). and ii) an avian host: and b) immunizing said host with said purified fusion protein so as to generate an antitoxin capable of neutralizing said Clostridium difficile toxin A. The method may further comprise step c) collecting said antitoxin from said host and step d) purifying said antitoxin.
The present invention also contemplates uses for the toxin fragments in vaccines and diagnostic assays. The fragments may be used separately as purified, soluble antigens or, alternatively, in mixtures or "cocktails." The present invention provides compositions comprising an avian neutralizing antitoxin directed against a portion of C. difficile toxin A and a portion of C. difficile toxin B. The -28 antitoxins find use in humans and other animals exposed to or at risk of exposure to C.
difficile. In one embodiment, the component of the avian neutralizing antitoxin directed against a portion C difficile toxin A is directed against a first fusion protein comprising a portion of C. difficile toxin A and a second fusion protein comprising a portion of C. difficile toxin B. In yet another embodiment, both first and second fusion proteins further comprise at least one non-toxin protein sequence. In a still further embodiment, the antitoxin is directed against a portion of C difficile toxin A comprising a portion of SEQ ID NO:6. In another embodiment, the antitoxin is directed against a portion of C. difficile toxin A. wherein the portion of SEQ ID NO:6 comprises a sequence selected from the group comprising SEQ ID NOS:7, 8 and 29. In yet another embodiment, the first and second fusion proteins comprise at least one non-toxin protein sequence. It is not intended that the present invention be limited by the nature of the non-toxin protein sequence. In one embodiment the non-toxin protein sequence comprises a poly-histidine tract. In yet another embodiment the non-toxin protein sequence comprises the maltose binding protein. In yet another embodiment the nontoxin protein sequence comprises a thioredoxin protein. In a still further embodiment, the antitoxin is directed against a portion of C. difficile toxin B comprising a portion of SEQ ID In another embodiment, the antitoxin is directed against a portion of C. difficile toxin B. wherein the portion of SEQ ID NO:10 comprises a sequence selected from the group comprising SEQ ID NOS:11. 12. 20. 21 and 30. In still another embodiment the 20 compositions comprising the avian antitoxins further comprise an enteric coating.
The invention also contemplates a method of treatment comprising: a) providing: i) a subject. ii) a first avian neutralizing antitoxin directed against a portion of Clostridium difficile toxin A sequence SEQ ID NO:6. and iii) a second avian neutralizing antitoxin directed against a portion of Clostridium difficile toxin B sequence SEQ ID NO:10: b) mixing the first and second antitoxins to create a therapeutic mixture: and c) administering the therapeutic mixture to a subject for a treatment period. The invention further contemplates a method of treatment s* which further comprises the step of, prior to step processing the therapeutic mixture to improve its enteric stability. In a preferred embodiment, this treating comprises encapsulating the antitoxins of the therapeutic mixture. In a particularly preferred embodiment the 29encapsulating step comprises overcoating the antitoxins in the therapeutic mixture with an enteric film.
The invention further contemplates the method of treatment wherein the subject has been exposed to at least one Clostridium difficile toxin prior to administration of antitoxin. In one embodiment, the exposed subject is suffering from the symptoms of intoxication and administering antitoxin results in the substantial elimination of symptoms beyond the treatment period. In another embodiment, the symptoms of intoxication comprise diarrhea.
The invention also contemplates the method of treatment wherein the subject has not been exposed to Clostridium difficile toxin prior to administration of antitoxin.
In one embodiment, the method of treatment provides a first avian antitoxin directed against a portion of Closiridium difficile toxin A comprising a protein sequence selected from the group comprising SEQ ID NOS:7. 8 and 29. In another embodiment, the method of treatment provides a second avian antitoxin directed against a portion of Clostridium difficile toxin B comprising a protein sequence selected from the group comprising SEQ ID NOS:I 1.
15 12. 20. 21 and The method of treatment is not limited by the method of administration of the antitoxin. In one embodiment. the method of treatment comprises administration of the antitoxins by oral administration. In another embodiment. the method of treatment comprises administration of the antitoxins by parenteral administration.
The invention further contemplates a method of vaccinating a subject to produce °neutralizing antitoxin directed against C. difficile toxin comprising: a) providing in any order: i) a subject. ii) a first purified soluble and substantially endotoxin-free protein comprising a portion of Clostridium difficile toxin A sequence SEQ ID NO:6, and iii) a second purified soluble and substantially endotoxin-free protein comprising a portion of Clostridium difficile toxin B sequence SEQ ID NO:10;b) mixing the first and second proteins to create a therapeutic vaccine: and c) vaccinating the subject with the therapeutic vaccine so as to generate neutralizing antitoxin. The method of vaccination is not limited by the nature or species of the subject. In one. embodiment the subject is a bird. In another embodiment the subject is a mammal. In yet another embodiment the subject is a human. In a still further embodiment, the method of vaccination the first and second toxin proteins further comprise at least one non-toxin protein sequence. The invention is not limited by the nature of the nontoxin protein sequence. In one embodiment, the non-toxin protein sequence comprises a polyhistidine tract. In another embodiment, the non-toxin protein sequence comprises the maltose binding protein. In yet another embodiment, the non-toxin protein sequence comprises a thioredoxin protein.
In one embodiment, the method of vaccinating uses a first purified and substantially endotoxin-free protein comprising SEQ ID NO:29. In another embodiment. the method of vaccinating uses a second purified and substantially endotoxin-free protein comprising SEQ ID The invention further provides a fusion protein comprising at least one non-toxin protein sequence and a portion of the Clostridium difficile toxin A sequence consisting of SEQ ID NO:29. In one embodiment. the non-toxin protein sequence comprises a thioredoxin protein. In yet another embodiment. the non-toxin protein sequence further comprises a polyhistidine tract.
15 The present invention provides a method for the detection of Clostridium difficile antigens in a sample, comprising providing, in any order a sample suspected of containing OClostridium difficile antigens. solid support conjugates comprising antibodies reactive with Clostridium difficile antigens bound to a solid suppoit: mixing the sample and solid support conjugates under conditions such that the conjugates are capable of binding to Clostridium difficile antigens: and detecting binding. In one embodiment, the antibodies reactive with Clostridium difficile antigens are avian antibodies. In a preferred embodiment. the avian antibodies reacts with Toxin A of Clostridium difficile. In a particularly preferred embodiment, the avian antibodies reacts with the A-6 interval of Toxin A. In an alternative preferred embodiment the avian antibodies react with Toxin B of Clostridium difficile. In another preferred embodiment, the avian antibodies react with the B-3 interval of Toxin B. In yet another preferred embodiment, the avian antibodies react with Toxin A and Toxin B. It is also contemplated that the solid support used in the method comprises polystyrene particles.
In one preferred embodiment. the mixing of step results in the formation of visible aggregates. In a preferred embodiment, the sample is human feces.
31 In an alternative embodiment, the present invention comprises a method of treatment comprising providing a subject exposed to Clostridium dfficile exhibiting symptoms comprising diarrhea. and antibody reactive with Clostridium difficile. wherein the antibody is present in a therapeutic amount that is administrable, and administering the antibody to the subject under conditions such that the subject ceases to exhibit symptoms and treatment can be terminated. In a particularly preferred embodiment, the subject exhibits long-term survival beyond the treatment period. In one preferred embodiment, the antibodies reactive with Clostridium difficile antigens are avian antibodies. It is contemplated that the antibodies will be reactive against various moieties or antigens, including, but not limited to Toxin A of Clostridium difficile. the A-6 interval of Toxin A. Toxin B of Clostridium difficile, the B-3 interval of Toxin B. and a combination of Toxin A and Toxin B.
The present invention also provides a method of purifying Clostridium difficile toxins S. from a culture, comprising providing in any order, a culture comprising Clostridium difficile organisms and a supernatant comprising toxins in solution, antibodies reactive with 15 Clostridium difficile toxins immobiled on a solid support, collecting the supernatant from the culture comprising toxins, adding the supematant to immobilized antibody under conditions such that antibodies are capable of binding to the toxins, eluting the toxins from the immobilized antibodies: and detecting any eluted toxins. In one preferred embodiment, the antibodies reactive with Clostridium difficile antigens are avian antibodies. It is contemplated that various antibodies will be used in this method, including antibodies reactive against various antigens or moieties, including, but not limited to Toxin A of Clostridium difficile, the o o A-6 interval of Toxin A. Toxin B of Clostridium difficile. the B-3 interval of Toxin B. and a combination of Toxin A and Toxin B.
o" DEI The present invention provides compositions comprising an avian antitoxin directed against a clostridial toxin protein. In a preferred embodiment these compositions are in a solid dosage form. The term "solid dosage form" means as dosage forms including tablets.
pills, capsules (including, a gel-cap as that term is commonly used in the pharmaceutical industry) and all extended-release variations thereof controlled-release. sustained-release.
timed-release, prolonged-action and the like). Moreover, the term "solid dosage form" can also include suspensions solid particles comprising avian antitoxin suspended within a 32liquid vehicle: the solid particles may further comprise an enteric coating) which may be delivered orally.
In one embodiment, the solid dosage form of the avian antitoxin further comprises an enteric coating. In a particularly preferred embodiment, the enteric coating dissolves at a pH about 7.0. The terms "at a pH about 7.0" and "about pH 7.0" are used interchangeably and refer to a pH range of 6.5 to 7.5. Particularly preferred enteric coating are those which remain essentially intact during transient of the enterically-coated tablet (pill, capsule, etc.) through the stomach (pH of about 0- 2.0) and small intestine (pH of about 5.0 6.5) but which dissolve when they reach the large intestine (pH of about Examples of suitable enteric coatings are methacrylic acid copolymers Eudragit L or S (Rohm Tech, Inc..
Maiden. cellulose acetate phthalate (CAP) Aquateric (FMC Corp., Philadelphia, PA) which dissolves at pH hydroxypropyl methylcellulose acetate succinate (HPMCAS) Aqoat Grade 3 (Shin-Esta Chemical Corp.. Japan) which dissolves at pH 7.0] and polyvinyl acetate phthalate (PVAP) Sureteric (Colorcaon. Inc.. West Point The I 5 term "essentially intact" when used in reference to an enterically-coated tablet (pill. capsule.
etc.) means less than 10% of the protein avian antitoxin) present in the coated tablet is released at pH below about In one embodiment, the avian antitoxin present in a solid dosage form comprises a tablet. The term tablet" refers to a solid dosage form containing medicinal substances avian antitoxin) with or without suitable diluents. excipients. fillers. etc. The tablet may vary in shape, size and Weight and may be classed according to the method of manufacture a o*o molded tablet a compressed tablet, etc.). The term tablet encompasses pills.
In another embodiment the compositions comprising avian antitoxins in a solid dosage form contain polyethylene glycol (PEG). Solid dosage forms tablets comprising lyophilized avian antibody antitoxin) preparations containing PEG may contain 0-60% (of the total weight of the tablet) PEG and more preferably 20-40% PEG. The tablets may also contain water (as well as fillers, binders, extenders, coloring agents. etc.). The water content may vary: the presence of water in the tablet may be due to absorption of water from the atmosphere during the handling of lyophilized avian antibody preparations prior to or .33 during the formation of the tablets. If desirable for the formation of the tablet. water may be deliberately added to the solid antibody preparations.
The present invention also provides a method of generating a solid dosage form of an avian antitoxin directed against a clostridial toxin protein, comprising: a) providing a composition comprising an avian antitoxin directed against a clostridial toxin protein in a dry form; and b) shaping said dry avian antitoxin into a tablet. The process of shaping dry antitoxin into a tablet encompasses any procedure capable of rendering the solid antitoxin preparation into a tablet form. including compression or molding of the antitoxin. The art is wellaware of methods for the formation of tablets from dry material. In a preferred embodiment the shaping of the dry antitoxin into a tablet is accomplished by compression of the dry antitoxin using a tablet press.
In vet a further embodiment, the method of generating a solid dosage form comprising avian antitoxin further comprises the step of applying an enteric coating to tablet. In a still further preferred embodiment, the method of generating a solid dosage form comprising avian S 15 antitoxin utilizes a composition comprising dry antitoxin which contains polyethylene glycol.
DESCRIPTION OF THE INVENTION The present invention contemplates vaccinating humans and other animals polypeptides derived from C botulinum neurotoxin which are substantially endotoxin-free. These botulinal peptides are also useful for the production of antitoxin. Anti-botulinal toxin antitoxin is useful for the treatment of patients effected by or at risk of symptoms due to the action of C botulinum toxins. The organisms. toxins and individual steps of the present invention are described separately below.
I. Clostridium Species, Clostridial Diseases And Associated Toxins A preferred embodiment of the method of the present invention is directed toward obtaining antibodies against Clostridium species, their toxins, enzymes or other metabolic byproducts. cell wall components. or synthetic or recombinant versions of any of these compounds. It is contemplated that these antibodies will be produced by immunization of humans or other animals. It is not intended that the present invention be limited to any 34 particular toxin or any species of organism. In one embodiment, toxins from all Clostridium species are contemplated as immunogens. Examples of these toxins include the neuraminidase toxin of C. buvyricum. C. sordellii toxins HT and LT. toxins A, B, C. D, E, F. and G of C.
botulinum and the numerous C perfringens toxins. In one preferred embodiment. t6xins A and B of C. difficile are contemplated as immunogens. Table 2 below lists various Clostridium species, their toxins and some antigens associated with disease.
TABLE 2 Clostridial Toxins Organism Toxins and Disease-Associated Antigens C botulinum A. B. D, E. F. G C. butvricum Neuraminidase A. B. Enterotoxin (not A nor Motility Altering Factor.
C. difficile Low Molecular Weight Toxin. Others C perfIingens a. P, c. t, y. 5, v. 6. K. A. C sordelli/ HT. LT. a, 1, y 15 C. bifermentans C. nowvi a, Y. 5 E v. 0 C septicum a. P. 8.
C histolvyicum a, i, y. e plus additional enzymes C. chauvoei a. 0, y It is not -ntended that antibodies produced against one toxin will only be used against that toxin. It is contemplated that antibodies directed against one toxin C. perfringens type A enterotoxin) may be used as an effective therapeutic against one or more toxin(s) produced by other members of the genus Clostridium or other toxin producing organisms Bacillus cereus. Staphylococcus aureus. Streptococcus mutans. Acinetobacter 35 calcoaceticus. Pseudomonas aeruginosa. other Pseudomonas species. etc.). It is further contemplated that antibodies directed against the portion of the toxin which binds to mammalian membranes C. perfringens enterotoxin A) can also be used against other organisms. It is contemplated that these membrane binding domains are produced synthetically and used as immunogens.
II. Obtaining Antibodies In Non-Mammals A preferred embodiment of the method of the present invention for obtaining antibodies involves immunization. However, it is also contemplated that antibodies could be obtained from non-mammals without immunization. In the case where no immunization is contemplated. the present invention may use non-mammals with preexisting antibodies to toxins as well as non-mammals that have antibodies to whole organisms by virtue of reactions with the administered antigen. An example of the latter involves immunization with synthetic peptides or recombinant proteins sharing epitopes with whole organism components.
S: 15 In a preferred embodiment, the method of the present invention contemplates immunizing non-mammals with bacterial toxin(s). It is not intended that the present invention be limited to any particular toxin. In one embodiment. toxin from all clostridial bacteria sources (see Table 2) are contemplated as immunogens. Examples of these toxins are C burvricum neuraminidase toxin, toxins A. B. C. D. E. F. and G from C botulinum 20 C. perfingens toxins a. P. E. and t. and C sordellii toxins HT and LT. In a preferred embodiment. C difficile toxins A and B are contemplated as immunogens.
A particularly preferred embodiment involves the use of bacterial toxin protein or fragments of toxin proteins produced by molecular biological means recombinant toxin proteins). In a preferred embodiment. the immunogen comprises interval 6 of C. difficile toxin A produced by recombinant DNA technology. In yet another preferred embodiment, the immunogen comprises interval 3 of C difficile toxin B produced by recombinant DNA technology. The recombinant C difficile toxin proteins may be used as immunogens separately or in combination to produce antibodies specific for either C difficile toxin A, C difficile toxin B or both C. difficile toxins A and B. Specifically. the recombinant C. difficile toxins A and B proteins may be mixed together and used as a single immunogen.
Alternatively. C. difficile toxin A proteins may be used separately as an immunogen in a first subject. Similarly. C difficile toxin B proteins may be used separately as an immunogen in a second subject. The antitoxin produced by separate immunization of two separate subjects 36 with C. difficile toxin A proteins or C difficile toxin B proteins may be combined to yield an antitoxin directed against both C. difficile toxins A and B.
The recombinant C difficile toxin proteins provided herein enables the production of antibodies which are specific for a single C. difficile toxin mono-specific antibodies).
This is in contrast to the biochemical purification of C difficile toxin A from natural sources results invariably in the isolation of a toxin A preparation contaminated with immunologically significant amounts of toxin B: similarly the biochemical purification of C. difficile toxin B from natural sources results in the isolation of a toxin B preparation contaminated with immunologically significant amounts of toxin A. Because, these preparations of nonrecombinant toxin A and or toxin B are cross-contaminated with either toxin B or A.
immunization of an animal will result in the production of polyclonal antibodies reactive *i against both toxins A and B.
As discussed below in section VI. accurate detection of the presence of C. difficile toxin A and/or B in a sample requires the availability of both pure preparations of toxin A S 15 and B and the availability of mono-specific antibodies. The use of recombinant C. difficile toxin proteins thus allows for the production of a polyclonal antibody preparation that can be used for accurate detection of individual C difficile toxins as well as C. difficile organisms.
When immunization is used. the preferred non-mammal is from the class Aves. All birds are contemplated duck. ostrich. emu. turkey, etc.). A preferred bird is a chicken.
20 Importantly. chicken antibody does not fix mammalian complement. [See H.N. Benson et al..
J. Immunol. 87:616 (1961).] Thus. chicken antibody will normally not cause a complementdependent reaction. Benedict and K. Yamaga. "Immunoglobulins and Antibody Production in Avian Species." in Comparative Immunology Marchaloni. pp. 335- 375, Blackwell, Oxford (1966).] Thus. the preferred antitoxins of the present invention will not exhibit complement-related side effects observed with antitoxins known presently.
When birds are used. it is contemplated that the antibody will be obtained from either the bird serum or the egg. A preferred embodiment involves collection of the antibody from the egg. Laying hens transport immunoglobulin to the egg yolk in concentrations equal to or exceeding that found in serum. [See R. Patterson et al.. J. Immunol. 89:272 (1962): and S.B. Carroll and B.D. Stollar. J. Biol. Chem. 258:24 (1983).] In addition, the large volume of egg yolk produced vastly exceeds the volume of serum that can be safely obtained from the bird over any given time period. Finally. the antibody from eggs is purer 37 and more homogeneous: there is far less non-immunoglobulin protein (as compared to serum) and only one class of immunoglobulin is transported to the yolk.
When considering immunization with toxins, one may consider modification of the toxins to reduce the toxicity. In this regard, it is not intended that the present invention be limited by immunization with modified toxin. Unmodified ("native") toxin is also contemplated as an immunogen.
It is also not intended that the present invention be limited by the type of modification if modification is used. The present invention contemplates all types of toxin modification.
including chemical and heat treatment of the toxin. The preferred modification. however, is formaldehyde treatment.
It is not intended that the present invention be limited to a particular mode of immunization: the present invention contemplates all modes of immunization. including subcutaneous. intramuscular. intraperitoneal. and intravenous or intravascular injection, as well as per os administration of immunogen.
15 The present invention further contemplates immunization with or without adjuvant.
(Adjuvant is defined as a substance known to increase the immune response to other antigens when administered with other antigens.) If adjuvant is used. it is not intended that the present invention be limited to any particular type of adjuvant or that the same adjuvant. once used.
be used all the time. While the present invention contemplates all types of adjuvant, whether used separately or in combinations, the preferred use of adjuvant is the use of Complete Freund's Adjuvant followed sometime later with Incomplete Freund's Adjuvant. Another "i preferred use of adjuvant is the use of Gerbu Adjuvant. The invention also contemplates the use of RIBI fowl adjuvant and Quil A adjuvant.
When immunization is used, the present invention contemplates a wide variety of immunization schedules. In one embodiment a chicken is administered toxin(s) on day zero and subsequently receives toxin(s) in intervals thereafter. It is not intended that the present invention be limited by the particular intervals or doses. Similarly. it is not intended that the present invention be limited to any particular schedule for collecting antibody. The preferred collection time is sometime after day 100.
Where birds are used and collection of antibody is performed by collecting eggs. the eggs may be stored prior to processing for antibody. It is preferred that eggs be stored at 4°C for less than one year.
38 It is contemplated that chicken antibody produced in this manner can be bufferextracted and used analytically. While unpurified, this preparation can serve as a reference for activity of the antibody prior to further manipulations immunoaffinity purification).
III. Increasing The Effectiveness Of Antibodies When purification is used. the present invention contemplates purifying to increase the effectiveness of both non-mammalian antitoxins and mammalian antitoxins. Specifically. the present invention contemplates increasing the percent of toxin-reactive immunoglobulin. The preferred purification approach for avian antibody is polyethylene glycol (PEG) separation.
The present invention contemplates that avian antibody be initially purified using simple. inexpensive procedures. In one embodiment. chicken antibody from eggs is purified by extraction and precipitation with PEG. PEG purification exploits the differential solubility of lipids (which are abundant in egg yolks) and yolk proteins in high concentrations of PEG 8000. [Polson et al.. Immunol. Comm. 9:495 (1980).] The technique is rapid, simple. and relatively inexpensive and yields an immunoglobulin fraction that is significantly purer in terms of contaminating non-immunoglobulin proteins than the comparable ammonium sulfate fractions of mammalian sera and horse antibodies. The majority of the PEG is removed from the precipitated chicken immunoglobulin by treatment with ethanol. Indeed. PEG-purified antibody is sufficiently pure that the present invention contemplates the use of PEG-purified 20 antitoxins in the passive immunization of intoxicated humans and animals.
The invention further contemplates increasing the effectiveness of compositions comprising antitoxins by enterically-coating a solid form of the antitoxin to improve the survival of the antitoxin in the gastrointestinal tract enteric stability) as discussed further below in section IV(C).
IV. Treatment The present invention contemplates antitoxin therapy for humans and other animals intoxicated by bacterial toxins. A preferred method of treatment is by oral administration of antitoxin. Another preferred method of treatment is by parenteral administration of antitoxin.
A. Therapeutic Preparations and Combinations The present invention contemplates using therapeutic compositions of antitoxins. The antitoxin compositions may comprise antitoxin in a solid or liquid form.
39- It is not intended that the present invention be limited by the particular nature of the therapeutic preparation. For example. such compositions can be provided together with physiologically tolerable liquid, gel or solid carriers, diluents, adjuvants and excipients. In addition, the antitoxins may be used together with other therapeutic agents. including antibiotics.
As noted above, these therapeutic preparations can be administered to mammals for veterinary use. such as with domestic animals, and clinical use in humans in a manner similar to other therapeutic agents. In general, the dosage required for therapeutic efficacy will vary according to the type of use and mode of administration, as well as the particularized requirements of individual hosts.
With respect to the mode of administration, the antitoxins may be employed for oral.
intravenous. intraperitoneal. intramuscular or intrathecal administration. Formulations for such administrations may comprise an effective amount of antitoxin in sterile water or physiological saline.
15 On the other hand. formulations may contain such normally employed additives such as binders, fillers, carriers, preservatives, stabilizing agents. emulsifiers. buffers and excipients as. for example, pharmaceutical grades of mannitol. lactose. starch. magnesium stearate.
sodium saccharin, cellulose, magnesium carbonate, and the like. These compositions typically contain 1%-95% of active ingredient, preferably 2%-70%.
The compositions are preferably prepared for oral administration, either as liquid solutions or suspensions: solid forms. including solid forms suitable for solution in. or suspension in. liquid prior to administration. may also be prepared. Solid forms of the antitoxins may further comprise an enteric coating. The compositions are also preferably prepared as injectables. either as liquid solutions or suspensions: solid forms suitable for solution in. or suspension in. liquid prior to administration may also be prepared.
The antitoxins of the present invention are often mixed with diluents or excipients which are physiological tolerable and compatible. Suitable diluents and excipients are, for example, water, saline. nutritional formulations Ensure®. Enfamil®. etc.) dextrose.
glycerol. or the like. and combinations thereof. In addition. if desired the compositions may contain minor amounts of auxiliary substances such as wetting or emulsifying agents, stabilizing or pH buffering agents.
B. Dosage Of Antitoxin It is noted by way of background that a balance must be struck when administering currently available antitoxin which is usually produced in large animals such as horses; sufficient antitoxin must be administered to neutralize the toxin, but not so much antitoxin as to increase the risk of untoward side effects. These side effects are caused by: i) patient sensitivity to foreign horse) proteins: ii) anaphylactic or immunogenic properties of nonimmunoglobulin proteins: iii) the complement fixing properties of mammalian antibodies; and/or iv) the overall burden of foreign protein administered. It is extremely difficult to strike this balance when. as noted above, the degree of intoxication (and hence the level of antitoxin therapy needed) can only be approximated.
The present invention contemplates significantly reducing side effects so that this balance is more easily achieved. Treatment according to the present invention contemplates reducing side effects by using PEG-purified antitoxin from birds.
In one embodiment, the treatment of the present invention contemplates the use of 15 PEG-purified antitoxin from birds. The use of yolk-derived. PEG-purified antibody as
S
antitoxin allows for the administration of: 1) non(mammalian)-complement-fixing. avian antibody; 2) a less heterogeneous mixture of non-immunoglobulin proteins: and 3) less total protein to deliver the equivalent weight of active antibody present in currently available antitoxins. The non-mammalian source of the antitoxin makes it useful for treating patients who are sensitive to horse or other mammalian sera.
As is true in cases of botulism, the degree of an individual's exposure to C. difficile toxin and the prognosis are often difficult to assess, and depend upon a number of factors the quantity of the inoculum, the toxigenicity and serotype of C. dfficile strain involved.
etc.). Thus, the clinical presentation of a patient is usually a more important consideration than a quantitative diagnostic test. for determination of dosage in antitoxin administration.
Indeed, for many toxin-associated diseases botulism, tetanus, diphtheria, etc.), there is no rapid, quantitative test to detect the presence of the toxin or organism. Rather, these toxinassociated diseases are medical emergencies which mandate immediate treatment.
Confirmation of the etiologic agent must not delay the institution of therapy, as the condition of an affected patient may rapidly deteriorate. In addition to the initial treatment with antitoxin, subsequent doses may be indicated, as the patient's disease progresses. The dosage and timing of these subsequent doses is dependent upon the signs and symptoms of disease in each individual patient.
-41 It is contemplated that when antitoxin is to be administered parentally, the administration of antitoxin to an affected individual would involve an initial injection of an approximately 10 ml dose of immune globulin (with less than approximately 1 gram of total protein). In one preferred embodiment, it is contemplated that at least 50% of the initial injection comprises immune globulin. It is also contemplated that more purified immune globulin be used for treatment. wherein approximately 90% of the initial injection comprises immune globulin. When more purified immune globulin purified IgY) is used, it is contemplated that the total protein will be less than approximately 100 milligrams. It is also contemplated that additional doses be given, depending upon the signs and symptoms associated with C. difficile associated disease progression.
It is contemplated that when antitoxin is to be administered orally, the administration of antitoxin to an affected individual would involve a treatment course initial and subsequent doses) comprising the administration of a therapeutic composition comprising about 50 gm of antitoxin and more preferably about 4-5 gm of antitoxin.
C. Delivery Of Antitoxin Although it is not intended to limit the route of delivery, the present invention contemplates a method for antitoxin treatment of bacterial intoxication in which delivery of antitoxin is parenteral or oral.
In one embodiment, antitoxin is parenterally administered to a subject in an aqueous Ssolution. It is not intended that the parenteral administration be limited to a particular route.
SIndeed. it is contemplated that all routes of parenteral administration will be used. In one embodiment, parenteral administration is accomplished via intramuscular injection. In an alternative embodiment, parenteral administration is accomplished via intravenous injection.
In one embodiment, antitoxin is delivered in a solid form tablets, capsules). In an alternative embodiment antitoxin is delivered in an aqueous solution. When an aqueous solution is used, the solution has sufficient ionic strength to solubilize antibody protein, yet is made palatable for oral administration. The delivery solution may also be buffered carbonate buffer pH 9.5) which can neutralize stomach acids and stabilize the antibodies when the antibodies are administered orally. In one embodiment the delivery solution is an aqueous solution. In another embodiment the delivery solution is a nutritional formula. Preferably, the delivery solution is infant formula. Yet another embodiment contemplates the delivery of lyophilized antibody encapsulated or microencapsulated inside acid-resistant compounds.
-42- Methods of applying enteric coatings to pharmaceutical compounds are well known to the art [companies specializing in the coating of pharmaceutical compounds are available; for example. The Coating Place (Verona. WI) and AAI (Wilmington. Enteric coatings which are resistant to gastric fluid and whose release dissolution of the coating to release the pharmaceutical compound) is pH dependent are commercially available [for example, the polymethacrylates Eudragit® L and Eudragit® S (Rihm GmbH)]. Eudragit® S is soluble in intestinal fluid from pH 7.0: this coating can be used to microencapsulate lyophilized antitoxin antibodies and the particles are suspended in a solution having a pH above or below pH for oral administration. The microparticles remain intact and undissolved until they reach the intestines where the intestinal pH causes them to dissolve thereby releasing the antitoxin.
The invention is directed to the improvement of the enteric stability of the therapeutic antitoxin [Enteric stability is defined as the stability of the antitoxin during passage through the gastrointestinal tract: the enteric stability is improved by increasing the amount of the orally administered antitoxin which is delivered to the desired site the intestines) in a S* 15 functional or active form]. Antibodies. and avian antibodies in particular. are known to be significantly denatured when exposed to acidic solutions gastric fluid). Denaturation of the antibody results in the loss of functionality loss of the ability to bind to the specific target). In addition to the denaturation of antibodies due to the low pH found in portions of the gastrointestinal tract proteolytic degradation of the antitoxin may occur due to digestion 20 with enzymes. The invention improves the enteric stability of the therapeutic antitoxins by coating the antitoxins with an enteric coating. The enteric coating prevents the acid-induced denaturation of the antitoxin and prevents exposure of the antitoxin to enzymes present in the upper portions of the gastrointestinal tract.
Application of acid resistant enteric coatings are shown herein to prevent release of microencapsulated antitoxin enterically-coated antitoxin) into simulated gastric solution while permitting release of the antitoxin in simulated intestinal solution. The enteric survival of the therapeutic antitoxins may also be improved through the use of excipients (more or less inert substances added to a therapeutic compound as a diluent or to give form or consistency when the compound is provided in tablet form). Excipients. such as carbonate buffers of about pH 9.5 or nutritional formulations Ensure®. Enfamil®. etc.) may indirectly reduce the denaturation of the antitoxin in the stomach by raising the stomach pH or by providing additional protein to compete for degradation by gastric enzymes.. In contrast. the use of enteric coatings on the antitoxin composition directly prevents the denaturation or digestion of 43 the antitoxin in the stomach by preventing the release of the antitoxin from the entericallycoated particle until the particle reaches the intestinal fluid which has a basic pH. The use of enteric coatings is a particularly preferred means of improving the acid stability of the therapeutic antitoxins of the invention.
The invention contemplates a method of treatment which can be administered for treatment of acute intoxication. In one embodiment, antitoxin is administered orally in either a delivery solution or in tablet form. in therapeutic dosage, to a subject intoxicated by the bacterial toxin which served as immunogen for the antitoxin.
The invention also contemplates a method of treatment which can be administered prophylactically. In one embodiment, antitoxin is administered orally, in a delivery solution.
in therapeutic dosage, to a subject, to prevent intoxication of the subject by the bacterial toxin which served as immunogen for the production of antitoxin. In another embodiment antitoxin is administered orally in solid form such as tablets or as microencapsulated particles.
Microencapsulation of lyophilized antibody using compounds such as Eudragit® (Rohm Tech, 15 Inc.) or polyethylene glycol. which dissolve at a wide range of pH units. allows the oral administration of solid antitoxin in a liquid form a suspension) to recipients unable to tolerate administration of tablets children or patients on feeding tubes). In a preferred embodiment, the lyophilized antibody is coated with Eudragit® L30D (R6hm Tech. Inc.). In one preferred embodiment the subject is an child. In another embodiment, antibody raised against whole bacterial organism is administered orally to a subject, in a delivery solution. in therapeutic dosage.
V. Vaccines Against Clostridial Species The invention contemplates the generation of mono- and multivalent vaccines for the protection of an animal (particularly humans) against several clostridial species. Of particular interest are vaccines which stimulate the production of a humoral immune response to C difficile. C. tetani and C botulinum in humans. The antigens comprising the vaccine preparation may be native or recombinantly produced toxin proteins from the clostridial species listed above. When toxin proteins are used as immunogens they are generally modified to reduce the toxicity. This modification may be by chemical or genetic recombinant DNA technology) means. In general genetic detoxification the expression of nontoxic fragments in a host cell) is preferred as the expression of nontoxic fragments in a host cell precludes the presence of intact. active toxin in the final preparation. However.
-44when chemical modification is desired, the preferred toxin modification is formaldehyde treatment.
The invention contemplates that recombinant C difficile toxin proteins be used as antigens in mono- and multivalent vaccine preparations. Soluble. substantially endotoxin-free recombinant C difficile toxin A and or toxin B proteins may be used alone or in conjunction with either recombinant or native toxins or toxoids from C. botulinum. C. difficile and C tetani as antigens for the preparation of these mono- and multivalent vaccines. It is contemplated that. due to the structural similarity of C. botulinum and C. tetani toxin proteins.
a vaccine comprising C. difficile and botulinum toxin proteins (native or recombinant or a mixture thereof) be used to stimulate an immune response against C. botulinum. C. tetani and C. difficile.
The adverse consequences of exposure to C difficile toxins would be avoided by immunization of subjects at risk of exposure to the toxin with nontoxic preparations which confer immunity such as chemically or genetically detoxified toxin.
15 Vaccines which confer immunity against one or more of the toxin types A and B would be useful as a means of protecting animals, including humans. from the deleterious effects of C difficile toxins. A subject may be immunized with compositions comprising one or more C difficile toxin proteins to generate neutralizing antibodies in the subject. A subject tnay be immunized with a first immunogen comprising C difficile toxin A proteins followed S 20 by a separate immunization with a second immunogen comprising C. difficile B toxin proteins to produce neutralizing antibodies directed against C difficile toxins A and B. Alternatively.
the subject may be immunized with a single immunogen comprising C difficile toxin A and B proteins.
In general, chemical detoxification of bacterial toxins using agents such as formaldehyde, glutaraldehyde or hydrogen peroxide is not optimal for the generation of vaccines or antitoxins. A delicate balance must be struck between too much and too little chemical modification. If the treatment is insufficient, the vaccine may retain residual toxicity. If the treatment is too excessive, the vaccine may lose potency due to destruction of native immunogenic determinants. Another major limitation of using botulinal toxoids for the generation of antitoxins or vaccines is the high production expense. For the above reasons.
the development of methods for the production of nontoxic but immunogenic C. difficile toxin proteins is desirable.
Recombinant C. difficile toxin proteins have be produced in a host cell such as E. coli in either a soluble or insoluble form. Insoluble recombinant proteins are found in inclusion bodies. Inclusion body protein must be solubilized prior to purification and/or administration to a host. The harsh treatment of inclusion body protein needed to accomplish this solubilization may reduce the immunogenicity of the purified protein. Ideally, recombinant proteins to be used as vaccines are expressed as soluble proteins at high levels greater than or equal to about 0.75% of total cellular protein) in E. coli or other host cells. This facilitates the production and isolation of sufficient quantities of the immunogen in a highly purified form substantially free of endotoxin or other pyrogen contamination). The ability to express recombinant toxin proteins as soluble proteins in E. coli is advantageous due to the low cost of growth compared to insect or mammalian tissue culture cells.
The subject invention provides soluble C difficile toxin proteins produced in economical host cells E. coli). Further. methods for the isolation of purified soluble C.
difficile toxin proteins which are suitable for immunization of humans and other animals are 15 provided. These soluble. purified preparations of C difficile toxin proteins provide the basis for improved vaccine preparations and facilitate the production of antitoxin.
When recombinant clostridial toxin proteins produced in gram-negative bacteria E. col) are used as vaccines, they are purified to remove endotoxin prior to administration to a host animal. In order to vaccinate a host, an immunogenically-effective amount-of purified substantially endotoxin-free recombinant clostridial toxin protein is administered in any of a number of physiologically acceptable carriers known to the art. When administered for the So purpose of vaccination, the purified substantially endotoxin-free recombinant clostridial toxin protein may be used alone or in conjunction with known adjutants. including potassium alum.
o, aluminum phosphate. aluminum hydroxide. Gerbu adjuvant (GMDP; C.C. Biotech Corp.), RIBI adjuvant (MPL: RIBI Immunochemical Research. Inc.). QS21 (Cambridge Biotech).
The alum and aluminum-based adjutants are particularly preferred when vaccines are to be' administered to humans. The route of immunization may be nasal. oral. intramuscular, intraperitoneal or subcutaneous.
The invention contemplates the use of soluble, substantially endotoxin-free preparations of fusion proteins comprising portions of C difficile toxins A and B as vaccines.
In one embodiment, the vaccine comprises a portion of a C. difficile toxin and a poly-histidine tract (also called a histidine tag). In a particularly preferred embodiment. a fusion protein comprising a portion of a C. difficile toxin protein and a poly-histidine tract is expressed -46 using the pET series of expression vectors (Novagen). The pET expression system utilizes a vector containing the T7 promoter which encodes the fusion protein and a host cell which can be induced to express the T7 DNA polymerase a DE3 host strain). The production of C difficile toxin fusion proteins containing a histidine tract is not limited to the use of a particular expression vector and host strain. Several commercially available expression vectors and host strains can be used to express the C difficile protein sequences as a fusion protein containing a histidine tract (For example, the pQE series (pQE-8. 12. 16, 17, 18, 31, 32. 40. 41. 42. 50. 51. 52. 60 and 70) of expression vectors (Qiagen) which are used with the host strains M15[pREP4] (Qiagen) and SG13009[pREP4] (Qiagen) can be used to express fusion proteins containing six histidine residues at the amino-terminus of the fusion protein).
VI. Detection Of Toxin The invention contemplates detecting bacterial toxin in a sample. The term "sample" in the present specification and claims is used in its broadest sense. On the one hand it is 15 meant to include a specimen or culture. On the other hand. it is meant to include both *biological and environmental samples.
Biological samples may be animal, including human, fluid, solid stool) or tissue; liquid and solid food products and ingredients such as dairy items, vegetables, meat and meat by-products, and waste. Environmental samples include environmental material such as surface matter. soil, water and industrial samples. as well as samples obtained from food and dairy processing instruments. apparatus. equipment. disposable and non-disposable items.
These examples are not to be construed as limiting the sample types applicable to the present invention.
As discussed above in section IV. toxin-associated diseases are medical emergencies which mandate immediate treatment. Because existing methodologies do not provide rapid, quantitative tests for the presence of C difficile toxins or organisms, treatment of subjects suspected of having C difficile associated disease is begun prior to a determination of the amount or nature of the toxin or organism present. If a rapid and quantitative test for C difficile toxins or organisms were available, the dosage of therapeutic compounds could be adjusted to provide maximum benefit to the intoxicated subject. The specific anti-C. difficile toxin A and B antibodies of the invention and the purified recombinant C. difficile toxin A and B proteins enable rapid and quantitative tests for C. difficile toxins or organisms.
-47 The invention contemplates detecting bacterial toxin by a competitive immunoassay method that utilizes recombinant toxin A and toxin B proteins, antibodies raised against recombinant bacterial toxin proteins. A fixed amount of the recombinant toxin proteins are immobilized to a solid support a microtiter plate) followed by the addition of a biological sample suspected of containing a bacterial toxin. The biological sample is first mixed with affinity-purified or PEG fractionated antibodies directed against the recombinant toxin protein. A reporter reagent is then added which is capable of detecting the presence of antibody bound to the immobilized toxin protein. The reporter substance may comprise an antibody with binding specificity for the antitoxin attached to a molecule which is used to S. 10 identify the presence of the reporter substance. If-toxin is present in the sample, this toxin i will compete with the immobilized recombinant toxin protein for binding to the antirecombinant antibody thereby reducing the signal obtained following the addition of the reporter reagent. A control is employed where the antibody is not mixed with the sample.
This gives the highest (or reference) signal.
The invention also contemplates detecting bacterial toxin by a "sandwich" immunoassay method that utilizes antibodies directed against recombinant bacterial toxin proteins. Affinity-purified antibodies directed against recombinant bacterial toxin proteins are immobilized to a solid support microtiter plates). Biological samples suspected of containing bacterial toxins are then added followed by a washing step to remove substantially all unbound antitoxin. The biological sample is next exposed to the reporter substance. which binds to antitoxin and is then washed free of substantially all unbound reporter substance.
oe The reporter substance may comprise an antibody with binding specificity for the antitoxin attached to a molecule which is used to identify the presence of the reporter substance.
Identification of the reporter substance in the biological tissue indicates the presence of the bacterial toxin.
It is also contemplated that bacterial toxin be detected by pouring liquids soups and other fluid foods and feeds including nutritional supplements for humans and other animals) over immobilized antibody which is directed against the bacterial toxin. It is contemplated that the immobilized antibody will be present in or on such supports as cartridges, columns, beads. or any other solid support medium. In one embodiment, following the exposure of the liquid to the immobilized antibody, unbound toxin is substantially removed by washing. The exposure of the liquid is then exposed to a reporter substance which detects the presence of bound toxin. In a preferred embodiment the reporter substance -48is an enzyme. fluorescent dye. or radioactive compound attached to an antibody which is directed against the toxin in a "sandwich" immunoassay). It is also contemplated that the detection system will be developed as necessary the addition of enzyme substrate in enzyme systems; observation using fluorescent light for fluorescent dye systems; and quantitation of radioactivity for radioactive systems).
EXPERIMENTAL
The following examples serve to illustrate certain preferred embodiments and aspects of the present invention and are not to be construed as limiting the scope thereof.
10 In the disclosure which follows, the following abbreviations apply: *C (degrees Centigrade); rpm (revolutions per minute); BBS-Tween (borate buffered saline containing Tween); BSA (bovine serum albumin); ELISA (enzyme-linked immunosorbent assay); CFA (complete Freund's adjuvant); IFA (incomplete Freund's adjuvant); IgG (immunoglobulin
G);
IgY (immunoglobulin IM (intramuscular); IP (intraperitoneal); IV (intravenous or 15 intravascular); SC (subcutaneous); H,O (water); HCI (hydrochloric acid); LDoo (lethal dose for 100% of experimental animals); aa (amino acid); HPLC (high performance liquid chromatography); kD (kilodaltons); gm (grams); pg (micrograms); mg (milligrams); ng (nanograms); pl (microliters); ml (milliliters); mm (millimeters); nm (nanometers); p.m (micrometer); M (molar); mM (millimolar); MW (molecular weight): sec (seconds); min(s) (minute/minutes); hr(s) (hour/hours); MgCI, (magnesium chloride): NaCI (sodium chloride); S' Na,CO, (sodium carbonate): OD,, (optical density at 280 nm): OD, (optical density at 600 nm): PAGE (polyacrylamide gel electrophoresis); PBS [phosphate buffered saline (150 mM NaCI. 10 mM sodium phosphate buffer, pH PEG (polyethylene glycol): PMSF (phenylmethylsulfonyl fluoride); SDS (sodium dodecyl sulfate); Tris (tris(hydroxymethyl)aminomethane); Ensure® (Ensure®. Ross Laboratories. Columbus OH); Enfamil® (Enfamil®, Mead Johnson); w/v (weight to volume); v/v (volume to volume); Accurate Chemical (Accurate Chemical Scientific Corp., Westbury, NY); Amicon (Amicon, Inc., Beverly, MA); Amresco (Amresco, Inc., Solon, OH); ATCC (American Type Culture Collection. Rockville, MD); BBL (Baltimore Biologics Laboratory, (a division of Becton Dickinson). Cockeysville. MD); Becton Dickinson (Becton Dickinson Labware. Lincoln Park, NJ); BioRad (BioRad, Richmond, CA); Biotech (C-C Biotech Corp., Poway, CA); Charles River (Charles River Laboratories. Wilmington. MA): Cocalico (Cocalico Biologicals Inc..
Reamstown. PA): CytRx (CvtRx Corp., Norcross. GA): Falcon Baxter Healthcare Corp..
49 McGaw Park. IL and Becton Dickinson); FDA (Federal Food and Drug Administration); Fisher Biotech (Fisher Biotech. Springfield. NJ); GIBCO (Grand Island Biologic Company/BRL, Grand Island, NY); Gibco-BRL (Life Technologies, Inc., Gaithersburg, MD); Harlan Sprague Dawley (Harlan Sprague Dawley, Inc., Madison. WI); Mallincirodt (a division of Baxter Healthcare Corp., McGaw Park, IL); Millipore (Millipore Corp., Marlborough, MA); New England Biolabs (New England Biolabs. Inc.. Beverly, MA); Novagen (Novagen, Inc.. Madison. WI); Pharmacia (Pharmacia, Inc.. Piscataway, NJ); Qiagen (Qiagen, Chatsworth. CA): RIBI (RIBI Immunochemical Research. Inc.. Hamilton. MT); Sasco (Sasco. Omaha. NE); Showdex (Showa Denko America, Inc.. New York, NY): Sigma 10 (Sigma Chemical Co.. St. Louis. MO): Sterogene (Sterogene. Inc., Arcadia. CA); Tech Lab (Tech Lab. Inc.. Blacksburg. VA); and Vaxcell (Vaxcell, Inc.. a subsidiary of CytRX Corp., o o Norcross. GA).
When a recombinant protein is described in the specification it is referred to in a short-hand manner by the amino acids in the toxin sequence present in the recombinant 15 protein rounded to the nearest 10. For example, the recombinant protein pMB1850-2360 contains amino acids 1852 through 2362 of the C difficile toxin B protein. The specification gives detailed construction details for all recombinant proteins such that one skilled in the art will know precisely which amino acids are present in a given recombinant protein.
EXAMPLE 1 Production Of Hieh-Titer Antibodies To Clostridium difficile Organisms In A Hen Antibodies to certain pathogenic organisms have been shown to be effective in treating diseases caused by those organisms. It has not been shown whether antibodies can be raised, against Clostridium difficile. which would be effective in treating infection by this organism.
Accordingly, C. difficile was tested as immunogen for production of hen antibodies.
To determine the best course for raising high-titer egg antibodies against whole C difficile organisms, different immunizing strains and different immunizing concentrations were examined. The example involved preparation of the bacterial immunogen, immunization, purification of anti-bacterial chicken antibodies. and detection of anti-bacterial antibodies in the purified IgY preparations.
a) Preparation Of Bacterial Immunogen C. difficile strains 43594 (serogroup A) and 43596 (serogroup C) were originally obtained from the ATCC. These two strains were selected because they represent two of the most commonly-occurring serogroups isolated from patients with antibiotic-associated pseudomembranous colitis. [Delmee et al., J. Clin. Microbiol.. 28(10):2210 (1990).] Additionally, both of these strains have been previously characterized with respect to their virulence in the Syrian hamster model for C. difficile infection. [Delmee et al.. J. Med Microbiol., 33:85 (1990).] The bacterial strains were separately cultured on brain heart infusion agar for 48 hours 10 at 37 0 C in a Gas Pack 100 Jar (BBL. Cockeysville. MD) equipped with a Gas Pack Plus anaerobic envelope (BBL). Forty-eight hour cultures were used because they produce better growth and the organisms have been found to be more cross-reactive with respect to their surface antigen presentation. The greater the degree of cross-reactivity of our IgY preparations. the better the probability of a broad range of activity against different strains/serogroups. [Toma et al., J. Clin. Microbiol., 26(3):426 (1988).] The resulting organisms were removed from the agar surface using a sterile dacron-tip swab. and were suspended in a solution containing 0.4% formaldehyde in PBS, pH 7.2. This S. :concentration of formaldehyde has been reported as producing good results for the purpose of preparing whole-organism immunogen suspensions for the generation of polyclonal anti-C.
difficile antisera in rabbits. [Delmee et al., J. Clin. Microbiol.. 21:323 (1985); Davies et al., Microbial Path.. 9:141 (1990).] In this manner, two separate bacterial suspensions were prepared, one for each strain. The two suspensions were then incubated at 4 0 C for 1 hour.
Following this period of formalin-treatment, the suspensions were centrifuged at 4,200 x g for min.. and the resulting pellets were washed twice in normal saline. The washed pellets, which contained formalin-treated whole organisms, were resuspended in fresh normal saline such that the visual turbidity of each suspension corresponded to a #7 McFarland standard.
Edelstein, "Processing Clinical Specimens for Anaerobic Bacteria: Isolation and Identification Procedures," in S.M. Finegold et al Bailey and Scott's Diagnostic Microbiology, pp. 477-507, C.V. Mosby Co., (1990). The preparation of McFarland nephelometer standards and the corresponding approximate number of organisms for each tube are described in detail at pp. 172-173 of this volume.] Each of the two #7 suspensions was then split into two separate volumes. One volume of each suspension was volumetrically adjusted, by the addition of saline, to correspond to the visual turbidity of a #1 McFarland 51 standard. The #1 suspensions contained approximately 3x10' organisms/ml. and the #7 suspensions contained approximately 2x109 organisms/ml. The four resulting concentration-adjusted suspensions of formalin-treated C. difficile organisms were considered to be "bacterial immunogen suspensions." These suspensions were used immediately after preparation for the initial immunization. [See section The formalin-treatment procedure did not result in 100% non-viable bacteria in the immunogen suspensions. In order to increase the level of killing, the formalin concentration and length of treatment were both increased for subsequent immunogen preparations, as described below in Table 3. (Although viability was decreased with the stronger formalin 10 treatment. 100% inviability of the bacterial immunogen suspensions was not reached.) Also, in subsequent immunogen preparations, the formalin solutions were prepared in normal saline instead of PBS. At day 49. the day of the fifth immunization, the excess volumes of the four previous bacterial immunogen suspensions were stored frozen at -70 0 C for use during all subsequent immunizations.
b) Immunization For the initial immunization. 1.0 ml volumes of each of the four bacterial immunogen suspensions described above were separately emulsified in 1.2 ml volumes of CFA (GIBCO).
For each of the four emulsified immunogen suspensions. two four-month old White Leghorn 20 hens (pre-laying) were immunized. (It is not necessary to use pre-laying hens: actively-laying hens can also be utilized.) Each hen received a total volume of approximately 1.0 ml of a single emulsified immunogen suspension via four injections (two subcutaneous and two intramuscular) of approximately 250 pl per site. In this manner, a total of four different immunization combinations, using two hens per combination, were initiated for the purpose of evaluating both the effect of immunizing concentration on egg yolk antibody (IgY) production, and interstrain cross-reactivity of IgY raised against heterologous strains. The four immunization groups are summarized in Table 3.
52 9* TABLE 3 Immunization Groups GROUP IMMUNIZING
APPROXIMATE
DESIGNATION STRAIN IMMUNIZING DOSE CD 43594, #1 C. difficile 1.5 x 10' organisms/hen strain 43594 CD 43594. #7 1.0 x 109 organisms/hen CD 43596, #1 C. difficile 1.5 x 10' organisms/hen strain 43596 CD 43596, #7 1.0 x 10' organisms/hen The time point for the first series of immunizations was designated as "day zero." All subsequent immunizations were performed as described above except that the bacterial immunogen suspensions were emulsified using IFA (GIBCO) instead of CFA. and for the later time point immunization, the stored frozen suspensions were used instead of freshlyprepared suspensions. The immunization schedule used is listed in Table 4.
TABLE 4 Immunization Schedule DAY OF FORMALIN
IMMUNOGEN
IMMUNIZATION TREATMENT PREPARATION USED 0 1 hr. freshly-prepared 14 overnight 21 overnight 48 hrs.
49 72 hrs.
stored frozen 105 53c) Purification Of Anti-Bacterial Chicken Antibodies Groups of four eggs were collected per immunization group between days 80 and 84 post-initial immunization, and chicken immunoglobulin (IgY) was extracted according to a modification of the procedure of A. Poison et al., Immunol. Comm.. 9:495 (1980). A gentle stream of distilled water from a squirt bottle was used to separate the yolks from the whites, and the yolks were broken by dropping them through a funnel into a graduated cylinder. The four individual yolks were pooled for each group. The pooled, broken yolks were blended with 4 volumes of egg extraction buffer to improve antibody yield (egg extraction buffer is 0.01 M sodium phosphate, 0.1 M NaCI, pH 7.5. containing 0.005% thimerosal), and PEG 10 8000 (Amresco) was added to a concentration of When all the PEG dissolved, the protein precipitates that formed were pelleted by centrifugation at 13.000 x g for 10 minutes.
The supernatants were decanted and filtered through cheesecloth to remove the lipid layer.
and the PEG was added to the supematants to a final concentration of 12% (the supernatants were assumed to contain 3.5% PEG). After a second centrifugation, the supernatants were discarded and the pellets were centrifuged a final time to extrude the remaining PEG. These crude IgY pellets were then dissolved in the original yolk volume of egg extraction buffer and stored at 4°C. As an additional control, a preimmune IgY solution was prepared as described above, using eggs collected from unimmunized hens.
d) Detection Of Anti-Bacterial Antibodies In The Purified IgY Preparations In order to evaluate the relative levels of specific anti-C. difficile activity in the IgY preparations described above, a modified version of the whole-organism ELISA procedure of N.V. Padhye el al., J. Clin. Microbiol. 29:99-103 (1990) was used. Frozen organisms of both C difficile strains described above were thawed and diluted to a concentration of approximately 1 x 10 7 organisms/ml using PBS, pH 7.2. In this way, two separate coating suspensions were prepared, one for each immunizing strain. Into the wells of 96-well microtiter plates (Falcon. Pro-Bind Assay Plates) were placed 100 pl volumes of the coating suspensions. In this manner, each plate well received a total of approximately 1 x 106 organisms of one strain or the other. The plates were then incubated at 4 0 C overnight. The next morning, the coating suspensions were decanted, and all wells were washed three times using PBS. In order to block non-specific binding sites. 100 pl of 0.5% BSA (Sigma) in PBS was then added to each well. and the plates were incubated for 2 hours at room temperature.
54- The blocking solution was decanted. and 100 pl volumes of the IgY preparations described above were initially diluted 1:500 with a solution of 0.1% BSA in PBS. and then serially diluted in 1:5 steps. The following dilutions were placed in the wells: 1:500. 1:2.500.
1:62.5000, 1:312.500, and 1:1,562.500. The plates were again incubated for 2 hours at room temperature. Following this incubation, the IgY-containing solutions were decanted, and the wells were washed three times using BBS-Tween (0.1 M boric acid. 0.025 M sodium borate.
M NaCI. 0.1% Tween-20). followed by two washes using PBS-Tween and finally, two washes using PBS only. To each well. 100 pl of a 1:750 dilution of rabbit anti-chicken IgG (whole-molecule)-alkaline phosphatase conjugate (Sigma) (diluted in 0.1% 10 BSA in PBS) was added. The plates were again incubated for 2 hours at room temperature.
The conjugate solutions were decanted and the plates were washed as described above.
substituting 50 mM Na.CO,. pH 9.5 for the PBS in the final wash. The plates were developed by the addition of 100 1p of a solution containing 1 mg/ml para-nitrophenyl phosphate (Sigma) dissolved in 50 mM Na,CO,. 10 mM MgCl,. pH 9.5 to each well, and incubating the plates at room temperature in the dark for 45 minutes. The absorbance of each well was measured at 410 nm using a Dynatech MR 700 plate reader. In this manner, each of the four IgY preparations described above was tested for reactivity against both of the immunizing C. difficile strains: strain-specific, as well as cross-reactive activity was determined.
55 TABLE Results Of The Anti-C. difficile Whole-Organism
ELISA
IgY DILUTION OF 43594-COATED 43596-COATED PREPARATION IgY PREP WELLS
WELLS
1:500 1.746 1.801 1:2.500 1.092 1.670 1:12.500 0.202 0.812 CD 43594. #1 1:62.500 0.136 0.179 1:312.500 0.012 0.080 1:1.562.500 0.002 0.020 1:500 1.780 1.771 1:2.500 1.025 1.078 1:12.500 0.188 0.382 CD 43594. #7 1:62500 0.052 0.132 1:312.500 0.022 0.043 1:1.562.500 0.005 0.024 S. 5555
I
CD 43596. #1 1:500 1:2.500 1:12.500 1:62.500 1:312.500 1:1.562.500 1:500 1:2.500 1:12.500 1:62.500 1:312.500 1:1.562.500 CD 43596. #7 1.526 0.832 0.247 0.050 0.010 0.000 1.702 0.706 0.250 0.039 0.002 0.000 0.142 0.032 0.006 0.002 0.004 0.002 1.790 1.477 0.452 0.242 0.067 0.036 1.505 0.866 0.282 0.078 0.017 0.010 0.309 0.077 0.024 0.012 0.010 0.014 Preimmune IgY 1:500 1:2.500 1:12.500 1:62.500 1:312.500 1:1.562.500 I Table 5 shows the results of the whole-organism ELISA. All four IgY preparations demonstrated significant levels of activity, to a dilution of 1:62.500 or greater against both of the immunizing organism strains. Therefore. antibodies raised against one strain were highly cross-reactive with the other strain, and vice versa. The immunizing concentration of organisms did not have a significant effect on organism-specific IgY production. as both concentrations produced approximately equivalent responses. Therefore, the lower 56 immunizing concentration of approximately 1.5 x 10' organisms/hen is the preferred immunizing concentration of the two tested. The preimmune IgY preparation appeared to possess relatively low levels of C. difficile-reactive activity to a dilution of 1:500. probably due to prior exposure of the animals to environmental clostridia.
An initial whole-organism ELISA was performed using IgY preparations made from single CD 43594. #1 and CD 43596. #1 eggs collected around day 50 (data not shown).
Specific titers were found to be 5 to 10-fold lower than those reported in Table 5. These results demonstrate that it is possible to begin immunizing hens prior to the time that they begin to lay eggs. and to obtain high titer specific IgY from the first eggs that are laid. In 10 other words, it is not necessary to wait for the hens to begin laying before the immunization schedule is started.
EXAMPLE 2 Treatment Of C difficile Infection With Anti-C difficile Antibody In order to determine whether the immune IgY antibodies raised against whole C.
difficile organisms were capable of inhibiting the infection of hamsters by C. difficile.
hamsters infected by these bacteria were utilized. [Lyerly el al.. Infect. Immun.. 59:2215- 2218 (1991).] This example involved: determination of the lethal dose of C. difficile S 20 organisms: and treatment of infected animals with immune antibody or control antibody in nutritional solution.
So* a) Determination Of The Lethal Dose Of C difficile Organisms Determination of the lethal dose of C difficile organisms was carried out according to the model described by D.M. Lyerly et al.. Infect. Immun.. 59:2215-2218 (1991). C. difficile strain ATCC 43596 (serogroup C. ATCC) was plated on BHI agar and grown anaerobically (BBL Gas Pak 100 system) at 37 0 C for 42 hours. Organisms were removed from the agar 57surface using a sterile dacron-tip swab and suspended in sterile 0.9% NaCI solution to a density of 108 organisms/ml.
In order to determine the lethal dose of C. difficile in the presence of control antibody and nutritional formula. non-immune eggs were obtained from unimmunized hens and a 12% PEG preparation made as described in Example This preparation was redissolved in one fourth the original yolk volume of vanilla flavor Ensure®.
Starting on day one. groups of female Golden Syrian hamsters (Harlan Sprague Dawley), 8-9 weeks old and weighing approximately 100 gm, were orally administered 1 ml of the preimmune/Ensure® formula at time zero. 2 hours. 6 hours, and 10 hours. At 1 hour, animals were orally administered 3.0 mg clindamycin HCI (Sigma) in 1 ml of water. This drug predisposes hamsters to C difficile infection by altering the normal intestinal flora. On day two. the animals were given I ml of the preimmune IgY/Ensure® formula at time zero, 2 hours. 6 hours, and 10 hours. At 1 hour on day two. different groups of animals were inoculated orally with saline (control), or 102. 104. 10 or 10' C difficile organisms in 1 ml of saline. From days 3-12. animals were given I ml of the preimmune IgY/Ensure® formula three times daily and observed for the onset of diarrhea and death. Each animal was housed in an individual cage and was offered food and water ad libitum.
Administration of 10" 10* organisms resulted in death in 3-4 days while the lower doses of 102 10 organisms caused death in 5 days. Cecal swabs taken from dead animals 20 indicated the presence of C. difficile. Given the effectiveness of the 102 dose. this number of organisms was chosen for the following experiment to see if hyperimmune anti-C difficile antibody could block infection.
o b) Treatment Of Infected Animals With Immune Antibody Or Control Antibody In Nutritional Formula The experiment in was repeated using three groups of seven hamsters each. Group A received no clindamvcin or C difficile and was the survival control. Group B received clindamycin. 102 C difficile organisms and preimmune IgY on the same schedule as the animals in above. Group C received clindamycin. 10' C difficile organisms, and hyperimmune anti-C difficile IgY on the same schedule as Group B. The anti-C dificile IgY was prepared as described in Example 1 except that the 12% PEG preparation was dissolved in one fourth the original yolk volume of Ensure®.
58 All animals were observed for the onset of diarrhea or other disease symptoms and death. Each animal was housed in an individual cage and was offered food and water ad libitum. The results are shown in Table 6.
TABLE 6 The Effect Of Oral Feeding Of Hyperimmune IgY Antibody on C difficile Infection TIME TO TIME TO ANIMAL GROUP DIARRHEA' DEATH" 10 A pre-immune IgY only no diarrhea no deaths B Clindamycin. C difficile preimmune IgY 30 hrs. 49 hrs.
C Clindamycin. C. difficile. immune IgY 33 hrs. 56 hrs.
Mean of seven animals.
Hamsters in the control group A did not develop diarrhea and remained healthy during the experimental period. Hamsters in groups B and C developed diarrheal disease. Anti-C.
15 difficile IgY did not protect the animals from diarrhea or death, all animals succumbed in the same time interval as the animals treated with preimmune IgY. Thus. while immunization with whole organisms apparently can improve sub-lethal symptoms with particular bacteria (see U.S. Patent No. 5.080.895 to H. Tokoro). such an approach does not prove to be productive to protect against the lethal effects of C. difficile.
EXAMPLE 3 Production of C. botulinum Type A Antitoxin in Hens In order to determine whether antibodies could be raised against the toxin produced by clostridial pathogens, which would be effective in treating clostridial diseases, antitoxin to C botulinum type A toxin was produced. This example involves: toxin modification: (b) immunization: antitoxin collection; antigenicity assessment; and assay of antitoxin titer.
-59w a) Toxin Modification C. botulinum type A toxoid was obtained from B. R. DasGupta. From this, the active type A neurotoxin approximately 150 kD) was purified to greater than 99% purity, according to published methods. DasGupta V. Sathyamoorthy. Toxicon. 22:415 (1984).] The neurotoxin was detoxified with formaldehyde according to published methods.
Singh B.R. DasGupta. Toxicon. 27:403 (1989).] b) Immunization C. botulinum toxoid for immunization was dissolved in PBS (1 mg/ml) and was emulsified with an approximately equal volume of CFA (GIBCO) for initial immunization or IFA for booster immunization. On day zero. two white leghorn hens. obtained from local breeders. were each injected at multiple sites (intramuscular and subcutaneous) with I ml inactivated toxoid emulsified in 1 ml CFA. Subsequent booster immunizations were made according to the following schedule for day of injection and toxoid amount: days 14 and 21 0.5 mg: day 171 0.75 mg: days 394. 401. 409 0.25 mg. One hen received an additional booster of 0.150 mg on day 544.
c) Antitoxin Collection Total yolk immunoglobulin (IgY) was extracted as described in Example l(c) and the 20 IgY pellet was dissolved in the original yolk volume of PBS with thimerosal.
d) Antigenicity Assessment Eggs were collected from day 409 through day 423 to assess whether the toxoid was sufficiently immunogenic to raise antibody. Eggs from the two hens were pooled and antibody was collected as described in the standard PEG protocol. [Example Antigenicity of the botulinal toxin was assessed on Western blots. The 150 kD detoxified type A neurotoxin and unmodified. toxic. 300 kD botulinal type A complex (toxin used for intragastric route administration for animal gut neutralization experiments; see Example 6) were separated on a SDS-polyacrylamide reducing gel. The Western blot technique was performed according to the method of Towbin. Towbin et al.. Proc. Natl. Acad. Sci.
USA. 76:4350 (1979).] Ten lg samples of C botulinum complex and toxoid were dissolved in SDS reducing sample buffer SDS. 0.5% 2-mercaptoethanol. 50 mM Tris. pH 6.8. glycerol. 0.025% w/v bromphenol blue. 10% P-mercaptoethanol), heated at 95 0 C for 10 min and separated on a 1 mm thick 5% SDS-polyacrylamide gel. Weber and M.
Osbom."Proteins and Sodium Dodecvl Sulfate: Molecular Weight Determination on Polvacrvlamide Gels and Related Procedures." in The Proteins. 3d Edition Neurath R.L. Hill. eds), pp. 179-223. (Academic Press. NY. 1975).] Part of the gel was cut off and the proteins were stained with Coomassie Blue. The proteins in the remainder of the gel were transferred to nitrocellulose using the Milliblot-SDE electro-blotting system (Millipore) according to manufacturer's directions. The nitrocellulose was temporarily stained with Ponceau S Carroll and A. Laughon. "Production and Purification ofPolvclonal Antibodies to the Foreign Segment of 6-galactosidase Fusion Proteins." in DNA Cloning: A 10 Practical Approach. Vol.111. Glover, pp. 89-111. IRL Press. Oxford. (1987)] to visualize the lanes. then destained by running a gentle stream of distilled water over the blot for several minutes. The nitrocellulose was immersed in PBS containing 3% BSA overnight at 4°C to block any remaining protein binding sites.
The blot was cut into strips and each strip was incubated with the appropriate primary antibody. The avian anti-C hotulinum antibodies [described in and pre-immune chicken antibody (as control) were diluted 1:125 in PBS containing 1 mg/mi BSA for 2 hours at room temperature. The blots were washed with two changes each of large volumes of PBS, BBS- Tween and PBS. successively (10 min/wash). Goat anti-chicken lgG alkaline phosphatase conjugated secondary antibody (Fisher Biotech) was diluted 1:500 in PBS containing 1 mg/ml 20 BSA and incubated with the blot for 2 hours at room temperature. The blots were washed with two changes each of large volumes of PBS and BBS-Tween. followed by one change of PBS and 0.1 M Tris-HCi. pH 9.5. Blots were developed in freshly prepared alkaline phosphatase substrate buffer (100 pg/ml nitroblue tetrazolium (Sigma). 50 pg/ml 5-bromo-4chloro-3-indolyl phosphate (Sigma). 5 mM MgCl in 50 mM Na 2
CO
3 pH The Western blots are shown in Figure 1. The anti-C botulinum IgY reacted to the toxoid to give a broad immunoreactive band at about 145-150 kD on the reducing gel. This toxoid is refractive to disulfide cleavage by reducing agents due to formalin crosslinking. The immune IgY reacted with the active toxin complex, a 97 kD C. botulinum type A heavy chain and a 53 kD light chain. The preimmune IgY was unreactive to the C. botulinum complex or toxoid in the Western blot.
61 e) Antitoxin Antibody Titer The IgY antibody titer to C botulinum type A toxoid of eggs harvested between day 409 and 423 was evaluated by ELISA. prepared as follows. Ninety-six-well Falcon Pro-bind plates were coated overnight at 4°C with 100 pl/well toxoid Singh B.R. Das Gupta, Toxicon 27:403 (1989)] at 2.5 ig/ml in PBS. pH 7.5 containing 0.005% thimerosal. The following day the wells were blocked with PBS containing 1% BSA for 1 hour at 37 0 C. The IgY from immune or preimmune eggs was diluted in PBS containing 1% BSA and 0.05% Tween 20 and the plates were incubated for 1 hour at 37 0 C. The plates were washed three times with PBS containing 0.05% Tween 20 and three times with PBS alone. Alkaline phosphatase-conjugated goat-anti-chicken IgG (Fisher Biotech) was diluted 1:750 in PBS containing 1% BSA and 0.05% Tween 20. added to the plates. and incubated 1 hour at 37 0
C.
The plates were washed as before. and p-nitrophenyl phosphate (Sigma) at 1 mg/ml in 0.05 M NaCO,. pH 9.5. 10 mM MgC, was added.
The results are shown in Figure 2. Chickens immunized with the toxoid generated high titers of antibody to the immunogen. Importantly. eggs from both immunized hens had significant anti-immunogen antibody titers as compared to preimmune control eggs. The anti- C. botulinum IgY possessed significant activity, to a dilution of 1:93.750 or greater.
EXAMPLE 4 20 Preparation Of Avian Egg Yolk Immunoglobulin In An Orally Administrable Form SIn order to administer avian IgY antibodies orally to experimental mice. an effective delivery formula for the IgY had to be determined. The concern was that if the crude IgY was dissolved in PBS. the saline in PBS would dehydrate the mice. which might prove harmful over the duration of the study. Therefore, alternative methods of oral administration of IgY were tested. The example involved: isola-tion of immune IgY; solubilization of IgY in water or PBS. including subsequent dialysis of the IgY-PBS solution with water to eliminate or reduce the salts (salt and phosphate) in the buffer: and comparison of the quantity and activity of recovered IgY by absorbance at 280 nm and PAGE. and enzymelinked immunoassay (ELISA).
62 a) Isolation Of Immune IgY In order to investigate the most effective delivery formula for IgY. we used IgY which was raised against Crotalus durissus terrificus venom. Three eggs were collected from hens immunized with the C durissus terrificus venom and IgY was extracted from the yolks using the modified Poison procedure described by Thalley and Carroll [Bio/Technology, 8:934-938 (1990)] as described in Example 1(c).
The egg yolks were separated from the whites. pooled, and blended with four volumes of PBS. Powdered PEG 8000 was added to a concentration of The mixture was centrifuged at 10.000 rpm for 10 minutes to pellet the precipitated protein, and the supernatant was filtered through cheesecloth to remove the lipid layer. Powdered PEG 8000 was added to the supernatant to bring the final PEG concentration to 12% (assuming a PEG concentration of 3.5% in the supernatant). The 12% PEG/IgY mixture was divided into two equal volumes and centrifuged to pellet the IgY.
b) Solubilization Of The IgY In Water Or PBS One pellet was resuspended in 1/2 the original yolk volume of PBS. and the other pellet was resuspended in 1/2 the original yolk volume of water. The pellets were then centrifuged to remove any particles or insoluble material. The IgY in PBS solution dissolved readily but the fraction resuspended in water remained cloudy.
20 In order to satisfy anticipated sterility requirements for orally administered antibodies.
the antibody solution needs to be filter-sterilized (as an alternative to heat sterilization which would destroy the antibodies). The preparation of IgY resuspended in water was too cloudy to pass through either a 0.2.or 0.45 lpm membrane filter. so 10 ml of the PBS resuspended fraction was dialyzed overnight at room temperature against 250 ml of water. The following morning the dialysis chamber was emptied and refilled with 250 ml of fresh H0O for a second dialysis. Thereafter. the yields of soluble antibody were determined at OD 2 ,o and are compared in Table 7.
63- TABLE 7 Dependence Of IgY Yield On Solvents ABSORBANCE OF 1:10 PERCENT FRACTN DILUTION AT 280 nm RECOVERY PBS dissolved 1.149 100% H,O dissolved 0.706 61% PBS dissolved/H,O dialyzed 0.885 77% Resuspending the pellets in PBS followed by dialysis against water recovered more antibody than directly resuspending the pellets in water (77% versus Equivalent volumes of the IgY preparation in PBS or water were compared by PAGE. and these results were in accordance with the absorbance values (data not shown).
c) Activity Of IgY Prepared With Different Solvents An ELISA was performed to compare the binding activity of the IgY extracted by each procedure described above. C durissus lerrificus venom at 2.5 pg/ml in PBS was used to coat each well of a 96-well microtiter plate. The remaining protein binding sites were blocked with PBS containing 5 mg/ml BSA. Primary antibody dilutions (in PBS containing 1 mg/ml BSA) were added in duplicate. After 2 hours of incubation at room temperature. the unbound primary antibodies were removed by washing the wells with PBS.
BBS-Tween. and PBS. The species specific secondary antibody (goat anti-chicken immunoglobulin alkaline-phosphatase conjugate (Sigma) was diluted 1:750 in PBS containing 1 mg/ml BSA and added to each well of the microtiter plate. After 2 hours of incubation at room temperature. the unbound secondary antibody was removed by washing the plate as before, and freshly prepared alkaline phosphatase substrate (Sigma) at I mg/ml in 50 mM Na,CO 3 10 mM MgCl,. pH 9.5 was added to each well. The color development was measured on a Dynatech MR 700 microplate reader using a 412 nm filter. The results are shown in Table 8.
64 TABLE 8 Antigen-Binding Activity of IgY Prepared with Different Solvents PBS H,O DILUTION PREIMMUNE DISSOLVED DISSOLVED PBS/H,O 1:500 0.005 1.748 1.577 1.742 1:2.500 0.004 0.644 0.349 0.606 1:12.500 0.001 0.144 0.054 0.090 1:62.500 0.001 0.025 0.007 0.016 1:312.500 0.010 0.000 0.000 0.002
S..
S
*SSS
*5 a.
S.
*SSS
10 The binding assay results parallel the recovery values in Table 7. with PBS-dissolved IgY showing slightly more activity than the PBS-dissolved/HO dialyzed antibody. The water-dissolved antibody had considerably less binding activity than the other preparations.
EXAMPLE 15 Survival Of Antibody Activity After Passage Throuah The Gastrointestinal Tract In order to determine the feasibility of oral administration of antibody. it was of interest to determine whether orally administered IgY survived passage through the gastrointestinal tract. The example involved: oral administration of specific immune antibody mixed with a nutritional formula: and assay of antibody activity extracted from feces.
a) Oral Administration Of Antibody The IgY preparations used in this example are the same PBS-dissolved/HO0 dialyzed antivenom materials obtained in Example 4 above, mixed with an equal volume of Enfamil®.
Two mice were used in this experiment. each receiving a different diet as follows: 1) water and food as usual: 2) immune IgY preparation dialyzed against water and mixed 1:1 with Enfamil®.
(The mice were given the corresponding mixture as their only source of food and water).
65 b) Antibody Activity After Ingestion After both mice had ingested their respective fluids, each tube was refilled with approximately 10 ml of the appropriate fluid first thing in the morning. By mid-morning there was about 4 to 5 ml of liquid left in each tube. At this point stool samples were collected from each mouse, weighed, and dissolved in approximately 500 pl PBS per 100 mg stool sample. One hundred and sixty mg of control stools (no antibody) and 99 mg of experimental stools (specific antibody) in 1.5 ml microfuge tubes were dissolved in 800 and 500 il PBS. respectively. The samples were heated at 37*C for 10 minutes and vortexed vigorously. The experimental stools were also broken up with a narrow spatula. Each sample was centrifuged for 5 minutes in a microfuge and the supernatants. presumably containing the antibody extracts, were collected. The pellets were saved at 2-8°C in case future extracts were needed. Because the supernatants were tinted, they were diluted five-fold in PBS .containing 1 mg/ml BSA for the initial dilution in the enzyme immunoassay (ELISA). The primary extracts were then diluted five-fold serially from this initial dilution. The volume of primary extract added to each well was 190 pl. The ELISA was performed exactly as described in Example 4.
TABLE 9 Specific Antibody Activity After Passage Through the Gastrointestinal Tract 20 *5*2 CONTROL FECAL EXP. FECAL DILUTION PREIMMUNE IgY EXTRACT EXTRACT 1:5 <0 0.000 0.032 1:25 0.016 <0 0.016 1:125 <0 <0 0.009 1:625 <0 0.003 0.001 1:3125 <0 <0 0.000 There was some active antibody in the fecal extract from the mouse given the specific antibody in Enfamil® formula, but it was present at a very low level. Since the samples were assayed at an initial 1:5 dilution, the binding observed could have been higher with less dilute samples. Consequently. the mice were allowed to continue ingesting either regular food and -66water or the specific IgY in Enfamil® formula. as appropriate, so the assay could be repeated.
Another ELISA plate was coated overnight with 5 Vg/ml of C.dt. venom in PBS.
The following morning the ELISA plate was blocked with 5 mg/ml BSA. and the fecal samples were extracted as before, except that instead of heating the extracts at 37 0 C. the samples were kept on ice to limit proteolysis. The samples were assayed undiluted initially.
and in 5X serial dilutions thereafter. Otherwise the assay was carried out as before.
TABLE Specific Antibody Survives Passage Through The Gastrointestinal Tract
S
C
9*
CONTROL
DILUTION PREIMMUNE IgY EXTRACT EXP. EXTRACT undiluted 0.003 <0 0.379 1:5 <0 <0 0.071 1:25 0.000 <0 0.027 1:125 0.003 <0 0.017 1:625 0.000 <0 0.008 1:3125 0.002 <0 0.002 The experiment confirmed the previous results. with the antibody activity markedly higher. The control fecal extract showed no anti-Cd.t. activity, even undiluted. while the fecal extract from the anti-Cd.t. IgY/Enfamil®-fed mouse showed considerable anti-Cd.t.
activity. This experiment (and the previous experiment) clearly demonstrate that active IgY antibody survives passage through the mouse digestive tract. a finding with favorable implications for the success of IgY antibodies administered orally as a therapeutic or prophylactic.
EXAMPLE 6 In Vivo Neutralization Of Type C. botulinum Type A Neurotoxin By Avian Antitoxin Antibody This example demonstrated the ability of PEG-purified antitoxin, collected as described in Example 3. to neutralize the lethal effect of C botulinum neurotoxin type A in 67 mice. To determine the oral lethal dose (LDI) of toxin A. groups of BALB/c mice were given different doses of toxin per unit body weight (average body weight of 24 grams). For oral administration, toxin A complex. which contains the neurotoxin associated with other non-toxin proteins was used. This complex is markedly more toxic than purified neurotoxin S when given by the oral route. Ohishi et al.. Infect. Immun.. 16:106 (1977).] C botulinum toxin type A complex. obtained from Eric Johnson (University Of Wisconsin. Madison) was 250 pg/ml in 50 mM sodium citrate. pH 5.5. specific toxicity 3 x 107 mouse LDs0/mg with parenteral administration. Approximately 40-50 ng/gm body weight was usually fatal within 48 hours in mice maintained on conventional food and water. When mice were given a diet ad libitum of only Enfamil® the concentration needed to produce lethality was approximately 2.5 times higher (125 ng/gm body weight). Botulinal toxin concentrations of approximately 200 ng/gm body weight were fatal in mice fed Enfamil® containing preimmune IgY (resuspended in Enfamil® at the original yolk volume).
S. The oral LDso o of C botulinum toxin was also determined in mice that received known amounts of a mixture of preimmune IgY-Ensure® delivered orally through feeding needles. Using a 22 gauge feeding needle. mice were given 250 jil each of a preimmune IgY-Ensure® mixture (preimmune IgY dissolved in 1/4 original yolk volume) 1 hour before and 1/2 hour and 5 hours after administering botulinal toxin. Toxin concentrations given orally ranged from approximately 12 to 312 ng/gm body weight (0.3 to 7.5 jlg per mouse).
20 Botulinal toxin complex concentration of approximately 40 ng/gm body weight (1 pg per mouse) was lethal in all mice in less than 36 hours.
Two groups of BALB/c mice. 10 per group, were each given orally a single dose of I pg each of botulinal toxin complex in 100 pi of 50 mM sodium citrate pH 5.5. The mice received 250 pl treatments of a mixture of either preimmune or immune IgY in Ensure® (1/4 original yolk volume) 1 hour before and 1/2 hour. 4 hours, and 8 hours after botulinal toxin administration. The mice received three treatments per day for two more days. The mice were observed for 96 hours. The survival and mortality are shown in Table 11.
68- TABLE 11 Neutralization Of Botulinal Toxin A In Vivo TOXIN DOSE NUMBER OF NUMBER OF MICE ANTIBODY TYPE
DEAD
ng/gm MICE ALIVE D E A D 41.6 non-immune 0 41.6 anti-botulinal toxin 10 0 All mice treated with the preimmune IgY-Ensure® mixture died within 46 hours posttoxin administration. The average time of death in the mice was 32 hours post toxin administration. Treatments of preimmune IgY-Ensure® mixture did not continue beyond 24 hours due to extensive paralysis of the mouth in mice of this group. In contrast. all ten mice treated with the immune anti-botulinal toxin IgY-Ensure® mixture survived past 96 hours.
Only 4 mice in this group exhibited symptoms of botulism toxicity (two mice about 2 days after and two mice 4 days after toxin administration). These mice eventually died 5 and 6 15 days later. Six of the mice in this immune group displayed no adverse effects to the toxin and remained alive and healthy long term. Thus. the avian anti-botulinal toxin antibody -demonstrated very good protection from the lethal effects of the toxin in the experimental S* mice.
0 EXAMPLE 7 Production Of An Avian Antitoxin Against Clostridium difficile Toxin A ~Toxin A is a potent cytotoxin secreted by pathogenic strains of C. difficile, that plays a direct role in damaging gastrointestinal tissues. In more severe cases of C. difficile intoxication. pseudomembranous colitis can develop which may be fatal. This would be prevented by neutralizing the effects of this toxin in the gastrointestinal tract. As a first step.
antibodies were produced against a portion of the toxin. The example involved: (a) conjugation of a synthetic peptide of toxin A to bovine serum albumin: immunization of hens with the peptide-BSA conjugate: and detection of antitoxin peptide antibodies by
ELISA.
-69 a) Conjugation Of A Synthetic Peptide Of Toxin A To Bovine Serum Albumin The synthetic peptide CQTIDGKKYYFN-NH, was prepared commercially (Multiple Peptide Systems. San Diego, CA) and validated to be >80% pure by high-pressure liquid chromatography. The eleven amino acids following the cysteine residue represent a consensus sequence of a repeated amino acid sequence found in Toxin A. [Wren et al.. Infect. Immun..
59:3151-3155 (1991).] The cysteine was added to facilitate conjugation to carrier protein.
In order to prepare the carrier for conjugation. BSA (Sigma) was dissolved in 0.01 M NaPO,, pH 7.0 to a final concentration of 20 mg/ml and n-maleimidobenzoyl-Nhydroxysuccinimide ester (MBS: Pierce) was dissolved in N.N-dimethyl formamide to a concentration of 5 mg/ml. MBS solution. 0.51 ml. was added to 3.25 ml of the BSA solution and incubated for 30 minutes at room temperature with stirring every 5 minutes. The MBSactivated BSA was then purified by chromatography on a Bio-Gel P-10 column (Bio-Rad: ml bed volume) equilibrated with 50 mM NaPO,. pH 7.0 buffer. Peak fractions were pooled (6.0 mi).
Lyophilized toxin A peptide (20 mg) was added to the activated BSA mixture, stirred until the peptide dissolved and incubated 3 hours at room temperature. Within 20 minutes.
the reaction mixture became cloudy and precipitates formed. After 3 hours, the reaction mixture was centrifuged at 10.000 x g for 10 min and the supernatant analyzed for protein content. No significant protein could be detected at 280 nm. The conjugate precipitate was washed three times with PBS and stored at 4°C. A second conjugation was performed with 15 mg of activated BSA and 5 mg of peptide and the conjugates pooled and suspended at a peptide concentration of 10 mg/ml in 10 mM NaPO 4 pH 7.2.
S 25 b) Immunization Of Hens With Peptide Conjugate Two hens were each initially immunized on day zero by injection into two subcutaneous and two intramuscular sites with I mg of peptide conjugate that was emulsified in CFA (GIBCO). The hens were boosted on day 14 and day 21 with I mg of peptide conjugate emulsified in IFA (GIBCO).
c) Detection Of Antitoxin Peptide Antibodies By ELISA IgY was purified from two eggs obtained before immunization (pre-immune) and two eggs obtained 31 and 32 days after the initial immunization using PEG fractionation as described in Example 1.
Wells of a 96-well microtiter plate (Falcon Pro-Bind Assay Plate) were coated overnight at 4°C with 100 pLg/ml solution of the toxin A synthetic peptide in PBS. pH 7.2 prepared by dissolving 1 mg of the peptide in 1.0 ml of H,O and dilution of PBS. The preimmune and immune IgY preparations were diluted in a five-fold series in a buffer containing 1% PEG 8000 and 0.1% Tween-20 in PBS. pH 7.2. The wells were blocked for 2 hours at room temperature with 150 il of a solution containing 5% Carnation® nonfat dry milk and 1% PEG 8000 in PBS. pH 7.2. After incubation for 2 hours at room temperature. the wells were washed, secondary rabbit anti-chicken IgG-alkaline phosphatase S (1:750) added. the wells washed again and the color development obtained as described in Example 1. The results are shown in Table 12.
15 TABLE 12 Reactivity Of IgY With Toxin Peptide
S..
S
2 ABSORBANCE AT 410 nm DILUTION OF PEG PREP IMMUNE ANTI- PREIMMUNE PEPTIDE
PEPTIDE
1:100 0.013 0.253 1:500 0.004 0.039 1:2500 0.004 0.005 Clearly. the immune antibodies contain titers against this repeated epitope of toxin A.
EXAMPLE 8 Production Of Avian Antitoxins Against Clostridium difficile Native Toxins A and B To determine whether avian antibodies are effective for the neutralization of C.
difficile toxins, hens were immunized using native C difficile toxins A and B. The resulting egg yolk antibodies were then extracted and assessed for their ability to neutralize toxins A 71 and B in vitro. The Example involved preparation of the toxin immunogens. (b) immunization. purification of the antitoxins, and assay of toxin neutralization activity.
a) Preparation Of The Toxin Immunogens Both C. difficile native toxins A and B. and C difficile toxoids. prepared by the treatment of the native toxins with formaldehyde, were employed as immunogens. C. difficile toxoids A and B were prepared by a procedure which was modified from published methods (Ehrich et al., Infect. Immun. 28:1041 (1980). Separate solutions (in PBS) of native C.
difficile toxin A and toxin B (Tech Lab) were each adjusted to a concentration of 0.20 mg/ml.
and formaldehyde was added to a final concentration of The toxin/formaldehyde solutions were then incubated at 37 0 C for 40 hrs. Free formaldehyde was then removed from the resulting toxoid solutions by dialysis against PBS at 4"C. In previously published reports.
this dialysis step was not performed. Therefore. free formaldehyde must have been present in their toxoid preparations. The toxoid solutions were concentrated. using a Centriprep 15 concentrator unit (Amicon). to a final toxoid concentration of 4.0 mg/ml. The two resulting preparations were designated as toxoid A and toxoid B.
C difficile native toxins were prepared by concentrating stock solutions of toxin A and toxin B (Tech Lab. Inc), using Centriprep concentrator units (Amicon). to a final concentration of 4.0 mg/ml.
b) Immunization The first two immunizations were performed using the toxoid A and toxoid B immunogens described above. A total of 3 different immunization combinations were employed. For the first immunization group. 0.2 ml of toxoid A was emulsified in an equal 25 volume of Titer Max adjuvant (CytRx). Titer Max was used in order to conserve the amount of immunogen used. and to simplify the immunization procedure. This immunization group was designated "CTA." For the second immunization group. 0.1 ml of toxoid B was emulsified in an equal volume of Titer Max adjuvant. This group was designated "CTB." For the third immunization group. 0.2 ml of toxoid A was first mixed with 0.2 ml of toxoid B. and the resulting mixture was emulsified in 0.4 ml of Titer Max adjuvant. This group was designated "CTAB." In this way. three separate immunogen emulsions were prepared, with each emulsion containing a final concentration of 2.0 mg/ml of toxoid A (CTA) or toxoid B (CTB) or a mixture of 2.0 mg/ml toxoid A and 2.0 mg/ml toxoid B (CTAB).
72 On day 0. White Leghorn hens. obtained from a local breeder. were immunized as follows: Group CTA. Four hens were immunized, with each hen receiving 200Pg of toxoid A, via two intramuscular injections of 501l of CTA emulsion in the breast area.
Group CTB. One hen was immunized with 200pg of toxoid B. via two I.M. injections of 501l of CTB emulsion in the breast area. Group CTAB. Four hens were immunized, with each hen receiving a mixture containing 200lg of toxoid A and 200utg of toxoid B. via two I.M. injections of 100l l of CTAB emulsion in the breast area. The second immunization was performed 5 weeks later. on day 35. exactly as described for the first immunization above.
In order to determine whether hens previously immunized with C difficile toxoids could tolerate subsequent booster immunizations using native toxins, a single hen from group CTAB was immunized for a third time. this time using a mixture of the native toxin A and native toxin B described in section above (these toxins were not formaldehyde-treated, and were used in their active form). This was done in order to increase the amount (titer) and affinity of specific antitoxin antibody produced by the hen over that achieved by immunizing S 15 with toxoids only. On day 62. 0.1 ml of a toxin mixture was prepared which contained 200gg of native toxin A and 200pg of native toxin B. This toxin mixture was then emulsified in 0.1 ml of Titer Max adjuvant. A single CTAB hen was then immunized with the resulting immunogen emulsion. via two I.M. injections of 100l l each. into the breast area.
This hen was marked with a wing band. and observed for adverse effects for a period of approximately 1 week. after which time the hen appeared to be in good health.
Because the CTAB hen described above tolerated the booster immunization with native toxins A and B with no adverse effects. it was decided to boost the remaining hens with native toxin as well. On day 70. booster immunizations were performed as follows: Group CTA. A 0.2 ml volume of the 4 mg/ml native toxin A solution was emulsified in an equal volume of Titer Max adjuvant. Each of the 4 hens was then immunized with 200lg of native toxin A. as described for the toxoid A immunizations above. Group CTB. A 50tl volume of the 4 mg/ml native toxin B solution was emulsified in an equal volume of Titer Max adjuvant. The hen was then immunized with 200ug of native toxin B. as described for the toxoid B immunizations above. Group CTAB. A 0.15 ml volume of the 4 mg/ml native toxin A solution was first mixed with a 0.15 ml volume the 4 mg/ml native toxin B solution.
The resulting toxin mixture was then emulsified in 0.3 ml of Titer Max adjuvant. The 3 remaining hens (the hen with the wing band was not immunized this time) were then immunized with 200gg of native toxin A and 200pg of native toxin B as described for the.- 73 toxoid A+ toxoid B immunizations (CTAB) above. On day 85. all hens received a second booster immunization using native toxins, done exactly as described for the first boost with native toxins above.
All hens tolerated both booster immunizations with native toxins with no adverse effects. As previous literature references describe the use of formaldehyde-treated toxoids.
this is apparently the first time that any immunizations have been performed using native C.
difficile toxins.
c) Purification Of Antitoxins Eggs were collected from the hen in group CTB 10-12 days following the second immunization with toxoid (day 35 immunization described in section above), and from the hens in groups CTA and CTAB 20-21 days following the second immunization with toxoid.
To be used as a pre-immune (negative) control. eggs were also collected from unimmunized hens from the same flock. Egg yolk immunoglobulin (IgY) was extracted from the 4 groups of eggs as described in Example 1 and the final IgY pellets were solubilized in the original yolk volume of PBS without thimerosal. Importantly. thimerosal was excluded because it would have been toxic to the CHO cells used in the toxin neutralization assays described in section below.
d) Assay Of Toxin Neutralization Activity The toxin neutralization activity of the IgY solutions prepared in section above was determined using an assay system that was modified from published methods. [Ehrich et al..
Infect. Immun. 28:1041-1043 (1992): and McGee et al. Microb. Path. 12:333-341 (1992).] As additional controls. affinity-purified goat anti-C difficile toxin A (Tech Lab) and affinity- 25 purified goat anti-C difficile toxin B (Tech Lab) were also assayed for toxin neutralization activity.
The IgY solutions and goat antibodies were serially diluted using F 12 medium (GIBCO) which was supplemented with 2% FCS (GIBCO)(this solution will be referred to as "medium" for the remainder of this Example). The resulting antibody solutions were then mixed with a standardized concentration of either native C difficile toxin A (Tech Lab), or native C difficile toxin B (Tech Lab). at the concentrations indicated below. Following incubation at 37 0 C for 60 min.. 100ul volumes of the toxin antibody mixtures were added to the wells of 96-well microtiter plates (Falcon Microtest III) which contained 2.5 x 104 74 W Chinese Hamster Ovary (CHO) cells per well (the CHO cells were plated on the previous day to allow them to adhere to the plate wells). The final concentration of toxin, or dilution of antibody indicated below refers to the final test concentration of each reagent present in the respective microtiter plate wells. Toxin reference wells were prepared which contained CHO cells and toxin A or toxin B at the same concentration used for the toxin plus antibody mixtures (these wells contained no antibody). Separate control wells were also prepared which contained CHO cells and medium only. The assay plates were then incubated for 18- 24 hrs. in a 37*C. humidified. 5% CO, incubator. On the following day. the remaining adherent (viable) cells in the plate wells were stained using 0.2% crystal violet (Mallinckrodt) dissolved in 2% ethanol. for 10 min. Excess stain was then removed by rinsing with water.
and the stained cells were solubilized by adding 100p.l of 1% SDS (dissolved in water) to each well. The absorbance of each well was then measured at 570 nm. and the percent cvtotoxicity of each test sample or mixture was calculated using the following formula: o CHO Cell Cytotoxicity [1 Abs. C ol X 100 Abs. Control Unlike previous reports which quantitate results visually by counting cell rounding by "microscopy. this Example utilized spectrophotometric methods to quantitate the C. difficile toxin bioassay. In order to determine the toxin A neutralizing activity of the CTA. CTAB.
and pre-immune IgY preparations. as well as the affinity-purified goat antitoxin A control.
dilutions of these antibodies were reacted against a 0.1 pg/ml concentration of native toxin A S 20 (this is the approx. 50% cytotoxic dose of toxin A in this assay system). The results are shown in Figure 3.
Complete neutralization of toxin A occurred with the CTA IgY (antitoxin A. above) at dilutions of 1:80 and lower, while significant neutralization occurred out to the 1:320 dilution.
The CTAB IgY (antitoxin A toxin B. above) demonstrated complete neutralization at the 1:320-1:160 and lower dilutions, and significant neutralization occurred out to the 1:1280 dilution. The commercially available affinity-purified goat antitoxin A did not completely neutralize toxin A at any of the dilutions tested, but demonstrated significant neutralization out to a dilution of 1:1.280. The preimmune IgY did not show any toxin A neutralizing activity at any of the concentrations tested. These results demonstrate that IgY purified from eggs laid by hens immunized with toxin A alone, or simultaneously with toxin A and toxin B.
is an effective toxin A antitoxin.
1W The toxin B neutralizing activity of the CTAB and pre-immune IgY preparations, and also the affinity-purified goat antitoxin B control was determined by reacting dilutions of these antibodies against a concentration of native toxin B of 0.1 ng/ml (approximately the cyxotoxic dose of toxin B in the assay system). The results are shown in Figure 4.
Complete neutralization of toxin B occurred with the CTAB IgY (antitoxin A toxin B. above) at the 1:40 and lower dilutions, and significant neutralization occurred out to the 1:320 dilution. The affinity-purified goat antitoxin B demonstrated complete neutralization at dilutions of 1:640 and lower, and significant neutralization occurred out to a dilution of 1:2.560. The preimmune IgY did not show any toxin B neutralizing activity at any of the concentrations tested. These results demonstrate that IgY purified from eggs laid by hens immunized simultaneously with toxin A and toxin B is an effective toxin B antitoxin.
In a separate study. the toxin B neutralizing activity of CTB. CTAB. and pre-immune IgY preparations was determined by reacting dilutions of these antibodies against a native toxin B concentration of 0.1 lug/ml (approximately 100% cytotoxic dose of toxin B in this S 15 assay system). The results are shown'in Figure SSignificant neutralization of toxin B occurred with the CTB IgY (antitoxin B. above) ""at dilutions of 1:80 and lower, while the CTAB IgY (antitoxin A toxin B. above) was found to have significant neutralizing activity at dilutions of 1:40 and lower. The preimmune IgY did not show any toxin B neutralizing activity at any of the concentrations tested. These results demonstrate that IgY purified from eggs laid by hens immunized with toxin B alone.
or simultaneously with toxin A and toxin B. is an effective toxin B antitoxin.
EXAMPLE 9 In vivo Protection Of Golden Syrian Hamsters From S 25 C. difficile Disease By Avian Antitoxins Atainst C difficile Toxins A and B The most extensively used animal model to study C. difficile disease is the hamster.
[Lyerly et al., Infect. Immun. 47:349-352 (1992).] Several other animal models for antibiotic-induced diarrhea exist. but none mimic the human form of the disease as closely as the hamster model. Fekety. "Animal Models of Antibiotic-Induced Colitis." in O. Zak and M. Sande Experimental Models in Antimicrobial Chemotherapy. Vol. 2. pp.
6 1-72, (1986).] In this model. the animals are first predisposed to the disease by the oral administration of an antibiotic. such as clindamycin. which alters the population of normallyoccurring gastrointestinal flora (Fekety. at 61-72). Following the oral challenge of these 76- W animals with viable C difficile organisms, the hamsters develop cecitis. and hemorrhage.
ulceration. and inflammation are evident in the intestinal mucosa. [Lyerly et al.. Infect.
Immun. 47:349-352 (1985).] The animals become lethargic, develop severe diarrhea, and a high percentage of them die from the disease. [Lyerly et Infect. lmmun. 47:349-352 (1985).] This model is therefore ideally suited for the evaluation of therapeutic agents designed for the treatment or prophylaxis of C. difficile disease.
The ability of the avian C difficile antitoxins, described in Example 1 above, to protect hamsters from C difficile disease was evaluated using the Golden Syrian hamster model of C. dificile infection. The Example involved preparation of the avian C difficile antitoxins. in vivo protection of hamsters from C. difficile disease by treatment with avian antitoxins, and long-term survival of treated hamsters.
a) Preparation Of The Avian C. difficile Antitoxins :i Eggs were collected from hens in groups CTA and CTAB described in Example 1 (b) above. To be used as a pre-immune (negative) control, eggs were also purchased from a local S: supermarket. Egg yolk immunoglobulin (IgY) was extracted from the 3 groups of eggs as described in Example 1 and the final IgY pellets were solubilized in one fourth the original yolk volume of Ensure® nutritional formula.
b) In vivo Protection Of Hamsters Against C difficile Disease By Treatment With Avian Antitoxins The avian C. difficile antitoxins prepared in section above were evaluated for their ability to protect hamsters from C difficile disease using an animal model system which was modified from published procedures. [Fekety. at 61-72: Borriello et al.. J. Med. Microbiol..
25 24:53-64 (1987); Kim et al.. Infect. Immun.. 55:2984-2992 (1987): Borriello et al.. J. Med.
Microbiol.. 25:191-196 (1988): Delmee and Avesani. J. Med. Microbiol.. 33:85-90 (1990); and Lyerly et al.. Infect. Immun.. 59:2215-2218 (1991).] For the study, three separate experimental groups were used. with each group consisting of 7 female Golden Syrian hamsters (Charles River). approximately 10 weeks old and weighing approximately 100 gms.
each. The three groups were designated "CTA," "CTAB" and "Pre-immune." These designations corresponded to the antitoxin preparations with which the animals in each group were treated. Each animal was housed in an individual cage. and was offered food and water ad libitum through the entire length of the study. On day 1, each animal was orally 77administered 1.0 ml of one of the three antitoxin preparations (prepared in section above) at the following timepoints: 0 hrs.. 4 hrs.. and 8 hrs. On day 2. the day 1 treatment was repeated. On day 3, at the 0 hr. timepoint. each animal was again administered antitoxin, as described above. At 1 hr.. each animal was orally administered 3.0 mg of clindamycin-HCl (Sigma) in 1 ml of water. This treatment predisposed the animals to infection with C difficile. As a control for possible endogenous C difficile colonization, an additional animal from the same shipment (untreated) was also administered 3.0 mg of clindamycin-HCI in the same manner. This clindamycin control animal was left untreated (and uninfected) for the remainder of the study. At the 4 hr. and 8 hr. timepoints, the animals were administered antitoxin as described above. On day 4. at the 0 hr. timepoint. each animal was again administered antitoxin as described above. At 1 hr.. each animal was orally challenged with 1 ml of C difficile inoculum. which contained approx. 100 C difficile strain 43596 organisms o in sterile saline. C difficile strain 43596. which is a serogroup C strain, was chosen because it is representative of one of the most frequently-occurring serogroups isolated from patients 15 with antibiotic-associated pseudomembranous colitis. [Delmee et al.. J. Clin. Microbiol..
28:2210-2214 (1985).] In addition. this strain has been previously demonstrated to be virulent in the hamster model of infection. [Delmee and Avesani. J. Med. Microbiol.. 33:85-90 (1990).] At the 4 hr. and 8 hr. timepoints, the animals were'administered antitoxin as described above. On days 5 through 13. the animals were administered antitoxin 3x per day as described for day I above, and observed for the onset of diarrhea and death. On the morning of day 14. the final results of the study were tabulated. These results are shown in Table 13.
TABLE 13 Treatment Results SNo. Animals No. Animals Treatment Group Surviving Dead Pre-Immune 1 6 CTA (Antitoxin A only) 5 2 CTAB (Antitoxin A Antitoxin B) 7 0 78 Representative animals from those that died in the Pre-Immune and CTA groups were necropsied. Viable C difficile organisms were cultured from the ceca of these animals, and the gross pathology of the gastrointestinal tracts of these animals was consistent with that expected for C. difficile disease (inflamed. distended. hemorrhagic cecum. filled with watery diarrhea-like material). In addition, the clindamycin control animal remained healthy throughout the entire study period, therefore indicating that the hamsters used in the study had not previously been colonized with endogenous C difficile organisms prior to the start of the study. Following the final antitoxin treatment on day 13. a single surviving animal from the CTA group. and also from the CTAB group. was sacrificed and necropsied. No pathology was noted in either animal.
Treatment of hamsters with orally-administered toxin A and toxin B antitoxin (group CTAB) successfully protected 7 out of 7 (100%) of the animals from C difficile disease.
Treatment of hamsters with orally-administered toxin A antitoxin (group CTA) protected out of 7 of these animals from C difficile disease. Treatment using pre-immune IgY 15 was not protective against C difficile disease. as only I out of 7 of these animals survived. These results demonstrate that the avian toxin A antitoxin and the avian toxin A toxin B antitoxin effectively protected the hamsters from C. difficile disease. These results also suggest that although the neutralization of toxin A alone-confers some degree of protection against C difficile disease. in order to achieve maximal protection. simultaneous 20 antitoxin A and antitoxin B activity is necessary.
c) Long-Term Survival Of Treated Hamsters It has been previously reported in the literature that hamsters treated with orallyadministered bovine antitoxin IgG concentrate are protected from C. difficile disease as long 25 as the treatment is continued, but when the treatment is stopped. the animals develop diarrhea and subsequently die within 72 hrs. [Lyerly et al.. Infect. Immun.. 59(6):2215-2218 (1991).] In order to determine whether treatment of C difficile disease using avian antitoxins promotes long-term survival following the discontinuation of treatment, the 4 surviving animals in group CTA. and the 6 surviving animals in group CTAB were observed for a period of 11 days (264 hrs.) following the discontinuation of antitoxin treatment described in section above. All hamsters remained healthy through the entire post-treatment period.
This result demonstrates that not only does treatment with avian antitoxin protect against the 79 onset of C difficile disease it is effective as a prophylactic). it also promotes long-term survival beyond the treatment period, and thus provides a lasting cure.
EXAMPLE In vivo Treatment Of Established C difficile Infection In Golden Syrian Hamsters With Avian Antitoxins Against C difficile Toxins A and B The ability of the avian C difficile antitoxins, described in Example 8 above, to treat an established C. difficile infection was evaluated using the Golden Syrian hamster model.
The Example involved preparation of the avian C. difficile antitoxins. in vivo treatment of hamsters with established C difficile infection, and histologic evaluation of cecal tissue.
a) Preparation Of The Avian C. difficile Antitoxins Eggs were collected from hens in group CTAB described in Example 8 above.
15 which were immunized with C difficile toxoids and native toxins A and B. Eggs purchased from a local supermarket were used as a pre-immune (negative) control. Egg yolk :'immunoglobulin (IgY) was extracted from the 2 groups of eggs as described in Example 1 and the final IgY pellets were solubilized in one-fourth the original yolk volume of Ensure® nutritional formula.
b) In vivo Treatment Of Hamsters With Established C. difficile Infection The avian C difficile antitoxins prepared in section above were evaluated for the ability to treat established C difficile infection in hamsters using an animal model system 25 which was modified from the procedure which was described for the hamster protection study .00 in Example 8(b) above.
For the study, four separate experimental groups were used. with each group consisting of 7 female Golden Syrian hamsters (Charles River), approx. 10 weeks old. weighing approximately 100 gms. each. Each animal was housed separately. and was offered food and water ad libitum through the entire length of the study.
On day 1 of the study. the animals in all four groups were each predisposed to C.
difficile infection by the oral administration of 3.0 mg of clindamycin-HCI (Sigma) in 1 ml of water.
On day 2. each animal in all four groups was orally challenged with 1 ml of C. difficile inoculum. which contained approximately 100 C difficile strain 43596 organisms in sterile saline. C. difficile strain 43596 was chosen because it is representative of one of the most frequently-occurring serogroups isolated from patients with antibiotic-associated pseudomembranous colitis. [Delmee et al.. J. Clin. Microbiol., 28:2210-2214 (1990).] In addition. as this was the same C. difficile strain used in all of the previous Examples above, it was again used in order to provide experimental continuity.
On day 3 of the study (24 hrs. post-infection), treatment was started for two of the four groups of animals. Each animal of one group was orally administered 1.0 ml of the CTAB IgY preparation (prepared in section above) at the following timepoints: 0 hrs.. 4 hrs.. and 8 hrs.' The animals in this group were designated "CTAB-24." The animals in the second group were each orally administered 1.0 ml of the pre-immune IgY preparation (also prepared in section above) at the same timepoints as for the CTAB group. These animals were designated "Pre-24." Nothing was done to the remaining two groups of animals on day 3.
On day 4. 48 hrs. post-infection, the treatment described for day 3 above was repeated for the CTAB-24 and Pre-24 groups. and was initiated for the remaining two groups at the same timepoints. The final two groups of animals were designated "CTAB-48" and "Pre-48" respectively.
20 On days 5 through 9. the animals in all four groups were administered antitoxin or pre-immune IgY. 3x per day. as described for day 4 above. The four experimental groups are summarized in Table 14.
81 TABLE 14 Experimental Treatment Groups Group Designation Experimental Treatment CTAB-24 Infected. treatment w/antitoxin IgY started 24 hrs. post-infection.
Infected. treatment w/pre-immune IgY started 24 hrs. post- Pre-24 infection.
CTAB-48 Infected. treatment w/antitoxin IgY started 48 hrs. post-infection.
Infected. treatment w/pre-immune IgY started 48 hrs. post- Pre-48 infection.
All animals were observed for the onset of diarrhea and death through the conclusion of the study on the morning of day 10. The results of this study are displayed in Table TABLE Experimental Outcome--Day Treatment Group No. Animals Surviving No. Animals Dead CTAB-24 6 1 Pre-24 0 7 CTAB-48 4 3 Pre-48 2 Eighty-six percent of the animals which began receiving treatment with antitoxin IgY at 24 hrs. post-infection (CTAB-24 above) survived, while 57% of the animals treated with antitoxin IgY starting 48 hrs. post-infection (CTAB-48 above) survived. In contrast none of the animals receiving pre-immune IgY starting 24 hrs. post-infection (Pre-24 above) survived.
and only 29% of the animals which began receiving treatment with pre-immune IgY at 48 hrs. post-infection (Pre-48 above) survived through the conclusion of the study. These results demonstrate that avian antitoxins raised against C difficile toxins A and B are capable of successfully treating established C. difficile infections in vivo.
82c) Histologic Evaluation Of Cecal Tissue In order to further evaluate the ability of the IgY preparations tested in this study to treat established C difficile infection, histologic evaluations were performed on cecal tissue specimens obtained from representative animals from the study described in section above.
Iimediately following death, cecal tissue specimens were removed from animals which died in the Pre-24 and Pre-48 groups. Following the completion of the study, a representative surviving animal was sacrificed and cecal tissue specimens were removed from the CTAB-24 and CTAB-48 groups. A single untreated animal from the same shipment as those used in the study was also sacrificed and a cecal tissue specimen was removed as a normal control. All tissue specimens were fixed overnight at 4°C in 10% buffered formalin.
The fixed tissues were paraffin-embedded. sectioned. and mounted on glass microscope slides.
The tissue sections were then stained using hematoxylin and eosin (H and E stain), and were examined by licht microscopy.
15 Upon examination, the tissues obtained from the CTAB-24 and CTAB-48 animals showed no pathology, and were indistinguishable from the normal control. This observation provides further evidence for the ability of avian antitoxins raised against C difficile toxins A and B to effectively treat established C difficile infection, and to prevent the pathologic consequences which normally occur as a result of C difficile disease.
In contrast. characteristic substantial mucosal damage and destruction was observed in the tissues of the animals from the Pre-24 and Pre-48 groups which died from C. difficile disease. Normal tissue architecture was obliterated in these two preparations. as most of the mucosal layer was observed to have sloughed away. and there were numerous large hemorrhagic areas containing massive numbers of erythrocytes.
EXAMPLE 11 Clonine And Expression Of C. difficile Toxin A Fragments The toxin A gene has been cloned and sequenced. and shown to encode a protein of predicted MW of 308 kd. (Dove et al.. Infect. Immun.. 58:480-488 (1990).] Given the expense and difficulty of isolating native toxin A protein, it would be advantageous to use simple and inexpensive procaryotic expression systems to produce and purify high levels of recombinant toxin A protein for immunization purposes. Ideally. the isolated recombinant 83 w protein would be soluble in order to preserve native antigenicity. since solubilized inclusion body proteins often do not fold into native conformations. To allow ease of purification, the recombinant protein should be expressed to levels greater than 1 mg/liter of E. coli culture.
To determine whether high levels of recombinant toxin A protein can be produced in E. coli. fragments of the toxin A gene were cloned into various prokaryotic expression vectors, and assessed for the ability to express recombinant toxin A protein in E. coli. Three prokaryotic expression systems were utilized. These systems were chosen because they drive expression of either fusion (pMALc and pGEX2T) or native (pET23a-c) protein to high levels in E. coli. and allow affinity purification of the expressed protein on a ligand containing column. Fusion proteins expressed from pGEX vectors bind glutathione agarose beads, and are eluted with reduced glutathione. pMAL fusion proteins bind amylose resin, and are eluted with maltose. A poly-histidine tag is present at either the N-terminal (pET16b) or C-terminal (pET23a-c) end of pET fusion proteins. This sequence specifically binds Ni,' chelate columns.
and is eluted with imidazole salts. Extensive descriptions of these vectors are available 15 (Williams et al. (1994) DNA Cloning: Expression Systems. in press). and will not be discussed in detail here. The Example involved cloning of the toxin A gene. (b) expression of large fragments of toxin A in various prokaryotic expression systems. (c) identification of smaller toxin A gene fragments that express efficiently in E. coli. (d) purification of recombinant toxin A protein by affinity chromatography. and demonstration 20 of functional activity of a recombinant fragment of the toxin A gene.
a) Cloning Of The Toxin A Gene A restriction map of the toxin A gene is shown in Figure 6. The encoded protein contains a carboxy terminal ligand binding region. containing multiple repeats of a carbohydrate binding domain. [von Eichel-Streiber and Sauerborn. Gene 96:107-113 (1990).] The toxin A gene was cloned in three pieces. by using either the polymerase chain reaction (PCR) to amplify specific regions. (regions 1 and 2. Figure 6) or by screening a constructed genomic library for a specific toxin A gene fragment (region 3. Figure The sequences of the utilized PCR primers are PI: 5' GGAAATT TAGCTGCAGCATCTGAC 3' (SEQ ID NO:1); P2: 5' TCTAGCAAATTCGCTTGT GTTGAA 3' (SEQ ID NO:2): P3: 5' CTCGCATATAGCATTAGACC 3' (SEQ ID NO:3); and P4: 5' CTATCTAGGCCTAAAGTAT 3' (SEQ ID NO:4).
84 These regions were cloned into prokaryotic expression vectors that express either fusion (pMALc and pGEX2T) or native (pET23a-c) protein to high levels in E. coli. and allow affinity purification of the expressed protein on a ligand containing column.
Clostridium difficile VPI strain 10463 was obtained from the ATCC (ATCC #43255) and grown under anaerobic conditions in brain-heart infusion medium (BBL). High molecular-weight C difficile DNA was isolated essentially as described by Wren and Tabaqchali (1987) J. Clin. Microbiol.. 25:2402. except proteinase K and sodium dodecyl sulfate (SDS) was used to disrupt the bacteria, and cetyltrimethylammonium bromide precipitation [as described in Ausubel el al.. Current Protocols in Molecular Biology (1989)] was used to remove carbohydrates from the cleared lysate. The integrity and yield of genomic DNA was assessed by comparison with a serial dilution of uncut lambda DNA after electrophoresis on an agarose gel.
SFragments I and 2 were cloned by PCR. utilizing a proofreading thermostable DNA polymerase (native pfit polymerase: Stratagene). The high fidelity of this polymerase reduces 15 the mutation problems associated with amplification by error prone polymerases Taq polymerase). PCR amplification was performed using the indicated PCR primers (Figure 6) in 50 ptl reactions containing 10 mM Tris-HCl(8.3). 50 mM KCI. 1.5 mM MgCl, 200 PM each dNTP. 0.2 pM each primer, and 50 ng C difficile genomic DNA. Reactions were overlaid with 100 pl mineral oil. heated to 94 0 C for 4 min. 0.5 pl native pfu polymerase (Stratagene) added, and the reaction cycled 30x at 94°C for I min. 50 0 C for I min. 72 0 C for 4 min. followed by 10 min at 72 0 C. Duplicate reactions were pooled. chloroform extracted, and ethanol precipitated. After washing in 70% ethanol. the pellets were resuspended in 50 pt TE buffer [10 mM Tris-HCL. 1 mM EDTA pH Aliquots of 10l each were restriction digested with either EcoRIIHincll (fragment 1) or EcoRlIPstl (fragment and the appropriate restriction fragments were gel purified using the Prep-A-Gene kit (BioRad). and ligated to either EcoRI/Smal-restricted pGEX2T (Pharmacia) vector (fragment or the EcoRIIPstl pMAlc (New England Biolabs) vector (fragment Both clones are predicted to produce in-frame fusions with either the glutathione-S-transferase protein (pGEX vector) or the maltose binding protein (pMAL vector). Recombinant clones were isolated, and confirmed by restriction digestion. using standard recombinant molecular biology techniques.
[Sambrook et al.. Molecular Cloning. A Laboratory Manual (1989). and designated 660 and pMA660-1100, respectively (see Figure 6 for description of the clone designations).] 85 JW Fragment 3 was cloned from a genomic library of size selected Psti digested C. difficile genomic DNA. using standard molecular biology techniques (Sambrook et al.).
Given that the fragment 3 internal PstI site is protected from cleavage in C difficile genomic DNA [Price ei al.. Curr. Microbiol.. 16:55-60 (1987)], a 4.7 kb fragment from Pstl restricted C. difficile genomic DNA was gel purified, and ligated to Pstl restricted. phosphatase treated pUC9 DNA. The resulting genomic library was screened with a oligonucleotide primer specific to fragment 3. and multiple independent clones were isolated. The presence of fragment 3 in several of these clones was confirmed by.restriction digestion. and a clone of the indicated orientation (Figure 6) was restricted with BamHIIHindIIl. the released fragment purified by gel electrophoresis. and ligated into similarly restricted pET23c expression vector DNA (Novagen). Recombinant clones were isolated, and confirmed by restriction digestion.
This construct is predicted to create both a predicted in frame fusion with the pET protein leader sequence, as well as a predicted C-terminal poly-histidine affinity tag. and is designated pPA1100-2680 (see Figure 6 for the clone designation).
l b) Expression Of Large Fragments Of Toxin A In E coli Protein expression from the three expression constructs made in was induced, and analyzed by Western blot analysis with an affinity purified, goat polyclonal antiserum directed against the toxin A toxoid (Tech Lab). The procedures utilized for protein induction. SDS- S 20 PAGE. and Western blot analysis are described in detail in Williams el al (1994). supra. In brief. 5 ml 2X YT (16 g tryptone. 10 g yeast extract. 5 g NaCI per liter. pH 7.5 100 pg/ml ampicillin were added to cultures of bacteria (BL21 for pMAl and pGEX plasmids. and BL21(DE3)LysS for pET plasmids) containing the appropriate recombinant clone which were induced to express recombinant protein by addition of IPTG to 1 mM. Cultures were grown 25 at 37*C. and induced when the cell density reached 0.5 OD Induced protein was allowed to accumulate for two hrs after induction. Protein samples were prepared by pelleting 1 ml aliquots of bacteria by centrifugation (1 min in a microfuge). and resuspension of the pelleted bacteria in 150 pl of 2x SDS-PAGE sample buffer [Williams el al. (1994), supra]. The samples were heated to 95°C for 5 min. the cooled and 5 or 10 pl aliquots loaded on SDS-PAGE gels. BioRad high molecular weight protein markers were also loaded, to allow estimation of the MW of identified fusion proteins. After electrophoresis, protein was detected either generally by staining gels with Coomassie blue. or specifically, by blotting to nitrocellulose for Western blot detection of specific immunoreactive protein. Western blots.
86 (performed as described in Example 3) which detect toxin A reactive protein in cell lysates of induced protein from the three expression constructs are shown in Figure 7. In this figure, lanes 1-3 contain cell lysates prepared from E. coli strains containing pPA1100-2860 in B121(DE3)lysE cells; lanes 4-6 contain cell lysates prepared from E. coli strains containing pPA1100-2860 in B121(DE3)lysS cells: lanes 7-9 contain cell lysates prepared from E. coli strains containing pMA30-660: lanes 10-12 contain cell lysates prepared from E. coli strains containing pMA660-l 100. The lanes were probed with an affinity purified goat antitoxin A polyclonal antibody (Tech Lab). Control lysates from uninduced cells (lanes 1. 7, and contain very little immunoreactive material compared to the induced samples in the remaining lanes. The highest molecular weight band observed for each clone is consistent with the predicted size of the full length fusion protein.
Each construct directs expression of high molecular weight (HMW) protein that is reactive with the toxin A antibody. The size of the largest immunoreactive bands from each sample is consistent with predictions of the estimated MW of the intact fusion proteins. This 15 demonstrates that the three fusions are in-frame, and that none of the clones contain cloning artifacts that disrupt the integrity of the encoded fusion protein. However, the Western blot S demonstrates that fusion protein from the two larger constructs (pGA30-660 and pPAI 100- 2680) are highly degraded. Also. expression levels of toxin A proteins from these two constructs are low, since induced protein bands are not visible by Coomassie staining (not 20 shown). Several other expression constructs that fuse large sub-regions of the toxin A gene to :i either pMALc or pET23a-c expression vectors, were constructed and tested for protein induction. These constructs were made by mixing gel purified restriction fragments, derived from the expression constructs shown in Figure 6. with appropriately cleaved expression vectors, ligating, and selecting recombinant clones in which the toxin A restriction fragments had ligated together and into the expression vector as predicted for in-frame fusions. The expressed toxin A interval within these constructs are shown in Figure 8. as well as the internal restriction sites utilized to make these constructs.
As used herein, the term "interval" refers to any portion any segment of the toxin which is less than the whole toxin molecule) of a clostridial toxin. In a preferred embodiment, "interval" refers to portions of C. difficile toxins such as toxin A or toxin B. It is also contemplated that these intervals will correspond to epitopes of immunologic importance, such as antigens or immunogens against which a neutralizing antibody response is effected. It is not intended that the present invention be limited to the particular intervals or 87sequences described in these Examples. It is also contemplated that sub-portions of intervals an epitope contained within one interval or which bridges multiple intervals) be used as compositions and in the methods of the present invention.
In all cases. Western blot analysis of each of these constructs with goat antitoxin A antibody (Tech Lab) detected HMW fusion protein of the predicted size (not shown). This confirms that the reading frame of each of these clones is not prematurely terminated, and is fused in the correct frame with the fusion partner. However, the Western blot analysis revealed that in all cases, the induced protein is highly degraded, and. as assessed by the absence of identifiable induced protein bands by Coomassie Blue staining, are expressed only at low levels. These results suggest that expression of high levels of intact toxin A recombinant protein is not possible when large regions of the toxin A gene are expressed in E. coli using these expression vectors.
c) High Level Expression Of Small Toxin A Protein Fusions In 15 E. coli Experience indicates that expression difficulties are often encountered when large (greater than 100 kd) fragments are expressed in E. coli. A number of expression constructs containing smaller fragments of the toxin A gene were constructed. to determine if small regions of the gene can be expressed to high levels without extensive protein degradation. A summary of these expression constructs are shown in Figure 9. All were constructed by inframe fusions of convenient toxin A restriction fragments to either the pMALc or pET23a-c vectors. Protein preparations from induced cultures of each of these constructs were analyzed by both Coomassie Blue staining and Western analysis as in above. In all cases. higher levels of intact, full length fusion proteins were observed than with the larger recombinants from section d) Purification Of Recombinant Toxin A Protein Large scale (500 ml) cultures of each recombinant from were grown. induced, and soluble and insoluble protein fractions were isolated. The soluble protein extracts were affinity chromatographed to isolate recombinant fusion protein, as described [Williams el al.
(1994). supra]. In brief, extracts containing tagged pET fusions were chromatographed on a nickel chelate column. and eluted using imidazole salts as described by the distributor (Novagen). Extracts containing soluble pMAL fusion protein were prepared and 88
C
chromatographed in column buffer (10 mM NaPO,. 0.5M NaCI. 10 mM P-mercaptoethanol.
pH 7.2) over an amylose resin column (New England Biolabs). and eluted with column buffer containing 10 mM maltose as described [Williams et al. (1994). supra]. When the expressed protein was found to be predominantly insoluble, insoluble protein extracts were prepared by the method described in Example 17. infra. The results are summarized in Table 16. Figure 10 shows the sample purifications of recombinant toxin A protein. In this figure.
lanes 1 and 2 contain MBP fusion protein purified by affinity purification of soluble protein.
TABLE 16 Purification Of Recombinant Toxin A Protein Yield Affinity Intact Yield Intact Clone S Protein Purified Soluble Soluble Fusion Insoluble Fusion Solubility Protein (b Protein'" Protein pMA30-270 Soluble 4 mg/500 mis 10% NA PMA30-300 Soluble 4 mg/500 mis 5-10% NA pMA300-660 Insoluble NA 10 mg/500 ml pMA660-1100 Soluble 4.5 mg/500 mis 50%
NA
pMA1100-1610 Soluble 18 mg/500 mis 10%
NA
pMA1610-1870 Both 22 mg/500 mis 90% 20 mg/500 ml pMA1450-1870 Insoluble NA 0.2 mg/500 ml pPAl100-1450 Soluble 0.1 mg/500 mis 90%
NA
pPA1100-1870 Soluble 0.02 mg/500 mis 90%
NA
pMA1870-2680 Both 12 mg/500 mis 80%/
NA
pPAl870-2680 Insoluble NA 10 mg/500 ml pP pET23 vector. pM=pMALc vector. A=toxin A.
Based on 1.5 ODgo I mg/ml (extinction coefficient of MBP).
4C) Estimated by Coomassie staining of SDS-PAGE gels.
Lanes 3 and 4 contain MBP fusion protein purified by solubilization of insoluble inclusion bodies. The purified fusion protein samples are pMA 870-2680 (lane pMA660-l 100 (lane pMA300-600 (lane 3) and pMA1450-1870 (lane 4).
89 Poor yields of affinity purified protein were obtained when poly-histidine tagged pET vectors were used to drive expression (pPA 100-1450. pPl 100-1870). However. significant protein yields were obtained from pMAL expression constructs spanning the entire toxin A gene. and yields of full-length soluble fusion protein ranged from an estimated 200-400 /500 ml culture (pMA30-300) to greater than 20 mg/500 ml culture (pMA1610-1870).
Only one interval was expressed to high levels as strictly insoluble protein (pMA300-660).
Thus. although high level expression was not observed when using large expression constructs from the toxin A gene. usable levels of recombinant protein spanning the entire toxin A gene were obtainable by isolating induced protein from a series of smaller pMAL expression constructs that span the entire toxin A gene. This is the first demonstration of the feasibility of expressing recombinant toxin A protein to high levels in E. coli.
e) Hemagglutination Assay Using The Toxin A Recombinant Proteins 15 The carboxy terminal end consisting of the repeating units contains the hemagglutination activity or binding domain of C difficile toxin A. To determine whether the expressed toxin A recombinants retain functional activity. hemagglutination assays were performed. Two toxin A recombinant proteins, one containing the binding domain as either soluble affinity purified protein (pMA1870-2680) or SDS solubilized inclusion body protein 20 (pPA1870-2680) and soluble protein from one region outside that domain (pMA1100-1610) were tested using a described procedure. Krivan et. al.. Infect. Immun.. 53:573 (1986).] Citrated rabbit red blood cells (RRBC)(Cocalico) were washed several times with .i Tris-buffer 0.1M Tris and. 50 mM NaCI by centrifugation at 450 x g for 10 minutes at 4* C. A 1% RRBC suspension was made from the packed cells and resuspended in Tris-buffer.
Dilutions of the recombinant proteins and native toxin A (Tcch Labs) were made in the Trisbuffer and added t duplicate to a round-bottomed 96-well microtiter plate in a final volume of 100 pl. To each well. 50 pi of the 1% RRBC suspension was added. mixed by gentle tapping. and incubated at 4 0 C for 3-4 hours. Significant hemagglutination occurred only in the recombinant proteins containing the binding domain (pMA 1870-2680) and native toxin A. The recombinant protein outside the binding domain (pMA 1100-1610) displayed no hemagglutination activity. Using equivalent protein concentrations. the hemagglutination titer for toxin A was 1:256. while titers for the soluble and insoluble recombinant proteins of the binding domain were 1:256 and about 1:5000. Clearly. the recombinant proteins tested retained functional activity and were able to bind RRBC's.
EXAMPLE 12 Functional Activity Of IgY Reactive Aeainst Toxin A Recombinants The expression of recombinant toxin A protein as multiple fragments in E.coli has demonstrated the feasibility of generating toxin A antigen through use of recombinant methodologies (Example 11). The isolation of these recombinant proteins allows the immunoreactivity of each individual subregion of the toxin A protein to be determined in a antibody pool directed against the native toxin A protein). This identifies the regions (if any) for which little or no antibody response is elicited when the whole protein is used as a i* immunogen. Antibodies directed against specific fragments of the toxin A protein can be purified by affinity chromatography against recombinant toxin A protein, and tested for *e 15 neutralization ability. This identifies any toxin A subregions that are essential for producing neutralizing antibodies. Comparison with the levels of immune response directed against these intervals when native toxin is used as an immunogen predicts whether potentially higher titers of neutralizing antibodies can be produced by using recombinant protein directed against a individual region. rather than the entire protein. Finally. since it is unknown whether 20 antibodies reactive to the recombinant toxin A proteins produced in Example II neutralize toxin A as effectively as antibodies raised against native toxin A (Examples 9 and 10). the protective ability of a pool of antibodies affinity purified against recombinant toxin A fragments was assessed for its ability to neutralize toxin A.
S::.This Example involved epitope mapping of the toxin A protein to determine the titre of specific antibodies directed against individual subregions of the toxin A protein when native toxin A protein is used as an immunogen. affinity purification of IgY reactive against recombinant proteins spanning the toxin A gene. toxin A neutralization assays with affinity purified IgY reactive to recombinant toxin A protein to identify subregions of the toxin A protein that induce the production of neutralizing antibodies. and determination of whether complete neutralization of toxin A can be elicited with a mixture of antibodies reactive to recombinant toxin A protein.
-91 1W a) Epitope Mapping Of The Toxin A Gene The affinity purification of recombinant toxin A protein specific to defined intervals of the toxin A protein allows epitope mapping of antibody pools directed against native toxin A.
This has not previously been possible. since previous expression of toxin A recombinants has been assessed only by Western blot analysis. without knowledge of the expression levels of the protein von Eichel-Streiber et al. J. Gen. Microbiol.. 135:55-64 (1989)]. Thus. high or low reactivity of recombinant toxin A protein on Western blots may reflect protein expression level differences. not immunoreactivity differences. Given that the purified recombinant protein generated in Example 11 have been quantitated. the issue of relative immunoreactivity of individual regions of the toxin A protein was precisely addressed.
For the purposes of this Example. the toxin A protein was subdivided into 6 intervals numbered from the amino (interval 1) to the carboxyl (interval 6) termini.
The recombinant proteins corresponding to these intervals were from expression clones (see Example I l(d) for clone designations) pMA30-300 (interval pMA300-660 (interval 15 pMA660-1100 (interval pPAI 100-1450 (interval pMA1450-1870 (interval 5) and pMA1870-2680 (interval These 6 clones were selected because they span the entire protein from amino acids numbered 30 through 2680. and subdivide the protein into 6 small intervals. Also. the carbohydrate binding repeat interval is contained specifically in one interval (interval allowing evaluation of the immune response specifically directed against 20 this reuion. Western blots of 7.5% SDS-PAGE gels. loaded and electrophoresed with defined quantities of each recombinant protein. were probed with either goat antitoxin A polyclonal antibody (Tech Lab) or chicken antitoxin A polyclonal antibody [pCTA IgY. Example The blots were prepared and developed with alkaline phosphatase as previously described [Williams et al. (1994). supra]. At least 90% of all reactivity, in either goat or chicken antibody pools. was found to be directed against the ligand binding domain (interval The remaining immunoreactivity was directed against all five remaining intervals, and was similar in both antibody pools. except that the chicken antibody showed a much lower reactivity against interval 2 than the goat antibody.
This clearly demonstrates that when native toxin A is used as an immunogen in goats or chickens. the bulk of the immune response is directed against the ligand binding domain of the protein, with the remaining response distributed throughout the remaining 2/3 of the protein.
92 b) Affinity Purification Of IgY Reactive Against Recombinant Toxin A Protein Affinity columns. containing recombinant toxin A protein from the 6 defined intervals in above, were made and used to affinity purify antibodies reactive to each individual interval from the CTA IgY preparation [Example and (ii) deplete interval specific antibodies from the CTA IgY preparation. Affinity columns were made by coupling 1 ml of PBS-washed Actigel resin (Sterogene) with region specific protein and 1/10 final volume of Aid-coupling solution (1M sodium cyanoborohydridc). The total region specific protein added to each reaction mixture was 2.7 mg (interval 3 mg (intervals 2 and 0.1 mg (interval 0.2 mg (interval 5) and 4 mg (interval Protein for intervals 1. 3, and 6 was affinity purified pMAI fusion protein in column buffer (see Example 11). Interval 4 was affinity purified poly-histidine containing pET fusion in PBS: intervals 2 and 5 were from inclusion body preparations of insoluble pMAL fusion protein, dialyzed extensively in PBS.
Aliquots of the supematants from the coupling reactions. before and after coupling, were 15 assessed by Coomassie staining of 7.5% SDS-PAGE gels. Based on protein band intensities.
in all cases greater than 50% coupling efficiencies were estimated. The resins were poured into 5 ml BioRad columns. washed extensively with PBS. and stored at 4*C.
Aliquots of the CTA IgY polyclonal antibody preparation were depleted for each individual region as described below. A 20 ml sample of the CTA IgY preparation [Example 20 was dialyzed extensively against 3 changes of PBS (1 liter for each dialysis). quantitated by absorbance at and stored at 4*C. Six I ml aliquots of the dialyzed IgY preparation were removed, and depleted individually for each of the six intervals. Each I ml aliquot was passed over the appropriate affinity column. and the eluate twice reapplied to the column.
The eluate was collected, and pooled with a 1 ml PBS wash. Bound antibody was eluted from the column by washing with 5 column volumes of 4 M Guanidine-HCI (in 10 mM Tris- HCI. pH The column was reequilibrated in PBS. and the depleted antibody stock reapplied as described above. The eluate was collected. pooled with a 1 ml PBS wash.
quantitated by absorbance at OD, 8 o, and stored at 4* C. In this manner. 6 aliquots of the CTA IgY preparation were individually depleted for each of the 6 toxin A intervals, by two rounds of affinity depletion. The specificity of each depleted stock was tested by Western blot analysis. Multiple 7.5% SDS-PAGE gels were loaded with protein samples corresponding to all 6 toxin A subregions. After electrophoresis. the gels were blotted, and protein transfer confirmed by Ponceau S staining [protocols described in Williams et al. (1994). supra]. After -93 W blocking the blots 1 hr at 20 0 C in PBS+ 0.1% Tween 20 (PBST) containing 5% milk (as a blocking buffer). 4 ml of either a 1/500 dilution of the dialyzed CTA IgY preparation in blocking buffer. or an equivalent amount of the six depleted antibody stocks (using OD,s 0 to standardize antibody concentration) were added and the blots incubated a further 1 hr at room temperature. The blots were washed and developed with alkaline phosphatase (using a rabbit anti-chicken alkaline phosphate conjugate as a secondary antibody) as previously described [Williams et al. (1994). supra]. In all cases, only the target interval was depleted for antibody reactivity, and at least 90% of the reactivity to the target intervals was specifically depleted.
Region specific antibody pools were isolated by affinity chromatography as described below. Ten mis of the dialyzed CTA IgY preparation were applied sequentially to each affinity column. such that a single 10 ml aliquot was used to isolate region specific antibodies specific to each of the six subregions. The columns were sequentially washed with volumes of PBS. 6 volumes of BBS-Tween. 10 volumes of TBS. and eluted with 4 ml Actisep elution media (Sterogene). The eluate was dialyzed extensively against several 15 changes of PBS. and the affinity purified antibody collected and stored at 4 0 C. The volumes of the eluate increased to greater than 10 mis during dialysis in each case. due to the high viscosity of the Actisep elution media. Aliquots of each sample were 20x concentrated using S" Centricon 30 microconcentrators (Amicon) and stored at 4 0 C. The specificity of each region specific antibody pool was tested. relative to the dialyzed CTA IgY preparation. by Western 20 blot analysis. exactly as described above, except that 4 ml samples of blocking buffer containing 100 ll region specific antibody (unconcentrated) were used instead of the depleted CTA IgY preparations. Each affinity purified antibody preparation was specific to the defined interval, except that samples purified against intervals 1-5 also reacted with interval 6. This may be due to non-specific binding to the interval 6 protein, since this protein contains the repetitive ligand binding domain which has been shown to bind antibodies nonspecifically.
[Lyerly et al.. Curr. Microbiol.. 19:303-306 (1989).] The reactivity of each affinity purified antibody preparation to the corresponding proteins was approximately the same as the reactivity of the 1/500 diluted dialyzed CTA IgY preparation standard. Given that the specific antibody stocks were diluted 1/40. this would indicate that the unconcentrated affinity purified antibody stocks contain 1/10-1/20 the concentration of specific antibodies relative to the starting CTA IgY preparation.
94 c) Toxin A Neutralization Assay Using Antibodies Reactive Toward Recombinant Toxin A Protein The CHO toxin neutralization assay [Example was used to assess the ability of the depleted or enriched samples generated in above to neutralize the cytotoxicity of toxin A. The general ability of affinity purified antibodies to neutralize toxin A was assessed by mixing together aliquots of all 6 concentrated stocks of the 6 affinity purified samples generated in above, and testing the ability of this mixture to neutralize a toxin A concentration of 0.1 Lg/ml. The results. shown in Figure 11. demonstrate almost complete neutralization of toxin A using the affinity purified (AP) mix. Some epitopes within the recombinant proteins utilized for affinity purification were probably lost when the proteins were denatured before affinity purification [by Guanidine-HCI treatment in above]. Thus, Sthe neutralization ability of antibodies directed against recombinant protein is probably underestimated using these affinity purified antibody pools. This experiment demonstrates that antibodies reactive to recombinant toxin A can neutralize cytotoxicity. suggesting that 15 neutralizing antibodies may be generated by using recombinant toxin A protein as immunogen.
In view of the observation that the recombinant expression clones of the toxin A gene divide the protein into 6 subregions, the neutralizing ability of antibodies directed against each individual region was assessed. The neutralizing ability of antibodies directed against the 20 ligand binding domain of toxin A was determined first.
In the toxin neutralization experiment shown in Figure 11. interval 6 specific antibodies (interval 6 contains the ligand binding domain) were depleted from the dialyzed PEG preparation, and the effect on toxin neutralization assayed. Interval 6 antibodies were depleted either by utilizing the interval 6 depleted CTA IgY preparation from above aff. depleted" in Figure 11). or by addition of interval 6 protein to the CTA IgY preparation (estimated to be a 10 fold molar excess.over anti-interval 6 immunoglobulin present in this preparation) to competitively compete for interval 6 protein prot depleted" in Figure 11).
In both instances, removal of interval 6 specific antibodies reduces the neutralization efficiency relative to the starting CTA IgY preparation. This demonstrates that antibodies directed against interval 6 contribute to toxin neutralization. Since interval 6 corresponds to the ligand binding domain of the protein, these results demonstrate that antibodies directed against this region in the PEG preparation contribute to the neutralization of toxin A in this assay. However, it is significant that after removal of these antibodies, the PEG preparation 95 M retains significant ability to neutralize toxin A (Figure 11). This neutralization is probably due to the action of antibodies specific to other regions of the toxin A protein, since at least of the ligand binding region reactive antibodies were removed in the depleted sample prepared in above. This conclusion was supported by comparison of the toxin neutralization of the affinity purified (AP) mix compared to affinity purified interval 6 antibody alone. Although some neutralization ability was observed with AP interval 6 antibodies alone, the neutralization was significantly less than that observed with the mixture of all 6 AP antibody stocks (not shown).
Given that the mix of all six affinity purified samples almost completely neutralized the cytotoxicity of toxin A (Figure 11), the relative importance of antibodies directed against toxin A intervals 1-5 within the mixture was determined. This was assessed in two ways.
First. samples containing affinity purified antibodies representing 5 of the 6 intervals were prepared. such that each individual region was depleted from one sample. Figure 12 demonstrates a sample neutralization curve. comparing the neutralization ability of affinity purified an:ibody mixes without interval 4 or 5 specific antibodies. relative to the mix of all 6 affinity purified antibody stocks (positive control). While the removal of interval 5 specific antibodies had no effect on toxin neutralization (or intervals 1-3. not shown). the *loss of interval 4 specific antibodies significantly reduced toxin neutralization (Figure 12).
Similar results were seen in a second experiment, in which affinity purified antibodies.
S: 20 directed against a single region. were added to interval 6 specific antibodies. and the effects on toxin neutralization assessed. Only interval 4 specific antibodies significantly enhanced neutralization when added to interval 6 specific antibodies (Figure 13). These results demonstrate that antibodies directed against interval 4 (corresponding to clone pPA1100-1450 in Figure 9) are important for neutralization of cytotoxicity in this assay. Epitope mapping has shown that only low levels of antibodies reactive to this region are" generated when native toxin A is used as an immunogen [Example It is hypothesized that immunization with recombinant protein specific to this interval will elicit higher titers of neutralizing antibodies.
In summary. this analysis has identified two critical regions of the toxin A protein against which neutralizing antibodies are produced. as assayed by the CHO neutralization assay.
96- Example 13 Production And Evaluation Of Avian Antitoxin Against C. difficile Recombinant Toxin A Polypeptide In Example 12, we demonstrated neutralization of toxin A mediated cytotoxicity by affinity purified antibodies reactive to recombinant toxin A protein. To determine whether antibodies raised against a recombinant polypeptide fragment of C. difficile toxin A may be effective in treating clostridial diseases, antibodies to recombinant toxin A protein representing the binding domain were generated.
Two toxin A binding domain recombinant polypeptides, expressing the binding domain in either the pMALc (pMA1870-2680) or pET 23(pPA1870-2680) vector, were used as immunogens. The pMAL protein was affinity purified as a soluble product [Example 12(d)] and the pET protein was isolated as insoluble inclusion bodies [Example 12(d)] and solubilized to an immunologically active protein using a proprietary method described in United States Patent No. 5,605,691. This.example involves (a) immunization, antitoxin collection, determination of antitoxin antibody titer, anti-recombinant toxin A neutralization of toxin A hemagglutination activity in vitro, and assay of in vitro toxin A 5 neutralizing activity.
a) Immunization The soluble and the inclusion body preparations each were used separately to immunize hens.
Both purified toxin A polypeptides were diluted in PBS and emulsified with approximately equal volumes of CFA for the initial immunization of IFA for subsequent booster immunizations. On day 20 zero, for each of the recombinant preparations, two egg laying white Leghom hens (obtained from local breeder) were each injected at multiple sites (intramuscular and subcutaneous) with 1ml of recombinant adjuvant mixture containing approximately 0.5 to 1.5mgs of recombinant toxin A.
Booster immunizations of 1.0mg were given on days 14 and day 28.
b) Antitoxin Collection 25 Total yolk immune IgY was extracted as described in the standard PEG protocol (as in Example 1) and the final IgY pellet was dissolved in sterile PBS at the original yolk volume. This material is designated "immune recombinant IgY" or "immune IgY".
[R:\LIBAA]07510.doc:TAB c) Antitoxin Antibody Titer To determine if the recombinant toxin A protein was sufficiently immunogenic to raise antibodies in hens, the antibody titer of a recombinant toxin A polypeptide was determined by ELISA. Eggs from both hens were collected on day 32. the yolks pooled and the antibody was isolated using PEG as described. The immune recombinant IgY antibody titer was determined for the soluble recombinant protein containing the maltose binding protein fusion generated in p-Mal (pMA1870-2680). Ninety-six well Falcon Pro-bind plates were coated overnight at 4°C with 100 pl /well of toxin A recombinant at 2.5 pg /pl in PBS containing 0.05% thimerosal. Another plate was also coated with maltose binding protein (MBP) at the same concentration. to permit comparison of antibody reactivity to the fusion partner. The next day, the wells were blocked with PBS containing 1% bovine serum aluimin"TBSA) for 1 hour at 37°C. IgY isolated from immune or preimmune eggs was diluted in antibody diluent (PBS containing 1% BSA and 0.05% Tween-20), and added to the blocked wells and incubated for I hour at 37 0 C The plates were washed three times with PBS with 0.05% S 15 Tween-20. then three times with PBS. Alkaline phosphatase conjugated rabbit anti-chicken IgG (Sigma) diluted 1:1000 in antibody diluent was added to the plate. and incubated for 1 hour at 37°C. The plates were washed as before and substrate was added. [p-nitrophenyl phosphate (Sigma)] at I mg/ml in 0.05M Na,CO 3 pH 9.5 and 10 mM MgCI,. The plates were evaluated quantitatively on a Dynatech MR 300 Micro EPA plate reader at 410 nm 20 about 10 minutes after the addition of substrate.
Based on these ELISA results, high antibody titers were raised in chickens immunized with the toxin A recombinant polypeptide. The recombinant appeared to be highly immunogenic. as it was able to generate high antibody titers relatively quickly with few immunizations. Immune IgY titer directed specifically to the toxin A portion of the recombinant was higher than the immune IgY titer to its fusion partner, the maltose binding protein, and significantly higher than the preimmune IgY. ELISA titers (reciprocal of the highest dilution of IgY generating a signal) in the preimmune IgY to the MBP or the recombinant was <1:30 while the immune IgY titers to MBP and the toxin A recombinant were 1:18750 and 1:93750 respectively. Importantly. the anti-recombinant antibody titers generated in the hens against the recombinant polypeptide is much higher, compared to antibodies to that region raised using native toxin A. The recombinant antibody titer to region 1870-2680 in the CTA antibody preparation is at least five-fold lower compared to the recombinant generated antibodies (1:18750 versus >1:93750). Thus. it appears a better -98 immune response can be generated against a specific recombinant using that recombinant as the immunogen compared to the native toxin A.
This observation is significant. as it shows that because recombinant portions stimulate the production of antibodies. it is not necessary to use native toxin molecules to produce antitoxin preparations. Thus. the problems associated with the toxicity of the native toxin are avoided and large-scale antitoxin production is facilitated.
d) Anti-Recombinant Toxin A Neutralization Of Toxin A Hemagglutination Activity In Vitro Toxin A has hemagglutinating activity besides cytotoxic and enterotoxin properties.
Specifically. toxin A agglutinates rabbit erythrocytes by binding to a trisacciaride-Tgal 1-3B1- 4GlcNAc) on the cell surface. Krivan et al.. Infect. Immun.. 53:573-581 (1986).] We examined whether the anti-recombinant toxin A (immune IgY. antibodies raised against the insoluble product expressed in pET) can neutralize the hemagglutination activity of toxin A in 15 vitro. The hemagglutination assay procedure used was described by H.C. Krivan et al.
Polyethylene glycol-fractionated immune or preimmune IgY were pre-absorbed with citrated rabbit erythrocytes prior to performing the hemagglutination assay because we have found that IgY alone can agglutinate red blood cells. Citrated rabbit red blood cells (RRBC's)(Cocalico) were washed twice by centrifugation at 450 x g with isotonic buffer (0.1 M Tris-HCI, 0.05 M .i 20 NaCI, pH RRBC-reactive antibodies in the IgY were removed by preparing a RRBC suspension (made by adding packed cells to immune or preimmune IgY) and incubating the mixture for 1 hour at 37*C. The RRBCs were then removed by centrifugation.
*Neutralization of the hemagglutination activity of toxin A by antibody was tested in roundbottomed 96-well microtiter plates. Twenty-five pl of toxin A (36 pg /ml) (Tech Lab) in isotonic buffer was mixed with an equal volume of different dilutions of immune or preimmuf.e IgY in isotonic buffer, and incubated for 15 minutes at room temperature. Then.
pl of a 1% RRBC suspension in isotonic buffer was added and the mixture was incubated for 3 hours at 4 0 C. Positive control wells containing the final concentration of 9 Pg/ml of toxin A after dilution without IgY were also included. Hemagglutination activity was assessed visually, with a diffuse matrix of RRBC's coating the bottom of the well representing a positive hemagglutination reaction and a tight button of RRBC's at the bottom of the well representing a negative reaction. The anti-recombinant immune IgY neutralized -99toxin A hemagglutination activity, giving a neutralization titer of 1:8. However. preimmune IgY was unable to neutralize the hemagglutination ability of toxin A.
e) Assay Of In Vitro Toxin A Neutralizing Activity The ability of the anti-recombinant toxin A IgY (immune IgY antibodies raised against pMA1870-2680, the soluble recombinant binding domain protein expressed in pMAL.
designated as Anti-tox. A-2 in Figure 14. and referred to as recombinant region 6) and preimmune IgY. prepared as described in Example 8(c) above, to neutralize the cytotoxic activity of toxin A was assessed in vitro using the CHO cell cytotoxicity assay. and toxin A (Tech Lab) at a concentration of 0.1 pg/ml. as described in Example 8(d) above. As additional controls. the anti-native toxin A IgY (CTA) and pre-immune IgY preparations described in Example 8(c) above were also tested. The results are shown in Figure 14.
The anti-recombinant toxin A IgY demonstrated only partial neutralization of the cytotoxic activity of toxin A. while the pre-immune IgY did not demonstrate any significant 15 neutralizing activity.
EXAMPLE 14 In vivo Neutralization Of C difficile Toxin A The ability of avian antibodies (IgY) raised against recombinant toxin A binding domain to neutralize the enterotoxin activity of C difficile toxin A was evaluated in vivo using Golden Syrian hamsters. The Example involved: preparation of the avian antirecombinant toxin A IgY for oral administration; in vivo protection of hamsters from C difficile toxin A enterotoxicity by treatment of toxin A with avian anti-recombinant toxin A IgY; and histologic evaluation of hamster ceca.
a) Preparation Of The Avian Anti-Recombinant Toxin A IgY For Oral Administration Eggs were collected from hens which had been immunized with the recombinant C.
difficile toxin A fragment pMA1870-2680 (described in Example 13. above). A second group of eggs purchased at a local supermarket was used as a pre-immune (negative) control. Egg yolk immunoglobulin (IgY) was extracted by PEG from the two groups of eggs as described in Example and the final IgY pellets were solubilized in one-fourth the original yolk.
100volume using 0.1M carbonate buffer (mixture of NaHCO, and NaCO 3 pH 9.5. The basic carbonate buffer was used in order to protect the toxin A from the acidic pH of the stomach environment.
b) In viva Protection Of Hamsters Against C difficile Toxin A Enterotoxicity By Treatment Of Toxin A With Avian Antirecombinant Toxin A IgY In order to assess the ability of the avian anti-recombinant toxin A IgY. prepared in section above to neutralize the in vive enterotoxin activity of toxin A. an in vivo toxin neutralization model was developed using Golden Syrian hamsters. This model was based on published values for the minimum amount of toxin A required to elicit diarrthea (0-08 mg Stoxin A/Kg body wt.) and death (0.16 mg toxin A/Kg body wt.) in hamsters when administered orally (Lyerly et al. Infect. Immun.. 47:349-352 (1985).
For the study. four separate experimental groups were used, with each group consisting S" 15 of 7 female Golden Syrian hamsters (Charles River). approx. three and one-half weeks old.
weighing approx. 50 gms each. The animals were housed as groups of 3 and 4. and were offered food and water ad libitum through the entire length of the study.
For each animal. a mixture containing either 0lg of toxin A (0.2 mg/Kg) or 30pg of toxin A (0.6 mg/Kg) (C difficile toxin A was obtained from Tech Lab and 1 ml of either the 20 anti-recombinant toxin A IgY or pre-immune IgY (from section above) was prepared.
These mixtures were incubated at 37*C for 60 min. and were then administered to the animals by the oral route. The animals were then observed for the onset of diarrhea and death for a period of 24 hrs. following the administration of the toxin A+IgY mixtures, at the end of which time, the following results were tabulated and shown in Table 17: 101 TABLE 17 Study Outcome At 24 Hours Study Outcome at 24 Hours Experimental Group Healthy' Diarrhea 2 Dead 3 pg Toxin A Antitoxin Against Interval-6 7 0 0 30 gg Toxin A Antitoxin Against Interval 6 7 0 0 pg Toxin A Pre-Immune Serum 0 5 2 30 pg Toxin A Pre-Immune 0 5 2
S
S
S..
0*
S
*5 Animals remained healthy through the entire 24 hour study period.
10 Animals developed diarrhea. but did not die.
Animals developed diarrhea. and subsequently died.
Pretreatment of toxin A at both doses tested. using the anti-recombinant toxin A IgY, prevented all overt symptoms of disease in hamsters. Therefore. pretreatment of C difficile 15 toxin A. using the anti-recombinant toxin A IgY. neutralized the in vivo enterotoxin activity of the toxin A. In contrast all animals from the two groups which received toxin A which had been pretreated using pre-immune IgY developed disease symptoms which ranged from diarrhea to death. The diarrhea which developed in the 5 animals which did not die in each of the two pre-immune groups, spontaneously resolved by the end of the 24 hr. study period.
c) Histologic Evaluation Of Hamster Ceca In order to further assess the ability of anti-recombinant toxin A IgY to protect hamsters from the enterotoxin activity of toxin A. histologic evaluations were performed on the ceca of hamsters from the study described in section above.
Three groups of animals were sacrificed in order to prepare histological specimens.
The first group consisted of a single representative animal taken from each of the 4 groups of surviving hamsters at the conclusion of the study described in section above. These animals represented the 24 hr. timepoint of the study.
The second group consisted of two animals which were not part of the study described above, but were separately treated with the same toxin A pre-immune IgY mixtures as described for the animals in section above. Both of these hamsters developed diarrhea.
102 and were sacrificed 8 hrs. after the time of administration of the toxin A pre-immune IgY mixtures. At the time of sacrifice, both animals were presenting symptoms of diarrhea.
These animals represented the acute phase of the study.
The final group consisted of a single untreated hamster from the same shipment of animals as those used for the two previous groups. This animal served as the normal control.
Samples of cecal tissue were removed from the 7 animals described above, and were fixed overnight at 4*C using 10% buffered formalin. The fixed tissues were paraffinembedded. sectioned, and mounted on glass microscope slides. The tissue sections were then stained using hematoxylin and eosin (H and E stain), and were examined by light microscopy.
The tissues obtained from the two 24 hr. animals which received mixtures containing either 10g or 30pg of toxin A and anti-recombinant toxin A IgY were indistinguishable from the normal control, both in terms of gross pathology, as well as at the microscopic level.
These observations provide further evidence for the ability of anti-recombinant toxin A IgY to effectively neutralize the in vivo enterotoxin activity of C difficile toxin A. and thus its ability 15 to prevent acute or lasting toxin A-induced pathology.
In contrast, the tissues from the two 24 hr. animals which received the toxin A preimmune IgY mixtures demonstrated significant pathology. In both of these groups, the mucosal layer was observed to be less organized than in the riormal control tissue. The cytoplasm of the epithelial cells had a vacuolated appearance. and gaps were present between 20 the epithelium and the underlying cell layers. The lamina propria was largely absent.
Intestinal villi and crypts were significantly diminished, and appeared to have been overgrown by a planar layer of epithelial cells and fibroblasts. Therefore. although these animals overtly appeared to recover from the acute symptoms of toxin A intoxication, lasting pathologic alterations to the cecal mucosa had occurred.
The tissues obtained from the two acute animals which received mixtures of toxin A and pre-immune IgY demonstrated the most significant pathology. At the gross pathological level, both animals were observed to have severely distended ceca which were filled with watery, diarrhea-like material. At the microscopic level, the animal that was given the mixture containing 10g of toxin A and pre-immune IgY was found to have a mucosal layer which had a ragged, damaged appearance, and a disorganized, compacted quality. The crypts were largely absent, and numerous breaks in the epithelium had occurred. There was also an influx of erythrocytes into spaces between the epithelial layer and the underlying tissue. The animal which had received the mixture containing 30ltg of toxin A and pre-immune IgY 103 demonstrated the most severe pathology. The cecal tissue of this animal had an appearance very similar to that observed in animals which had died from C difficile disease. Widespread destruction of the mucosa was noted. and the epithelial layer had sloughed. Hemorrhagic areas containing large numbers of erythrocytes were very prevalent. All semblance of normal tissue architecture was absent from this specimen. In terms of the presentation of pathologic events, this in vivo hamster model of toxin A-intoxication correlates very closely with the pathologic consequences of C difficile disease in hamsters. The results presented in this Example demonstrate that while anti-recombinant toxin A (Interval 6) IgY is capable of only partially neutralizing the cytotoxic activity of C difficile toxin A. the same antibody effectively neutralizes 100% of the in vivo enterotoxin activity of the toxin. While it is not *intended that this invention be limited to this mechanism. this may be due to-the cytotoxicity o and enterotoxicity of C. difficile Toxin A as two separate and distinct biological functions.
EXAMPLE In Vivo Neutralization Of C Difficile Toxin A By Antibodies Against Recombinant Toxin A Polvpeptides The ability of avian antibodies directed against the recombinant C difficile toxin A fragment 1870-2680 (as expressed by pMA1870-2680: see Example 13) to neutralize the 20 enterotoxic activity of toxin A was demonstrated in Example 14. The ability of avian antibodies (IgYs) directed against other recombinant toxin A epitopes to neutralize native toxin A in vivo was next evaluated. This example involved: the preparation of IgYs against recombinant toxin A polypeptides; in vivo protection of hamsters against toxin A by treatment with anti-recombinant toxin A IgYs and quantification of specific antibody concentration in CTA and Interval 6 IgY PEG preparations.
The nucleotide sequence of the coding region of the entire toxin A protein is listed in SEQ ID NO:5. The amino acid sequence of the entire toxin A protein is listed in SEQ ID NO:6. The amino acid sequence consisting of amino acid residues 1870 through 2680 of toxin A is listed in SEQ ID NO:7. The amino acid sequence consisting of amino acid residues 1870 through 1960 of toxin A is listed in SEQ ID NO:8. The amino acid sequence of residues 1873 through 2684 of toxin A is listed in SEQ ID NO:29.
104 a) Preparation Of IgY's Against Recombinant Toxin A Polypeptides Eggs were collected from Leghorn hens which have been immunized with recombinant C. difficile toxin A polypeptide fragments encompassing the entire toxin A protein. The polypeptide fragments used as immunogens were: 1) pMA 1870-2680 (Interval 2) pPA 1100-1450 (Interval and 3) a mixture of fragments consisting of pMA 30-300 (Interval 1), pMA 300-660 (Interval pMA 660-1100 (Interval 3) and pMA 1450-1870 (Interval This mixture of immunogens is referred to as Interval 1235. The location of each interval within the toxin A molecule is shown in Figure 15A. In Figure 15A. the following 10 abbreviations are used: pP refers to the pET23 vector (New England BioLabs): pM refers to S.t. the pMALu'-c vector (New England BioLabs): A refers to toxin A: the numbers refer to the amino acid interval expressed in the clone. (For example, the designation pMA30-300 indicates that the recombinant clone encodes amino acids 30-300 of toxin A and the vector used was pMAL'-c).
The recombinant proteins were generated as described in Example 11. The IgYs were extracted and solubilized in 0.1M carbonate buffer pH 9.5 for oral administration as described in Example 14(a). The IgY reactivities against each individual recombinant interval was evaluated by ELISA as described in Example 13(c).
*o 20 b) In Vivo Protection Of Hamsters Against Toxin A By Treatment With Anti-Recombinant Toxin A Antibodies The ability of antibodies raised against recombinant toxin A polypeptides to provide in vivo protection against the enterotoxic activity of toxin A was examined in the hamster model system. This assay was performed as described in Example 14(b). Briefly. for each 40-50 gram female Golden Syrian hamster (Charles River). I ml of IgY 4X resuspended in 1/4 of the original yolk volume) PEG prep against Interval 6. Interval 4 or Interval 1235 was mixed with 30 pg (LDo oral dose) of C difficile toxin A (Tech Lab). Preimmune IgY mixed with toxin A served as a negative control. Antibodies raised against C difficile toxoid A (Example 8) mixed with toxin A (CTA) served as a positive control. The mixture was incubated for 1 hour at 37 0 C then orally administered to lightly etherized hamsters using an 18G feeding needle. The animals were then observed for the onset of diarrhea and death for a period of approximately 24 hours. The results are shown in Table 18.
105 TABLE 18 Study Outcome After 24 Hours Treatment group Healthy' Diarrhea' Dead 3 Preimmune 0 0 7 CTA 5 0 0 Interval 6 6 1 0 Interval 4 0 1 6 Interval 1235 0 0 7 e S 10 Animal shows no sign of illness.
Animal developed diarrhea. but did not die.
3 Animal developed diarrhea and died.
Pre-treatment of toxin A with IgYs against Interval 6 prevented diarrhea in 6 of 7 15 hamsters and completely prevented death in all 7. In contrast as with preimmune IgY, IgYs against Interval 4 and Interval 1235 had no effect on the onset of diarrhea and death in the hamsters.
c) Quantification Of Specific Antibody Concentration In CTA 20 And Interval 6 IgY PEG Preparations To determine the purity of IgY PEG preparations, an aliquot of a pMA1870-2680 (Interval 6) IgY PEG preparation was chromatographed using HPLC and a KW-803 sizing column (Shodex). The resulting profile of absorbance at 280 nm is shown in Figure 16. The single large peak corresponds to the predicted MW of IgY. Integration of the area under the single large peak showed that greater than 95% of the protein eluted from the column was present in this single peak. This result demonstrated that the majority of the material absorbing at 280 nm in the PEG preparation corresponds to IgY. Therefore. absorbance at 280 nm can be used to determine the total antibody concentration in PEG preparations.
To determine the concentration of Interval 6-specific antibodies (expressed as percent of total antibody) within the CTA and pMA1870-2680 (Interval 6) PEG preparations, defined quantities of these antibody preparations were affinity purified on a pPA1870-2680(H) (shown schematically in Figure 15B) affinity column and the specific antibodies were quantified. In 106 Figure 15B the following abbreviations are used: pP refers to the pET23 vector (New England BioLabs): pM refers to the pMALM-c vector (New England BioLabs): pG refers to the pGEX vector (Pharmacia); pB refers to the PinPoint T Xa vector (Promega): A refers to toxin A: the numbers refer to the amino acid interval expressed in the clone. The solid black ovals represent the MBP: the hatched ovals represent glutathione S-transferase: the hatched circles represent the biotin tag; and HHH represents the poly-histidine tag.
An affinity column containing recombinant toxin A repeat protein was made as follows. Four ml of PBS-washed Actigel resin (Sterogene) was coupled with 5-10 mg of pPA1870-2680 inclusion body protein [prepared as described in Example (17) and dialyzed into PBS] in a 15 ml tube (Falcon) containing 1/10 final volume Aid-coupling solution (1 M Ssodium cyanoborohydride). Aliquots of the supernatant from the coupling reactions, before and after coupling, were assessed by Coomassie staining of 7.5% SDS-PAGE gels. Based .upon protein band intensities, greater than 6 mg of recombinant protein was coupled to the resin. The resin was poured into a 10 ml column (BioRad). washed extensively with PBS.
pre-eluted with 4 M guanidine-HCI (in 10 mM Tris-HC1. pH 8.0: 0.005% thimerosal) and reequilibrated with PBS. The column was stored at 4°C.
Aliquots of a pMA1870-2680 (Interval 6) or a CTA IgY polyclonal antibody preparation (PEG prep) were affinity purified on the above affinity column as follows. The column was attached to an UV monitor (ISCO) and washed with PBS. For pMA1870-2680 20 IgY purification, a 2X PEG prep (filter sterilized using a 0.45 ta filter: approximately 500 mg total IgY) was applied. The column was washed with PBS until the baseline was reestablished (the column flow-through was saved), washed with BBSTween to elute nonspecifically binding antibodies and re-equilibrated with PBS. Bound antibody was eluted from the column in 4 M guanidine-HCI (in 10 mM Tris-HCI. pH 8.0: 0.005% thimerosal).
The entire elution peak was collected in a 15 ml tube (Falcon). The column was reequilibrated and the column eluate was re-chromatographed as described above. The antibody preparation was quantified by UV absorbance (the elution buffer was used to zero the spectrophotometer). Total purified antibody was approximately 9 mg and 1 mg from the first and second chromatography passes. respectively. The low yield from the second pass indicated that most specific antibodies were removed by the first round of chromatography.
The estimated percentage of Interval 6 specific antibodies in the pMA1870-2680 PEG prep is approximately 2%.
107- 1W The percentage of Interval 6 specific antibodies in the CTA PEG prep was determined (utilizing the same column and methodology described above) to be approximately 0.5% of total IgY.
A 4X PEG prep contains approximately 20 mg/ml IgY. Thus in b) above.
approximately 400 pg specific antibody in the Interval 6 PEG prep neutralized 30 lig toxin A in vivo.
EXAMPLE 16 In Vivo Treatment Of C. difficile Disease In Hamsters By Recombinant Interval 6 Antibodies The ability of antibodies directed against recombinant Interval 6 of toxin A-to protect hamsters in vivo from C difficile disease was examined. This example involved: (a) o prophylactic treatment of C difficile disease and therapeutic treatment of C difficile disease.
a) Prophylactic Treatment Of C difficile Disease This experiment was performed as described in Example Three groups each consisting of 7 female 100 gram Syrian hamsters (Charles River) were prophylactically treated with either preimmune IgYs. IgYs against native toxin A and B [CTAB: see Example 8 (a) 20 and or IgYs against Interval 6. IgYs were prepared as 4X PEG preparations as described in Example 9(a).
The animals were orally dosed 3 times daily. roughly at 4 hour intervals, for 12 days with 1 ml antibody preparations diluted in Ensure®. Using estimates of specific antibody concentration from Example 15(c), each dose of the Interval 6 antibody prep contained approximately 400 pg of specific antibody. On day 2 each hamster was predisposed to C.
difficile infection by the oral administration of 3.0 mg of Clindamycin-HCI (Sigma) in 1 ml of water. On day 3 the hamsters were orally challenged with 1 ml of C difficile inoculum strain ATCC 43596 in sterile saline containing approximately 100 organisms. The animals were then observed for the onset of diarrhea and subsequent death during the treatment period. The results are shown in Table 19.
108 TABLE 19 Lethality After 12 Days Of Treatment Treatment Group Number Animals Alive Number Animals Dead Preimmune 0 7 CTAB 6 1 Interval 6 7 0 Treatment of hamsters with orally-administered IgYs against Interval 6 successfully protected 7 out of 7 (100%) of the animals from C difficile disease. One of the hamsters in 10 this group presented with diarrhea which subsequently resolved during the course of treatment. As shown previously in Example 9. antibodies to native toxin A and toxin B were highly protective. In this Example. 6 out of 7 animals survived in the CTAB treatment group.
All of the hamsters treated with preimmune sera came down with diarrhea and died. The survivors in both the CTAB and Interval 6 groups remained healthy throughout a 12 day post- 15 treatment period. In particular. 6 out of 7 Interval 6-treated hamsters survived at least 2 weeks after termination of treatment which suggests that these antibodies provide a longlasting cure. These results represent the first demonstration that antibodies generated against a recombinant region of toxin A can prevent CDAD when administered passively to animals.
These results also indicate that antibodies raised against Interval 6 alone may be sufficient to protect animals from C difficile disease when administered prophylactically.
Previously others had raised antibodies against toxin A by actively immunizing hamsters against a recombinant polypeptide located within the Interval 6 region [Lyerly, et al. (1990) Curr. Microbiol. 21:29]. Figure 17 shows schematically the location of the Lyerly. et al. intra-Interval 6 rdeombinant protein (cloned into the pUC vector) in comparison with the complete Interval 6 construct (pMA1870-2680) used herein to generate neutralizing antibodies directed against toxin A. In Figure 17. the solid black oval represents the MBP which is fused to the toxin A Interval 6 in pMAl870-2680.
The Lyerly, et al. antibodies (intra-Interval 6) were only able to partially protect hamsters against C. difficile infection in terms of survival (4 out of 8 animals survived) and furthermore, these antibodies did not prevent diarrhea in any of the animals. Additionally, 109rW animals treated with the intra-Interval 6 antibodies [Lyerly. et al. (1990), supra] died when treatment was removed.
In contrast. the experiment shown above demonstrates that passive administration of anti-Interval 6 antibodies prevented diarrhea in 6 out of 7 animals and completely prevented death due to CDAD. Furthermore, as discussed above, passive administration of the anti- Interval 6 antibodies provides a long lasting cure treatment could be withdrawn without incident).
b) Therapeutic Treatment Of C difficile Disease: In Vivo Treatment Of An Established C. difficile Infection In Hamsters With Recombinant Interval 6 Antibodies The ability of antibodies against recombinant interval 6 of toxin A to therapeutically treat C difficile disease was examined. The experiment was performed essentially as described in Example 10(b). Three groups. each containing seven to eight female Golden Syrian hamsters (100 g each: Charles River) were treated with either preimmune IgY, IgYs against native toxin A and toxin B (CTAB) and IgYs against Interval 6. The antibodies were prepared as described above as 4X PEG preparations.
The hamsters were first predisposed to C. difficile infection with a 3 mg dose of Clindamycin-HCI (Sigma) administered orally in 1 ml of water. Approximately 24 hrs later, 20 the animals were orally challenged with 1 ml of C difficile strain ATCC 43596 in sterile saline containing approximately 200 organisms. One day after infection, the presence of toxin :A and B was determined in the feces of the hamsters using a commercial immunoassay kit (Cytoclone A+B EPA. Cambridge Biotech) to verify establishment of infection. Four members of each group were randomly selected and tested. Feces from an uninfected hamster was tested as a negative control. All infected animals tested positive for the presence of toxin according to the manufacturer's procedure. The initiation of treatmnent then started approximately 24 hr post-infection.
The animals were dosed daily at roughly 4 hr intervals with 1 ml antibody preparation diluted in Ensure® (Ross Labs). The amount of specific antibodies given per dose (determined by affinity purification) was estimated to be about 400 pg of anti-Interval 6 IgY (for animals in the Interval 6 group) and 100 ±g and 70 jtg of anti-toxin A (Interval 6specific) and anti-toxin B (Interval 3-specific: see Example 19). respectively, for the CTAB preparation. The animals were treated for 9 days and then observed for an additional 4 days 110- W for the presence of diarrhea and death. The results indicating the number of survivors and the number of dead 4 days post-infection are shown in Table TABLE In vivo Therapeutic Treatment With Interval 6 Antibodies Treatment Group Number Animals Alive Number Animals Dead Preimmune 4 3 CTAB 8 0 Interval 6 8 0 S 10 Antibodies directed against both Interval 6 and CTAB successfully prevented death from C difficile when therapeutically administered 24 hr after infection. This result is significant since many investigators begin therapeutic treatment of hamsters with existing drugs vancomycin. phenelfamycins. tiacumicins. etc.) 8 hr post-infection [Swanson. et al. (1991) Antimicrobial Agents and Chemotherapy 35:1108 and (1989) J. Antibiotics 42:94].
15 Forty-two percent of hamsters treated with preimmune IgY died from CDAD. While the anti-Interval 6 antibodies prevented death in the treated hamsters. they did not eliminate all symptoms of CDAD as 3 animals presented with slight diarrhea. In addition. one CTABo• treated and one preimmune-treated animal also had diarrhea 14 days post-infection. These results indicate that anti-Interval 6 antibodies provide an effective means of therapy for 20 CDAD.
EXAMPLE 17 Induction Of Toxin A Neutralizine Antibodies Requires Soluble Interval 6 Protein As shown in Examples l l(d) and 15. expression of recombinant proteins in E. coli may result in the production of either soluble or insoluble protein. If insoluble protein is produced. the recombinant protein is solubilized prior to immunization of animals. To determine whether, one or both of the soluble or insoluble recombinant proteins could be used to generate neutralizing antibodies to toxin A. the following experiment was performed. This example involved a) expression of the toxin A repeats and subfragments of these repeats in E.
coli using a variety of expression vectors, b) identification of recombinant toxin A repeats and 111 V sub-regions to which neutralizing antibodies bind: and c) determination of the neutralization ability of antibodies raised against soluble and insoluble toxin A repeat immunogen.
a) Expression Of The Toxin A Repeats And Subfragments Of These Repeats In E. coli Using A Variety Of Expression Vectors The Interval 6 immunogen utilized in Examples 15 and 16 was the pMA1870-2680 protein, in which the toxin A repeats are expressed as a soluble fusion protein with the MBP (described in Example 11). Interestingly, expression of this region (from the Spel site to the end of the repeats. see Figure 15B) in three other expression constructs, as either native (pPA 870-2680). poly-His tagged [pPA1870-2680 or biotin-tagged (pBA 1870=2680) proteins resulted in completely insoluble protein upon induction of the bacterial host (see Figure 15B). The host strain BL21 (Novagen) was used for expression of pBA1870-2680 and S host strain BL21(DE3) (Novagen) was used for expression of pPA 1870-2680 and pPA1870- 2680(H). These insoluble proteins accumulated to high levels in inclusion bodies. Expression of recombinant plasmids in E. coli host cells grown in 2X YT medium was performed as described [Williams. et al. (1994), supra].
As summarized in Figure 15B. expression of fragments of the toxin A repeats (as either N-terminal SpeI-EcoRI fragments. or C-terminal EcoRI-end fragments) also yielded 20 high levels of insoluble protein using pGEX (pGA1870-2190). PinPointm-Xa (pBA1870-2190 and pBA2250-2680) and pET expression systems (pPA1870-2190). The pGEX and pET expression systems are described in Example 11. The PinPointM-Xa expression system drives the expression of fusion proteins in E. coli. Fusion proteins from PinPointM-Xa vectors contain a biotin tag at the amino-terminal end and can be affinity purified SoftLink M Soft Release avidin resin (Promega) under mild denaturing conditions (5 mM biotin).
The solubility of expressed proteins from the pPG1870-2190 and pPA1870-2190 expression constructs was determined after induction of recombinant protein expression under conditions reported to enhance protein solubility [These conditions comprise growth of the host at reduced temperature (30 0 C) and the utilization of high (1 mM IPTG) or low (0.1 mM IPTG) concentrations of inducer [Williams et al. (1994), supra]. All expressed recombinant toxin A protein was insoluble under these conditions. Thus. expression of these fragments of the toxin A repeats in pET and pGEX expression vectors results in the production of insoluble recombinant protein even when the host cells are grown at reduced temperature and using 112lower concentrations of the inducer. Although expression of these fragments in pMal vectors yielded affinity purifiable soluble fusion protein, the protein was either predominantly insoluble (pMA 1870-2190) or unstable (pMA2250-2650). Attempts to solubilize expressed protein from the pMA1870-2190 expression construct using reduced temperature or lower inducer concentration (as described above) did not improve fusion protein solubility.
Collectively, these results demonstrate that expression of the toxin A repeat region in E. coli results in the production of insoluble recombinant protein, when expressed as either large (aa 1870-2680) or small (aa 1870-2190 or aa 2250-2680) fragments. in a variety of expression vectors (native or poly-his tagged pET. pGEX or PinPoint'- Xa vectors), utilizing 10 growth conditions shown to enhance protein solubility. The exception to this rule were fusions with the MBP. which enhanced protein solubility. either partially (plvAl 870-2190) or fully (pMA1870-2680).
S* b) Identification Of Recombinant Toxin A Repeats And Sub- Regions To Which Neutralizing Antibodies Bind Toxin A repeat regions to which neutralizing antibodies bind were identified by utilizing recombinant toxin A repeat region proteins expressed as soluble or insoluble proteins to deplete protective antibodies from a polyclonal pool of antibodies against native C difficile S toxin A. An in vivo assay was developed to evaluate proteins for the ability to bind 20 neutralizing antibodies.
The rational for this assay is as follows. Recombinant proteins were first pre-mixed with antibodies against native toxin A (CTA antibody: generated in Example 8) and allowed to react. Subsequently. C. difficile toxin A was added at a concentration lethal to hamsters and the mixture was administered to hamsters via IP injection. If the recombinant protein contains neutralizing epitopes. the CTA antibodies would lose their ability to bind toxin A resulting in diarrhea and/or death of the hamsters.
The assay was performed as follows. The lethal dose of toxin A when delivered orally to nine 40 to 50 g Golden Syrian hamsters (Sasco) was determined to be 10 to 30 pg. The PEG-purified CTA antibody preparation was diluted to 0.5X concentration the antibodies were diluted at twice the original yolk volume) in 0.1 M carbonate buffer, pH 9.5. The antibodies were diluted in carbonate buffer to protect them from acid degradation in the stomach. The concentration of 0.5X was used because it was found to be the lowest effective concentration against toxin A. The concentration of Interval 6-specific antibodies in the 113- W CTA prep was estimated to be 10-15 pg/ml (estimated using the method described in Example The inclusion body preparation [insoluble Interval 6 protein: pPA1870-2680(H)] and the soluble Interval 6 protein [pMA1870-2680: see Figure 15] were both compared for their ability to bind to neutralizing antibodies against C difficil toxin A (CTA). Specifically. 1 to 2 mg of recombinant protein was mixed with 5 ml of a 0.5X CTA antibody prep (estimated to contain 60-70 pg of Interval 6-specific antibody). After incubation for I hr at 37 0 C. CTA (Tech Lab) at a final concentration of 30 pg/ml was added and incubated for another 1 hr at 37 0 C. One ml of this mixture containing 30 pg of toxin A (and 10-15 pg of Interval 6specific antibody) was administered orally to 40-50 g Golden Syrian hamsters (Sasco).
Recombinant proteins that result in the loss of neutralizing capacity of the CTA antibody would indicate that those proteins contain neutralizing epitopes. Preimmune and CTA antibodies (both at 0.5X) without the addition of any recombinant protein served as negative S* and positive controls. respectively.
15 Two other inclusion body preparations. both expressed as insoluble products in the pET vector, were tested: one containing a different insert (toxin B fragment) other than Interval 6 called pPB1850-2070 (see Figure 18) which serves as a control for insoluble Interval 6. the other was a truncated version of the Interval 6 region called pPA1870-2190 (see Figure 15B). The results of this experiment are shown in Table 21.
ai 114- .o 10 TABLE 21 Binding Of Neutralizing Antibodies By Soluble Interval 6 Protein Study Outcome After 24 Hours Treatment Group' Healthy' Diarrhea 3 Dead 4 Preimmune Ab 0 3 2 CTA Ab 4 1 0 CTA Ab Int 6 (soluble) 1 2 2 CTA Ab Int 6 (insoluble) 5 0 0 CTA Ab pPB1850-2070 5 0 0 CTA Ab pPA1870-2190 5 0 0 C. difficile toxin A (CTA) was added to each group.
S Animals showed no signs of illness.
Animals developed diarrhea but did not die.
15 4 Animals developed diarrhea and died.
Preimmune antibody was ineffective against toxin A. while anti-CTA antibodies at a dilute 0.5X concentration almost completely protected the hamsters against the enterotoxic effects of CTA. The addition of recombinant proteins pPB 1850-2070 or pPA 1870-2190 to the anti-CTA antibody had no effect upon its protective ability, indicating that these recombinant proteins do not bind to neutralizing antibodies. On the other hand. recombinant Interval 6 protein was able to bind to neutralizing anti-CTA antibodies and neutralized the in vivo protective effect of the anti-CTA antibodies. Four out of five animals in the group treated with anti-CTA antibodies mixed with soluble Interval 6 protein exhibited toxin associated toxicity (diarrhea and death). Moreover, the results showed that interval 6 protein must be expressed as a soluble product in order for it to bind to neutralizing anti-CTA antibodies since the addition of insoluble Interval 6 protein had no effect on the neutralizing capacity of the CTA antibody prep.
115c) Determination Of Neutralization Ability Of Antibodies Raised Against Soluble And Insoluble Toxin A Repeat Immunogen To determine if neutralizing antibodies are induced against solubilized inclusion bodies. insoluble toxin A repeat protein was solubilized and specific antibodies were raised in chickens. Insoluble pPA1870-2680 protein was solubilized using the method described in Williams el al. (1994), supra. Briefly. induced cultures (500 ml) were pelleted by centrifugation at 3.000 X g for 10 min at 4*C. The cell pellets were resuspended thoroughly in 10 ml of inclusion body sonication buffer (25 mM HEPES pH 7.7. 100 mM KCI, 12.5 mM MgCl,. 20% glycerol. 0.1% Nonidet P-40. 1 mM DTT). The suspension was transferred oo to a 30 ml non-glass centrifuge tube. Five hundred pl of 10 mg/ml lysozyme was-added and the tubes were incubated on ice for 30 min. The suspension was then frozen at -70 0 C for at least 1 hr. The suspension was thawed rapidly in a water bath at room temperature and then placed on ice. The suspension was then sonicated using at least eight 15 sec bursts of the microprobe (Branson Sonicator Model No. 450) with intermittent cooling on ice.
The sonicated suspension was transferred to a 35 ml Oakridge tube and centrifuged at 6,000 X g for 10 min at 4 0 C to pellet the inclusion bodies. The pellet was washed 2 times by pipetting or vortexing in fresh. ice-cold RIPA buffer SDS. 1% Triton X-100. 1% sodium deoxycholate in TBS (25 mM Tris-Cl pH 7.5. 150 mM NaCI)]. The inclusion bodies 20 were recentrifuged after each wash. The inclusion bodies were dried and transferred using a small metal spatula to a 15 ml tube (Falcon). One ml of 10% SDS was added and the pellet was solubilized by gently pipetting the solution up and down using a 1 ml micropipettor. The solubilization was facilitated by heating the sample to 95*C when necessary.
Once the inclusion bodies were in solution. the samples were diluted with 9 volumes of PBS. The protein solutions were dialyzed overnight against a 100-fold volume of PBS containing 0.05% SDS at room temperature. The dialysis buffer was then changed to PBS containing 0.01% SDS and the samples were dialyzed for several hours to overnight at room temperature. The samples were stored at 4 0 C until used. Prior to further use. the samples were warmed to room temperature to allow any precipitated SDS to go back into solution.
The inclusion body preparation was used to immunize hens. The protein was dialyzed into PBS and emulsified with approximately equal volumes of CFA for the initial immunization or IFA for subsequent booster immunizations. On day zero. for each of the recombinant recombinant preparations. two egg laying white Leghorn hens were each injected 116at multiple sites (IM and SC) with 1 ml of recombinant protein-adjuvant mixture containing approximately 0.5-1.5 mg of recombinant protein. Booster immunizations of 1.0 mg were given of days 14 and day 28. Eggs were collected on day 32 and the antibody isolated using PEG as described in Example 14(a). High titers of toxin A specific antibodies were present (as assayed by ELISA. using the method described in Example 13). Titers were determined for both antibodies against recombinant polypeptides pPA1870-2680 and pMA1870-2680 and were found to be comparable at 1:62.500.
Antibodies against soluble Interval 6 (pMA1870-2680) and insoluble Interval 6 [(inclusion body), pPA1870-2680] were tested for neutralizing ability against toxin A using the in vivo assay described in Example 15(b). Preimmune antibodies and antibodies against toxin A (CTA) served as negative and positive controls. respectively. The results are shown in Table 22.
S. TABLE 22 Neutralization Of Toxin A By Antibodies Against Soluble Interval 6 Protein Study Outcome After 24 Hours S 0 0 S S Antibody Treatment Group Healthy' Diarrhea 2 Dead 3 Preimmune 1 0 4 CTA 5 0 0 Interval 6 (Soluble) 4 5 0 0 Interval 6 (Insoluble) 0 2 3 Animals showed no sign of illness.
S Animal developed diarrhea but did not die.
3 Animal developed diarrhea and died.
4 400 g/ml.
Antibodies raised against native toxin A were protective while preimmune antibodies had little effect. As found using the in vitro CHO assay [described in Example where antibodies raised against the soluble Interval 6 could partially neutralize the effects of toxin A.
here they were able to completely neutralize toxin A in vivo. In contrast, the antibodies raised against the insoluble Interval 6 was unable to neutralize the effects of toxin A in vivo as shown above (Table 22) and in vitro as shown in the CHO assay [described in Example 117- These results demonstrate that soluble toxin A repeat immunogen is necessary to induce the production of neutralizing antibodies in chickens. and that the generation of such soluble immunogen is obtained only with a specific expression vector (pMal) containing the toxin A region spanning aa 1870-2680. That is to say. insoluble protein that is subsequently solubilized does not result in a toxin A antigen that will elicit a neutralizing antibody.
EXAMPLE 18 Cloning And Expression Of The C. difficile Toxin B Gene 10 The toxin B gene has been cloned and sequenced: the amino acid sequence deduced from the cloned nucleotide sequence predicts a MW of 269.7 kD for toxin B-[Baneso et at..
Nucl. Acids Res. 18:4004 (1990)]. The nucleotide sequence of the coding region of the entire toxin B gene is listed in SEQ ID NO:9. The amino acid sequence of the entire toxin B protein is listed in SEQ ID NO:10. The amino acid sequence consisting of amino acid residues 1850 through 2360 of toxin B is listed in SEQ ID NO:11. The amino acid sequence consisting of amino acid residues 1750 through 2360 of toxin B is listed in SEQ ID NO:12.
The amino acid sequence consisting of amino acid residues 1754 through 2362 of toxin B is listed in SEQ ID Given the expense and difficulty of isolating native toxin B protein, it would be 20 advantageous to use simple and inexpensive procaryotic expression systems to produce and purify high levels of recombinant toxin B protein for immunization purposes. Ideally. the isolated recombinant protein would be soluble in order to preserve native antigenicity. since solubilized inclusion body proteins often do not fold into native conformations. Indeed as shown in Example 17. neutralizing antibodies against recombinant toxin A were only obtained when soluble recombinant toxin A polypeptides were used as the immunogen. To allow ease of purification, the recombinant protein should be expressed to levels greater than 1 mg/liter of E. coli culture.
To determine whether high levels of recombinant toxin B protein could be produced in E. coli. fragments of the toxin B gene were cloned into various prokaryotic expression vectors, and assessed for the ability to express recombinant toxin B protein in E. coli. This Example involved cloning of the toxin B gene and expression of the toxin B gene in E. coli.
-118a) Cloning Of The Toxin B Gene The toxin B gene was cloned using PCR amplification from C. difficile genomic DNA.
Initially, the gene was cloned in two overlapping fragments, using primer pairs P5/P6 and P7/P8. The location of these primers along the toxin B gene is shown schematically in Figure 18. The sequence of each of these primers is: 5' TAGAAAAAATGGCAAATGT 3' (SEQ ID NO:l 1); P6: 5'TTTCATCTTGTA GAGTCAAAG 3' (SEQ ID NO:12): P7: 5' GATGCCACAAGATGATTTAGTG 3' (SEQ ID NO:13): and P8: 5' CTAATTGAGCTGTATCAGGATC 3' (SEQ ID NO:14).
Figure 18 also shows the location of the following primers along the toxin B gene: P9 which consists of the sequence 5' CGGAATTCCTAGAAAAAATGGCAA ATG y(SEQ ID NO:15); PI0 which consists of the sequence 5' GCTCTAGAATGA CCATAAGCTAGCCA 3' (SEQ ID NO:16): P I which consists of the sequence 5' CGGAATTCGAGTTGGTAG- AAAGGTGGA 3' (SEQ ID NO:17): P13 which consists of the sequence 5' CGGAATTCGG- 15 TTATTATCTTAAGGATG 3' (SEQ ID NO:18): and P14 which consists of the sequence CGGAATTCTTGATAACTGGAT TTGTGAC 3' (SEQ ID NO:19). The amino acid sequence consisting of amino acid residues 1852 through 2362 of toxin B is listed in SEQ ID NO:20. The amino acid sequence consisting of amino acid residues 1755 through 2362 of toxin B is listed in SEQ ID NO:21. The amino acid sequence consisting of amino acid residues 1754 through 2362 of toxin B is listed in SEQ ID •Clostridium difficile VPI strain 10463 was obtained from the American Type Culture Collection (ATCC 43255) and grown under anaerobic conditions in brain-heart infusion medium (Becton Dickinson). High molecular-weight C difficile DNA was isolated essentially as described [Wren and Tabaqchali (1987) J. Clin. Microbiol.. 25:2402], except 1) 100 pg/ml proteinase K in 0.5% SDS was used to disrupt the bacteria and 2) cetytrimethylammonium bromide (CTAB) precipitation [as described by Ausubel et al.. Eds.. Current Protocols in Molecular Biology, Vol. 2 (1989) Current Protocols] was used to remove carbohydrates from the cleared lysate. Briefly, after disruption of the bacteria with proteinase K and SDS, the solution is adjusted to approximately 0.7 M NaCI by the addition of a 1/7 volume of NaCI. A 1/10 volume of CTAB/NaCI (10% CTAB in 0.7 M NaCI) solution was added and the solution was mixed thoroughly and incubated 10 min at 65*C. An equal volume of chloroform/isoamyl alcohol (24:1) was added and the phases were thoroughly mixed. The organic and aqueous phases were separated by centrifugation in a microfuge for 5 min. The 119aqueous supernatant was removed and extracted with phenol/chloroform/ isoamyl alcohol (25:24:1). The phases were separated by centrifugation in a microfuge for 5 min. The supernatant was transferred to a fresh tube and the DNA was precipitated with isopropanol.
The DNA precipitate was pelleted by brief centrifugation in a microfuge. The DNA pellet was washed with 70% ethanol to remove residual CTAB. The DNA pellet was then dried and redissolved in TE buffer (10 mM Tris-HCI pH8.0. 1 mM EDTA). The integrity and yield of genomic DNA was assessed by comparison with a serial dilution of uncut lambda DNA after electrophoresis on an agarose gel.
Toxin B fragments were cloned by PCR utilizing a proofreading thermostable DNA polymerase (native Pfu polymerase (Stratagene)]. The high fidelity of this polymerase reduces the mutation problems associated with amplification by error prone-polyrrases Taq polymerase). PCR amplification was performed using the PCR primer pairs P5 (SEQ ID NO:11) with P6 (SEQ ID NO:12) and P7 (SEQ ID NO:13) with P8 (SEQ ID NO:14) in il reactions containing 10 mM Tris-HCl pH8.3. 50 mM KCI. 1.5 mM MgCI,. 200 pM of 15 each dNTP. 0.2 pM each primer, and 50 ng C. difficile genomic DNA. Reactions were overlaid with 100 pl mineral oil. heated to 94*C for 4 min, 0.5p1 native Pfu polymerase (Stratagene) was added, and the reactions were cycled 30 times at 94 0 C for I min. 50 0 C for 1 min. 72*C (2 min for each kb of sequence to be amplified), followed by 10 min at 72*C.
Duplicate reactions were pooled. chloroform extracted, and ethanol precipitated. After S 20 washing in 70% ethanol. the pellets were resuspended in 50 pl TE buffer (10 mM Tris-HCI pH8.0. 1 mM EDTA).
The P5/P6 amplification product was cloned into pUC19 as outlined below. 10l1 aliquots of DNA were gel purified using the Prep-a-Gene kit (BioRad). and ligated to Smal restricted pUC19 vector. Recombinant clones were isolated and confirmed by restriction digestion using standard recombinant molecular biology techniques (Sambrook et al.. 1989).
Inserts from two independent isolates were identified in which the toxin B insert was oriented such that the vector BamHI and Sad sites were 5' and 3' oriented. respectively (pUCB 1530). The insert-containing BamHI/Sacl fragment was cloned into similarly cut pET23a-c vector DNA. and protein expression was induced in small scale cultures (5 ml) of identified clones.
Total protein extracts were isolated, resolved on SDS-PAGE gels. and toxin B protein identified by Western analysis utilizing a goat anti-toxin B affinity purified antibody (Tech Lab). Procedures for protein induction. SDS-PAGE. and Western blot analysis were 120performed as described in Williams et al. (1994), supra. In brief. 5 ml cultures of bacteria grown in 2XYT containing 100 pg/ml ampicillin containing the appropriate recombinant clone were induced to express recombinant protein by addition of IPTG to ImM. The E. coli hosts used were: BL21(DE3) or BL21(DE3)LysS (Novagen) for pET plasmids.
Cultures were induced by the addition of IPTG to a final concentration of 1.0 mM when the cell density reached 0.5 OD6, and induced protein was allowed to accumulate for two hrs after iiduction. Protein samples were prepared by pelleting 1 ml aliquots of bacteria by centrifugation (1 min in microfuge), and resuspension of the pelleted bacteria in 150 l of 2X SDS-PAGE sample buffer (0.125 mM Tris-HCI pH 6.8. 2 mM EDTA. 6% SDS. glycerol. 0.025% bromophenol blue: 3-mercaptoethanol is added to 5% before use). The samples were heated to 95*C for 5 min. then cooled and 5 or 10 pls loaded on 7.5% SDS- PAGE gels. High molecular weight protein markers (BioRad) were also loaded, to allow estimation of the MW of identified fusion proteins. After electrophoresis. protein was detected either generally by staining the gels with Coomassie Blue. or specifically, by blotting 15 to nitrocellulose for Western blot detection of specific immunoreactive protein. The MW of induced toxin B reactive protein allowed the integrity of the toxin B reading frame to be determined.
The pET23b recombinant (pPB 10-1530) expressed high MW recombinant toxin B reactive protein, consistent with predicted values. This confirmed that frame terminating 20 errors had not occurred during PCR amplification. A pET23b expression clone containing the 10-1750aa interval of the toxin B gene was constructed. by fusion of the EcoRV-Spel fragment of the P7/P8 amplification product to the P5-EcoRV interval of the P5/P6 S"amplification product (see Figure 18) in pPB0-1530. The integrity of this clone (pPBIO- 1750) was confirmed by restriction mapping, and Western blot detection of expressed recombinant toxin B protein. Levels of induced protein from both pPB10-1530 and pPBI0- 1750 were too low to facilitate purification of usable amounts of recombinant toxin B protein.
The remaining 1750-2360 aa interval was directly cloned into pMAL or pET expression vectors from the P7/P8 amplification product as described below.
b) Expression Of The Toxin B Gene i) Overview Of Expression Methodologies Fragments of the toxin B gene were expressed as either native or fusion proteins in E.
coli. Native proteins were expressed in either the pET23a-c or pET16b expression vectors 121 (Novagen). The pET23 vectors contain an extensive polylinker sequence in all three reading frames (a-c vectors) followed by a C-terminal poly-histidine repeat. The pET16b vector contains a N-terminal poly-histidine sequence immediately 5' to a small polylinker. The poly-histidine sequence binds to Ni-Chelate columns and allows affinity purification of tagged target proteins [Williams et al. (1994). supra]. These affinity tags are small (10 aa for pET16b. 6 aa for pET23) allowing the expression and affinity purification of native proteins with only limited amounts of foreign sequences.
An N-terminal histidine-tagged derivative of pET16b containing an extensive cloning cassette was constructed to facilitate cloning of N-terminal poly-histidine tagged toxin B expressing constructs. This was accomplished by replacement of the promoter region of the pET23a and b vectors with that of the pETI6b expression vector. :Each vector was restricted with BglI and Ndel. and the reactions resolved on a 1.2 agarose gel. The pET16b promoter region (contained in a 200 bp BgllI-NdeI fragment) and the promoter-less pET23 a or b vectors were cut from the gel. mixed and Prep-A-Gene (BioRad) purified. The eluted 15 DNA was ligated, and transformants screened for promoter replacement by NcoI digestion of purified plasmid DNA (the pET16b promoter contains this site, the pET23 promoter does not). These clones (denoted pETHisa or b) contain the pET16b promoter (consisting of a pT7-lac promoter, ribosome binding site and poly-histidine (lOaa) sequence) fused at the Ndel site to the extensive pET23 polylinker.
20 All MBP fusion proteins were constructed and expressed in the pMAL™-c or pMALM-p2 vectors (New England Biolabs) in which the protein of interest is expressed as a C-terminal fusion with MBP. All pET plasmids were expressed in either the BL21(DE3) or BL21(DE3)LysS expression hosts. while pMal plasmids were expressed in the BL21 host.
Large scale (500 mis to 1 liter) cultures of each recombinant were grown in 2X YT broth, induced, and soluble protein fractions were isolated as described [Williams, et al.
(1994). supra]. The soluble protein extracts were affinity chromatographed to isolate recombinant fusion protein, as described [Williams et al., (1994) supra]. In brief, extracts containing tagged pET fusions were chromatographed on a nickel chelate column, and eluted using imidazole salts or low pH (pH 4.0) as described by the distributor (Novagen or Qiagen).
Extracts containing soluble pMAL fusion protein were prepared and chromatographed in PBS buffer over an amylose resin (New England Biolabs) column, and eluted with PBS containing mM maltose as described [Williams et al. (1994), supra].
122 ii) Overview Of Toxin B Expression In both large expression constructs described in above, only low level less than 1 mg/liter of intact or nondegraded recombinant protein) expression of recombinant protein was detected. A number of expression constructs containing smaller fragments of the toxin B gene were then constructed. to determine if small regions of the gene can be expressed to high levels greater than 1 mg/liter intact protein) without extensive protein degradation. All were constructed by in frame fusions of convenient toxin B restriction fragments to either the pMAL-c. pET23a-c. pET16b or pETHisa-b expression vectors, or by engineering restriction sites at specific locations using PCR amplification [using the same conditions described in above]. In all cases, clones were verified by restriction mapping, and. where indicated. DNA sequencing.
Protein preparations from induced cultures of each of these constructs were analyzed.
by SDS-PAGE. to estimate protein stability (Coomassie Blue staining) and immunoreactivity against anti-toxin B specific antiserum (Western analysis). Higher levels of intact 15 nondegraded). full length fusion proteins were observed with the smaller constructs as compared with the larger recombinants. and a series of expression constructs spanning the entire toxin B gene were constructed (Figures 18. 19 and 20 and Table 23).
Constructs that expressed significant levels of recombinant toxin B protein (greater than 1 mg/liter intact recombinant protein) in E. coli were identified and a series of these S 20 clones that spans the toxin B gene are shown in Figure 19 and summarized in Table 23.
S" These clones were utilized to isolate pure toxin B recombinant protein from the entire toxin B gene. Significant protein yields were obtained from pMAL expression constructs spanning the entire toxin B gene. and yields of full length soluble fusion protein ranged from an estimated 1 mg/liter culture (pMB 1100-1530) to greater than 20 mg/liter culture (pMB 1750-2360).
Representative purifications of MBP and poly-histidine-tagged toxin B recombinants are shown in Figures 21 and 22. Figure 21 shows a Coomassie Blue stained 7.5% SDS- PAGE gel on which various protein samples extracted from bacteria harboring pMB1850- 2360 were electrophoresed. Samples were loaded as follows: Lane 1: protein extracted from uninduced culture: Lane 2: induced culture protein: Lane 3: total protein from induced culture after sonication: Lane 4: soluble protein; and Lane 5: eluted affinity purified protein. Figure 22 depicts the purification of recombinant proteins expressed in bacteria harboring either pPB1850-2360 (Lanes 1-3) or pPB1750-2360 (Lanes Samples were loaded as follows: uninduced total protein (Lanes 1 and induced total protein (Lanes 2 and and eluted 123 affinity purified protein (Lanes 3 and The broad range molecular weight protein markers (BioRad) are shown in Lane 7.
Thus, although high level expression was not attained using large expression constructs from the toxin B gene. usable levels of recombinant protein were obtained by isolating induced protein from a series of smaller pMAL expression constructs that span the entire toxin B gene.
Theseresults represent the first demonstration of the feasibility of expressing recombinant toxin B protein to high levels in E. coli. As well. expression of small regions of the putative ligand binding domain (repeat region) of toxin B as native protein yielded insoluble protein, while large constructs, or fusions to MBP were soluble (Figure 19).
demonstrating that specific methodologies are necessary to produce soluble fusion-protein from this interval.
0 iii) Clone Construction And Expression Details 15 A portion of the toxin B gene containing the toxin B repeat region [amino acid residues 1852-2362 of toxin B (SEQ ID NO:20)] was isolated by PCR amplification of this interval of the toxin B gene from C difficile genomic DNA. The sequence, and location within the toxin B gene. of the two. PCR primers [P7 (SEQ ID NO:13) and P8 (SEQ ID NO:14)] used to amplify this region are shown in Figure 18.
20 DNA from the PCR amplification was purified by chloroform extraction and ethanol precipitation as described above. The DNA was restricted with Spel. and the cleaved DNA was resolved by agarose gel electrophoresis. The restriction digestion with Spel cleaved the 3.6 kb amplification product into a 1.8 kb doublet band. This doublet band was cut from the gel and mixed with appropriately cut, gel purified pMALc or pET23b vector. These vectors were prepared by digestion with HindII. filling in the overhanging ends using the Klenow enzyme, and cleaving with Xbal (pMALc) or Nhel (pET23b). The gel purified DNA fragments were purified using the Prep-A-Gene kit (BioRad) and the DNA was ligated, transformed and putative recombinant clones analyzed by restriction mapping.
pET and pMal clones containing the toxin B repeat insert (aa interval 1750-2360 of toxin B) were verified by restriction mapping, using enzymes that cleaved specific sites within the toxin B region. In both cases fusion of.the toxin B Spel site with either the compatible Xbal site (pMal) or compatible Nhel site (pET) is predicted to create an in frame fusion. This was confirmed in the case of the pMB 1750-2360 clone by DNA sequencing of the clone 124junction and 5' end of the toxin B insert using a MBP specific primer (New England Biolabs). In the case of the pET construct. the fusion of the blunt ended toxin B 3' end to the filled HindIll site should create an in-frame fusion with the C-terminal poly-histidine sequence in this vector. The pPB1750-2360 clone selected had lost as predicted, the HindIII site at this clone junction: this eliminated the possibility that an additional adenosine residue was added to the 3' end of the PCR product by a terminal transferase activity of the Pfu polymerase. since fusion of this adenosine residue to the filled HindIll site would regenerate the restriction site (and was observed in several clones).
One liter cultures of each expression construct were grown. and fusion protein purified by affinity chromatography on either an amylose resin column (pMAL constructs: resin supplied by New England Biolabs) or Ni-chelate column (pET constructs: resin supplied by Qiagen or Novagen) as described [Williams et al. (1994). supra]. The integrity and purity of the fusion proteins were determined by Coomassie staining of SDS-PAGE protein gels as well as Western blot analysis with either an affinity purified goat polyclonal antiserum (Tech Lab).
S. 15 or a chicken polyclonal PEG prep. raised against the toxin B protein (CTB) as described above in Example 8. In both cases. affinity purification resulted in yields in excess of 20 mg protein per liter culture. of which greater than 90% was estimated to be full-length recombinant protein. It should be noted that the poly-histidine affinity tagged protein was released from the Qiagen Ni-NTA resin at low imidazole concentration (60 mM), 20 necessitating the use of a 40 mM imidazole rather than a 60 mM imidazole wash step during purification.
A periplasmically secreted version of pMB1750-2360 was constructed by replacement of the promoter and MBP coding region of this construct with that from a related vector (pMALM-p2; New England Biolabs) in which a signal sequence is present at the N-terminus of the MBP. such that fusion protein is exported. This was accomplished by substituting a BglII-EcoRV promoter fragment from pMAL-p2 into pMB1750-2360. The yields of secreted.
affinity purified protein (recovered from osmotic shock extracts as described by Riggs in Current Protocols in Molecular Biology, Vol. 2. Ausubel. et al.. Eds. (1989). Current Protocols. pp. 16.6.1 16.6.14] from this vector (pMBpl750-2360) were 6.5 mg/liter culture.
of which 50% was estimated to be full-length fusion protein.
The interval was also expressed in two non-overlapping fragments. pMB1750-1970 was constructed by introduction of a frameshift into pMB1750-2360. by restriction with HindII. filling in the overhanging ends and religation of the plasmid. Recombinant clones 4 125 Swere selected by loss of the Hindlll site. and further restriction map analysis. Recombinant protein expression from this vector was more than 20 mg/liter of greater than 90% pure protein.
The complementary region was expressed in pMB1970-2360. This construct was created by removal of the 1750-1970 interval of pMB 1750-2360. This was accomplished by restriction of this plasmid with EcoRI (in the pMalc polylinker 5' to the insert) and III, filling in the overhanging ends. and religation of the plasmid. The resultant plasmid, pMB1970-2360.
was made using both intracellularly and secreted versions of the pMB1750-2360 vector.
No fusion protein was secreted in the pMB1970-2360 version. perhaps due to a conformational constraint that prevents export of the fusion protein. However, the intracellularly expressed vector produced greater than 40mg/liter of greater than .90% fulllength fusion protein.
Constructs to precisely express the toxin B repeats in either pMalc (pMB1850-2360) or pET16b (pPB1850-2360) were constructed as follows. The DNA interval including the toxin 15 B repeats was PCR amplified as described above utilizing PCR primers P14 (SEQ ID NO:19) and P8 (SEQ ID NO:14). Primer P14 adds a EcoRI site immediately flanking the start of the toxin B repeats.
The amplified fragment was cloned into the pT7 Blue T-vector (Novagen) and recombinant clones in which single copies of the PCR fragment were inserted in either S 20 orientation were selected (pT71850-2360) and confirmed by restriction mapping. The insert was excised from two appropriately oriented independently isolated pT71850-2360 plasmids.
with EcoRI end of repeats) and PstI (in the flanking polylinker of the vector), and cloned into EcoRI/PstI cleaved pMalc vector. The resulting construct (pMB1850-2360) was confirmed by restriction analysis, and yielded 20 mg/I of soluble fusion protein [greater than 90% intact nondegraded)] after affinity chromatography. Restriction of this plasmid with HindIII and religation of the vector resulted in the removal of the 1970-2360 interval. The resultant construct (pMB1850-1970) expressed greater than 70 mg/liter of 90% full length fusion protein.
The pPB1850-2360 construct was made by cloning a EcoRI (filled with Klenow)- BamHI fragment from a pT71850-2360 vector (opposite orientation to that used in the pMB1850-2360 construction) into Ndel (filled)/BamHI cleaved pET16b vector. Yields of affinity purified soluble fusion protein were 15 mg/liter. of greater than 90% full length fusion protein.
126- Several smaller expression constructs from the 1750-2070 interval were also constructed in His-tagged pET vectors, but expression of these plasmids in the BL21 (DE3) host resulted in the production of high levels of mostly insoluble protein (see Table 23 and Figure 19). These constructs were made as follows.
pPB1850-1970 was constructed by cloning a BgIII-HindIII fragment of pPB1850-2360 into BglII/HindlII cleaved pET23b vector. pPB1850-2070 was constructed by cloning a Bgll-Pvuill fragment of pPB1850-2360 into Bgill/HinclI cleaved pET23b vector. pPB1750- 1970(c) was constructed by removal of the internal HindIII fragment of a pPB1750-2360 vector in which the vector Hindlll site was regenerated during cloning (presumably by the addition of an A residue to the amplified PCR product by terminal transferase activity of Pfu polymerase). The pPB1750-1970(n) construct was made by insertion of the insert-eentaining the Ndel-Hindll fragment of pPB1750-2360 into identically cleaved pETHisb vector. All constructs were confirmed by restriction digestion.
An expression construct that directs expression of the 10-470 aa interval of toxin B 15 was constructed in the pMalc vector as follows. A Nhel (a site 5' to the insert in the pET23 vector)-AflII (filled) fragment of the toxin B gene from pPB10-1530 was cloned into Xbal (compatible with Nhel)/HindIII (filled) pMalc vector. The integrity of the construct (pMBlO- 470) was verified by restriction mapping and DNA sequencing of the 5' clone junction using a MBP specific DNA primer (New England Biolabs). However, all expressed protein was 20 degraded to the MBP monomer MW.
A second construct spanning this interval (aa 10-470) was constructed by cloning the PCR amplification product from a reaction containing the P9 (SEQ ID NO:15) and P10 (SEQ ID NO:16) primers (Figure 18) into the pETHisa vector. This was accomplished by cloning the PCR product as an EcoRI-blunt fragment into EcoRI-Hincll restricted vector DNA; recombinant clones were verified by restriction mapping. Although this construct (pPB 520) allowed expression and purification (utilizing the N-terminal polyhistidine affinity tag) of intact fusion protein, yields were estimated at less than 500 .g per liter culture.
Higher yield of recombinant protein from this interval (aa 10-520) were obtained by expression of the interval in two overlapping clones. The 10-330aa interval was cloned in both pETHisa and pMalc vectors, using the BamHI-AfIIl (filled) DNA fragment from pPBl0- 520. This fragment was cloned into BamHI-HindlII (filled) restricted pMalc or BamHI-Hincll restricted pETHisa vector. Recombinant clones were verified by restriction mapping. High level expression of either insoluble (pET) or soluble (pMal) fusion protein was obtained. Total I 127yields of fusion protein from the pMB10-330 construct (Figure 18) were 20 mg/liter culture.
of which 10% was estimated to be full-length fusion protein. Although yields of this interval were higher and >90% full-length recombinant protein produced when expressed from the pET construct. the pMal fusion was utilized since the expressed protein was soluble and thus more likely to have the native conformation.
The pMB260-520 clone was constructed by cloning EcoRI-Xbal cleaved amplified DNA from a PCR reaction containing the P I (SEQ ID NO:17) and P10 (SEQ ID NO:16) DNA primers (Figure 18) into similarly restricted pMalc vector. Yields of affinity purified protein were 10 mg/liter. of which approximately 50% was estimated to be full-length recombinant protein.
The aa510-1 110 interval was expressed as described below. This entire inteval was expressed as a pMal fusion by cloning the Nhel-Hindl fragment of pUCBl0-1530 into XbaI- HindIII cleaved pMalc vector. The integrity of the construct (pMB510-1 110) was verified by o. restriction mapping and DNA sequencing of the 5' clone junction using a MBP specific DNA S* 15 primer. The yield of affinity purified protein was 25 mg/liter culture. of which 5% was estimated to be full-length fusion protein (1 mg/liter).
To attempt to obtain higher yields. this region was expressed in two fragments (aa510- 820. and 820-1110) in the pMalc vector. The pMB510-820 clone was constructed by insertion of a Sacl (in the pMalc polylinker 5' to the insert)-Hpal DNA fragment from 20 pMB510-1110 into Sacl/Stul restricted pMalc vector. The pMB820-1110 vector was constructed by insertion of the Hpal-HindlII fragment of pUCBl0-1530 into BamHI (filled)/HindlII cleaved pMalc vector. The integrity of these constructs were verified by restriction mapping and DNA sequencing of the 5' clone junction using a MBP specific DNA primer.
Recombinant protein expressed from the pMB510-820 vector was highly unstable.
However, high levels (20 mg/liter) of >90% full-length fusion protein were obtained from the pMB820-1105 construct. The combination of partially degraded pMB510-1110 protein (enriched for the 510-820 interval) with the pMB820-1 110 protein provides usable amounts of recombinant antigen from this interval.
The aal100-1750 interval was expressed as described below. The entire interval was expressed in the pMalc vector from a construct in which the AccI(filled)-Spel fragment of pPBIO-1750 was inserted into StullXbal (Xbal is compatible with Spel: Stul and filled Accl sites are both blunt ended) restricted pMalc. The integrity of this construct (pMB1100-1750) 128- Swas verified by restriction mapping and DNA sequencing of the clone junction using a MBP specific DNA primer. Although 15 mg/liter of affinity purified protein was isolated from cells harboring this construct, the protein was greater than 99% degraded to MBP monomer size.
A smaller derivative of pMBI 100-1750 was constructed by restriction of pMBI 100- 1750 with AfJll and Sail (in the pMalc polylinker 3' to the insert), filling in the overhanging ends. and religaTing the plasmid. The resultant clone (verified by restriction digestion and DNA sequencing) has deleted the aal530-1750 interval, was designated pMBI 100-1530.
pMB 1100-1530 expressed recombinant protein at a yield of greater than 40 mg/liter. of which 5% was estimated to be full-length fusion protein.
Three constructs were made to express the remaining interval. Initially.a BspHI S(filled)-Spel fragment from pPBlO-1750 was cloned into EcoRI(filled)lXbal cleaved pMalc vector. The integrity of this construct (pMB1570-1750) was verified by restriction mapping and DNA sequencing of the 5' clone junction using a MBP specific DNA primer. Expression g 15 of recombinant protein from this plasmid was very low. approximately 3 mg affinity purified protein per liter, and most was degraded to MBP monomer size. This region was subsequently expressed from a PCR amplified DNA fragment. A PCR reaction utilizing primers P13 [SEQ ID NO:18; P13 was engineered to introduce an EcoRI site 5' to amplified toxin B sequences] and P8 (SEQ ID NO:14) was performed on C. difficile genomic DNA as 20 described above. The amplified fragment was cleaved with EcoRI and SpeI. and cloned into EcoRIIXbaI cleaved pMalc vector. The resultant clone (pMB1530-1750) was verified by restriction map analysis, and recombinant protein was expressed and purified. The yield was greater than 20 mg protein per liter culture and it was estimated that 25% was full-length fusion protein: this was a significantly higher yield than the original pMB1570-1750 clone.
The insert of pMB1530-1750 (in a EcoRI-Sall fragment) was transferred to the pETHisa vector (EcoRI/XhoI cleaved. Xhol and Sail ends are compatible). No detectable fusion protein was purified on Ni-Chelate columns from soluble lysates of cells induced to express fusion protein from this construct.
129- S. 55
S
S.
S
S S *5 S.
S.
5' 5O *5 TABLE 23 Summary Of Toxin B Expression Constructs' Clone Affinity Tag Yield (mg/liter) Full Length (estimated from9 pPBIO1750noneWestern blot hyb.) 1530 none low (as above) pMB10-470 MBP 15mg/I 0% pPB 10-520 poly-his 0.5mg/I pPBI10-330 poly-his >20mg/I (insoluble) pMBJ0-330 MB? 20mg/I pMB26O-520 MBP I10mg/I pMBSIO-J 110 MBP 25mg/I pMB5 10-820 MBP degraded (by Western blot hyb) pMB82O-I1JO MBP 20mg/i pMB 1100- 1750 MBP 15mg/I 0% pMlBIJOO-1530 MBP 40mg/I pMBI57O-1750 MBP 3mg/I pPBI53O-1750 poly-his no purified 9) protein detected pMB]S3O-1 750 MB? 20mg/i pMBI 750-2360 MBP >20mg/I pMBpl750-2360 MBP 6.5mg/I (secreted) PPB1750-2360 poly-his >20mg/I pMB175O-1970 MBP >20mg/I 130 TABLE 23 Summary Of Toxin B Expression Constructs" Clone Affinity Tag Yield (mg/liter) Full Length pMB1970-2360 MBP 40mg/l pMBp197Q-2360 MBP (no secretion) NA pMB1850-2360 MBP 20mg/l pPB 1850-2360 poly-his 15mg/I pMB1850-1970 MBP 70mg/I >90% pPB1850-!970 poly-his >10mg/l (insoluble) pPB 1850-2070 poly-his >10mg/l (insoluble) pPB1750-1970(c) poly-his >1 0mg/i (insoluble) pPB1750-1970(n) poly-his >10mg/1 (insoluble) Oe 0W 00 S 0 e
S
0 0 0 0@@ 0*00 S. S
S
00 0 Clones in italics are clones currently utilized to purify recombinant protein from each selected interval.
EXAMPLE 19 Identification. Purification And Induction Of Neutralizing Antibodies Against Recombinant C. difficile Toxin B Protein To determine whether recombinant toxin B polypeptide fragments can generate neutralizing antibodies, typically animals would first be immunized with recombinant proteins and anti-recombinant antibodies-are generated. These anti-recombinant protein antibodies are then tested for neutralizing ability in vivo or in vitro. Depending on the immunogenic nature of the recombinant polypeptide. the generation of high-titer antibodies against that protein may take several months. To accelerate this process and identify which recombinant polypeptide(s) may be the best candidate to generate neutralizing antibodies, depletion studies were performed. Specifically, recombinant toxin B polypeptide were pre-screened by testing whether they have the ability to bind to protective antibodies from a CTB antibody preparation and hence deplete those antibodies of their neutralizing capacity. Those 131 W recombinant polypeptides found to bind CTB. were then utilized to generate neutralizing antibodies. This Example involved: a) identification of recombinant sub-regions within toxin B to which neutralizing antibodies bind; b) identification of toxin B sub-region specific antibodies that neutralize toxin B in vivo; and c) generation and evaluation of antibodies reactive to recombinant toxin B polypeptides.
a) Identification Of Recombinant Sub-Regions Within Toxin B To Which Neutralizing Antibodies Bind Sub-regions within toxin B to which neutralizing antibodies bind were identified by utilizing recombinant toxin B proteins to deplete protective antibodies from a polyclonal pool of antibodies against native C. difficile toxin B. An in vivo assay was developed te-evaluate protein preparations for the ability to bind neutralizing antibodies. Recombinant proteins were first pre-mixed with antibodies directed against native toxin B (CTB antibody: see Example 8) and allowed to react for one hour at 37 0 C. Subsequently. C. difficile toxin B (CTB: Tech 15 Lab) was added at a concentration lethal to hamsters and incubated for another hour at 37°C.
After incubation this mixture was injected intraperitoneally (IP) into hamsters. If the i: recombinant polypeptide contains neutralizing epitopes. the CTB antibodies will lose its ability to protect the hamsters against death from CTB. If partial or complete protection occurs with the CTB antibody-recombinant mixture. that recombinant contains only weak or S* 20 non-neutralizing epitopes of toxin B. This assay was performed as follows.
Antibodies against CTB were generated in egg laying Leghorn hens as described in Example 8. The lethal dosage (LD of C difficile toxin B when delivered I.P. into female Golden Syrian hamsters (Charles River) was determined to be 2.5 to 5 lg. Antibodies generated against CTB and purified by PEG precipitation could completely protect the hamsters at the I.P. dosage determined above. The minimal amount of CTB antibody needed to afford good protection against 5 ug of CTB when injected I.P. into hamsters was also determined (IX PEG prep). These experiments defined the parameters needed to test whether a given recombinant protein could deplete protective CTB antibodies.
132 The cloned regions tested for neutralizing ability cover the entire toxin B gene and were designated as Intervals (INT) 1 through 5 (see Figure 19). Approximately equivalent final concentrations of each recombinant polypeptide were tested. The following recombinant polypeptides were used: 1) a mixture of intervals I and 2 (INT-1. 2) a mixture of Intervals 4 and 5 (INT-4. 5) and 3) Interval 3 (INT-3). Recombinant proteins (each at about 1 mg total protein) were first preincubated with a final CTB antibody concentration of IX pellet dissolved in original yolk volume as described in Example in a final volume of mis for 1 hour at 37*C. Twenty-five pg of CTB (at a concentration of 5 pg/ml), enough CTB to kill 5 hamsters. was then added and the mixture was then incubated for 1 hour at 37 0 C. Five, 40g female hamsters (Charles River) in each treatment group were then each given 1 ml of the mixture I.P. using a tuberculin syringe with a 27 gauge needle. -The results of this experiment are shown in Table 24.
*TABLE 24 SBinding Of Neutralizing Antibodies By INT 3 Protein Binding Of Neutralizing Antibodies By INT 3 Protein 15 Treatment Group' Number Of Animals Alive Number Of Animals Dead CTB antibodies 3 2 CTB antibodies INT1.2 3 2 CTB antibodies INT4.5 3 2 CTB antibodies INT 3 0 C. difficile toxin B (CTB) was added to each group.
As shown in Table 24, the addition of recombinant proteins from INT-1. 2 or INT-4. 'had no effect on the in vivo protective ability of the CTB antibody preparation compared to the CTB antibody preparation alone. In contrast. INT-3 recombinant polypeptide was able to remove all of the toxin B neutralizing ability of the CTB antibodies as demonstrated by the death of all the hamsters in that group.
133 The above experiment was repeated. using two smaller expressed fragments (pMB 1750-1970 and pMB 1970-2360. see Figure 19) comprising the INT-3 domain to determine if that domain could be further subdivided into smaller neutralizing epitopes. In addition, fulllength INT-3 polypeptide expressed as a nickel tagged protein (pPB1750-2360) was tested for neutralizing ability and compared to the original INT-3 expressed MBP fusion (pMB1750- 2360). The results are shown in Table TABLE Removal Of Neutralizing Antibodies By Repeat Containing Proteins 10 Treatment Group Number Of Number Of Animals Alive Animals-Dead CTB antibodies 5 0 CTB antibodies pPB1750-2360 0 CTB antibodies pMB1750-2360 0 CTB antibodies pMB1970-2360 3 2 CTB antibodies pMB1750-1970 2 3 C. difficile toxin B (CTB) was added to each group.
The results summarized in Table 25 indicate that the smaller polypeptide fragments within the INT-3 domain. pMB1750-1970 and pMB1970-2360. partially lose the ability to bind to and remove neutralizing antibodies from the CTB antibody pool. These results demonstrate that the full length INT-3 polypeptide is required to completely deplete the CTB antibody pool of neutralizing antibodies. This experiment also shows that the neutralization epitope of INT-3 can be expressed in alternative vector systems and the results are independent of the vector utilized or the accompanying fusion partner.
Other Interval 3 specific proteins were subsequently tested for the ability to remove neutralizing antibodies within the CTB antibody pool as described above. The Interval 3 specific proteins used in these studies are summarized in Figure 23. In Figure 23 the following abbreviations are used: pP refers to the pET23 vector: pM refers to the pMALc vector: B refers to toxin B: the numbers refer to the amino acid interval expressed in the clone. The solid black ovals represent the MBP: and HHH represents the poly-histidine tag: I 134- Only recombinant proteins comprising the entire toxin B repeat domain (pMB1750- 2360. pPB1750- 236 0 and pPB 850-2360) can bind and completely remove neutralizing antibodies from the CTB antibody pool. Recombinant proteins comprising only a portion of the toxin B repeat domain were not capable of completely removing neutralizing antibodies from the CTB antibody pool (pMB1750-1970 and pMB1970-2360 could partially remove neutralizing antibodies while pMB1850-1970 and pPB1850-2070 failed to remove any neutralizing antibodies from the CTB antibody pool).
The above results demonstrate that only the complete ligand binding domain (repeat region) of the toxin B gene can bind and completely remove neutralizing antibodies from the 10 CTB antibody pool. These results demonstrate that antibodies directed against the entire toxin B repeat region are necessary for in vivo toxin neutralization (see Figure 23: only the recombinant proteins expressed by the pMB1750-2360. pPB1750-2360 and pPB1850-2360 vectors are capable of completely removing the neutralizing antibodies from the CTB antibody pool).
These results represent the first indication that the entire repeat region of toxin B
S
would be necessary for the generation of antibodies capable of neutralizing toxin B. and that sub-regions may not be sufficient to generate maximal titers of neutralizing antibodies.
b) Identification Of Toxin B Sub-Region Specific Antibodies That Neutralize Toxin B In Vivo To determine if antibodies directed against the toxin B repeat region are sufficient for neutralization, region specific antibodies within the CTB antibody preparation were affinity purified, and tested for in vivo neutralization. Affinity columns containing recombinant toxin B repeat proteins were made as described below. A separate affinity column was prepared using each of the following recombinant toxin B repeat proteins: pPB1750-2360. pPB1850- 2360. pMB1750-1970 and pMB1970-23 6 0.
For each affinity column to be made. four ml of PBS-washed Actigel resin (Sterogene) was coupled overnight at room temperature with 5-10 mg of affinity purified recombinant protein (first extensively dialyzed into PBS) in 15 ml tubes (Falcon) containing a 1/10 final volume Aid-coupling solution (1 M sodium cyanoborohydride). Aliquots of the supernatants from the coupling reactions, before and after coupling, were assessed by Coomassie staining of 7.5% SDS-PAGE gels. Based on protein band intensities, in all cases greater than coupling efficiencies were estimated. The resins were poured into 10 ml columns (BioRad).
135 washed extensively with PBS. pre-eluted with 4 M guanidine-HCI (in 10 mM Tris-HCl, pH and reequilibrated in PBS. The columns were stored at 4*C.
Aliquots of a CTB IgY polyclonal antibody preparation (PEG prep) were affinity purified on each of the four columns as described below. The columns were hooked to a UV monitor (ISCO). washed with PBS and 40 ml aliquots of a 2X PEG prep (filter sterilized using a 0.45 p filter) were applied. The columns were washed with PBS until the baseline value was re-established. The columns were then washed with BBStween to elute nonspecifically binding antibodies. and reequilibrated with PBS. Bound antibody was eluted from the column in 4M guanidine-HCI (in 10mM Tris-HC1. pH8.0). The eluted antibody was immediately dialyzed against a 100-fold excess of PBS at 4 0 C for 2 hrs. The samples were then dialyzed extensively against at least 2 changes of PBS. and affinity purifiediantibody was i collected and stored at 4*C. The antibody preparations were quantified by UV absorbance.
The elution volumes were in the range of 4-8 ml. All affinity purified stocks contained similar total antibody concentrations. ranging from 0.25-0.35% of the total protein applied to the columns.
The ability of the affinity purified antibody preparations to neutralize toxin B in vivo was determined using the assay outlined in a) above. Affinity purified antibody was diluted 1:1 in PBS before testing. The results are shown in Table 26.
ooo 136- TABLE 26 Neutralization Of Toxin B By Affinity Purified Antibodies Number of Number of Treatment group' Number of Treatment group' Animals Aliveb Animals Deadb Preimmune' 0 CTB': 400 pg 5 0 CTB (AP on pPB1750-2360):- 875 pg 5 0 CTB (AP on pMB1750-1970); 2 875 pg 5 0 CTB (AP on pMB1970-2360):- 500 pg 5 0 10 C difficile toxin B (CTB) (Tech Lab: at 5 pg/ml. 25 pg total) at lethal concentration to hamsters is added to antibody and incubated for one hour at 37*C. After incubation this mixture is injected intraperitoneally (IP) into hamsters. Each treatment group received toxin premixed with antibody raised against the indicated protein, as either: 4X antibody PEG prep or 'affinity purified (AP) antibody (from CTB PEG prep. on indicated columns). The amount of specific antibody in each prep is indicated: the amount is directly determined for affinity purified preps and is estimated for the 4X CTB as described in Example h The numbers in each group represent numbers of hamsters dead or alive. 2 hr post IP administration of toxin/antibody mixture.
In all cases similar levels of toxin neutralization was observed, such that lethality was delayed in all groups relative to preimmune controls. This result demonstrates that antibodies reactive to the repeat region of the toxin B gene are sufficient to neutralize toxin B in vivo.
The hamsters will eventually die in all groups. but this death is maximally delayed with the CTB PEG prep antibodies. Thus neutralization with the affinity purified (AP) antibodies is not as complete as that observed with the CTB prep before affinity chromatography. This result may be due to loss of activity during guanidine denaturation (during the elution of the antibodies from the affinity column) or the presence of antibodies specific to other regions of the toxin B gene that can contribute to toxin neutralization (present in the CTB PEG prep).
137 The observation that antibodies affinity purified against the non-overlapping pMB 1750-1970 and pMB1970-2360 proteins neutralized toxin B raised the possibility that either 1) antibodies specific to repeat sub-regions are sufficient to neutralize toxin B or 2) sub-region specific proteins can bind most or all repeat specific antibodies present in the CTB polvclonal pool. This would likely be due to conformational similarities between repeats.
since homology in the primary amino acid sequences between different repeats is in the range of only 25-75% [Eichel-Streiber. et al. (1992) Molec. Gen. Genetics 233:260]. These possibilities were tested by affinity chromatography.
The CTB PEG prep was sequentially depleted 2X on the pMB1750-1970 column: only a small elution peak was observed after the second chromatography. indicating that most 'reactive antibodies were.removed. This interval depleted CTB preparation was then chromatographed on the pPB1850-2360 column: no antibody bound to the column. The reactivity of the CTB and CTB (pMB1750-1970 depleted) preps to pPB1750-2360. pPB1850- 2360. pMB1750-1970 and pMB1970-2360 proteins was then determined by ELISA using the 15 protocol described in Example 13(c). Briefly. 96-well microtiter plates (Falcon. Pro-Bind Assay Plates) were coated with recombinant protein by adding 100 ul volumes of protein at 1-2 tg/ml in PBS containing 0.005% thimerosal to each well and incubating overnight at 4 0
C.
The next morning. the coating suspensions were decanted and the wells were washed three times using PBS. In order to block non-specific binding sites. 100 ll of 1.0% BSA (Sigma) S" 20 in PBS (blocking solution) was then added to each well. and the plates were incubated for I hr. at 37*C. The blocking solution was decanted and duplicate samples of 150 gl of diluted antibody was added to the first well of a dilution series. The initial testing serum dilution was (1/200 for CTB prep. (the concentration of depleted CTB was standardized by OD,t) in blocking solution containing 0.5% Tween 20. followed by 5-fold serial dilutions into this solution. This was accomplished by serially transferring 30 pl aliquots to 120 pl buffer.
mixing, and repeating the dilution into a fresh well. After the final dilution. 30 pi was removed from the well such that all wells contained 120 ll final volume. A total of 5 such dilutions were performed (4 wells total). The plates were incubated for 1 hr at 37*C.
Following this incubation, the serially diluted samples were decanted and the wells were washed three times using PBS containing 0.5% Tween 20 (PBST), followed by two 5 min washes using BBS-Tween and a final three washes using PBST. To each well. 100 pi of 1/1000 diluted secondary antibody [rabbit anti-chicken IgG alkaline phosphatase (Sigma) diluted in blocking solution containing 0.5% Tween 20] was added, and the plate was 138 incubated 1 hr at 37 0 C. The conjugate solutions were decanted and the plates were washed 6 times in PBST. then once in 50 mM NaCO. 10 mM MgCl,. pH 9.5. The plates were developed by the addition of 100 pl of a solution containing I mg/mi para-nitro phenyl phosphate (Sigma) dissolved in 50 mM Na.CO3, 10 mM MgCl,. pH 9.5 to each well. The plates were then incubated at room temperature in the dark for 5-45 min. The absorbency of each well was measured at 410 nm using a Dynatech MR 700 plate reader.
As preaicted by the affinity chromatography results. depletion of the CTB prep on the pMBB1750-!970 column removed all detectable reactivity to the pMB1970-2360 protein. The reciprocal purification of a CTB prep that was depleted on the pMB 970-2360 column yielded no bound antibody when chromatographed on the pMB1750-1970 column. These S" results demonstrate that all repeat reactive antibodies in the CTB polyclonal pool 'etcognize a conserved structure that is present in non-overlapping repeats. Although it is possible that this conserved structure represents rare conserved linear epitopes. it appears more likely that the neutralizing antibodies recognize a specific protein conformation. This conclusion was also suggested by the results of Western blot hybridization analysis of CTB reactivity to these recombinant proteins.
Western blots of 7.5% SDS-PAGE gels. loaded and electrophoresed with defined quantities of each recombinant protein, were probed with the CTB polyclonal antibody .preparation. The blots were prepared and developed with alkaline phosphatase as described in 20 Example 3. The results are shown in Figure 24.
Figure 24 depicts a comparison of immunoreactivity of IgY antibody raised against either native or recombinant toxin B antigen. Equal amounts of pMB1750-1970 (lane 1).
pMB1970-2360 (lane pPB1850-2360 (lane 3) as well as a serial dilution of pPB1750-2360 (lanes 4-6 comprising IX. 1/10X and 1/100X amounts, respectively) proteins were loaded in duplicate and resolved on a 7.5% SDS-PAGE gel. The gel was blotted and each half was hybridized with PEG prep IgY antibodies from chickens immunized with either native CTB or pPB1750-2360 protein. Note that the full-length pMB1750-1970 protein was identified only by antibodies reactive to the recombinant protein (arrows).
Although the CTB prep reacts with the pPB1750-2360, pPB1850-2360. and pMB1970- 2360 proteins, no reactivity to the pMB1750-1970 protein was observed (Figure 24). Given that all repeat reactive antibodies can be bound by this protein during affinity chromatography. this result indicates that the protein cannot fold properly on Western blots.
Since this eliminates all antibody reactivity, it is unlikely that the repeat reactive antibodies.in 139 the CTB prep recognize linear epitopes. This may indicate that in order to induce protective antibodies, recombinant toxin B protein will need to be properly folded.
c) Generation And Evaluation Of Antibodies Reactive To Recombinant Toxin B Polypeptides i) Generation Of Antibodies Reactive To Recombinant Toxin B Proteins Antibodies against recombinant proteins were generated in egg laying Leghorn hens as described in Example 13. Antibodies were raised [using Freund's adjuvant (Gibco) unless otherwise indicated] against the following recombinant proteins: 1) a mixture of Interval 1+2 proteins (see Figure 18); 2) a mixture of interval 4 and 5 proteins (see Figure 18);3) pMB1970-2360 protein: 4) pPB1750-2360 protein: 5) pMB1750-2360: 6) pMB1750-2360 [Titermax adjuvant (Vaxcell)]; 7) pMB1750-2360 [Gerbu adjuvant (Biotech)]; 8) pMBpl750- 2360 protein; 9) pPB1850-2360: and 10) pMB1850-2360.
15 Chickens were boosted at least 3 times with recombinant protein until ELISA reactivity [using the protocol described in b) above with the exception that the plates were coated with pPB1750-2360 protein] of polyclonal PEG preps was at least equal to that of the CTB polyclonal antibody PEG prep. ELISA titers were determined for the PEG preps from all of the above immunogens and were found to be comparable ranging from 1:12500 to 20 1:62500. High titers were achieved in all cases except in 6) pMB1750-2360 in which strong titers were not observed using the Titermax adjuvant, and this preparation was not tested further.
ii) Evaluation Of Antibodies Reactive To Recombinant Proteins By Western Blot Hybridization Western blots of 7.5% SDS-PAGE gels. loaded and electrophoresed with defined quantities of recombinant protein (pMB1750-1970, pPB1850-2360. and pMB1970-2360 proteins and a serial dilution of the pPB1750-2360 to allow quantification of reactivity), were probed with the CTB, pPB1750-2360, pMB1750-2360 and pMB1970-2360 polyclonal antibody preparations (from chickens immunized using Freund's adjuvant). The blots were prepared and developed with alkaline phosphatase as described above in b).
140- As shown in Figure 24. the CTB and pMB1970-2360 preps reacted strongly with the pPB 1750-2360. pPB1850-2360. and pMB1970-2360 proteins while the pPB1750-2360 and pMB1970-2360 (Gerbu) preparations reacted strongly with all four proteins. The Western blot reactivity of the pPB1750-2360 and pMB1970-2360 (Gerbu) preparations were equivalent to that of the CTB preparation. while reactivity of the pMB1970-2360 preparation was that of the CTB prep. Despite equivalent ELISA reactivities only weak reactivity (approximately" to the recombinant proteins were observed in PEG preps from two independent groups immunized with the pMB1750-2360 protein and one group immunized with the pMB1750-2360 preparation using Freund's adjuvant.
Affinity purification was utilized to determine if this difference in immunoreactivity by Western blot analysis reflects differing antibody titers. Fifty ml 2X PEG preparations from chickens immunized with either pMB1750-2360 or pMB1970-2360 protein were chromatographed on the pPB1750-2360 affinity column from b) above, as described. The yield of affinity purified antibody total protein in preparation) was equivalent to the yield 15 obtained from a CTB PEG preparation in b) above. Thus. differences in Western reactivity.
reflect a qualitative difference in the antibody pools. rather than quantitative differences..
These results demonstrate that certain recombinant proteins are more effective at generating high affinity antibodies (as assayed by Western blot hybridization).
20 iii) In Vivo Neutralization Of Toxin B Using Antibodies Reactive To Recombinant Protein The in vivo hamster model [described in Examples 9 and 14(b)] was utilized to assess the neutralizing ability of antibodies raised against recombinant toxin B proteins. The results from three experiments are shown below in Tables 27-29.
The ability of each immunogen to neutralize toxin B in vivo has been compiled and is shown in Table 30. As predicted from the recombinant protein-CTB premix studies (Table 24) only antibodies to Interval 3 (1750-2366) and not the other regions of toxin B intervals 1-5) are protective. Unexpectedly, antibodies generated to INT-3 region expressed in pMAL vector (pMB 1750-2360 and pMB1750-2360) using Freund's adjuvant were nonneutralizing. This observation is reproducible. since no neutralization was observed in two independent immunizations with pMB 1750-2360 and one immunization with pMpB1750- 2360. The fact that 5X quantities of affinity purified toxin B repeat specific antibodies from pMB1750-2360 PEG preps cannot neutralize toxin B while IX quantities of affinity purified- 141 anti-CTB antibodies can (Table 28) demonstrates that the differential ability of CTB antibodies to neutralize toxin B is due to qualitative rather than quantitative differences in these antibody preparations. Only when this region was expressed in an alternative vector (pPB1750-2360) or using an alternative adjuvant with the pMB1750-2360 protein were neutralizing antibodies generated. Importantly, antibodies raised using Freund's adjuvant to pPB1850-2360. which contains a fragment that is only 100 amino acids smaller than recombinant pP1B750-2360. are unable to neutralize toxin B in vivo (Table 27): note also that the same vector is used for both pPB1850-2360 and pPB1750-2360.
TABLE 27 In Vivo Neutralization Of Toxin B o 15 15 Treatment Group' Number Animals Aliveb Number Animals Deadb Preimmune 0 CTB 5 0 INTI+2 0 INT 4+5 0 pMB1750-2360 0 pMB1970-2360 0 pPB1750-2360 5 0 C. difficile toxin B (CTB) (at 5 ig/ml: 25 jpg total: Tech Lab) at lethal concentration to hamsters is added to antibody and incubated for one hour at 37*C. After incubation this mixture is injected intraperitoneally (IP) into hamsters. Each treatment group received toxin premixed with antibody raised against the indicated protein, as a 4X antibody PEG prep.
b The numbers in each group represent numbers of hamsters dead or alive. 2 hours post IP administration of toxin/antibody mixture.
142 0 0* 4 TABLE 28 In Vivo Neutralization Of Toxin B Using Affinity Purified Antibodies Treatment Group' Number Animals Aliveb Number Animals Deadb Preimmune(l) 0 'CTB(l) 5 0 pPB1750-2360(1) 5 0 mg anti-pMB1750-2360(2) 1 4 1.5 mg anti-pMB1970-2360(2) 0 300 pg anti-CTB(2) 5 0 C. difficile toxin B (CTB) (at 5 pg/ml: 25 pg total:Tech Lab) at lethal concentration to hamsters is added to antibody and incubated for one hour at 37 0 C. After incubation. 1 ml of this mixture is injected intraperitoneally (IP) into hamsters. Each treatment group received toxin premixed with antibody 15 raised against the indicated protein, as either 4X antibody PEG prep or (2) affinity purified antibody (on a pPB1750-2360 resin), either 1.5 mg/group (anti-pMB1750-2360 and anti-pMB1970-2360: used undiluted affinity purified antibody) or 350 pg/group (anti-CTB. repeat specific; used 1/5 diluted anti- CTB antibody).
h The numbers in each group represent numbers of hamsters dead or alive. 2 hr post-IP administration of toxin/antibody mixture.
143 TABLE 29 Generation Of Neutralizing Antibodies Utilizing The Gerbu Adjuvant Treatment Group' Number Animals Aliveb Number Animals Deadb Preimmune 0 6TB 5 0 pMBi970-2360 0 pMB1850-2360 0 pPB1850-2360 0 pMB 1750-2360 (Gerbu adj) 5 0 C. difficile toxin B (CTB) (Tech Lab) at lethal concentration to hamsters is added to antibody and incubated for one hour at 37°C. After incubation this mixture is injected intraperitoneally (IP) into hamsters. Each treatment group received toxin premixed with antibody raised against the indicated protein, as a 4X antibody PEG prep.
b The numbers in each group represent numbers of hamsters dead or alive. 2 hrs post IP administration of toxin/antibody mixture.
-144- 10 TABLE In Vivo Neutralization Of Toxin B Immunogen Adjuvant Tested Antigen In viva Preparation' Utilized For AP Neutralization" Preimmune NA' PEG NA no CTB (nativ e) Titerniax PEG NA yes CTB (native) Titerinax AP pPB 1750-2360 yes CTB (native) Titermax AP pPB 1850-2360 yes GTB (native) Titermax AP pPB175O-1970 yes CTB (native) Titermax AP pPB 1970-2360 yes pMBI750-2360 Freunds PEG NA no pMB 1750-2360 Freunds AP pPBI750-2360 no pMBI750-2360 Gerbu PEG NA yes pMBI970-2360 Freunds; PEG NA no pMB197O-2360 Freunds AP pPB1750-2360 no pPBI1750-2360 Freunds PEG NA yes pPB1850-2360 Freunds PEG NA no pMBI85O-236O Freunds PEG NA no INT 1+2 Freunds PEG NA no INT 4+5 Freunds PEG NA no Either PEG preparation (PEG) or affinity purified antibodies (AP).
b 'Yes' denotes complete neutralization (0/5 dead) while 'no' denotes no neutralization (5/5 dead) of toxin B. 2 hours post-administration of mixture.
'NA' denotes not applicable.
S. S
*SSSS.
*S S 145 The pPB1750-2360 antibody pool confers significant in vivo protection. equivalent to that obtained with the affinity purified CTB antibodies. This correlates with the observed high affinity of this antibody pool (relative to the pMB1750-2360 or pMB1970-2360 pools) as assayed by Western blot analysis (Figure 24). These results provide the first demonstration that in vivo neutralizing antibodies can be induced using recombinant toxin B protein as immunogen.
The failure of high concentrations of antibodies raised against the pMB1750-2360 protein (using Freunds adjuvant) to neutralize, while the use of Gerbu adjuvant and pMB1750-23 6 0 protein generates a neutralizing response. demonstrates that conformation or presentation of this protein is essential for the induction of neutralizing antibodies. These results are consistent with the observation that the neutralizing antibodies produced when native CTB is used as an immunogen appear to recognize conformational epitopes [see section i b) above]. This is the first demonstration that the conformation or presentation of recombinant toxin B protein is essential to generate high titers of neutralizing antibodies.
EXAMPLE Determination Of Quantitative And Qualitative Differences Between pMB1750-2360.
:M:17S- 2 3 6 0 (Gerbu) Or pPB 750-2360 IgY Polvclonal Antibody Preparations In Example 19. it was demonstrated that toxin B neutralizing antibodies could be generated using specific recombinant toxin B proteins (pPB1750-2360) or specific adjuvants.
Antibodies raised against pMB1750-2360 were capable of neutralizing the enterotoxin effect of toxin B when the recombinant protein was used to immunize hens in conjunction with the "Gerbu adjuvant, but not when Freunds adjuvant was used. To determine the basis for these 25 antigen and adjuvant restrictions, toxin B-specific antibodies present in the neutralizing and non-neutralizing PEG preparations were isolated by affinity chromatography and tested for qualitative or quantitative differences. The example involved a) purification of anti-toxin
B
specific antibodies from pMB1750-2360 and pPB1750-2360 PEG preparations and b) in vivo neutralization of toxin B using the affinity purified antibody.
a) Purification Of specific Antibodies From pMB1750-2360 And pPB1750- 23 6 0 PEG Preparations To purify and determine the concentration of specific antibodies (expressed as the percent of total antibody) within the pPB1750-2360 (Freunds and Gerbu) and pPB1750-2360 146- PEG preparations. defined quantities of these antibody preparations were chromatographed on an affinity column containing the entire toxin B repeat region (pPB1750-2360). The amount of affinity purified antibody was then quantified.
An affinity column containing the recombinant toxin B repeat protein, pPB1750-2360, was made as follows. Four ml of PBS-washed Actigel resin (Sterogene) was coupled with mg of pPB1750-2360 affinity purified protein (dialyzed into PBS: estimated to be greater than full length fusion protein) in a 15 ml tube (Falcon) containing 1/10 final volume Aidcoupling solution (I M sodium cyanoborohydride). Aliquots of the supernatant from the coupling reactions. before and after coupling, were assessed by Coomassie staining of SDS-PAGE gels. Based on protein band intensities, greater than 95% (approximately 5 mg) of recombinant protein was coupled to the resin. The coupled resin was poured into a 10 ml column (BioRad). washed extensively with PBS. pre-eluted with 4M guanidine-HCI (in mM Tris-HCL. pH 8.0: 0.005% thimerosal) and re-equilibrated in PBS and stored at 4 0
C.
Aliquots of pMB1750-2360. pMB1750-2360 (Gerbu) or pPB1750-2360 IgY polyclonal 15 antibody preparations (PEG preps) were affinity purified on the above column as follows.
The column was attached to an UV monitor (ISCO). and washed with PBS. Forty ml aliquots of 2X PEG preps (filter sterilized using a 0.45 p filter and quantified by OD,o before chromatography) was applied. The column was washed with PBS until the baseline was reestablished (the column flow-through was saved), washed with BBSTween to elute 20 nonspecifically binding antibodies and re-equilibrated with PBS. Bound antibody was eluted from the column in 4M guanidine-HCI (in 10 mM Tris-HCL. pH 8.0. 0.005% thimerosal) and the entire elution peak collected in a 15 ml tube (Falcon). The column was re-equilibrated.
and the column eluate re-chromatographed as described above. The antibody preparations were quantified by UV absorbance (the elution buffer was used to zero the spectrophotometer). Approximately 10 fold higher concentrations of total purified antibody was obtained upon elution of the first chromatography pass relative to the second pass. The low yield from the second chromatography pass indicated that most of the specific antibodies were removed by the first round of chromatography.
Pools of affinity purified specific antibodies were prepared by dialysis of the column elutes after the first column chromatography pass for the pMB1750-2360. pMB1750-2360 (Gerbu) or pPB1750-2360 IgY polyclonal antibody preparations. The elutes were collected on ice and immediately dialyzed against a 100-fold volume of PBS at 4°C for 2 hrs. The samples were then dialyzed against 3 changes of a 65-fold volume of PBS at 4 0 C. Dialysis- 147was performed for a minimum of 8 hrs per change of PBS. The dialyzed samples were collected, centrifuged to remove insoluble debris, quantified by OD,, and stored at 4 0
C.
The percentage of toxin B repeat-specific antibodies present in each preparation was determined using the quantifications of antibody yields from the first column pass (amount of specific antibody recovered after first pass/total protein loaded). The yield of repeat-specific affinity purified antibody (expressed as the percent of total protein in the preparation) in: 1) the pMB1750-2360 PEG prep was approximately 2) the pMB1750-2360 (Gerbu) prep was approximately and 3) the pPB1750-2360 prep was approximately 0.4%.
Purification of a CTB IgY polyclonal antibody preparation on the same column demonstrated that the concentration of toxin B repeat specific antibodies in the CTB preparation was 0.35%.
These results demonstrate that 1) the use of Gerbu adjuvant enhanced the titer of specific antibody produced against the pMB1750-2360 protein 5-fold relative to immunization using Freunds adjuvant. and 2) the differences seen in the in vivo neutralization ability of the pMB1750-2360 (not neutralizing) and pPB1750-2360 (neutralizing) and CTB (neutralizing) 15 PEG preps seen in Example 19 was not due to differences in the titers of repeat-specific antibodies in the three preparations because the titer of repeat-specific antibody was similar for all three preps: therefore the differing ability of the three antibody preparations to neutralize toxin B must reflect qualitative differences in the induced toxin B repeat-specific antibodies. To confirm that qualitative differences exist between antibodies raised in hens 20 immunized with different recombinant proteins and/or different adjuvants. the same amount of affinity purified anti-toxin B repeat (aa 1870-2360 of toxin B) antibodies from the different preparations was administered to hamsters using the in vivo hamster model as described below.
b) In vivo Neutralization Of Toxin B Using Affinity Purified Antibody The in vivo hamster model was utilized to assess the neutralizing ability of the affinity purified antibodies raised against recombinant toxin B proteins purified in above. As well, a 4X IgY PEG preparation from a second independent immunization utilizing the pPB1750- 2360 antigen with Freunds adjuvant was tested for in vivo neutralization. The results are shown in Table 31.
148 TABLE 31 In vivo Neutralization Of Toxin B Using Affinity Purified Antibodies Treatment Group' Number Animals Aliveb Number Animals Dead b Preimmune' 0 CTB (300 pg) 2 5 0 CTB (100 pg) I 4 pMB1750-2360 (5 mg) 2 5 0 pMB1750-2360 (1.5 mg) 5 0 pMB1750-2360 (300 pg) 2 5 0 pMB1750-2360 (1.5 mg) 0 pPBl750-2360 (1.5 mg) 5 0 pPBI750-2360 (300 jg)- 1 4 CTB (100 g) 3 2 3 pPB1750-2360 (500 pg)' 5 0 C difficile toxin B (CTB) (Tech Lab) at lethal concentration to hamsters pg) was added to the antibody (amount of specific antibody is indicated) and incubated for one hour at 37 0 C. After incubation, this mixture was injected IP into hamsters (1/5 total mix injected per hamster). Each treatment group received toxin premixed with antibody raised against the indicated protein (G=gerbu adjuvant. F=Freunds adjuvant). 'indicates the antibody was a 4X IgY PEG prep; 2 indicates the antibody was affinity purified on a pPB1850- 2360 resin; and 3 indicates that the antibody was a IX IgY PEG prep.
The numbers in each group represent numbers of hamsters dead or alive. 2 hrs post IP administration of toxin/antibody mixture.
149- The results shown in Table 31 demonstrate that: 1) as shown in Example 19 and reproduced here. 1.5 mg of affinity purified antibody from pMB 1750-2360 immunized hens using Freunds adjuvant does not neutralize toxin B in vivo. However. 300 pg of affinity purified antibody from similarly immunized hens utilizing Gerbu adjuvant demonstrated complete neutralization of toxin B in vivo. This demonstrates that Gerbu adjuvant, in addition to enhancing the titer of antibodies reactive to the pMBI750-2360 antigen relative to Freunds adjuvant (demonstrated in above), also enhances the yield of neutralizing antibodies to this antigen, greater than 5 fold.
2) Complete in vivo neutralization of toxin .B was observed with 1.5 mg of affinity purified antibody from hens immunized with pPB1750-2360 antigen, but not with pMB1750-2360 antigen, when Freunds adjuvant was used. This demonstrates.
using standardized toxin B repeat-specific antibody concentrations, that neutralizing antibodies were induced when pPB1750-2360 but not pMB1750-2360 was used as the antigen with Freunds adjuvant.
3) Complete in vivo neutralization was observed with 300 pg of pMB1750-2360 (Gerbu) antibody, but not with 300 jlg of pPB1750-2360 (Freunds) antibody. Thus the pMB1750-2360 (Gerbu) antibody has a higher titer of neutralizing antibodies than the pPB1750-2360 (Freunds) antibody.
20 4) Complete neutralization of toxin B was observed using 300 pg of CTB antibody [affinity purified but not 100 pg CTB antibody (AP or PEG prep).
This demonstrates that greater than 100 pg of toxin B repeat-specific antibody (anti- CTB) is necessary to neutralize 25 pg toxin B in vivo in this assay, and that affinity purified antibodies specific to the toxin B repeat interval neutralize toxin B as effectively as the PEP prep of IgY raised against the entire CTB protein (shown in this assay).
As was observed with the initial pPB1750-2360 (IgY) PEG preparation (Example 19). complete neutralization was observed with a IgY PEG preparation isolated from a second independent group of pPB1750-2360 (Freunds) immunized hens. This demonstrates that neutralizing antibodies are reproducibly produced when hens are immunized with pPB1750-2360 protein utilizing Freund's adjuvant.
150 EXAMPLE 21 Diagnostic Enzyme Immunoassavs For C. difficile Toxins A and B The ability of the recombinant toxin proteins and antibodies raised against these recombinant proteins (described in the above examples) to form the basis of diagnostic assays for the detection of clostridial toxin in a sample was examined. Two immunoassay formats were tested to quantitatively detect C. difficile toxin A and toxin B from a biological specimen. The first format involved a competitive assay in which a fixed amount of recombinant toxin A or B was immobilized on a solid support microtiter plate wells) followed by the addition of a toxin-containing biological specimen mixed with affinitypurified or PEG fractionated antibodies against recombinant toxin A or B. If toxin is present in a specimen, this toxin will compete with the immobilized recombinant toxin protein for binding to the anti-recombinant antibody thereby reducing the signal obtained following the addition of a reporter reagent. The reporter reagent detects the presence of antibody bound to 15 the immobilized toxin protein.
In the second format. a sandwich immunoassay was developed using affinity-purified antibodies to recombinant toxin A and B. The affinity-purified antibodies to recombinant toxin A and B were used to coat microtiter wells instead of the recombinant polypeptides (as was done in the competitive assay format). Biological samples containing toxin A or B were 20 then added to the wells followed by the addition of a reporter reagent to detect the presence of bound toxin in the well.
a) Competitive Immunoassay For The Detection Of C. difficile Toxin Recombinant toxin A or B was attached to a solid support by coating 96 well microtiter plates with the toxin protein at a concentration of lgg/ml in PBS. The plates were incubated overnight at 2-8*C. The following morning. the coating solutions were removed and the remaining protein binding sites on the wells were blocked by filling each well with a PBS solution containing 0.5% BSA and 0.05% Tween-20. Native C. difficile toxin A or B (Tech Lab) was diluted to 4 ig/ml in stool extracts from healthy Syrian hamsters (Sasco).
The stool extracts were made by placing fecal pellets in a 15 ml centrifuge tube; PBS was added at 2 mlpellet and the tube was vortexed to create a uniform suspension. The tube was then centrifuged at 2000 rpm for 5 min at room temperature. The supernatant was removed; 151 this comprises the stool extract. Fifty pl of the hamster stool extract was pipetted into each well of the microtiter plates to serve as the diluent for serial dilutions of the 4 pg/ml toxin samples. One hundred pi of the toxin samples at 4 pg/ml was pipetted into the first row of wells in the microtiter plate. and 50 pi aliquots were removed and diluted serially down the plate in duplicate. An equal volume of affinity purified anti-recombinant toxin antibodies [1 ng/well of anti-pMA1870-2680 antibody was used for the detection of toxin A: 0.5 ng/well of anti-pMB1750-2360(Gerbu) was used for the detection of toxin B] were added to appropriate wells, and the plates were incubated at room temperature for 2 hours with gentle agitation.
Wells serving as negative control contained antibody but no native toxin to compete for binding.
Unbound toxin and antibody were removed by washing the plates 3 to 5 times with PBS containing 0.05% Tween-20. Following the wash step. 100 pl of rabbit anti-chicken IgG antibody conjugated to alkaline phosphatase (Sigma) was added to each well and the plates ro :were incubated for 2 hours at room temperature. The plates were then washed as before to 15 remove unbound secondary antibody. Freshly prepared alkaline phosphatase substrate (1 mg/ml p-nitrophenyl phosphate (Sigma) in 50 mM Na,CO3, pH 9.5: 10 mM MgCI,) was added to each well. Once sufficient color developed, the plates were read on a Dynatech MR700 microtiter plate reader using a 410 nm filter.
The results are summarized in Tables 32 and 33. For the results shown in Table 32, I 20 the wells were coated with recombinant toxin A protein (pMA1870-2680). The amount of native toxin A added (present as an addition to solubilized hamster stool) to a given well is indicated (0 to 200 ng). Antibody raised against the recombinant toxin A protein. pMA1870- 2680. was affinity purified on the an affinity column containing pPA1870-2680 (described in Example 20). As shown in Table 32. the recombinant toxin A protein and affinity-purified antitoxin can be used for the basis of a competitive immunoassay for the detection of toxin A in biological samples.
Similar results were obtained using the recombinant toxin B. pPB1750-2360. and antibodies raised against pMB1750-2360(Gerbu). For the results shown in Table 33, the wells were coated with recombinant toxin B protein (pPB1750-2360). The amount of native toxin B added (present as an addition to solubilized hamster stool) to a given well is indicated (0 to 200 ng). Antibody raised against the recombinant toxin B protein. pMB1750- 2360(Gerbu). was affinity purified on the an affinity column containing pPBl850-2360 (described in Example 20). As shown in Table 33. the recombinant toxin B protein and 152a affinity-purified antitoxin can be used for the basis of a competitive immunoassay for the detection of toxin B in biological samples.
In this competition assay, the reduction is considered significant over the background levels at all points: therefore the assay can be used to detect samples containing less than 12.5 ng toxin A/well and as little as 50-100 ng toxin B/well.
TABLE 32 Competitive Inhibition Of Anti-C. difficile Toxin A By Native Toxin A ng Toxin A/Well Readout 200 0.176 100 0.253 0.240 0.259 12.5 0.309 6.25 0.367 3.125 0.417 0 0.590 TABLE 33 Competitive Inhibition Of Anti-C. difficile Toxin B By Native Toxin B ng Toxin B/Well OD 4 ,o Readout 200 0.392 100 0.566 0.607 0.778 12.5 0.970 6.25 0.902 3.125 1.040 0 1.055 153 These competitive inhibition assays demonstrate that native C. difficile toxins and recombinant C. difficile toxin proteins can compete for binding to antibodies raised against recombinant C. difficile toxins demonstrating that these anti-recombinant toxin antibodies provide effective diagnostic reagents.
b) Sandwich Immunoassay For The Detection Of C. difficile Toxin Affinity-purified antibodies against recombinant toxin A or toxin B were immobilized to 96 well microtiter plates as follows. The wells were passively coated overnight at 4°C with affinity purified antibodies raised against either pMA1870-2680 (toxin A) or pMB1750- 2360(Gerbu) (toxin The antibodies were affinity purified as described in Example The antibodies were used at a concentration of 1 pg/ml and 100 pi was added to each microtiter well. The wells were then blocked with 200 pl of 0.5% BSA in PBS for 2 hours at room temperature and the blocking solution was then decanted. Stool samples from healthy Syrian hamsters were resuspended in PBS. pH 7.4 (2 ml PBS/stool pellet was used to resuspend the pellets and the sample was centrifuged as described above). The stool suspension was then spiked with native C difficile toxin A or B (Tech Lab) at 4 g/ml. The stool suspensions containing toxin (either toxin A or toxin B) were then serially diluted twofold in stool suspension without toxin and 50 pl was added in duplicate to the coated 20 microtiter wells. Wells containing stool suspension without toxin served as the negative control.
The plates were incubated for 2 hours at room temperature and then were washed three times with PBS. One hundred pi of either goat anti-native toxin A or goat anti-native toxin B (Tech Lab) diluted 1:1000 in PBS containing 1% BSA and 0.05% Tween 20 was added to each well The plates were incubated for another 2 hours at room temperature.
The plates were then washed as before and 100 pl of alkaline phosphatase-conjugated rabbit anti-goat IgG (Cappel. Durham. was added at a dilution of 1:1000. The plates were incubated for another 2 hours at room temperature. The plates were washed as before then developed by the addition of 100 pl/well of a substrate solution containing 1 mg/ml pnitrophenyl phosphate (Sigma) in 50 mM Na,CO,. pH 9.5: 10 mM MgCI,. The absorbance of each well was measured using a plate reader (Dynatech) at 410 nm. The assay results are shown in Tables 34 and 154- TABLE 34 S.
S
*5
C
S.
C difficile Toxin A Detection In Stool Using Affinity-Purified Antibodies Against Toxin A ng Toxin A/Well
OD,
4 1 Readout 200 0.9 100 0.8 0.73 0.71 12.5 0.59 6.25 0.421 0 0 TABLE C. difficile Toxin B Detection In Stool Using Affinity-Purified Antibodies Against Toxin B ng Toxin B/Well
OD,
1 0 Readout 200 1.2 100 0.973 50 0.887 25 0.846 12.5 0.651 6.25 0.431 0 0.004 The results shown in Tables 34 and 35 show that antibodies raised against recombinent toxin A and toxin B fragments can be used to detect the presence of C difficile toxin in stool samples. These antibodies form the basis for a sensitive sandwich immunoassay which is capable of detecting as little as 6.25 ng of either toxin A or B in a 50 Vl stool sample. As shown above in Tables 34 and 35. the background for this sandwich immunoassay is extremely low: therefore, the sensitivity of this assay is much lower than 6.25 ng toxin/well.
It is likely that toxin levels of 0.5 to 1.0 pg/well could be detected by this assay.
155- The results shown above in Tables 32-35 demonstrate clear utility of the recombinant reagents in C difficile toxin detection systems.
EXAMPLE 22 Construction And Expression Of C. bolulinum C Fragment Fusion Proteins The C botulinum type A neurotoxin gene has been cloned and sequenced [Thompson, et al.. Eur. J. Biochem. 189:73 (1990)]. The nucleotide sequence of the toxin gene is available from the EMBL/GenBank sequence data banks under the accession number X52066; the nucleotide sequence of the coding region is listed in SEQ ID NO:27. The amino acid sequence of the C botulinum type A neurotoxin is listed in SEQ ID NO:28. The type A S neurotoxin gene is synthesized as a single polypeptide chain which is processed to form a dimer composed of a light and a heavy chain linked via disulfide bonds. The 50 kD carboxyterminal portion of the heavy chain is referred to as the C fragment or the H. domain.
S 15 Previous attempts by others to express polypeptides comprising the C fragment of C botulinum type A toxin as a native polypeptide not as a fusion protein) in E. coli have been unsuccessful LaPenotiere. et al. in Botulinum and Tetanus Neurotoxins, DasGupta, Ed.. Plenum Press. New York (1993), pp. 463-466]. Expression of the C fragment as a fusion with the E. coli MBP was reported to result in the production of insoluble protein S 20 LaPenotiere. et al.. supra).
In order to produce soluble recombinant C fragment proteins in E. coli. fusion proteins comprising a synthetic C fragment gene derived from the C. botulinum type A toxin and either a portion of the C. difficile toxin protein or the MBP were constructed. This example involved a) the construction of plasmids encoding C fragment fusion proteins and b) expression of C. botulinum C fragment fusion proteins in E. coli.
a) Construction Of Plasmids Encoding C Fragment Fusion Proteins In Example 11. it was demonstrated that the C. difficile toxin A repeat domain can be efficiently expressed and purified in E. coli as either native (expressed in the pET 23a vector in clone pPA1870-2680) or fusion (expressed in the pMALc vector as a fusion with the E.
coli MBP in clone pMA 870-2680) proteins. Fusion proteins comprising a fusion between the MBP, portions of the C. difficile toxin A repeat domain (shown to be expressed as a 156soluble fusion protein) and the C fragment of the C. botulinum type A toxin were constructed.
A fusion protein comprising the C fragment of the C. botulinum type A toxin and the MBP was also constructed.
Figure 25 provides a schematic representation of the botulinal fusion proteins along with the donor constructs containing the C difficile toxin A sequences or C. botulinum C fragment sequences which were used to generate the botulinal fusion proteins. In Figure the solid boxes represent C. difficile toxin A gene sequences, the open boxes represent C.
botulinum C fragment sequences and the solid black ovals represent the E. coli MBP. When the name for a restriction enzyme appears inside parenthesis, this indicates that the restriction site was destroyed during construction. An asterisk appearing with the name for a restriction enzyme indicates that this restriction site was recreated at the cloning junction.
In Figure 25. a restriction map of the pMAI 870-2680 and pPAI 100-2680 constructs (described in Example 11) which contain sequences derived from the C. dificile toxin A
C
repeat domain are shown: these constructs were used as the source of C difficile toxin A gene 15 sequences for the construction of plasmids encoding fusions between the C botulinum C fragment gene and the C. dfficile toxin A gene. The pMAl870-2680 expression construct expresses high levels of soluble, intact fusion protein (20 mg/liter culture) which can be affinity purified on an amylose column (purification described in Example I Id).
The pAlterBot construct (Figure 25) was used as the source of C. botulinum C 20 fragment gene sequences for the botulinal fusion proteins. pAlterBot was obtained from J.
i Middlebrook and R. Lemley at the U.S. Department of Defense. pAlterBot contains a synthetic C botulinum C fragment inserted in to the pALTER-I® vector (Promega). This synthetic C fragment gene encodes the same amino acids as does the naturally occurring C fragment gene. The naturally occurring C fragment sequences. like most clostridial genes. are extremely A/T rich (Thompson et al.. supra). This high A/T content creates expression difficulties in E. coli and yeast due to altered codon usage frequency and fortuitous polyadenylation sites. respectively. In order to improve the expression of C fragment proteins in E. coli. a synthetic version of the gene was created in which the non-preferred codons were replaced with preferred codons.
The nucleotide sequence of the C. botulinum C fragment gene sequences contained within pAlterBot is listed in SEQ ID NO:22. The first six nucleotides (ATGGCT) encode a methionine and alanine residue. respectively. These two amino acids result from the insertion of the C botulinum C fragment sequences into the pALTER® vector and provide the initiator 157 methionine residue. The amino acid sequence of the C hotulinum C fragment encoded by the sequences contained within pAlterBot is listed in SEQ ID NO:23. The first two amino acids (Met Ala) are encoded by vector-derived sequences. From the third amino acid residue onward (Arg), the amino acid sequence is identical to that found in the C botulinum type A toxin gene.
The pMA1870-2680. pPA 1100-2680 and pAlterBot constructs were used as progenitor plasmids to make expression constructs in which fragments of the C. difficile toxin A repeat domain were expressed as genetic fusions with the C botulinum C fragment gene using the pMAL-c expression vector (New England BioLabs). The pMAL-c expression vector generates fusion proteins which contain the MBP at the amino-terminal end of the protein. A construct. pMBot. in which the C. botulinum C fragment gene was expressed as a fusion with only the MBP was constructed (Figure 25). Fusion protein expression was induced from E.
coli strains harboring the above plasmids. and induced protein was affinity purified on an **00 amylose resin column.
i) Construction Of pBlueBot In order to facilitate the cloning of the C botulinum C fragment gene sequences into a number of desired constructs. the botulinal gene sequences were removed from pAlterBot and were inserted into the pBluescript plasmid (Stratagene) to generate pBlueBot (Figure 20 pBlueBot was constructed as follows. Bacteria containing the pAlterBot plasmid were grown I: in medium containing tetracycline and plasmid DNA was isolated using the QIAprep-spin Plasmid Kit (Qiagen). One microgram of pAlterBot DNA was digested with Ncol and the resulting 3' recessed sticky end was made blunt using the Klenow fragment of DNA polymerase I (here after the Klenow fragment). The pAlterBot DNA was then digested with HindIII to release the botulinal gene sequences (the Bot insert) as a blunt (filled NcoI site)- HindlII fragment. pBluescript vector DNA was prepared by digesting 200 ng of pBluescript DNA with Smal and HindII. The digestion products from both plasmids were resolved on an agarose gel. The appropriate fragments were removed from the gel, mixed and purified utilizing the Prep-a-Gene kit (BioRad). The eluted DNA was then ligated using T4 DNA ligase and used to transform competent DH5a cells (Gibco-BRL). Host cells were made competent for transformation using the calcium chloride protocol of Sambrook et al., supra at 1.82-1.83. Recombinant clones were isolated and confirmed by restriction digestion using standard recombinant molecular biology techniques (Sambrook et al. supra). The resultant 158 clone. pBlueBot. contains several useful unique restriction sites flanking the Bot insert the C botulinum C fragment sequences derived from pAlterBot) as shown in Figure ii) Construction Of C. difficile C. botulinum MBP Fusion Proteins Constructs encoding fusions between the C. difficile toxin A gene and the C. botulinum C fragment gene and the MBP were made utilizing the same recombinant DNA methodology outlined above: these fusion proteins contained varying amounts of the C difficile toxin A repeat domain.
The pMABot clone contains a 2.4 kb insert derived from the C. difficile toxin A gene fused to the Bot insert the C botulinum C fragment sequences derived from pAlterBot).
pMABot (Figure 25) was constructed by mixing gel-purified DNA from Notl/Hindll digested pBlueBot (the 1.2 kb Bot fragment). SpeI/Notl digested pPA1100-2680 (the 2.4 kb C. diffcile toxin A repeat fragment) and XbalIHindll digested pMAL-c vector. Recombinant clones 15 were isolated. confirmed by restriction digestion and purified using the QIAprep-spin Plasmid Kit (Qiagen). This clone expresses the toxin A repeats and the botulinal C fragment protein sequences as an in-frame fusion with the MBP.
The pMCABot construct contains a 1.0 kb insert derived from the C. difficile toxin A gene fused to the Bot insert the C botulinum C fragment sequences derived from 20 pAlterBot). pMCABot was constructed by digesting the pMABot clone with EcoRI to oo• remove the 5' end of the C difficile toxin A repeat (see Figure 25. the pMAL-c vector contains a EcoRI site 5' to the C difficile insert in the pMABot clone). The restriction sites S"were filled and religated together after gel purification. The resultant clone (pMCABot.
Figure 25) generated an in-frame fusion between the MBP and the remaining 3' portion of the C. difficile toxin A repeat domain fused to the Bot gene.
The pMNABot clone contains the 1 kb SpelicoRl (filled) fragment from the C.
difficile toxin A repeat domain (derived from clone pPA1100-2680) and the 1.2 kb C.
botulinum C fragment gene as a NcoI (filled)/HindllI fragment (derived from pAlterBot).
These two fragments were inserted into the pMAL-c vector digested with Xbal/HindIIl. The two insert fragments were generated by digestion of the appropriate plasmid with EcoRI (pPA1100-2680) or NcoI (pAlterBot) followed by treatment with the Klenow fragment. After treatment with the Klenow fragment. the plasmids were digested with the second enzyme (either Spel or HindIII). All three fragments were gel purified. mixed and Prep-a-Gene 159 purified prior to ligation. Following ligation and transformation. putative recombinants were analyzed by restriction analysis: the EcoRI site was found to be regenerated at the fusion junction, as was predicted for a fusion between the filled EcoRI and Ncol sites.
A construct encoding a fusion protein between the botulinal C fragment gene and the MBP gene was constructed this fusion lacks any C difficile toxin A gene sequences) and termed pMBot. The pMBot construct was made by removal of the C difficile toxin A sequences from the pMABot construct and fusing the C fragment gene sequences to the MBP.
This was accomplished by digestion of pMABot DNA with Stul (located in the pMALc polylinker 5' to the Xbal site) and Xbal (located 3' to the Notl site at the toxA-Bot fusion junction). filling in the Xbal site using the Klenow fragment, gel purifying the desired restriction fragment, and ligating the blunt ends to circularize the plasmid. Following ligation and transformation. putative recombinants were analyzed by restriction mapping of the Bot insert the C botulinum C fragment sequences).
15 b) Expression Of C botulinum C Fragment Fusion Proteins In E. coli Large scale (1 liter) cultures of the pMAL-c vector, and each recombinant construct described above in were grown. induced, and soluble protein fractions were isolated as described in Example 18. The soluble protein extracts were chromatographed on amylose 20 affinity columns to isolate recombinant fusion protein. The purified recombinant fusion proteins were analyzed by running samples on SDS-PAGE gels followed by Coomassie staining and by Western blot analysis as described [Williams el al. (1994) supra]. In brief.
extracts were prepared and chromatographed in column buffer (10 mM NaPO, 0.5 M NaCI.
10 mM p-mercaptoethanol. pH 7.2) over an amylose resin (New England Biolabs) column.
and eluted with column buffer containing 10 mM maltose as described [Williams. et al.
(1994), supra]. An SDS-PAGE gel containing the purified protein samples tained with Coomassie blue is shown in Figure 26.
In Figure 26. the following samples were loaded. Lanes 1-6 contain protein purified from E. coli containing the pMAL-c. pPA1870-2680, pMABot. pMNABot, pMCABot and pMBot plasmids. respectively. Lane 7 contains broad range molecular weight protein markers (BioRad).
The protein samples were prepared for electrophoresis by mixing 5 pl of eluted protein with 5 pl of 2X SDS-PAGE sample buffer (0.125 mM Tris-HC1. pH 6.8. 2 mM EDTA, 6% 160- SDS. 20% glycerol. 0.025% bromophenol blue; P-mercaptoethanol is added to 5% before use). The samples were heated to 95 0 C for 5 min. then cooled and loaded on a 7.5% agarose SDS-PAGE gel. Broad range molecular weight protein markers were also loaded to allow estimation of the MW of identified fusion proteins. After electrophoresis. protein was detected generally by staining the gel with Coomassie blue.
In all cases the yields were in excess of 20 mg fusion protein per liter culture (see Table 36) and. with the exception of the pMCABot protein, a high percentage greater than 20-50% of total eluted protein) of the eluted fusion protein was of a MW predicted for the full length fusion protein (Figure 26). It was estimated (by visual inspection) that less than 10% of the pMCABot fusion protein was expressed as the full length fusion protein.
TABLE 36 Yield Of Affinity Purified C. botulinum C Fragment MBP Fusion Proteins 1 Yield Percentage Of Total Construct(mg/liter of Culture) Soluble Protein pMABot 24 pMCABot 34 pMNABot 40 pMBot 22 pMA1870-2680 40 4.8 These results demonstrate that high level expression of intact C botulinum C fragment/C. difficile toxin A fusion proteins in E. coli is feasible using the pMAL-c expression system. These results are in contrast to those reported by H. F. LaPenotiere. et al.
(1993), supra. In addition, these results show that it is not necessary to fuse the botulinal C fragment gene to the C difficile toxin A gene in order to produce a soluble fusion protein using the pMAL-c system in E. coli.
In order to determine whether the above-described botulinal fusion proteins were recognized by anti-C botulinum toxin A antibodies. Western blots were performed. Samples containing affinity-purified proteins from E. coli containing the pMABot. pMCABot.
pMNABot, pMBot. pMA1870-2680 or pMALc plasmids were analyzed. SDS-PAGE gels acrylamide) were loaded with protein samples purified from each expression construct.
161 After electrophoresis. the gels were blotted and protein transfer was confirmed by Ponceau S staining (as described in Example 12b).
Following protein transfer, the blots were blocked by incubation for 1 hr at 20 0 C in blocking buffer [PBST (PBS containing 0.1% Tween 20 and 5% dry milk)]. The blots were then incubated in 10 ml of a solution containing the primary antibody; this solution comprised a 1/500 dilution of an anti-C botulinum toxin A IgY PEG prep (described in Example 3) in blocking buffer. The blots were incubated for 1 hr at room temperature in the presence of the primary antibody. The blots were washed and developed using a rabbit anti-chicken alkaline phosphatase conjugate (Boehringer Mannheim) as the secondary antibody asfollows. The rabbit anti-chicken antibody was diluted to I pg/ml in blocking buffer (10 ml final volume per blot) and the blots were incubated at room temperature for 1 hour in the presence of the S. secondary antibody. The blots were then washed successively with PBST, BBS-Tween and o. 50 mM Na,CO3, pH 9.5. The blots were then developed in freshly-prepared alkaline phosphatase substrate buffer (100 ig/ml nitro blue tetrazolium. 50 pg/ml 15 indolylphosphate. 5 mM MgCI, in 50 mM Na,CO,. pH Development was stopped by flooding the blots with distilled water and the blots were air dried.
This Western blot analysis detected anti-C botulinum toxin reactive proteins in the pMABot, pMCABot, pMNABot and pMBot protein samples (corresponding to the predicted full length proteins identified above by Coomassie staining in Figure 26). but not in the 20 pMA 1100-2680 or pMALc protein samples.
These:results demonstrate that the relevant fusion proteins purified on an amylose resin as described above in section a) contained immunoreactive C botulinum C fragment protein as predicted.
EXAMPLE 23 Generation Of Neutralizing Antibodies By Nasal Administration Of pMBot Protein The ability of the recombinant botulinal toxin proteins produced in Example 22 to stimulate a systemic immune response against botulinal toxin epitopes was assessed. This example involved: a) the evaluation of the induction of serum IgG titers produced by nasal or oral administration of botulinal toxin-containing C difficile toxin A fusion proteins and b) the in vivo neutralization of C. botulinum type A neurotoxin by anti- recombinant C.
botulinum C fragment antibodies.
162 a) Evaluation Of The Induction Of Serum IgG Titers Produced By Nasal Or Oral Administration Of Botulinal Toxin- Containing C. difficile Toxin A Fusion Proteins Six groups containing five 6 week old CF female rats (Charles River) per group were immunized nasally or orally with one of the following three combinations using protein prepared in Example 22: 250 pg pMBot protein per rat (nasal and oral); 2) 250 ipg pMABot protein per rat (nasal and oral); 3) 125 pg pMBot admixed with 125 pg pMA1870- 2680 per rat (nasal and oral). A second set of 5 groups containing 3 CF female rats/group were immunized nasally or orally with one of the following combinations 250 pg pMNABot protein per rat (nasal and oral) or 5) 250 pg pMAL-c protein per rat (nasal and oral).
The fusion proteins were prepared for immunization as follows. The proteins (in column buffer containing 10 mM maltose) were diluted in 0.1 M carbonate buffer. pH 9.5 and administered orally or nasally in a 200 pl volume. The rats were lightly sedated with ether prior to administration. The oral dosing was accomplished using a 20 gauge feeding needle.
The nasal dosing was performed using a P-200 micro-pipettor (Gilson). The rats were boosted 14 days after the primary immunization using the techniques described above and were bled 7 days later. Rats from each group were lightly etherized and bled from the tail.
The blood was allowed to clot at 37 0 C for 1 hr and the serum was collected.
The serum from individual rats was analyzed using an ELISA to determine the anti-C botulinum type A toxin IgG serum titer. The ELISA protocol used is a modification of that described in Example 13c. Briefly. 96-well microtiter plates (Falcon. Pro-Bind Assay Plates) were coated with C. botulinum type A toxoid (prepared as described in Example 3a) by placing 100 pl volumes of C botulinum type A toxoid at 2.5 pg/ml in PBS containing 0.005% thimerosal in each well and incubating overnight at 4 0 C. The next morning, the coating suspensions were decanted and all wells were washed three times using PBS.
In order to block non-specific binding sites. 100 pl of blocking solution BSA in PBS] was then added to each well and the plates were incubated for 1 hr at 37 0 C. The blocking solution was decanted and duplicate samples of 150 pl of diluted rat serum added to the first well of a dilution series. The initial testing serum dilution was 1:30 in blocking solution containing 0.5% Tween 20 followed by 5-fold dilutions into this solution. This was accomplished by serially transferring 30 pl aliquots to 120 pl blocking solution containing Tween 20. mixing, and repeating the dilution into a fresh well. After the final dilution.
163 pl was removed from the well such that all wells contained 120 Al final volume. A total of 3 such dilutions were performed (4 wells total). The plates were incubated 1 hr at 37 0
C.
Following this incubation, the serially diluted samples were decanted and the wells were washed six times using PBS containing 0.5% Tween 20 (PBST). To each well. 100 pl of a rabbit anti-Rat IgG alkaline phosphatase (Sigma) diluted (1/1000) in blocking buffer containing 0.5% Tween 20 was added and the plate was incubated for 1 hr at 37C. The conjugate solutions were decanted and the plates were washed as described above, substituting mM Na,C03, pH 9.5 for the PBST in the final wash. The plates were developed by the addition of 100 il of a solution containing 1 mg/ml para-nitro phenyl phosphate (Sigma) dissolved in 50 mM Na,CO,. 10 mM MgCl,, pH 9.5 to each well. and incubating the plates at room temperature in the dark for 5-45 min. The absorbency of each well was measured at .410 nm using a Dynatech MR 700 plate reader. The results are summarized in Tables 37 and 38 and represent mean serum reactivities of individual mice.
TABLE 37 Determination Of Anti-C. botulinum Type A Toxin Serum IgG Titers Following Immunization With C. botulinum C Fragment-Containing Fusion Proteins Route of Immunization Nasal Oral pMBot pMBot& Immunoen PRE pMBot pMA1870- pMABot pMBot pMAI870- pMABot IMMUNE 2680 2680 Dilution 1:30 0.080 1.040 1.030 0.060 0.190 0.080 0.120 1:150 0.017 0.580 0.540 0.022 0.070 0.020 0.027 1:750 0.009 0.280 0.260 0.010 0.020 0.010 0.014 1:3750 0.007 0.084 0.090 0.009 0.009 0.010 0.007 SRats 5 5 5 5 2 Tested Numbers represent the average values obtained from two ELISA plates.
standardized utilizing the preimmune control.
-164- TABLE 38 Determination Of Anti-C botulinum Type A Toxin Serum IgG Titers Following Immunization With C. botulinum C Fragment-Containing Fusion Proteins Route of Immunization Nasal Oral Immunogen PRE-IMMUNE pMBot pMABot pMNABot pMNABot Dilution 1:30 0.040 0.557 0.010 0.015 0.010 1:150 0.009 0383 0.001 0.003 0.002 1:750 0.001 0.140 0.000 0.000 0.000 1:3750 0.000 0.040 0.000 0.000 0.000 Rats Tested 1 I 3 3 The above ELISA results demonstrate that reactivity against the botulinal fusion proteins was strongest when the route of administration was nasal: only weak responses were stimulated when the botulinal fusion proteins were given orally. Nasally delivered pMbot and pMBot admixed with pMA1870-2680 invoked the greatest serum IgG response. These results show that only the pMBot protein is necessary to induce this response. since the addition of the pMA1870-2680 protein did not enhance antibody response (Table 37). Placement of the 20 C. difficile toxin A fragment between the MBP and the C. botulinum C fragment protein dramatically reduced anti-bot IgG titer (see results using pMABot. pMCABot and pMNABot proteins).
This study demonstrates that the pMBot protein induces a strong serum IgG response directed against C botulinum type A toxin when nasally administered.
b) In Vivo Neutralization Of C. botulinum Type A Neurotoxin By Anti- Recombinant C. botulinum C Fragment Antibodies The ability of the anti-C. botulinum type A toxin antibodies generated by nasal administration of recombinant botulinal fusion proteins in rats (Example 22) to neutralize C.
botulinum type A toxin was tested in a mouse neutralization model. The mouse model is the art accepted method for detection of botulinal toxins in body fluids and for the evaluation of anti-botulinal antibodies Schantz and D.A. Kautter. J. Assoc. Off. Anal. Chem. 61:96 (1990) and Investigational New Drug (BB-IND-3703) application by the Surgeon General of.
165 the Department of the Army to the Federal Food and Drug Administration]. The anti-C botulinum type A toxin antibodies were prepared as follows.
Rats from the group given pMBot protein by nasal administration were boosted a second time with 250 Vg pMBot protein per rat and serum was collected 7 days later. Serum from one rat from this group and from a preimmune rat was tested for anti-C botulinum type A toxin neutralizing activity in the mouse neutralization model described below.
The LD.o of a solution of purified C botulinum type A toxin complex. obtained from Dr. Eric Johnson (University of Wisconsin Madison). was determined using the intraperitoneal (IP) method of Schantz and Kautter Assoc. Off. Anal. Chem. 61:96 (1978)] using 18-22 gram female ICR mice and was found to be 3500 LDo/ml. The determination of the LDo was performed as follows. A Type A toxin standard was prepared by dissolving purified type A toxin complex in 25 mM sodium phosphate buffer. pH 6.8 to yield a stock toxin solution of 3.15 x 10 7 LDomg. The of the solution was determined and the concentration was adjusted to 10-20 pg/ml. The toxin solution was then diluted 1:100 in gel-phosphate (30 mM 15 phosphate. pH 6.4: 0.2% gelatin). Further dilutions of the toxin solution were made as shown below in Table 39. Two mice were injected IP with 0.5 ml of each dilution shown and the mice were observed for symptoms of botulism for a period of 72 hours.
TABLE 39 Determination Of The LDo Of Purified C. botulinum Type A Toxin Complex Dilution Number Dead At 72 hr 1:320 2/2 1:640 2/2 1:1280 2/2 1:2560 0/2 (sick after 72 hr) 1:5120 0/2 (no symptoms) From the results shown in Table 39. the toxin titer was assumed to be between 2560 LDo/ml and 5120 LDo/ml (or about 3840 LD o/ml). This value was rounded to 3500 LDo/ml for the sake of calculation.
The amount of neutralizing antibodies present in the serum of rats immunized nasally with pMBot protein was then determined. Serum from two rats boosted with pMBot protein as described above and preimmune serum from one rat was tested as follows. The toxin 166standard was diluted 1:100 in gel-phosphate to a final concentration of 350 LD.o/ml. One milliliter of the diluted toxin standard was mixed with 25 il of serum from each of the three rats and 0.2 ml of gel-phosphate. The mixtures were incubated at room temperature for min with occasional mixing. Each of two mice were injected with IP with 0.5 ml of the mixtures. The mice were observed for signs of botulism for 72 hr. Mice receiving serum from rats immunized with pMBot protein neutralized this challenge dose. Mice receiving preimmune rat'serum died in less than 24 hr.
The amount of neutralizing anti-toxin antibodies present in the serum of rats immunized with pMBot protein was then quantitated. Serum antibody titrations were performed by mixing 0.1 ml of each of the antibody dilutions (see Table 40) with 0.1 ml of a 1:10 dilution of stock toxin solution (3.5 x 10' LDS/ml) with 1.0 ml of gel-phosphate and injecting 0.5 ml IP into 2 mice per dilution. The mice were then observed for signs of botulism for 3 days (72 hr). The results are tabulated in Table 39.
As shown in Table 40 pMBot serum neutralized C botulinum type A toxin complex 15 when used at a dilution of 1:320 or less. A mean neutralizing value of 168 IU/ml was obtained for the pMBot serum (an IU is defined as 10.000 mouse LDo). This value translates to a circulating serum titer of about 3.7 IU/mg of serum protein. This neutralizing titer is comparable to the commercially available bottled concentrated (Connaught Laboratories. Ltd.) horse anti-C hotulinum antiserum. A 10 ml vial of Connaught antiserum contains about 200 20 mg/ml of protein:each ml can neutralize 750 IU of C botulinum type A toxin. After administration of one vial to a human, the circulating serum titer of the Connaught preparation would be approximately 25 IU/ml assuming an average serum volume of 3 liters).
Thus. the circulating anti-C botulinum titer seen in rats nasally immunized with pMBot protein (168 IU/ml) is 6.7 time higher than the necessary circulation titer of anti-C botulinum antibody needed to be protective in humans.
167 10 TABLE Quantitation Of Neutralizing Antibodies In pMBot Sera 1:20 2/2 2/2 1:40 2/2 2/2 1:80 2/2 2/2 1:160 2/2 2/2 1:320 2 /2b 2/2b 1:640 0/2 0/2 1:1280 0/2 0/2 1:2560 0/2 0/2 Numbers represent the number of mice surviving at 72 hours which received serum taken from rats immunized with the pMBot protein.
15 These mice survived but were sick after 72 hr.
These results demonstrate that antibodies capable of neutralizing C. botulinum type A toxin are induced when recombinant C hotulinum C fragment fusion protein produced in E.
coli is used as an immunogen.
EXAMPLE 24 Production Of Soluble C botulinum C Fragment Protein Substantially Free Of Endotoxin Contamination Example 23 demonstrated that neutralizing antibodies are generated by immunization with the pMBot protein expressed in E. coli. These results showed that the pMBot fusion protein is a good vaccine candidate. However. immunogens suitable for use as vaccines should be pyrogen-free in addition to having the capability of inducing neutralizing antibodies. Expression clones and conditions that facilitate the production of C botulinum C fragment protein for utilization as a vaccine were developed.
The example involved: determination of pyrogen content of the pMBot protein; generation of C. botulinum C fragment protein free of the MBP: expression of C 168botulinum C fragment protein using various expression vectors: and purification of soluble C botulinum C fragment protein substantially free of significant endotoxin contamination.
a) Determination Of The Pyrogen Content Of The pMBot Protein In order to use a recombinant antigen as a vaccine in humans or other animals, the antigen preparation must be shown to be free of pyrogens. The most significant pyrogen present in preparations of recombinant proteins produced in gram-negative bacteria, such as E.
coli. is endotoxin Pearson. Pyrogens: endotoxins. L4L testing and depyrogeniaion, (1985) Marcel Dekker. New York. pp. 23-56]. To evaluate the utility of the pMBot protein as a vaccine candidate. the endotoxin content in MBP fusion proteins was determined.
The endotoxin content of recombinant protein samples was assayed utilizing the Limulus assay (LAL kit: Associates of Cape Cod according to the manufacturer's instructions. Samples of affinity-purified pMal-c protein and pMA1870-2680 were found to 15 contain high levels of endotoxin [>50.000 EU/mg protein: EU (endotoxin unit)]. This suggested that MBP- or toxin A repeat-containing fusions with the botulinal C fragment should also contain high levels of endotoxin. Accordingly, removal of endotoxin from affinity-purified pMal-c and pMBot protein preparations was attempted as follows.
Samples of pMal-c and pMBot protein were depyrogenated with polymyxin to 20 determine if the endotoxin could be easily removed. The following amount of protein was treated: 29 ml at 4.8 OD,,/ml for pMal-c and 19 mis at 1.44 OD, 8 /ml for pMBot. The protein samples were dialyzed extensively against PBS and mixed in a 50 ml tube (Falcon) with 0.5 ml PBS-equilibrated polymyxin B (Affi-Prep Polymyxin. BioRad). The samples were allowed to mix by rotating the tubes overnight at 4 0 C. The polymyxin was pelleted by centrifugation for 30 min in a bench top centrifuge at maximum speed (approximately 2000 x g) and the supernatant was removed. The recovered protein (in the supernatant) was quantified by OD, 8 o, and the endotoxin activity was assayed by LAL. In both cases only approximately 1/3 of the input protein was recovered and the polymyxin-treated protein retained significant endotoxin contamination (approximately 7000 EU/mg of pMBot).
The depyrogenation experiment was repeated using an independently purified pMal-c protein preparation and similar results were obtained. From these studies it was concluded that significant levels of endotoxin copurifies with these MBP fusion proteins using the 169 amylose resin. Furthermore, this endotoxin cannot be easily removed by polymyxin treatment.
These results suggest that the presence of the MBP sequences on the fusion protein complicated the removal of endotoxin from preparations of the pMBot protein.
b) Generation Of C botulinum C Fragment Protein Free Of The MBP It was demonstrated that the pMBot fusion protein could not be easily purified from contaminating endotoxin in section a) above. The ability to produce a pyrogen-free endotoxin-free) preparation of soluble botulinal C fragment protein free of the MBP tag was next investigated. The pMBot expression construct was designed to facilitate purification of the botulinal C fragment from the MBP tag by cleavage of the fusion protein by utilizing an engineered Factor Xa cleavage site present between the MBP and the botulinal C fragment.
The Factor Xa cleavage was performed as follows.
15 Factor Xa (New England Biolabs) was added to the pMBot protein (using a 0.1-1.0% Factor Xa/pMBot protein ratio) in a variety of buffer conditions PBS-NaCI (PBS containing 0.5 M NaCI), PBS-NaCI containing 0.2% Tween 20. PBS. PBS containing 0.2% Tween 20, PBS-C (PBS containing 2 mM CaCI,), PBS-C containing either 0.1 or 0.5 Tween 20. PBS-C containing either 0.1 or 0.5% NP-40, PBS-C containing either 0.1 or S 20 Triton X-100. PBS-C containing 0.1% sodium deoxycholate. PBS-C containing 0.1% SDS].
The Factor Xa digestions were incubated for 12-72 hrs at room temperature.
The extent of cleavage was assessed by Western blot or Coomassie blue staining of proteins following electrophoresis on denaturing SDS-PAGE gels, as described in Example 22. Cleavage reactions (and control samples of uncleaved pMBot protein) were centrifuged for 2 min in a microfuge to remove insoluble protein prior to loading the samples on the gel.
The Factor Xa treated samples were compared with uncleaved, uncentrifuged pMBot samples on the same gel. The results of this analysis is summarized below.
1) Most (about 90%) pMBot protein could be removed by centrifugation. even when uncleaved control samples were utilized. This indicated that the pMBot fusion protein was not fully soluble it exists as a suspension rather than as a solution). [This result was consistent with the observation that most affinity-purified pMBot protein precipitates after long term storage weeks) at 4*C. Additionally, the majority 75%) of induced pMBot protein remains in the pellet after sonication and clarification of the induced E. coli.
170- Resuspension of these insoluble pellets in PBS followed by sonication results in partial solubilization of the insoluble pMBot protein in the pellets.] 2) The portion of pMBot protein that is fully in solution (about 10% of pMBot protein) is completely cleaved by Factor Xa. but the cleaved (released) botulinal C fragment is relatively insoluble such that only the cleaved MBP remains fully in solution.
3) None of the above reaction conditions enhanced solubility without also reducing effective cleavage. Conditions that effectively solubilized the cleaved botulinal C fragment were not identified.
4) The use of 0.1% SDS in the buffer used for Factor Xa cleavage enhanced the solubility of the pMBot protein (all of pMBot protein was soluble). However, the presence of the SDS prevented any cleavage of the fusion protein with Factor Xa.
5) Analysis of pelleted protein from the cleavage reactions indicated that both full length pMBot uncleaved) and cleaved botulinal C fragment protein precipitated during incubation.
15 These results demonstrate that purification of soluble botulinal C fragment protein after cleavage of the pMBot fusion protein is complicated by the insolubility of both the pMBot protein and the cleaved botulinal C fragment protein.
S
c) Expression Of C. botulinum C Fragment Using Various 20 Expression Vectors In order to determine if the solubility of the botulinal C fragment was enhanced by o. expressing the C fragment protein as a native protein, an N-terminal His-tagged protein or as a fusion with glutathione-S-transferase (GST). alternative expression plasmids were constructed. These expression constructs were generated utilizing the methodologies described in Example 22. Figure 27 provides a schematic representation of the vectors described below.
In Figure 27. the following abbreviations are used. pP refers to the pET23 vector.
pHIS refers to the pETHisa vector. pBlue refers to the pBluescript vector. pM refers to the pMAL-c vector and pG refers to the pGEX3T vector (described in Example 11). The solid black lines represent C botulinum C fragment gene sequences: the solid black ovals represent the MBP: the hatched ovals represent GST; "HHHHH" represents the poly-histidine tag. In Figure 27. when the name for a restriction enzyme appears inside parenthesis, this indicates that the restriction site was destroyed during construction. An asterisk appearing with the 171 name for a restriction enzyme indicates that this restriction site was recreated at a cloning junction.
i) Construction Of pPBot In order to express the C. botulinum C fragment as a native non-fused) protein, the pPBot plasmid (shown schematically in Figure 27) was constructed as follows. The C fragment sequences present in pAlterBot (Example 22) were removed by digestion of pAlterBot with Ncol and HindII. The NcoI/HindIII C fragment insert was ligated to pETHisa vector (described in Example 18b) which was digested with Ncol and HindIII. This ligation creates an expression construct in which the Ncol-encoded methionine of the botulinal C fragment is the initiator codon and directs expression of the native botulinal C fragment.
The ligation products were used to transform competent BL21(DE3)pLysS cells (Novagen).
Recombinant clones were identified by restriction mapping.
15 ii) Construction Of pHisBot In order to express the C botulinum C fragment containing a poly-histidine tag at the "amino-terminus of the recombinant protein, the pHisBot plasmid (shown schematically in Figure 27) was constructed as follows. The NcollHindI botulinal C fragment insert from pAlterbot was ligated into the pETHisa vector which was digested with Nhel and HindIII.
20 The Ncol (on the C fragment insert) and Nhel (on the pETHisa vector) sites were filled in using the Klenow fragment prior to ligation: these sites were then blunt end ligated (the NdeI site was regenerated at the clone junction as predicted). The ligation products were used to transform competent BL21(DE3)pLysS cells and recombinant clones were identified by restriction mapping.
The resulting pHisBot clone expresses the botulinal C fragment protein with a histidine-tagged N-terminal extension having the following sequence: MetGlyHisHisHisHisHisHisHisHisHisHisSerSerGlyHislleGluGlyArgHisMetAla (SEQ ID NO:24); the amino acids encoded by the botulinal C fragment gene are underlined and the vector encoded amino acids are presented in plain type. The nucleotide sequence present in the pETHisa vector which encodes the pHisBot fusion protein is listed in SEQ ID The amino acid sequence of the pHisBot protein is listed in SEQ ID NO:26.
172 iii) Construction Of pGBot The botulinal C fragment protein was expressed as a fusion with the glutathione-Stransferase protein by constructing the pGBot plasmid (shown schematically in Figure 27).
This expression construct was created by cloning the Noll/Sall C fragment insert present in pBlueBot (Example 22) into the pGEX3T vector which was digested with Smal and Xhol.
The NotI site (present on the botulinal fragment) was made blunt prior to ligation using the Klenow fragment. The ligation products were used to transform competent BL21 cells.
Each of the above expression constructs were tested by restriction digestion to confirm the integrity of the constructs.
Large scale (I liter) cultures of pPBot [BL21(DE3)pLysS host], pHisBot [BL21(DE3)pLysS host] and pGBot (BL21 host) were grown in 2X YT medium and induced (using IPTG to 0.8-1.0 mM) for 3 hrs as described in Example 22. Total, soluble and insoluble protein preparations were prepared from 1 ml aliquots of each large scale culture [Williams et al. (1994). supral and analyzed by SDS-PAGE. No obvious induced band was 15 detectable in the pPBot or pHisBot samples by Coomassie staining, while a prominent insoluble band of the anticipated MW was detected in the pGBot sample. Soluble lysates of the pGBot large scale (resuspended in PBS) or pHisBot large scale [resuspended in Novagen IX binding buffer (5 mM imidazole. 0.5 M NaCI, 20 mM Tris-HCI. pH cultures were prepared and used to affinity purify soluble affinity-tagged protein as follows.
20 The pGBot lysate was affinity purified on a glutathione-agarose resin (Pharmacia) exactly as described in Smith and Corcoran [Current Protocols in Molecular Biology.
Supplement 28 (1994). pp. 16.7.1-16.7.7]. The pHisBot protein was purified on the His-Bind resin (Novagen) utilizing the His-bind buffer kit (Novagen) exactly as described by manufacturer.
Samples from the purification of both the pGBot and pHisBot proteins (including uninduced. induced, total, soluble. -nd affinity-purified eluted protein) were resolved on SDS- PAGE gels. Following electrophoresis, proteins were analyzed by Coomassie staining or by Western blot detection utilizing a chicken anti-C botulinum Type A toxoid antibody (as described in Example 22).
These studies showed that the pGBot protein was almost entirely insoluble under the utilized conditions, while the pHisBot protein was soluble. Affinity purification of the pHisBot protein on this first attempt was inefficient, both in terms of yield (most of the 173 immunoreactive botulinal protein did not bind to the His-bind resin) and purity (the botulinal protein was estimated to comprise approximately 20% of the total eluted protein).
d) Purification Of Soluble C. botulinum C Fragment Protein Substantially Free Of Endotoxin Contamination The above studies showed that the pHisBot protein was expressed in E. coli as a soluble protei. However. the affinity purification of this protein on the His-bind resin was very inefficient. In order to improve the affinity purification of the soluble pHisBot protein (in terms of both yield and purity), an alternative poly-histidine binding affinity resin (Ni- NTA resin: Qiagen) was utilized. The Ni-NTA resin was reported to have a superior binding affinity (Kd= 1 x 10- 1 3 at pH 8.0: Qiagen user manual) relative to the His-bind resin.
A soluble lysate (in Novagen IX binding buffer) from an induced 1 liter 2X YT culture was prepared as described above. Briefly. the culture of pHisBot (B121(DE3)pLysS host] was grown at 37*C to an OD, of 0.7 in 1 liter of 2X YT medium containing 100 15 pg/ml ampicillin. 34 pg/ml chloramphenicol and 0.2% glucose. Protein expression was induced by the addition of IPTG to 1 mM. Three hours after the addition of the IPTG. the cells were cooled for 15 min in a ice water bath and then centrifuged 10 min at 5000 rpm in a JA10 rotor (Beckman) at 4°C. The pellets were resuspended in a total volume of 40 mis Novagen IX binding buffer (5 mM imidazole. 0.5 M NaCI. 20 mM Tris-HCI, pH 7.9), 20 transferred to two 35 ml Oakridge tubes and frozen at -70 0 C for at least I hr. The tubes °were thawed and the cells were lysed by sonication (4 X 20 second bursts using a Branson Sonifier 450 with a power setting of 6-7) on ice. The suspension was clarified by centrifugation for 20 min at 9.000 rpm (10.000 x g) in a JA-17 rotor (Beckman).
The soluble lysate was brought to 0.1% NP40 and then was batch absorbed to 7 ml of a 1:1 slurry of Ni-NTA resin:binding buffer by stirring for 1 hr at 4 0 C. The slurry was poured into a column having an internal diameter of 1 or 2.5 cm (BioRad). The column was then washed sequentially with 15 mis of Novagen IX binding buffer containing 0.1% ml of Novagen IX binding buffer. 15 ml wash buffer (60 mM imidazole, 0.5 M NaCl, mM Tris-HCl. pH 7.9) and 15 ml NaHPO, wash buffer (50 mM NaHPO 4 pH 7.0, 0.3 M NaCI. 10 glycerol). The bound protein was eluted by protonation of the resin using elution buffer (50 mM NaHPO 4 pH 4.0. 0.3 M NaCI. 10 glycerol). The eluted protein was stored at 4 0
C.
174 Samples of total. soluble and eluted protein were resolved by SDS-PAGE. Protein samples were prepared for electrophoresis as described in Example 22b. Duplicate gels were stained with Coomassie blue to visualize the resolved proteins and C. botulinum type A toxinreactive protein was detected by Western blot analysis as described in Example 22b. A representative Coomassie stained gel is shown in Figure 28. In Figure 28. the following samples were loaded on the 12.5% acrylamide gel. Lanes 1-4 contain respectively total protein, solubl? protein, soluble protein present in the flow-through of the Ni-NTA column and affinity-purified pHisBot protein protein released from the Ni-NTA resin by protonation). Lane 5 contains high molecular weight protein markers (BioRad).
The purification of pHisBot protein resulted in a yield of 7 mg of affinity purified protein from a 1 liter starting culture of BL21(DE3)pLysS cells harboring the pHisBot plasmid. The yield of purified pHisBot protein represented approximately 0.4% of the total soluble protein in the induced culture. Analysis of the purified pHisBot protein by SDS- PAGE revealed that at least 90-95% of the protein was present as a single band (Figure 28) of S: 15 the predicted MW (50 kD). This 50 kD protein band was immunoreactive with anti-C.
botulinum type A toxin antibodies. The extinction coefficient of the protein preparation was determined to be 1.4 (using the Pierce BCA assay) or 1.45 (using the Lowry assay) OD,so per I mg/ml solution.
Samples of pH neutralized eluted pHisBot protein were resolved on a KB 803 HPLC column (Shodex). Although His-tagged proteins are retained by this sizing column (perhaps S0 due to the inherent metal binding ability of the proteins), the relative mobility of the pHisBot protein was consistent with that expected for a non-aggregated protein in solution. Most of the induced pHisBot protein was determined to be soluble under the growth and solubilization conditions utilized above greater than 90% of the pHisBot protein was found to be soluble as judged by comparison of the levels of pHisBot protein seen in total and soluble protein samples prepared from BL21(DE3)pLysS cells containing the pHisBot plasmid).
SDS-PAGE analysis of samples obtained after centrifugation. extended storage at -20 0 C, and at least 2 cycles of freezing and thawing detected no protein loss (due to precipitation), indicating that the pHisBot protein is soluble in the elution buffer 50 mM NaHPO 4 pH 4.0. 0.3 M NaCl. 10 glycerol).
Determination of endotoxin contamination in the affinity purified pHisBot preparation (after pH neutralization) using the LAL assay (Associates of Cape Cod) detected no significant endotoxin contamination. The assay was performed using the endpoint 175 chromogenic method (without diazo-coupling) according to the manufacturer's instructions.
This method can detect concentrations of endotoxin greater than or equal to 0.03 EU/ml (EU refers to endotoxin units). The LAL assay was run using 0.5 ml of a solution comprising mg pHisBot protein in 50 mM NaHPO4, pH 7.0, 0.3 M NaCI, 10 glycerol: 30-60 EU were detected in the 0.5 ml sample. Therefore. the affinity purified pHisBot preparation contains 60-120 EU/mg of protein. FDA Guidelines for the administration of parenteral drugs require that a composition to be administered to a human contain less than 5 EU/kg body weight (The average human body weight is 70 kg; therefore up to 349 EU units can be delivered in a parental dose.). Because very small amount of protein are administered in a vaccine preparation (generally in the range of 10-500 ag of protein), administration of affinity purified pHisBot containing 60-120 EU/mg protein would result in delivery of only a small percentage of the permissible endotoxin load. For example. administration of 10-500 pg of purified pHisBot to a 70 kg human, where the protein preparation contains 60 EU/mg protein.
results in the introduction of only 0.6 to 30 EU 0.2 to 8.6% of the maximum allowable 15 endotoxin burden per parenteral dose (less than 5 EU/kg body weight)].
The above results demonstrate that endotoxin (LPS) does not copurify with the pHisBot protein using the above purification scheme. Preparations of recombinantly produced pHisBot protein containing lower levels of endotoxin (less than or equal to 2 EU/ mg recombinant protein) may be produced by washing the Ni-NTA column with wash buffer until 20 the OD,o0 returns to baseline levels until no more UV-absorbing material comes off of the column).
The above results illustrate a method for the production and purification of soluble.
botulinal C fragment protein substantially free of endotoxin.
EXAMPLE Optimization Of The Expression And Purification Of pHisBot Protein The results shown in Example 24d demonstrated that the pHisBot protein is an excellent candidate for use as a vaccine as it could be produced as a soluble protein in E. coli and could be purified free of pyrogen activity. In order to optimize the expression and purification of the pHisBot protein, a variety of growth and purification conditions were tested.
176a) Growth Parameters i) Host Strains The influence of the host strain utilized upon the production of soluble pHisBot protein was investigated. A large scale purification of pHisBot was performed [as described in Example 24d above] using the BL21(DE3) host (Novagen) rather than the BL21(DE3)pLysS host. The deletion of the pLysS plasmid in the BL21(DE3) host yielded higher levels of expression due to de-repression of the plasmid's T7-lac promoter. However, the yield of affinity-purified soluble recombinant protein was very low (approximately 600 pg/ liter culture) when purified under conditions identical to those described in Example 24d above. This result was due to the fact that expression in the BL21(DE3) host yielded very high level expression of the pHisBot protein as insoluble inclusion bodies as shown by SDS- PAGE analysis of protein prepared from induced BL21(DE3) cultures (Figure 29. lanes 1-7, described below). These results demonstrate that the pHisBot protein is not inherently toxic to E. coli cells and can be expressed to high levels using the appropriate promoter/host 15 combination.
Figure 29 shows a Coomassie blue stained SDS-PAGE gel (12.5% acrylamide) onto which extracts prepared from BL21(DE3) cells containing the pHisBot plasmid were loaded.
Each lane was loaded with 2.5 pl protein sample mixed with 2.5 pl of 2X SDS sample buffer.
The samples were handled as described in Example 22b. The following samples were applied to the gel. Lanes 1-7 contain protein isolated from the BL21(DE3) host. Lanes 8-14 contain proteins isolated from the BL21(DE3)pLysS host. Total protein was loaded in lanes 1. 2. 4.
6. 8, 10 and 12. Soluble protein was loaded in Lanes 3. 5. 7. 9. 11 and 13. Lane I contains protein from uninduced host cells. Lanes 2-13 contain protein from host cells induced for 3 hours. IPTG was added to a final concentration of 0.1 mM (Lanes 0.3 mM (Lanes or 1.0 mM (Lanes 2, 3. 8-13). The cultures were grown in LB broth (Lanes 2X YT broth (Lanes 10-11) or terrific broth (Lanes 1-7, 12-13). The pHisBot protein seen in Lanes 3. 5 and 7 is insoluble protein which spilled over from Lanes 2. 4 and 6, respectively. High molecular weight protein markers (BioRad) were loaded in Lane 14.
A variety of expression conditions were tested to determine if the BL21(DE3) host could be utilized to express soluble pHisBot protein at suitably high levels about mg/ml). The conditions altered were temperature (growth at 37 or 30 0 culture medium (2X YT, LB or Terrific broth) and inducer levels 0.3 or 1.0 mM IPTG). All combinations of these variables were tested and the induction levels and solubility was then 177 assessed by SDS-PAGE analysis of total and soluble extracts [prepared from 1 ml samples as described in Williams et al.. (1994). supra].
All cultures were grown in 15 ml tubes (Falcon #2057). All culture medium was prewarmed overnight at the appropriate temperature and were supplemented with 100 gg/ml ampicillin and 0.2% glucose. Terrific broth contains 12 g/l bacto-tryptone. 24 g/1 bacto-yeast extract and 100 ml/I of a solution comprising 0.17 M KIHPO 4 0.72 M KHPO 4 Cultures were grown in'a incubator on a rotating wheel (to ensure aeration) to an OD. of approximately 0.4. and induced by the addition of IPTG. In all cases, high level expression of insoluble pHisBot protein was observed, regardless of temperature. medium or inducer concentration.
The effect of varying the concentration of IPTG upon 2X YT cultures grown at 23"C was then investigated. IPTG was added to a final concentration of either 1 mM, 0.1 mM.
0.05 mM or 0.01 mM. At this temperature. similar levels of pHis Bot protein was induced in the presence of either 1 or 0.1 mM IPTG: these levels of expression was lower than that S 15 observed at higher temperatures. Induced protein levels were reduced at 0.05 mM IPTG and absent at 0.01 mM IPTG (relative to 1.0 and 0.1 mM IPTG inductions at 23 0 However.
no conditions were observed in which the induced pHisBot protein was soluble in this host.
Thus. although expression levels are superior in the BL21(DE3) host (as compared to the BL21(DE3)pLysS host), conditions that facilitate the production of soluble protein in this host could not be identified.
These results demonstrate that production of soluble pHisBot protein was achieved using the BL21(DE3)pLysS host in conjunction with the T7-lac promoter.
ii) Effect Of Varying Temperature, Medium And IPTG Concentration And Length Of Induction The effect growing the host cells in various mediums upon the expression of recombinant botulinal protein from the pHisBot expression construct [in the BL21(DE3)pLysS host] was investigated. BL21(DE3)pLysS cells containing the pHisBot plasmid were grown in either LB. 2X YT or Terrific broth at 37 0 C. The cells were induced using 1 mM IPTG for a 3 hr induction period. Expression of pHisBot protein was found to be the highest when the cells were grown in 2X YT broth (see Figure 29. lanes 8-13).
The cells were then grown at 30 0 C in 2X YT broth and the concentration of IPTG was varied from 1.0. 0.3 or 0.1 mM and. the length of induction was either 3 or 5 hours.
178- Expression of pHisBot protein was similar at all 3 inducer concentrations utilized and the levels of induced protein were higher after a 5 hr induction as compared to a 3 hr induction.
Using the conditions found to be optimal for the expression of pHisBot protein, a large scale culture was grown in order to provide sufficient material for a large scale purification of the pHisBot protein. Three 1 liter cultures were grown in 2X YT medium containing 100 pg/ml ampicillin. 34 pg/ml chloramphenicol and 0.2% glucose. The cultures were grown at 0 C and were induced with 1.0 mM IPTG for a 5 hr period. The cultures were harvested and a soluble lysate were prepared as described in Example 18. A large scale purification was performed as described in Example 24d with the exception that except the soluble lysate was batch absorbed for 3 hours rather than for 1 hour. The final yield was 13 mg pHisBot protein/liter culture. The pHisBot protein represented 0.75% of the total soluble protein.
The above results demonstrate growth conditions under which soluble pHisBot protein is produced use of the BL21(DE3)pLysS host. 2X YT medium. 30 0 C. 1.0 mM IPTG for 5 hours).
b) Optimization Of Purification Parameters For optimization of purification conditions, large scale cultures (3 X 1 liter) were "grown at 30 0 C and induced with 1 mM IPTG for 5 hours as described above. The cultures were pooled, distributed to centrifuge bottles, cooled and pelleted as described in Example 24d. The cell pellets were frozen at -70 0 C until used. Each cell pellet represented 1/3 of a liter starting culture and individual bottles were utilized for each optimization experiment described below. This standardized the input bacteria used for each experiment, such that the yields of affinity purified pHisBot protein could be compared between different optimization experiments.
i) Binding Specificity (pH Protonation) A lysate of pHisBot culture was prepared in PBS (pH 8.0) and applied to a 3 ml Ni- NTA column equilibrated in PBS (pH 8.0) using a flow rate of 0.2 ml/min (3-4 column volumes/hr) using an Econo chromatography system (BioRad). The column was washed with PBS (pH 8.0) until the absorbance (OD,, 0 of the elute was at baseline levels. The flow rate was then increased to 2 ml/min and the column was equilibrated in PBS (pH A pH gradient (pH 7.0 to 4.0 in PBS) was applied in order to elute the bound pHisBot protein from the column. Fractions were collected and aliquots were resolved on SDS-PAGE gels. The 179- PAGE gels were subjected to Western blotting and the pHisBot protein was detected using a chicken anti-C. botulinum Type A toxoid antibody as described in Example 22.
From the Western blot analysis it was determined that the pHisBot protein begins to elute from the Ni-NTA column at pH 6.0. This is consistent with the predicted elution of a His-tagged protein monomer at pH 5.9.
These results demonstrate that the pH at which the pHisBot protein is protonated (released) from Ni-NTA resin in PBS buffer is pH ii) Binding Specificity (Imidazole Competition) In order to define purification conditions under which the native E. coli proteins could be removed from the Ni-NTA column while leaving the pHisBot protein bound to the column. the following experiment was performed. A lysate of pHisBot culture was prepared in 50 mM NaHPO. 0.5 M NaCI. 8 mM imidazole (pH This lysate was applied to a 3 ml Ni-NTA column equilibrated in 50 mM NaHPO, 0.5 M NaCI (pH 7.0) using an Econo S 15 chromatography system (BioRad). A flow rate of 0.2 ml/min (3-4 column volumes/hr) was utilized. The column was washed with 50 mM NaHPO,, 0.5 M NaCI (pH 7.0) until the absorbance of the elute returned to baseline. The flow rate was then increased to 2 ml/min.
The column was eluted using an imidazole step gradient [in 50 mM NaHPO 4 0.5 M NaCI (pH Elution steps were 20 mM, 40 mM. 60 mM. 80 mM. 100 mM. 200 mM, M imidazole. followed by a wash using 0.1 mM EDTA (to strip the nickel from the column and remove any remaining protein). In each step. the wash was continued until the ODso returned to baseline. Fractions were resolved on SDS-PAGE gels. Western blotted, and pHisBot protein detected using a chicken anti-C botulinum Type A toxoid antibody as described in Example 22. Duplicate gels were stained with Coomassie blue to detect eluted protein in each fraction.
The results of the PAGE analysis showed that most of the non-specifically binding bacterial protein was removed by the 20 mM imidiazole wash. with the remaining bacterial proteins being removed in the 40 and 60 mM imidazole washes. The pHisBot protein began to elute at 100 mM imidazole and was quantitatively eluted in 200 mM imidazole.
These results precisely defined the window of imidazole wash stringency that optimally removes E. coli proteins from the column while specifically retaining the pHisBot protein in this buffer. These results provided conditions under which the pHisBot protein can be purified free of contaminating host proteins.
180iii) Purification Buffers And Optimized Purification Protocols A variety of purification parameters were tested during the development of an optimized protocol for batch purification of soluble pHisBot protein. The results of these analyses are summarized below.
Batch purifications were performed (as described in Example 24d) using several buffers to determine if alternative buffers could be utilized for binding of the pHisBot protein to the Ni-NTA column. It was determined that quantitative binding of pHisBot protein to the Ni-NTA resin was achieved in either Tris-HCl (pH 7.9) or NaHPO, (pH 8.0) buffers.
Binding of the pHisBot protein in NaHPO, buffer was not inhibited using 5 mM. 8 mM or mM imidazole. Quantitative elution of bound pHisBot protein was obtained in buffers containing 50 mM NaHPO,, 0.3 M NaCI (pH with or without 10% glycerol.
However. quantitation of soluble affinity purified pHisBot protein before and after a freeze thaw (following several weeks storage of the affinity purified elute at -20 0 C) revealed that 15 94% of the protein was recovered using the glycerol-containing buffer, but only 68% of the protein was recovered when the buffer lacking glycerol was employed. This demonstrates that glycerol enhanced the solubility of the pHisBot protein in this low pH buffer when the eluted protein was stored at freezing temperatures Neutralization of pH by addition of NaH,PO 4 buffer did not result in obvious protein precipitation.
20 It was determined that quantitative binding of pHisBot protein using the batch format S* occurred after 3 hrs (Figure 30). but not after I hr of binding at 4 0 C (the resin was stirred during binding). Figure 30 depicts a Coomaisse blue stained SDS-PAGE gel S. acrylamide) containing samples of proteins isolated during the purification of pHisBot protein from lysate prepared from the BL21(DE3)pLysS host. Each lane was loaded with 5 p of protein sample mixed with 5 pl of 2X sample buffer and processed as described in Example 22b. Lane I contains high molecular weight protein markers (BioRad). Lanes 2 and 3 contain protein eluted from the Ni-NTA resin. Lane 4 contains soluble protein after a 3 hr batch incubation with the Ni-NTA resin. Lanes 5 and 6 contain soluble and total protein, respectively. Figure 30 demonstrates that the pHisBot protein is completely soluble [compare Lanes 5 and 6 which show that a similar amount of the 50 kD pHisBot protein is seen in both: if a substantial amount (greater than 20%) of the pHisBot protein were partially insoluble in the host cell, more pHisBot protein would be seen in lane 6 (total protein) as compared to lane 5 (soluble protein)]. Figure 30 also demonstrates that the pHisBot protein is 181 completely removed from the lysate after batch absorption with the Ni-NTA resin for 3 hours (compare Lanes 4 and The reported high affinity interaction of the Ni-NTA resin with His-tagged proteins (Kd= I x 10"1 at pH 8.0) suggested that it should be possible to manipulate the resin-protein complexes without significant release of the bound protein. Indeed. it was determined that after the recombinant protein was bound to the Ni-NTA resin, the resin-pHisBot protein complex was highly stable and remained bound following.repeated rounds of centrifugation of the resin for 2 min at 1600 x g. When this centrifugation step was performed in a 50 ml tube (Falcon). a tight resin pellet formed. This allowed the removal of spent soluble lysate biy pouring off the supernatant followed by resuspension of the pellet in wash buffer. Further washes can be performed by centrifugation. The ability to perform additional washes permits the development of protocols for batch absorption of large volumes of lysate with removal of the lysate being performed simply by centrifugation following binding of the recombinant protein to the resin.
A simplified, integrated purification protocol was developed as follows. A soluble lysate was made by resuspending the induced cell pellet in binding buffer [50 mM NaHPO 4 M NaCI. 60 mM imidazole (pH sonicating 4 x 20 sec and centrifuging for 20 min at 10.000 x g. NP-40 was added to 0.1% and Ni-NTA resin (equilibrated in binding buffer) was added. Eight milliliters of a 1:1 slurry (resin:binding buffer) was used per liter of 20 starting culture. The mixture was stirred for 3 hrs at 4°C. The slurry was poured into a column having a 1 cm internal diameter (BioRad). washed with binding buffer containing 0.1% NP40. then binding buffer until baseline was established (these steps may alternatively be performed by centrifugation of the resin, resuspension in binding buffer containing followed by centrifugation and resuspension in binding buffer). Imidazole was removed by washing the resin with 50 mM NaHPO,, 0.3M NaCI (pH Protein bound to the resin was eluted using the same buffer (50 mM NaHPO., 0.3M NaCI) having a reduced pH (pH A pilot purification was performed following this protocol and yielded 18 mg/liter affinity-purified pHisBot. The pHisBot protein was greater than 90% pure as estimated by Coomassie staining of an SDS-PAGE gel. This represents the highest observed yield of soluble affinity-purified pHisBot protein and this protocol eliminates the need for separate iridazole-containing binding and wash buffers. In addition to providing a simplified and efficient protocol for the affinity purification of recombinant pHisBot protein, the above 182results provide a variety of purification conditions under which pHisBot protein can be isolated.
EXAMPLE 26 The pHisBot Protein Is An Effective Immunogen In Example 23 it was demonstrated that neutralizing antibodies are generated in mouse serum after nasal immunization with the pMBot protein. However, the pMBot protein was found to copurify with significant amounts of endotoxin which could not be easily removed.
The pHisBot protein, in contrast, could be isolated free of significant endotoxin contamination making pHisBot a superior candidate for vaccine production. To further assess the suitability of pHisBot as a vaccine, the immunogenicity of the pHisBot protein was determined and a comparison of the relative immunogenicity of pMBot and pHisBot proteins in mice was Sperformed as follows.
S 15 Two groups of eight BALBc mice were immunized with either pMBot protein or pHisBot protein using Gerbu GMDP adjuvant (CC Biotech). pMBot protein (in PBS containing 10 mM maltose) or pHisBot protein (in 50 mM NaHPO, 0.3 M NaCI. glycerol. pH 4.0) was mixed with Gerbu adjuvant and used to immunize mice. Each mouse received an IP injection of 100 pl antigen/adjuvant mix (50 lg antigen plus 1 pg adjuvant) on day 0. Mice were boosted as described above with the exception that the route of administration was IM on day 14 and 28. The mice were bled on day 77 and anti-C.
botulinum Type A toxoid titers were determined using serum collected from individual mice in each group (as described in Example 23). The results are shown in Table 41.
183 TABLE 41 Anti-C holulinurn Type A Toxoid Serum IgG Titers In Individual Mice Immunized With pMBot or pHisBot Protein _____1Preimmune' pMBot: pHisBoe Mouse I Sample Dilution Sample Dilution Sample Dilution :5 I. 250 1:1250 1:6250 1:50 1:250 1: 120 1:6250 1:50 11:250 1:1250 1:620 10.678 0.190 00.v3 0.007 1.574 0.799 0.320 0.093 2 .161 0.931 015~4 0.075 1.513 0.829 0.409 0.134 32.364 0.458 0.195 0.041 1.596 1.028 0.453 0.122 4 1.622 1.189 0.334 0.067 1.552 0.840 0.348 0.090 5 1.612 1.030 0.289 0.067 1.629 1.590 0.895 (0.233 6 0.913 0.242 0.069 0.013 1.485 95i2 0.477 0.145 7 0.910 0.235 0.058 0.024 1.524 0.725 0.269 0.069 8 0.747 0.234 0.058 0.024 .274 0.427 0.1126 0.029 Mea 0.48 .02 0.22 0.002 .233 0.64 026 037 1.2 096 0.4212 0.2124 Titer I_ The preimmune sample represents the average from 2 sets of duplicate wells containing sen.'n from a individual mouse immunized with recombinant Staphylococcus enterotoxin B (SEB) antigen. This antigen is immunologically unrelated to C hotulintun toxin and provides a control serum.
Average of duplicate wells.
The results shown above in Table 41 demonstrate that both the pMBot and pHisBot proteins are immunogenic in-mnice as 100% of the mice in each group seroconverted from non-immune to immune status. The results also show that the average titer of anti-C bolulinum Type A toxoid IgG is 2-3 fold higher after immunization with the pHisBot protein relative to immunization with the pMBot protein. This suggests that the pHisBot protein may be a superior immunogen to the pMlol protein.
184 EXAMPLE 27 Immunization With The Recombinant pHisBot Protein Generates Neutralizing Antibodies The results shown in Example 26 demonstrated that both the pHisBot and pMBot proteins were capable of inducing high titers of anti-C. botulinum type A toxoid-reactive antibodies in immunized hosts. The ability of the immune sera from mice immunized with either the pHIsBot or pMBot proteins to neutralize C. botulinum type A toxoid in vivo was determined using the mouse neutralization assay described in Example 23b.
The two groups of eight BALBc mice immunized with either pMBot protein or pHisBot protein in Example 26 were boosted again one week after the bleeding on day 77.
The boost was performed by mixing pMBot protein (in PBS containing 10 mM maltose) or pHisBot protein (in 50 mM NaHPO,. 0.3 M NaCI. 10% glycerol. pH 4.0) with Gerbu adjuvant as described in Example 26. Each mouse received an IP injection of 100 pl antigen/adjuvant mix (50 pg antigen plus I pg adjuvant). The mice were bled 6 days after 15 this boost and the serum from mice within a group was pooled. Serum from preimmune mice was also collected (this serum is the same serum described in the footnote to Table The presence of neutralizing antibodies in the pooled or preimmune serum was detected by challenging mice with 5 LD.o units of type A toxin mixed with 100 pl of pooled serum. The challenge was performed by mixing (per mouse to be injected) 100 pl of serum from each pool with 100 pl of purified type A toxin standard (50 LD, /ml prepared as described in Example 23b) and 500 pl of gel-phosphate. The mixtures were incubated for min at room temperature with occasional mixing. Each of four mice were injected IP with the mixtures (0.7 ml/mouse). The mice were observed for signs of botulism for 72 hours.
Mice receiving toxin mixed with serum from mice immunized with either the pHisBot or pMBot proteins showed no signs of botulism intoxication. In contrast. mice receiving preimmune serum died in less than 24 hours.
These results demonstrate that antibodies capable of neutralizing C botulinum type A toxin are induced when either of the recombinant C. botulinum C fragment proteins pHisBot or pMBot are used as immunogens.
185 EXAMPLE 28 Expression and Purification of Recombinant C. dificile Toxin A Proteins Containing the 1870-2680. 1870-2190 and 1960-2680 Interval Previously others had raised antibodies against C. difficile toxin A by actively immunizing hamsters against a recombinant polypeptide located within the Interval 6 region [Lyerly. et al. (1990) Curr. Microbiol. 21:29]. The structure of the recombinant clone used by Lyerly el al. [(1990) Curr. Microbiol. 21:29] is shown schematically in Figure 31 as pUC 1960-2680.
In Figure 31. the following abbreviations are used. pP refers to the pET23 vector: pM refers to the pMal-c vector: pGEX refers to the pGEX vector: Trx refers to thioredoxin: pUC refers to the pUC9 vector: A refers to C. dificile toxin A. The numbers refer to the amino Sacid interval expressed in a given construct. The solid black boxes represent coding regions: the open box at the 3' end of the pUC1960-2680 construct represents a portion of a-peptide o 15 of the lacZ gene which is encoded by vector sequences. The solid ovals represent the MBP.
"HHH" represents the poly-histidine tract. The open circles represent thioredoxin. The hatched ovals represent GST.
Using a hamster model of C. difficile associated disease (CDAD) where antibodies are given. prophylactically. the Lyerly. et al. antibodies (intra-Interval 6: pUC1960-2680) were only able to partially protect hamsters against C difficile infection in terms of survival (4 out of 8 animals survived) and furthermore. these antibodies did not prevent diarrhea in any of the animals. Additionally. animals treated with the intra-lnterval 6 antibodies [Lyerly. et al.
(1990). supra] died when treatment was removed. In contrast Example 16 demonstrated that passive administration of anti-Interval 6 antibodies (anti-pMA1870-2680) prevented diarrhea in 6 out of 7 animals and completely prevented death due to CDAD in the prophylactic treatment model system. Furthermore passive administration of the anti-Interval 6 antibodies provided a long lasting cure treatment could be withdrawn without incident).
While the antibodies of Lyerly. et al. were reported to provide some protection against CDAD. the integrity and purity of the recombinant protein expressed from the pUC1960-2680 construct was not reported. The pUC1960-2680 construct potentially expresses the 1960-2680 aa interval of C. difficile toxin A in the pUC9 vector: this interval is nested within the pMA1870-2680 clone (see Figure 31).
This example involved: construction of pUC1960-2680 and characterization of the expressed protein by Western blot analysis: cloning and expression of the 1960-2680 186 interval as an affinity tagged protein in pET and pMal vectors and affinity purification and characterization of soluble MBP tagged proteins from clones expressing the 1870-2680. 1870- 2190 or 1960-2680 intervals.
a) Construction of pUC1960-2680 and Characterization of Expressed Protein by Western Blot Analysis The"pUC1960-2680 construct contains a 2.1 kb C. difficile toxin A gene fragment encoding 33 of the 38 repeat units: this construct was generated to provide the same recombinant protein utilized by Lyerly el al. [(1990) Curr.'Microbiol. 21:29] for the generation of anti-C difficile toxin A antibodies. pUC1960-2680 was constructed as follows.
Briefly, the 2.1 kb Pstl fragment containing the C difficile toxin A repeats was removed from pPA1100-2680 (Example 11) and was cloned into pUC9 which had been digested with Pstl and dephosphorylated. The dephosphoryation reaction was performed using calf intestinal alkaline phosphatase (CIP) according to the manufacturers instructions (New England 15 Biolabs). Following restriction digestion and CIP-treatment, the reaction products were resolved on an agarose gel, and the appropriate fragments were excised, mixed, and purified utilizing the Prep-a-Gene kit (BioRad). The eluted DNA was ligated, transformed into JM109 competent cells and recombinant clones isolated, and confirmed by restriction digestion using standard techniques of molecular biology. Plasmid DNA was isolated using the QIA-prep spin plasmid kit (Qiagen).
JM 109 containing the pUC1960-2680 construct were grown, induced and total and soluble extracts were prepared as described [Lyerly et al.(1990) Curr. Microbiol. 21:29].
Briefly. a 500 ml culture of Terrific broth was inoculated with pUC1960-2680 (in JM109) and grown at 37 0 C to early stationary phase (0.8 IPTG was added to a final concentration of 1 mM and the culture was induced for 2 hrs. A 1 ml aliquot of the culture was withdrawn prior to the addition of IPTG and served as the uninduced sample. Following growth in the presence of the IPTG for 2 hr. another 1 ml aliquot of the culture was withdrawn and served as the induced sample. These 1 ml uninduced and induced samples were treated as follows.
The bacteria were pelleted by centrifugation. The cell pellets were resuspended in 100 pl 2X sample buffer (0.125 mM Tris-HCI, pH 6.8, 2 mM EDTA 6% SDS, 20% glycerol. 0.25% bromophenol blue: p-mercaptoethanol was added to 5% before use).
The remaining culture was then processed to prepare total and soluble extracts for analysis. The culture was distributed into 500 ml centrifuge bottles. The bottles were cooled 187 for 15 min in a ice water bath and the cells were pelleted by centrifugation at 5.000 rpm in a Beckman JA10 rotor. The cell pellet was resuspended in 1/10 initial culture volume ml) of TBS (0.05 M Tris-HCI. 0.15 M NaCI, pH 7.5) and distributed to two 35 ml Oakridge tubes. One and one forth milliliters of a 10 mg/ml solution of lysozyme (in TBS) was added to each tube and the mixtures were incubated on ice for 20 min. The two tubes were stored at -70°C overnight. One of the two tubes from the induced culture was then thawed and sonicated in ice water using four twenty second bursts (Branson Sonifier Model 450 set at level The sample was clarified by centrifugation for 20 min at 9000 rpm (Beckman J2- 21 rotor), and the soluble lysate filter sterilized through a 0.45 pm filter. Total (before centrifugation) and soluble (after filter sterilization) extracts were prepared for electrophoresis by mixing 4 pll extract with 16 pl PBS and 20 pl 2X sample buffer.
The protein samples were resolved by electrophoresis on a 12.5% SDS-PAGE gel and :i the proteins detected either by Coomassie blue staining (detects all proteins) and Western blot analysis (detects specific proteins) utilizing a goat anti-toxin A specific antibody (TechLabs) S: 15 as follows. The 12.5% SDS-PAGE gels were loaded with the protein samples. After electrophoresis. the gel was bisected. One half was stained with Coomassie blue and the proteins on the other half were transferred to a solid support for Western blot analysis.
Protein transfer was confirmed by Ponceau S staining (as described in Example 12b). The blot was then incubated for 1 hr at 20 0 C in PBS containing 0.1% Tween 20 (PBST) and 20 milk (blocking buffer). Then 10 ml of a solution comprising a 1/1000 dilution of an affinity purified goat anti-C difficile toxin A antibody (Tech Labs) in blocking buffer was added and the blot was incubated for 1 hr at room temperature. The blot was then washed and the presence of the bound anti-C difficile antibody was detected using a rabbit anti-goat alkaline phosphatase conjugate as secondary antibody as described in Example 3. The resulting Coomassie blue-stained gel and developed Western blot are shown in Figure 32.
In Figure 32. the Coomassie blue-stained gel is shown on the left (lanes 1-5) and the Western blot is shown on the right (lanes The following abbreviations are used: uninduced induced total soluble and broad range molecular weight markers BioRad). The size of the MW markers is indicated by the numbers to the right of lane 5. Figure 32 shows that no induced bands corresponding to the size expected for the recombinant pUC1960-2680 protein were detectable by Coomassie blue staining. However.
Western blot detection of C. difficile toxin A-reactive material revealed a predominant, inducible protein species of the predicted MW for the full length recombinant C. difficile 188 toxin A protein. Although some induced protein is soluble. the majority of the protein is insoluble [compare the amount of protein reactive with the antibody present in lanes 8 (total) and 9 (soluble)]. The recombinant protein produced by pUC1960-2680 was also clearly unstable. since breakdown products were detected even in the uninduced (lane 6) or induced (lane 7) culture samples.
b) Cloning and Expression of the 1960-2680 Interval as an Affinity Tagged Protein in pET and pMal Vectors As shown above, the protein produced by the pUC1960-2680 construct was unstable prone to proteolytic degradation) and furthermore. it lacks an affinity tag. The instability of the pUC1960-2680 protein may be due to the presence of the a-peptide of the lacZ gene at the C terminus of the fusion protein: the presence of these sequences on a fusion protein is known to results in the production of an unstable protein. In order to determine whether soluble. stable. affinity purified fusion protein representing the pUC1960-2680 15 interval could be isolated, the following two constructs were made. The pPA1960-2680 construct contains the 1960-2680 interval of C. difficile toxin A in the pET23c vector (Novagen). The pET23 series of vectors permits the expression of inserted genes as a fusion protein containing a poly-histidine tag or tract at either the C- or N-terminus of the fusion Iprotein: the pPAI960-2680 construct expresses the C. difficile toxin A repeat region as a fusion protein containing a C-terminal poly-histidine tract. The pMA1960-2680 construct S" contains the 1960-2680 interval of C. difficile toxin A in the pMal-c vector (New England S* BioLabs) and expresses a fusion protein comprising the MBP at the N-terminus of the fusion protein.
The pPA1960-2680 construct was made as follows. A pUC1960-2680 clone in which the 2.1 kb Pstl fragment was in the opposite transcriptional orientation (relative to the direction of transcription through the LacZ sequences on the vector) was isolated using the methods described in section The C. difficile toxin A insert was excised by digestion with BamH1 and HindIlI and the insert was cloned into the pET23c vector (Novagen) digested with BamHI and HindIII as described in section a).
The pMA1960-2680 construct was created by cloning the C difficile toxin A repeat region of pPA1960-2680 as an Nhel-HindlIl fragment into the pMal-c vector cleaved with Xbal (Xbal ends are compatible with Nhel ends) and HindIII.
189 Expression of recombinant protein from these two plasmids was assessed in small scale cultures grown at 30°C. utilizing the BL21(DE3) pLysS (pPA1960-2680) or BL21pLysS (pMA1960-2680) hosts. The following conditions were varied: culture broth (2X YT, LB.
Terrific broth) and inducer levels 0.3 or 1.0 mM IPTG). All combinations of these variables were tested [except in Terrific broth. in which a single concentration (1 mM) of IPTG was tested]. The level of recombinant protein expressed upon induction and the solubility of the recombinant protein was assessed by SDS-PAGE analysis of total and soluble extracts (prepared from I ml samples as described in Example 25). All cultures were grown in 15 ml tubes (Falcon 2057): all culture medium was prewarmed overnight at the appropriate temperature, and supplemented with 100 pg/ml ampicillin and glucose to Cultures were grown in a incubator on a rotating wheel (to ensure aeration) to an OD6o of approximately 0.5-0.7 and induced with the indicated concentration of IPTG.
In all cases, high level expression of insoluble pPA1960-2680 protein was observed.
regardless of the broth or inducer concentration employed. The pMA1960-2680 protein was 15 partially soluble under all tested conditions, with maximal levels of soluble protein produced in 2X YT media at the lower inducer concentrations 0.1 and 0.3 mM IPTG).
These results demonstrate that the expression of the 1960-2680 interval of C. difficile toxin A in the pPA1960-2680 construct results in the production of insoluble recombinant protein under the conditions tested. The expression of this interval in the pMA1960-2680 20 construct permitted the expression of some soluble recombinant protein.
e) Affinity Purification and Characterization of Soluble MBP-Tagged Proteins From Constructs Expressing the 1870-2680, 1870-2190 or 1960e 2680 Intervals of C. difficile Toxin A Large scale (1 liter) cultures of the pMal-c vector vector lacking an insert), and each of the following recombinant constructs were grown, induced, and soluble protein fractions isolated: pMA1870-2190 (Example 17), pMA1960-2680 (Example 28b) and pMA1870-2680 [Example 11: Interval 6: Interval 6 contains amino acid residues 1873 through 2684 (SEQ ID NO:29) of the C difficile toxin A protein]. The large scale cultures were grown at 32°C in 2X YT broth and recombinant protein expression was induced by the addition of IPTG to 0.3 mM at ODo of 0.6. The cultures were induced for 4-5 hrs and then the cells were harvested. Soluble protein extracts were prepared and subjected to affinity 190 W^ chromatography to isolate recombinant fusion protein (Example lid). and analyzed by Coomassie staining and Western analysis as described (Example 1 b).
Briefly, soluble extracts were prepared and applied in PBS to an amylose resin (New England Biolabs) column. The column was eluted with PBS containing 10 mM maltose.
Protein yields were 40 mg per 1 liter starting volume I liter cultures) for each recombinant. Protein samples were analyzed by electrophoresis on 7.5% SDS-PAGE gels followed by'taining with Coomassie blue and Western blot analysis as described in section Protein samples were prepared for electrophoresis by mixing 1 .tl total or soluble (S) protein with 4 pi PBS and 5 pl 2X sample buffer. or 5 pl eluted protein and 5 pl 2X sample buffer or 0.5 pl eluted protein. 4.5 pl PBS and 5 pl 2X sample buffer (1/10E).
Samples of pMA1870-2680 and pPA1870-2680 (inclusion body preparations described in Example 11) were also resolved on the gel. The samples were heated to 95°C for 5 min. then cooled and loaded on a 7.5% SDS-PAGE gel. Broad range molecular weight protein markers (BioRad) were also loaded to allow estimation of the MW of identified fusion proteins.
15 After electrophoresis. protein was detected by staining the gel with Coomassie blue or the proteins were subjected to Western blotting using a goat anti-toxin A antibody (Tech Labs) as described in section a) above. The resulting gel and Western blot are shown in Figure 33.
In Figure 33. the Coomaisse blue-stained gel is shown on the left (lanes 1-10) and the Western blot is shown on the right (lanes Lanes 1-10 and l'-10' are mirror images of one another and contain the following samples: lanes I and 1' contain pMA1870-2190 lanes 2 and 2' contain pMA1870-2190 lanes 3 and 3' contain pMA1960-2680 lanes 4 and 4' contain pMA1960-2680 lanes 5 and 5' contain pMA1960-2680 lanes 6 and 6' contain pMA1960-2680 (I/10E); lanes 7 and 7' contain pMA1870-2680 lanes 8 25 and 8' contain pMA1870-2680 (1/10E); lanes 9 and 9' contain pPA1870-2680(N/C) (E) [pPAI870-2680(N/C) is described in Examples 15 and 29d]; and lanes 10 and 10' contain molecular weight markers.
The results shown in Figure 33 demonstrate: 1) That the pMA1870-2190 protein was unstable but was at least partially soluble under the growth conditions utilized. The affinity purified pMA1870-2190 preparation does however contain significant concentrations of full length fusion protein (Fig. 33. lane 2).
191 2) The pMA1960-2680 protein was partially soluble (compare lanes 3' and 4' in Fig.
33) and the integrity of the affinity purified protein (Fig. 33. lanes 5' and was comparable to that of the pMAI870-2680 preparation (Fig. 33. lane 2).
3) The full-length pMA1960-2680, pMA1870-2680 and pPA1870-2680 proteins bind the anti-C difficile toxin A antibody. while the full-length pMA1870-2190 protein does not (however. smaller breakdown products of the pMA 1870-2190 protein do bind to the antibody as shown in Fig. 33. lanes I' and This implies that either the epitopes identified by the antibody are present only in the C terminal end of the repeats. or that the antibodies recognize a conformation that cannot form with the N terminal fragment'represented in pMA1870-2190.
This observation is similar to the lack of reactivity of N-terminal fragments of the C. difficile toxin B gene (pMB1750-1970) with anti-toxin B antibody (Tech Labs) on Western blots seen in Example 19b (Figure 24).
The results shown above provide a method for the production of affinity purified recombinant C difficile toxin A protein from the 1870-2190 and 1960-2680 intervals. These results are in contrast to those obtained when using the pUC1960-2680 construct. which was prepared according to the description of Lyerly et al. [(1990) Curr. Microbiol. 21:29]. The protein expressed by the pUC1960-2680 construct was mainly insoluble and could not be affinity purified due to the absence of an affinity tag on the recombinant protein.
EXAMPLE 29 Purification of Soluble. Substantially Endotoxin-Free pPA1870-2680(N/C) Protein o• For potential utilization as a human vaccine to induce active immunity) or as an antigen in a host animal to induce protective antibodies antitoxin) for passive immunization of humans, a protein antigen should be 1) easily purified. 2) well characterized and of a high purity. 3) pyrogen poor.(when used as a human vaccine). 4) immunogenic and capable of inducing a protective immune response. In the case of the C. difficile toxin A repeat antigen. the protein must be soluble and capable of assuming a conformation which will induce a protective response. As was shown in Example 17. when pPAI870-2680(N/C) protein, which was expressed as insoluble protein inside inclusion bodies, was solubilized with SDS and then used to immunize chickens. no protective anti-toxin A antibodies were produced.
192 In this example, the recombinant C. difficile toxin A proteins were expressed and evaluated as vaccine candidates using the criteria stated above. This example involved a) evaluation of the utility of affinity purified pMA 1870-2680 protein as a vaccine antigen, b) construction, purification and evaluation of the pGA1870-2680 protein. c) development of a protocol for production of soluble pPA1870-2680. d) construction of pPA1870-2680(N) and large scale purification of N. C and N/C his-tagged 1870-2680 protein, e) construction of pPTrxAl870-2680(N) and and large scale purification of N. C and N/C his-tagged Trx 1870-2680 proteins, f) large scale affinity purification of pPA1870-2680 and pPB1750- 2360 proteins and determination of endotoxin levels and g) construction, large scale affinity purification of pPB1750-2360(N/C) and determination of endotoxin levels.
a) Evaluation of the Utility of Affinity Purified pMA1870-2680 Protein as a Vaccine Antigen Although the pMA1870-2680 protein (Example 11) was shown to be easily purified.
15 immunogenic and capable of inducing a protective response (Example 17). the ability to use this protein as a vaccine is limited by the poor purity of the affinity purified protein (see Figure 33. lanes 7' and It was estimated that only 50% of the affinity purified protein represents full-length fusion protein. The remainder of the proteins in the affinity purified preparation was found to be primarily MBP alone and contaminating E. coli proteins.
20 In order to assess whether affinity purified pMA1870-2680 protein could be used as a vaccine candidate, the endotoxin content in two independently affinity purified preparations of pMA1870-2680 protein was determined. Pyrogen content in the samples was assayed •*o utilizing the Limulus assay (LAL kit: Associates of Cape Cod) as described in Example 24d.
Both samples of affinity purified pMA1870-2680 were found to contain high levels of 25 endotoxin (>50.000 EU/mg purified recombinant protein). As seen in Examples 24a and b, high endotoxin load was determined to be a general feature of affinity purified MBP fusion proteins, or MBP alone. The above results indicate that. using current purification protocols, affinity purified MBP-C difficile toxin A fusion proteins are not suitable for use as vaccine antigens.
The pMA1870-2680 expression construct was designed to facilitate purification of the toxin A protein from the MBP tag by cleavage of the fusion protein at the engineered Factor Xa cleavage site located between the MBP and toxin A protein domains. The feasibility of obtaining substantially endotoxin-free. soluble recombinant C difficile toxin A protein by 193 purification of cleaved C difficile toxin A protein from the MBP-toxin A fusion protein was assessed. Factor Xa (New England Biolabs) was added to the affinity purified pMA1870- 2680 protein 0.2. 0.5. 1.0 and 2.5% Factor Xa/pMA1870-2680 protein ratio) in PBS containing 10 mM maltose and the mixtures were incubated for 5.5 and 20 hrs at room temperature. The extent of cleavage was assessed by Coomassie blue staining proteins after electrophoresis on SDS-PAGE gels.
The results demonstrated that some cleavage was observed in the 2.5% Factor Xa sample after 20 hrs. but cleavage was only partial. This indicates that cleavage of pMA1870- 2680 is not an efficient purification strategy to obtain soluble endotoxin-free C. difficile toxin A repeat protein using the above tested reaction conditions.
b) Construction, Purification and Evaluation of pG1870-2680 Protein In order to facilitate evaluation of the GST-containing proteins as a means of large scale production of antigens. the C difficile toxin A repeats were expressed as a fusion with 15 GST. The C difficile toxin A repeats were isolated by cleavage of pPAI 100-2680 (Example 11) with Spel followed by treatment with the Klenow fragment to fill in the ends: the DNA was then digested with Xhol. The Spel (Klenow filled)-XhoI fragment was cloned into EcoRI (Klenow filled)-Ahol cleaved pGEX3T vector (Pharmacia) to yield the pGA1870-2680 expression construct.
20 A large scale (1 liter) 2X YT culture of pGA1870-2680 [in BL21 host cells (Novagen)] was grown in 2X YT medium containing 50 pg/ml ampicillin and induced (using IPTG to 1.0 mM) for 3 hrs at 30 0 C as described in Example 28. A soluble lysate of the pGA1870-2680 large scale culture (resuspended in PBS) was prepared, and used to affinity purify soluble affinity tagged protein. The pGAl 870-2680 lysate was affinity purified on 25 Glutathione-agarose resin (Pharmacia) as described in [Smith and Corcoran. Current Protocols in Molecular Biology, Suppl. 28 (1994) pp. 16.7.1-16.7.7] with the exception that binding of protein to resin was for 1 hr at 4"C.
Briefly, following induction of the 1 liter culture for 3 hrs, the cells were collected by centrifugation for 10 min at 5.000 x g at room temperature. The cell pellet was resuspended in 10 ml ice-cold PBS. The cells were then disrupted by sonication as described in Example 24d. Triton X-100 was added to a final concentration of 1% and the sample was well mixed.
Insoluble debris was removed by centrifugation of the sample for 5 min at 10,000 x g at 4 0
C.
The supernatant was carefully removed and added to 1 ml of 50% slurry of glutathione- 194 agarose beads (Pharmacia). The mixture was allowed incubate for 1 hr at 4 0 C to allow the GST-tagged fusion protein to bind to the resin. The glutathione-agarose beads were then washed by adding 50 ml of ice-cold PBS. mixing and centrifuging for 10 sec at 500 x g at room temperature. The wash step was repeated twice (for a total of 3 washes). The resin was resuspended in 1 ml of ice-cold PBS and transferred to a 1.5 ml microcentrifuge tube.
The resin was pelleted by centrifugation for 10 sec at 500 x g at room temperature. The supernatant was removed and the fusion protein was eluted from the washed resin by adding 1 ml of 50 mM Tris-HCI (pH 8.0) and 5 mM reduced glutathione. The tube was mixed gently for 2 min then centrifuged for 10 sec at 500 x g at room temperature. The elution was repeated twice and the supernatants were pooled. The pooled supernatant. containing the eluted fusion protein, was stored in a solution containing 50 mM Tris-HCI (pH 5 mM reduced glutathione and 10% glycerol. Endotoxin content of the purified fusion protein was determined using the LAL kit as described in Example 24d.
Samples from the growth. induction and purification steps (uninduced. induced, total.
S: 15 soluble, and affinity purified elution) were resolved on SDS-PAGE gels. and proteins detected by staining with Coomassie blue (as described in Example 28). The fusion protein was found to be partially soluble most protein remained in the pellet) and approximately T mg/liter starting culture of mostly full length protein was affinity purified. The affinity purified preparation contained approximately 5000 EU/mg of affinity purified fusion protein.
These results demonstrate that under the above conditions. the pGEX expression system did not facilitate high level production of recombinant C. dfficile toxin A fusion protein, and that the recovered protein contained significant endotoxin contamination.
c) Development of a Protocol for Production of Soluble pPA1870-2680 In Example 11 it was shown that, when produced by growth at 37°C. induced pPA1870-2680 protein is almost entirely insoluble. To determine if expression at a lower temperature could enhance solubility, a culture of pPA1870-2680(N/C) was grown at and the level of soluble affinity purifiable protein determined. A soluble lysate (in Novagen IX binding buffer) from an induced I liter 2X YT culture was prepared as described below.
Briefly. a culture of pPA1870-2680(N/C) [in the BL21(DE3)pLysS host] was grown at 0 C to an OD, of 0.9 in 1 liter of 2X YT medium containing 100 pg/ml ampicillin. 34 lg/ml chloramphenicol and 0.2% glucose. Protein expression was induced by the addition of IPTG to 1 mM. After a 5 hr induction, the cells were cooled 15 min in a ice water bath and 195then centrifuged 10 min at 5.000 rpm in a JAI0 rotor (Beckman) at 4°C. The pellets were resuspended in a total volume of 40 mis Novagen IX binding buffer (5 mM imidazole. 0.5 M NaCI. 20 mM Tris-HCI. pH transferred to two 35 ml Oakridge tubes and frozen at -70 0
C
for at least 1 hr. The tubes were thawed and the cells were lysed by sonication (4 x second bursts) on ice using a Branson Sonifier 450 with a power setting of 6-7. The suspension was clarified by centrifugation for 20 min at 9,000 rpm (10.000 x g) in a JA-17 rotor. The-soluble lysate (after addition of NP40 to was batch absorbed to 7 ml of a 1:1 slurry of NiNTA resin (Qiagen): binding buffer [50 mM NaHPO,, 0.5 M NaCl. 60 mM imidazole (pH by stirring the mixture for 3 hr at 4*C. The slurry was poured into a' column having an internal diameter of 1 cm (BioRad). and washed with the following solutions in succession: 15 mis binding buffer containing 0.1%NP40. 15 ml binding buffer. 1.
ml wash buffer (40 mM imidazole. 0.5 M NaCI. 20 mM Tris-HCl. pH The bound protein was eluted in 200 mM imidazole. 0.5 M NaCI. 20 mM Tris-HCl. pH 7.9.
S* Samples of total. soluble. and eluted protein were resolved by SDS-PAGE. Total 15 protein was detected by staining the gel with Coomassie blue. The purification resulted in a yield of 34 mg of affinity purified protein from a I liter starting culture of the total soluble extract), of which at least 90-95% of the protein was found to migrated as a single band of the predicted MW (90 kd) for the recombinant C. difficile toxin A fusion protein the pPA1870-2680(N/C) protein].
These results provide a method. utilizing reduced growth temperature. that facilitates the efficient purification of high levels of soluble recombinant C difficile toxin A protein utilizing the pPA1870-2680(N/C) expression plasmid.
d) Construction of pPA1870-2680(N) and Large Scale Purification of N, C and N/C His-Tagged 1870-2680 Protein Expression plasmids that facilitated expression of the 1870-2680 interval of C difficile toxin A with either a N-terminal his-tag [pPA1870-2680 a C terminal his-tag [pPA1870- 2680(C)] or with both N- and C-terminal his-tags [pPA1870-2680(N/C)] were evaluated for large scale production and affinity purification of C. difficile toxin A repeat protein.
The features of the pPA1870-2680(C) and pPA1870-2680(N/C) expression vectors was described in Examples 11 and 15. In Example 11. pPAI870-2680(C) was termed pPA1870- 2680 and in Example 15. pPA1870-2680(N/C) was termed pPA1870-2680(H). In order to more clearly identify the location of the poly-histidine tract (his-tag) the plasmids are 196hereinafter referred to with the and suffixes. These three expression plasmids were constructed as follows.
pPA1870-2680(C) was constructed by insertion of the C difficile toxin A repeat containing Spel-HindlII fragment from pPA000-2680 (Example I la) into the pET23b vector (Novagen) cleaved with NheI and HindlII.
The pPA 870-2680(N/C) plasmid was constructed by replacement of the pPA 1870- 2680(C) promoter region. contained on a Bgll-Ndel fragment. with the corresponding Bgill- NdeI fragment from.the pET16b vector (Novagen).
The pPA1870-2680(N) vector was created by digestion of pPA1870-2680(N/C) at the C-terminal Hindlll site followed by treatment with the Klenow enzyme to fill-in the cut ends.
The blunted plasmid was then circularized by ligation to create pPA1870-2680(N).
Large scale cultures of pPA 1870-2680(N) and pPA 870-2680(C) were grown (using the BL21(DE3)pLysS host), induced and soluble protein was affinity purified and eluted as described in section c) above. In each case 10-20 mg affinity purified protein was recovered 15 and the purified protein was found to be greater than 50% full length fusion protein as estimated by SDS-PAGE analysis. However, the bulk of the pPA1860-2680(C) protein eluted in the 40 mM wash buffer. In an attempt to identify wash conditions which did not result in l the elution of significant amounts of the pPA1860-2680(C) protein, the following experiment was performed.
20 A one liter culture of pPA 870-2680(C) was grown as described above and purified utilizing a phosphate buffer based binding, wash and elution buffers. Cells were resuspended in phosphate binding buffer (50 mM NaPO,. 0.5 M NaCI. 5 mM imidazole. pH 8.0) and sequentially washed in phosphate binding buffer containing either 20. 40. or 200 mM imidazole. Recombinant protein eluted in all three washes (5.5 mg. 12.5 mg and 12 mg total protein, respectively) indicating that the C-terminal his-tagged protein is not retained by the resin at 40 mM imidazole concentrations in either buffer system utilized.
The above results demonstrated that soluble, affinity purified C. difficile toxin A protein was isolated using any of the pPA1870-2680 or expression plasmids.
197 e) Construction of pPTrxA1870-2680(N) and and Large Scale Purification of N, C and N/C His-Tagged Trx 1870-2680 Proteins The thioredoxin (Trx) expression system (Invitrogen) has been developed to facilitate soluble expression of normally insoluble or difficult to express proteins. Genes are cloned into the pTrxFus vector and expressed as fusions with the E. coli thioredoxin protein; this linkage often confers the solubility properties of thioredoxin to the fusion protein [La Vallic, et al.
(1993) Bio/Technology 11:187]. However, the pTrxFus vector has several undesirable properties for an expression vector. All plasmids must be grown in specific strains and growth media since fusion protein expression in this system is inducible by tryptophan. As well, the promoter is not stringently controlled, such that low level expression of fusion protein occurs at reduced temperatures (iLe., 30*C). Finally, the expression vector does not contain an affinity tag to facilitate high level affinity purification of soluble fusion protein.
To facilitate construction of IPTG-inducible, affinity tagged Trx fusion proteins, the 15 pETHisTrx vector was constructed. The thioredoxin gene of pTrxFus (Invitrogen) was excised as an NdeI-BamHI DNA fragment and was cloned into NdeI-BamHI digested pETHisb vector (Example 18) to created the pETHisTrx vector.
In the pETHisTrx vector, the Trx gene is expressed from the pET16b promoter and contains the pET16b N-terminal leader and his-tag sequence upstream of Trx, and the pET23b 20 polylinker (from the BamHI site) downstream of the Trx gene for construction of C-terminal genetic fusions. Three expression constructs which facilitate expression of a Trx-toxin A 1870-2680 interval fusion, as N, C or N/C terminal his-tags were constructed as follows.
The pPTrxA1870-2680(N/C) construct was constructed by ligation of the Ndel-BamHI (filled) Trx gene (isolated from the pTrxFus vector) and a SpeI (filled)-XhoI fragment S 15 containing the C difficile toxin A 1870-2680 gene [isolated from pPAl 100-2680 construct (Example 11)] into the NdeI-XhoI cleaved pETHisb vector (the filled BamHI and Spel sites blunt end ligate together and create an in-frame Trx-C difficile toxin A fusion).
The above Trx-C. difficile toxin A fusion was excised as an Ndel-HindIII fragment and inserted into NdeI-HindIII cleaved pET23a vector (Novagen) to create pPTrxA1870- 2680(C).
The HindIII site of pPTrxAl870-2680(N/C) was cleaved, filled-in by treatment with the Klenow enzyme and religatcd to create pPTrxAl870-2680(N).
198- The above constructions were carried using standard techniques of molecular biology as described in Example 29.
Large scale cultures of all three TrxA1870-2680 fusions pPTrxA1870-2680(C), pPTrxA1870-2680(N) and pPTrxAl 870-2680(N/C)] were grown and soluble affinity purified protein was isolated as described in section c) above. In all cases, affinity purified Trx fusion protein yields were similar in terms of solubility, mg/liter culture yields, and purity to the parallel pPA1870-2680 N. C. or N/C constructs described in section d) above.
i) Large Scale Affinity Purification of pPA1870-2680 and pPB1750-2360 Proteins and Determination of Endotoxin Levels Preparations of affinity purified pPA1870-2680(N/C) (Example 15) and pPB1750-2360 (Example 15b) protein were generated to determine the endotoxin levels in the purified samples. All buffers were filter sterilized and gloves were worn through the preparation of the buffers to reduce buffer-mediated endotoxin contamination of the purified recombinant S 15 protein samples. Large scale purifications of pPA1870-2680(N/C) and pPB1750-2360 proteins were performed as follows.
Briefly. cultures of pPA1870-2680(N/C) and pPB1750-2360 [in the BL21(DE3)pLysS host] was grown at 30 0 C to an ODo of 1.0 in I liter of2X YT medium containing 100 g/ml ampicillin. 34 pg/ml chloramphenicol and 0.2% glucose. Expression of the recombinant proteins was induced by the addition of IPTG to 0.3 mM. After 5-6 hrs of induction, the cells were for cooled 15 min in a ice water bath and then centrifuged 10 min at 5.000 rpm in a JAIO rotor (Beckman) at 4*C. The cell pellets were frozen at -70 0 C overnight and then thawed and resuspended in a total volume of 40 mis binding buffer (5 mM "imidazole. 0.5 M NaCI. 50 mM NaPO 4 pH 8.0) and transferred to two 35 ml Oakridge tubes. The cells lysed by sonication (8 x 20 second bursts) on ice using as described in Example 29c. The suspension was clarified by centrifugation for 30 min at 9,000 rpm (10,000 x g) in a JA-17 rotor (Beckman). The supernatant was removed (this constitutes the soluble lysate) and NP40 was added to a final concentration of The soluble lysate (after addition of NP40 to was batch absorbed to 8 ml of a 1:1 slurry of NiNTA resin (Qiagen): binding buffer by stirring for 3 hr at 4°C. The slurry was centrifuged for 1 min at 500 x g in 50 ml tube (Falcon). resuspended in 5 mls binding buffer containing 0.1% and poured into a 2.5 cm diameter column (BioRad). The resin was then washed with 20 mis binding buffer containing 0.1% NP40. The column was attached to a UV monitor (ISCO) 199and was washed with binding buffer until the baseline was established. Following establishment of the baseline absorbance. the column was washed with wash buffer [20 mM (pPB1750-2360) or 40 mM (pPA1870-2680) imidazole, 0.5 M NaCI. 50 mM NaPO,, pH until baseline was reestablished. Imidazole was removed by washing the column with 50 mM NaPO,, 0.3 M NaCI, 10% glycerol. pH 7.0. and the bound proteins were eluted in 50 mM NaPO,, 0.3 M NaCI. 10% glycerol. pH 3.5-4.0. Proteins samples from various stages in the purification process were resolved by electrophoresis on an SDS-PAGE gel:the resulting gel is shown in Figure 34.
Figure 34 shows a Coomassie blue-stained gel showing the steps of the purification.
Protein samples were prepared for electrophoresis by mixing 1 pi total or soluble or soluble protein after binding to NiNTA resin and centrifugation protein-with-4 pl PBS and 5 pd 2X SDS-PAGE sample buffer, or 5 l.1 eluted protein and 5 pl 2X sample buffer.
The samples were heated to 95°C for 5 min. then cooled and loaded onto a 7.5% SDS-PAGE gel. Broad range molecular weight protein markers BioRad) were also loaded, to allow 15 the estimation of the MW of identified fusion proteins. After electrophoresis. protein was detected generally by staining the gel with Coomassie blue. In Figure 34. lanes 1-4 contain protein from the purification of the pPA1870-2680 protein and lanes 5-8 contain protein from the purification of the pPB1750-2360 protein.
The purification resulted in a yield of approximately 30 mg/liter of affinity purified protein from 1 liter starting cultures (2-2.5 of the total soluble extract) for both proteins, of which at least 90-95% of the protein migrated as a single band of the predicted MW (90 kD) for the recombinant C. difficile toxin A protein. In both cases. most greater than 90 of the induced protein was soluble. and bound the resin quantitatively under the purification conditions utilized.
The endotoxin levels of the purified recombinant proteins was determined using the LAL kit (Example 24d) and was found to be less than 1.0 EU/mg purified protein for pPA1870-2680(N/C). and greater than 250 EU/mg purified protein for pPB1750-2360. The difference in endotoxin levels between these two purified recombinant proteins may reflect the lower stringency wash utilized during the purification of the pPB1750-2360 protein.
-200g) Construction, Large Scale Affinity Purification of pPB1750-2360(N/C) and Determination of Endotoxin Levels As shown above, the affinity purified pPB1750-2360 protein contained higher levels of endotoxin than did the purified pPA1870-2680(N/C) protein. The pPB1750-2360 protein contains a poly-histidine tract at the carboxy-terminus while pPA1870-2680(N/C) contains a poly-histidine tract at both the amino- and carboxy-termini. The presence of a poly-histidine tract at both ends of the fusion protein permitted higher stringency wash conditions to be employed during the affinity purification of pPA1870-2680(N/C) as compared to pPB1750- 2360 (40 mM imidazole versus 20 mM imidazole. respectively).
In order to produce a fusion protein comprising the 1750-2360 interval of C difficile toxin B containing poly-histidine tracts at both the amino- and carboxy-termini-pPB1750- 2360(N/C) was constructed as follows. pPB1750-2360 (Example 15b) was digested with NdeI and Xhol and the 1.5 kb Ndel/Xhol fragment was isolated and inserted into pETHisb vector (Example 18) digested with Ndel and Xhol. Routine procedures were employed for this 15 construction as described in the preceding Examples.
Large scale purification of pPB 750-2360(N/C) was conducted as described above in section f) for the purification of pPB1750-2360 with the exception that the wash buffer contained 40 mM imidazole. 0.5 M NaCI. 50 mM NaPO 4 pH 8.0. Following the wash step, imidazole was removed by washing the column with 50 mM NaPO 4 0.3 M NaCI. glycerol. pH 7.0. The column was then washed with 50 mM NaPO 4 M NaCl. glycerol. pH 3.0 in an attempt to elute the bound protein. No pPB1750-2360(N/C) was eluted under these conditions.
The large scale purification was then repeated as described above with the exception that following the wash step using the wash buffer containing 40 mM imidazole. 0.5 M NaCI, 50 mM NaPO 4 pH 8.0. the bound protein was eluted using a solution containing 200 mM imidazole, 0.5 M NaCI. 50 mM NaPO 4 pH 8.0. The imidazole was removed from the eluted protein by dialysis against PBS.
Analysis of the eluted pPB1750-2360(N/C) on SDS-PAGE gels stained with Coomassie blue revealed a single band of the MW expected for the full-length fusion protein.
The endotoxin levels of the purified pPB1750-2360(N/C) protein was determined using the LAL kit (Example 24d). Three separate determinations were conducted and the endotoxin level was found to be 80. 300 or 450 EU/mg of purified recombinant protein. While not limited to any particular mechanism, it is believed that the inconsistent LAL assay results seen 201 with pPB1750-2360(N/C) and the high reading obtained with pPB1750-2360 (see section f) are due to non-specific agglutination of the LAL components by carbohydrate binding moieties present on the C. dificile toxin B sequences present on these proteins. Regardless of whether the actual endotoxin level is 80 or 450 EU/mg purified protein, the affinity purified pPB1750-2360(N/C) preparation represents a substantially endotoxin-free preparation of recombinant protein (Administration of 10 to 500 pg of purified pPB1750-2360(N/C) would result in the introduction of only 4.5 to 225 EU: in a 70 kg human this amount of endotoxin is 1.3 to 64.5% of the maximum permissible dose).
The above results provide a protocol for the affinity purification of substantially endotoxin-free preparations of recombinant C difficile toxin A and B repeat proteins in high yields.
Example o* Purification of Soluble pPA1870-2680(N/C). pPA1960-2680 and pPA1870-2190 Proteins In Example 29. methods for the production of soluble. substantially endotoxin-free samples of pPA1870-2680(N), or were provided which produced proteins that met the initial criteria set for antigen production, that is the proteins were 1) easily purified 2) well characterized and of a high purity and 3) substantially endotoxin-free. In this example.
the ability to produce similarly pure antigen from the pPA1870-2190 or pPA1960-2680 expression constructs was examined. This example involved a) large scale purification of soluble pPA1870-2190 and pPA1960-2680 proteins and b) construction of the pPTrxA1870- 2190 vector and large scale purification of soluble pPTrxA1870-2190 protein.
oa a) Large Scale Purification of Soluble pPA1870-2190 and pPA1960-2680 Proteins Previous attempts to produce soluble affinity purified protein utilizing the pPA1870- 2190 (Example 17a) or pPA1960-2680 (Example 28) vectors were unsuccessful. as assessed by analysis of total and soluble protein produced in small scale cultures. However. the solubility properties of a protein determined utilizing small scale or minicultures may not translate to large scale cultures. due to differences in buffers. sonication conditions. etc.
Indeed, the successful expression of soluble, substantially endotoxin-free C. difficile toxin A repeat protein utilizing the pPA1870-2680 N. C or N/C constructs suggested that the -202 conditions utilized to solubilize these proteins might also enhance solubility of the pPA1870- 2190 and pPA1960-2680 proteins. This hypothesis was tested as follows.
Large scale cultures of pPA1870-2190 and pPA1960-2680 were grown and soluble protein affinity purified on Ni-NTA resin as described in Example 29c. Both the BL21(DE3) and BL21(DE3)pLysS hosts for pPA1960-2680. and the BL21(DE3)pLysS host for pPA1870- 2190 were utilized. The culture of pPA 870-2680(N/C) [in the BL21(DE3)pLysS host] was grown at 30 0 C to an OD, of 0.9 in 1 liter of 2X YT medium containing 100 pg/ml ampicillin and 0.2% glucose: when the host utilized harbored the pLysS plasmid. 34 pg/ml chloramphenicol was added to the above medium. Protein expression was induced by addition of IPTG to I mM. After 5 hrs of induction, the cells were cooled for 15 min in a ice water bath and then centrifuged for 10 min at 5.000 rpm in a JAIO rotor (Beckman) at 4C. The pellets were resuspended in a total volume of 40 mls Novagen IX binding buffer mM imidazole. 0.5 M NaCI. 20 mM Tris-HCI. pH transferred to two 35 ml Oakridge tubes and frozen at -70 0 C for at least 1 hr. The tubes were thawed and the cells were lysed 15 by sonication (4 x 20 second bursts using a Branson Sonifer 450 with a power setting of 6-7) on ice. The suspension was clarified by centrifugation for 20 min at 9.000 rpm (10,000 x g) in a JA-17 rotor (Beckman) at 4*C. The soluble lysate (after addition of NP40 to was batch absorbed to 7 ml of a 1:1 slurry of NiNTA resin (Qiagen): Novagen IX binding buffer by stirring for 3 hr at 4"C. The slurry was poured into a 1 cm internal diameter column (BioRad). and washed with the following solutions in succession: 15 mis Novagen IX binding buffer containing 0.1%NP40. 15 mi Novagen IX binding buffer. 15 ml wash buffer (40 mM imidazole. 0.5 M NaCI. 20 mM Tris-HCl, pH The bound protein was eluted in 200 mM imidazole. 0.5 M NaCI, 20 mM Tris-HCI. pH 7.9.
Samples of total, soluble, and eluted protein (both the 40 mM and 200 mM wash and elution buffers) were resolved by SDS-PAGE. Total protein was detected by Coomassie staining, and C. difficile toxin A-reactive protein (in the case of pPA1960-2680) detected by Western blot detection, utilizing affinity purified goat anti-toxin A antibody as described in Example 28.
The results of these analyses showed that for the pPA1870-2190 protein, only 600 Vg protein/liter culture was purified in the 200 mM imidazole elution. The C. difficile toxin A protein was expressed to high levels with this construct. but most of the induced protein was insoluble. As well. the pPA1870-2190 protein represented less than 10% of the total eluted protein. For the pPA1960-2680 construct, total yields of soluble affinity purified protein was 203 either I mg [Bl21(DE3)pLysS host] or 200 ipg [BL21(DE3) host] in the 200 mM elution fraction. Coomassie and Western analysis demonstrated that the pPA1960-2680 protein was expressed to high levels, but that most of the induced protein was insoluble, and that eluted protein preparations contained only approximately 20% C. difficile toxin A-reactive protein.
The above results demonstrate that the conditions utilized to solubilize the pPA1870- 2680 protein were not successful in generating solubilized C difficile toxin A repeat protein expressed by either the pPA1960-2680 or pPA1870-2190 constructs.
b) Construction of the pPTrxA1870-2190 Plasmid and Large Scale Purification of Soluble Protein To determine if the solubility of recombinant proteins comprising 1870-2680 interval of C difficile toxin A could be enhanced by utilizing the solubilizing properties of the Trx protein, a fusion construct in which the 1870-2680 interval was expressed as a fusion to thioredoxin (Trx) was constructed.
15 The pPTrxA1870-2190 construct was made in two steps. First. the 1870-2190 interval was cloned into the pTrxFus vector (Invitrogen). This was accomplished by ligating the Kpnl-SalI fragment from pMA1870-2190 which contains the 1870-2190 interval of C. difficile toxin A into the KpnI-SalI cleaved pTrxFus vector. A recombinant clone containing the appropriate DNA fragments was selected and the sequences encoding the Trx-C difficile toxin A fusion protein were excised utilizing Ndel and Sall. and cloned into the pETHisb vector (Example 18) cleaved with Ndel and Xhol. The resultant construct. pPTrxA1870-2190.
*OOQO
contains an N-terminal his-tagged Trx-C difficile toxin A fusiottdriven by the pET16b Spromoter.
Purification of soluble affinity purified Trx-C difficile toxin A protein from the pPTrxA1870-2190 construct was performed from a large scale culture as described in section a) above. Total. soluble and elution samples were resolved on a 12.5% SDS-PAGE gel and protein was detected by staining with Coomassie blue.
The results of this analysis revealed that the total yield of affinity purified recombinant protein was 2 mg of greater than 50% pure protein in the 200 mM imidazole elution. This yield of I mg specific protein (50% of 2 mg total purified protein) represents a ten fold increase over the yield obtained with the pPA1870-2190 construct (10% of 600 pg, or less than 100 pg specific protein) and demonstrates the solubilizing property of the Trx protein.
However, the majority of induced protein was insoluble with both constructs -204 pPTrxAl 870-2190 and pPA1870-2190) and the overall affinity purifiable protein yield with the pPTrxA1870-2190 vector was still less than 20 fold lower that obtained with the pPA1870-2680 constructs.
EXAMPLE 31 Protection of Hamsters Against C. difficile Disease with Avian Antibodies (IgY) Against Recombinant C difficile Toxin A and Toxin B In this example. experiments were performed to determine if orally administered IgY against a recombinant fragment of C. difficile toxin A and/or recombinant C. difficile toxin B can effectively prevent hamsters against C. difficile disease. This example involved a) the immunization of hens with recombinant C difficile toxin A or B protein. b) purification.
detection and quantification of anti-recombinant C. difficile toxin A and toxin B IgY and c) in vivo protection infection study using either anti-recombinant C difficile toxin A IgY or a 15 mixture of anti-recombinant C. difficile toxin A IgY and anti-recombinant C difficile toxin B IgY.
a) Immunization of Hens with Recombinant C difficile Toxin A or B Proteins Egg-laying Leghorn hens were each immunized with C difficile toxin A recombinant protein pMA1870-2680 (the 1870-2680 interval of C difficile toxin A is referred to as Interval A-6) or C difficile toxin B recombinant pPB1750-2360 (the 1750-2360 interval of C difficile toxin B is referred to as Interval Both recombinant proteins were expressed as soluble products and purified as described in Example 28 (pMA 1870-2680) and Example 29 (pPB1750-2360). About I mg of each recombinant protein was mixed with complete Freund's adjuvant (prepared as described in Example I) and subcutaneously administered to the hens at multiple sites. The hens were immunized ten times. The first four immunizations were given on Day 1. 14. 21 and 35. The remaining immunizations were then given at 4 week intervals.
b) Purification, Detection and Quantification of Anti-Recombinant
C.
difficile Toxin A and Toxin B IgY Eggs were collected about I week after the last boost and IgYs were extracted using PEG as described in Example 1. The anti-recombinant C difficile toxin A and B IgYs were resuspended as a 4X PEG concentrate resuspended in 1/4 of the original yolk volume) in -205 0.1 M carbonate buffer. pH 9.5. The total protein concentration of both of the 4X IgY concentrates was 20 mg/ml as judged by absorbance at 280 nm. The relative levels of IgY specific for reactivity with the immunogens were detected by ELISA as follows.
Microtiter plates were coated at 100 il/well with either 0.05 .g/ml of the recombinant S C. difficile toxin A protein. pPAI870-2680 (Example 11) or 1 g/iml of the recombinant C.
difficile toxin B. pPB1750-2360 (Example 18b). The ELISA was performed as described in Example 13c. The results of this analysis revealed that the antibody titers were both greater than 1:125.000. (Antibody titer is defined as the reciprocal of the highest antibody dilution that gives an ELISA signal that is at least 3-fold over pre-immune IgY.) The amount of specific anti-recombinant toxin A and anti-recombinant toxin B IgY was determined by affinity purification as described in Example 15c. The amount of specific-anti-recombinant C.
difficile toxin A and B antibodies present in the anti-pMA1870-2680 and anti-pPB1750-2360 preparations was determined to be about 160 pg/ml and 200 g/iml. respectively.
15 c) In Vivo Protection Infection Study Using Either Anti-Recombinant C difficile Toxin A IgY or a Mixture of Anti-Recombinant C difficile Toxin A IgY and Anti-Recombinant C difficile Toxin B IgY An in vivo protection study using antibodies raised against pMA1870-2680 (Example 15) and pPB1750-2360 (Example 18b) was performed using the C difficile-hamster model.
20 This study employed a hamster model which was modified from that used in Example 9. as follows.
Hamsters were predisposed to infection with C difficile by I.P. administration of 1 mg/100 gm body weight of Clindamycin phosphate (Biomol) in I ml of sterile water. The Clindamycin was administered I.P. using a 1 ml tuberculin syringe (Terumo). About 20-24 hours later, the hamsters were each infected orally with 1 ml of saline containing I x 104 C.
difficile (ATCC 43596). The C. difficile was grown for about 48 hours on CCFA (C difficile selective agar) plates (BBL) prior to infection.
Using the above modifications in the hamster model. the time course of infection (in particular. the time of onset of disease) in the hamsters was much more consistent and rapid as compared to the results obtained using the conditions described in Example 9. For the present study, 3 groups of hamsters (Sasco). 8 per group were treated with 2 mis of a 4X concentrate of preimmune or anti-recombinant C. difficile toxin A IgY containing 40 mg of total IgY: the amount of specific anti-recombinant C difficile toxin A was approximately 400 -206 pg. The third group was treated with 2 mis of an equal mixture of 4X concentration of IgYs to both recombinant C difficile toxin A and B giving a final specific concentration to each of 2X (the amount of specific anti-recombinant toxin A and B IgY was approximately 200 pg each). The third group. therefore has one-half the amount of specific antibodies to the recombinant C difficile toxin A compared to the anti-recombinant C. difficile toxin A only treatment.
Hamsters were treated 3 times daily at roughly 4 hour intervals starting 24-hours prior to infection. The hamsters were treated for 5 days. This was about I week less than the treatment period employed in Example 9. The outcome of the present prophylactic treatment study is shown in Figure In Figure 35. the percentage cumulative mortality is displayed along-the-erdinate and the time (in days) is displayed along the abscissa. The treatment period is indicated by the use of the bar between days 0 and 4. The administration of Clindamycin and the inoculation with C. difficile (marked as "Infection" in Fig. 35) is indicated. The solid black squares 15 represent hamsters which received pre-immune IgY: the open squares represent hamsters which received anti-recombinant C difficile toxin A IgY (anti-Interval A-6) and the solid black diamonds represent hamsters which received a mixture of anti-recombinant C dfficile toxins A and B IgY (anti-Interval A-6/B-3).
The results shown in Figure 35 demonstrate that under these model conditions, all of 20 the hamsters treated with pre-immune IgY developed diarrhea less than 24-hours post inoculation. One day post inoculation all of the animals were dead in that group. In contrast.
using the conditions employed in Example 9. the group treated with pre-immune IgY took several days before the onset of illness was apparent and often not all of the members died from the disease.
As shown in Figure 35. the hamsters treated with either the anti-recombinant C.
difficile toxin A IgY (anti-pMA1870-2680) or anti-recombinant C dificile toxin A (antipMA1870-2680) and toxin B (anti-pPB1750-2360) mixture were protected from death; 62 and 88 survived from each group. respectively. Chi-squared analysis of the results in the anti-recombinant C difficile toxin A and the mixture treated groups was significant compared to the pre-immune treated group. with P values of less than 0.05 and less than 0.005, respectively. Although the results comparing death as an endpoint between two test groups was not significant (P 0.75). diarrhea in the animals receiving the anti-recombinant C.
207 difficile toxin A and B IgY mix was less severe than that seen in the pre-immune control group.
The above results. using a highly aggressive hamster model of CDAD, show that IgYs against a recombinant C difficile toxin A protein (pMA1870-2680) was protective, but the addition of antibodies against the recombinant C difficile toxin B (pPB1750-2360) provided additional protection a lessening of the severity of the disease symptoms).
EXAMPLE 32 Treatment of Hamsters with an Established C difficile Infection with Avian Antibodies (IgY) Against Recombinant C difficile Toxin A and Toxin B In order to determine if orally administered IgY against a recombinant C difficile toxin A protein and /or recombinant C difficile toxin B can effectively treat hamsters infected with C difficile. the following experiments were performed. The example involved a) the 15 immunization of hens with recombinant C. difficile toxin A or B proteins b) purification and detection of anti-recombinant C difficile toxin A and B chicken IgYs c) an in vivo infection study where hfamsters were treated with IgYs against either recombinant C difficile toxin A or recombinant toxin B (Infection study In addition. a mixture of IgY. containing both antirecombinant toxin A and B was also used to treat hamsters after infection with C. difficile 20 (Infection study The conditions used in infection study #2 were repeated to yield Infection study #3.
a) Immunization of Hens with Recombinant C. difficile Toxin A or B proteins Egg-laying Leghorn hens were each immunized with the recombinant C difficile toxin A recombinant protein pMA1870-2680 (Interval A-6) or the C difficile toxin B recombinant pPB1750-236Q (Interval Each recombinant comprises the repeat regions of C difficile toxin A and toxin B. Both recombinant proteins were expressed as soluble proteins utilizing the pMal vector for the toxin A recombinant (Example 15) and pET for the toxin B recombinant (Example 18b).
About 1 mg of each recombinant protein was mixed with 500 jg of Fowl adjuvant (RIBI Immunochemical Research) for the C. difficile toxin A recombinant and or Freund's adjuvant (prepared as described in Example 1) for the C dfficile toxin B recombinant. Each hen was subcutaneously immunized about 7 times at roughly two to four week intervals.
-208 b) Purification and Detection of Anti-Recombinant C difficile Toxin A and B Chicken IgYs Eggs were collected about I week after the last boost and antibodies were extracted using PEG as described (Example The IgYs were resuspended as a 8X or 4X concentrate resuspension at 1/8 or 1/4 yolk volume in 0.1 M carbonate buffer. pH The relative levels of specific antibodies to the recombinant immunogens was detected by ELISA as described in Example 13c with the following modifications. The 96-well microtiter plate was coated with 0.05 tg/ml of recombinant toxin A protein pPTrxA1870-2680N/C (Example 29e) or 1 pg/ml of toxin B recombinant pPB1750-2360 (Example 18b) at 100 l/well. The 10 standard ELISA format to detect anti-recombinant C difficile toxin A or B was performed (Example 13c). Antibody titers by ELISA were both determined to be greater than 1:125.000.
*.o c) In vivo Infection Study Three infection studies. #2 and #3 were performed using the hamster model described in Example 31.
i) Infection Study #1 In the infection study three separate experimental groups, each consisting of 12 Golden Syrian hamsters (Sasco) weighing approximately 80-90 grams each were used. The 20 animals were housed at 3 per cage and were offered food and water ad libitum throughout the study. The hamster model was conducted as described in Example 31. At the start of the study. each hamster was predisposed to infection by the intra-peritoneal administration of Clindamycin-phosphate (Biomol) at I mg/100 gm body weight in 1 ml of water using a I ml tuberculin syringe (27 gauge needle). Approximately 24 hours later, each animal was orally challenged, using an 18 gauge feeding needle. with 1 ml of C difficile. (strain ATCC 43596) with approximately 10 3 to 10' organisms in sterile saline. The organisms were grown for 48 hours on CCFA plates (BBL) prior to infection.
Three hours after inoculation (Day treatment was initiated for both groups. The groups were each orally treated using an 18 gauge feeding needle to administer 2 mls of a 4X concentrate of either pre-immune IgY or specific immune IgY against either the recombinant C difficile toxin A (pMA1870-2680: Interval A-6) or toxin B (pPB1750-2360: Interval B-3).
On Day 1, the hamsters were treated additionally two more times at 2 hour intervals. On Day 2. through 4 the hamsters were each treated with 2 mis of the respective antibody preparations 209 3 times daily roughly at 4 hour intervals. Each 2 ml dose contained about 40 mg of IgY of which about 400 pig is specific IgY (determined by affinity purification as described in Example 15c) to the recombinant toxin protein or about 1200 pg of specific anti-C. difficile toxin protein per day. All animals were observed for the onset of diarrhea and death during and after the treatment period. The results are shown in Figure 36.
In Figure 36. the percentage cumulative mortality is displayed along the ordinate and the time (in days) is displayed along the abscissa. The treatment period is indicated by the use of the bar between days 1 and 4. The administration of Clindamycin and C. difficile organisms ("Infection") is indicated. The solid black squares represent hamsters which received pre-immune IgY: the open squares represent hamsters which received a 4X preparation of anti-recombinant C difficile toxin A IgY (anti-Interval A-6)-and the-solid black diamonds represent hamsters which received a 4X preparation of anti-recombinant C difficile toxin B IgY (anti-Interval B-3).
S The results shown in Figure 36 demonstrate that half of the hamsters (6/12) treated after infection with antibodies against the C difficile toxin A recombinant were protected from death from CDAD. The degree of protection in the anti-recombinant C difficile toxin A group was statistically significant at P< 0.025 using Chi-square analysis. Most of the hamsters (10/12) in that group presented with diarrhea. It appeared that at the concentration tested. antibodies against the C difficile toxin A recombinant was unable to prevent diarrhea 20 in the hamsters. In contrast. all of the pre-immune and anti-recombinant C difficile toxin B
F
treated hamsters developed diarrhea and died shortly afterward.
The above results demonstrated that IgYs raised against a recombinant C. difficile toxin A protein (pMA1870-2680) can protect the hamsters from death due to CDAD.
ii) Infection Study #2 A second experiment was conducted basically as described above with the exception that a mixture of antibodies to both recombinant C. difficile toxins A and B was tested for the ability to protect hamsters from CDAD. Equal volumes of an 8X concentration of IgYs to both recombinants (pMA1870-2680 and pPB 1750-2360) were mixed to give a final concentration to each recombinant equal to 4X. Each dose (2 ml) contained approximately mg/ml protein containing about 400 pug of specific IgY specific anti-C difficile toxin protein as compared to the total) to each recombinant. The amount of specific antirecombinant IgY to each toxin recombinant was determined by affinity purification using the respective recombinant protein. The resulting preparation therefore contains the same final 210concentration of anti-recombinant toxin A used in the previous experiment (section c(i) above) except it contains twice the amount of protein. Because of this difference, an additional test group was set-up and treated with equal volumes of two 8X concentration of anti-recombinant C difficile toxin A and pre-immune IgY. As a control, a third group of hamsters were treated with an 8X concentrate of only pre-immune IgY. Nine hamsters per group were infected with I x 10" C difficile organisms (ATCC 43596) and then were treated 4 hours later with 2 mis of either preimmune IgY, anti-recombinant C difficile toxin A IgY mixed with preimmune IgY or a mixture of anti-recombinant C difficile toxin A and B IgYs. The animals were treated as described (section c(i) above) at 3 times a day for 4 days. The outcome of this experiment is shown in Figure 37.
In Figure 37. the percentage cumulative mortality is displayed along the ordinate and the time (in days) is displayed along the abscissa. The treatment period is indicated by the use of the bar between days I and 4. The administration of Clindamycin and C difficile organisms ("Infection") is indicated. The solid black squares represent hamsters which received an 8X preparation of pre-immune IgY: the open squares represent hamsters which received a mixture of 8X preparations of pre-immune sera and anti-recombinant C. difficile toxin A IgY (anti-Interval A-6) and the solid black diamonds represent hamsters which received a mixture of 8X preparations of anti-recombinant C difficile toxins A and B IgY (anti-Interval A-6 and B-3).
20 The results shown in Figure 37 demonstrate that a mixture of IgYs to both recombinant C. difficile toxin A and B (pMA1870-2680 and pPB1750-2360) completely protected all the hamsters from death from CDAD. Only 1/3 (3 out of 9) of the animals treated with the mixture of anti C. difficile toxin A and B antibodies exhibited diarrhea (one had a very mild case). Hamsters treated with a mixture of anti-recombinant C difficile toxin A antibodies (anti-Interval A-6) and pre-immune IgY were partially protected with a 56 survival rate. All except one hamster in the anti-Interval A-6/pre-immune IgY group presented with diarrhea. The survival rate in this group, was comparable to the rate seen in infection study #1 (50 using only anti-recombinant C. difficile toxin A IgY without the addition of pre-immune IgY. This indicated that the addition of preimmune IgY probably had little or no effect (in terms of non-specific protection from proteases in the GI tract) on the effectiveness of the anti-recombinant C. difficile toxin A IgY. As usual. treatment of animals with pre-immune antibodies alone did not protect the hamsters from C. difficile infection and all the hamsters died within 2 days post-infection. The survival rates seen due to -211 administration of the anti-recombinant C difficile toxin A IgY and the anti-recombinant C.
difficile toxins A and B were both statistically significant compared to pre-immune IgY with P values of less than 0.05 and 0.001. respectively. The P-value comparing both recombinant treated groups was less than 0.10.
The survivors in both infection studies #1 and #2 survived lived long-term for a period of greater than or equal to 30 days after withdrawal of treatment: animals were euthanized about one month after withdrawal of treatment when the experiment was terminated). Furthermore. no relapse was observed in these animals (relapse is commonly seen in animals. including humans. treated with drugs such as vancomycin or metronidazole to combat C. difficile infection). These results represent the first time antibodies raised against recombinants proteins derived from C. difficile toxins A and B have been shown to be completely effective in animals given a lethal infection with C difficile.
iii) Infection Study #3 15 After several more immunizations of the hens with the recombinant C difficile toxin A alone (pMA1870-2680) and C. difficile toxin A/B recombinants (a mixture of pMA1870-2680 and pPB1750-2360), the in vivo therapeutic study described above (infection study using the mixture of both antibodies was repeated (infection study Three groups of hamsters, each group consisting of 10 members were treated 4 hours post-infection with either pre- 20 immune IgY. anti-recombinant C. difficile toxin A or a mixture of anti-recombinant C difficile toxin A and B IgYs at the same dosages and times outlined above. The results of this study is shown in Figure 38.
In Figure 38. the percentage cumulative mortality is displayed along the ordinate and the time (in days) is displayed along the abscissa. The treatment period is indicated by the use of the bar between days I and 4. The administration of Clindamycin and C. difficile organisms ("Infection") is indicated. The solid black squares represent hamsters which received an 8X preparation of pre-immune IgY: the open squares represent hamsters which received a mixture of 8X preparations of pre-immune sera and anti-recombinant C difficile toxin A IgY (anti-Interval A-6) and the solid black diamonds represent hamsters which received a mixture of 8X preparations of anti-recombinant C. difficile toxins A and B IgY (anti-Interval A-6 and B-3).
As shown in Figure 38. the hamsters treated with the antibody mixture to both recombinant C. difficile toxins A and B were completely protected from death as shown in the 212 previous experiment (infection study but in addition none of the treated (anti-recombinant toxins A and B) animals presented with diarrhea. While hamsters treated with antirecombinant C difficile toxin A were also protected from mortality (only one of ten died) all but one had diarrhea. All hamsters treated with preimmune IgY developed diarrhea and died within 48-hours of infection.
Prevention against mortality using antibodies to recombinant C dfficile toxin A and both C. difficile toxins A and B was statistically significant (P <0.001). compared to the results obtained using pre-immune antibody. Also. was shown in previous Examples (16 and sections i and ii above), all the treated hamsters survived long-term with no signs of relapse.
The prevention of morbidity in the hamsters. which includes presence of diarrhea and weight loss, by treating with anti-recombinant A and B IgY is shown in Table 42.
Table 42 Interval A-6 and B-3 Antibodies Reduce CDAD Morbidity .5 Animals with P Weight Loss P Treatment Group Diarrhea Pre-Immune 100 NAb pmA1870-2 6 8 0 90 NSc 16 <0.001 pmAl870- 2 6 80 plus 0 <0.00 1 NS pPB 1750-2360 (A-6/B-3) Weight loss of survivors calculated as the difference between the starting weight and that at termination of treatment.
h NA. not applicable.
c NS. not significant.
As shown in Table 42. the percent weight loss in the survivors treated with the antirecombinant C. difficile toxin A IgY alone (pMA1870-26 8 0: A-6) compared to the mean weight before infection was about 16%. The hamsters treated with both antibodies to both recombinants (pMAl870-2680 and pPB1750-2360: A-6/B-3) only lost 1% of their mean starting weight. These results demonstrate that the antibodies raised against the C. difficile toxin A recombinant protected the hamsters from the fatal stage of CDAD but the addition of antibodies to the C. difficile toxin B recombinant was necessary for the prevention of the diarrheal stage associated with CDAD.
-213 EXAMPLE 33 Relapse During In Vivo Treatment of Hamsters Infected with C. difficile Using Vancomvcin Therapy To determine if relapse of C. difficile disease occurs after vancomycin treatment under conditions used in the previous treatment studies, the following experiment was performed.
The conditions employed for the hamster model were identical to the conditions used in Example 32. Three groups of hamsters (Sasco). each group containing 6 members, were treated with 0.2. 1 or 5 mg/kg of vancomycin (Vancomycin HCI. Lilly) in one ml of sterile water. Animals were dosed once per day for 5 days. An additional untreated group was tested as a control. Hamsters were each orally infected with 1 x 103 C difficile organisms (ATCC 43596) and then vancomycin treatment was begun 3 hours post-infection. The outcome of the experiment twenty days after infection, is shown in Figure 39.
In Figure 39. the percentage cumulative mortality is displayed along the ordinate and 15 the time (in days) is displayed along the abscissa. The treatment period is indicated by the use of the bar between days I and 5. The administration of Clindamycin and the inoculation with C difflcile organisms (marked as "Infection" in Fig. 39) is indicated. The solid black squares represent hamsters which received no treatment (untreated): the open squares represent hamsters which received 0.2 mg/kg vancomycin: the solid black diamonds represent hamsters which received 1.0 mg/kg vancomycin: and the open diamonds represent hamsters which received 5.0 mg/kg vancomycin.
.o* The results shown in Figure 39 demonstrate that the hamsters treated with 0.2 mg/kg of vancomycin all died during the course of treatment. Hamsters treated with 1 mg/kg or mg/kg of vancomycin were protected during the period of treatment, but quickly relapsed and most died shortly after the termination of treatment. All of the treated hamsters developed diarrhea and 83% of the hamsters treated with 1 mg/kg vancomycin or 100% of the hamsters treated with 5 mg/kg vancomycin died 7 days or 9 days after withdrawal of treatment.
This relapse effect using vancomycin as illustrated here or using metronidazole to treat C difficile infections in the hamster model or in humans is a common occurrence that has been reported frequently. Up to 100% of hamsters and about 25% of humans treated with either of these two drugs relapse. This relapse effect is in marked contrast to the effect shown in the present invention when treating hamsters infected with C. difficile with IgYs raised against either native or recombinant C difficile toxin A or B. Relapse rarely or never -214 occurs when animals are treated with anti-C difficile toxin IgY. Thus, the prevention of relapse by the administration of anti-C. difficile toxin IgY represents an important therapeutic advantage over conventional antibiotic treatments.
EXAMPLE 34 Comparison of C diffcile Toxin A Neutralization In Vivo Using IgYs Against Three Different C. dificile Toxin A Repeat-Containing Recombinant Proteins Three C difficile toxin A recombinants proteins from the repeat region of C. difficile toxin A were expressed in the pMal-c vector. Antibodies against each were generated and compared for their ability to neutralize C difficile toxin A in hamsters. The.example involved a) immunization of hens. b) purification and detection of anti-recombinant toxin A IgYs and c) C difficile toxin A neutralization study in hamsters using anti-recombinant toxin A IgYs.
a) Immunization of Hens Three groups of egg-laying Leghorn hens were immunized with different toxin A recombinants proteins produced in.the pMal vector. All were expressed as MBP fusions.
They were pMA1870-2190 (Example 17), pMA1960-26 8 0 (Example 28) and pMAI870-26 8 0 (Example 11). The first two recombinants proteins comprise overlapping sub-fragments within the interval contained in the recombinant pMA 1870-2680.
Approximately I mg of each recombinant protein was given with Freund's adjuvant to each hen. The standard immunization procedure using this adjuvant was performed as .described Example 1. The hens were immunized four times at multiple sites using the time intervals described in Example 32a.
b) Purification and Detection of Anti-Recombinant C difficile Toxin A IgYs Antibodies were extracted using PEG from eggs collected after at least one week after the last boost. Anti-C difficile toxin A (CTA) and pre-immune IgYs were also prepared as a controls (as described in Examples and 1. respectively). The IgYs were resuspended in 0.1 M carbonate buffer (pH 9.5) at 4X concentration (1/4 the original yolk volume). The levels of specific antibodies from each group was determined by ELISA. Reactivity was determined against the soluble recombinant toxin A protein pPTrxl870- 2 68 0. The pPTrxl870-2680 215protein does not contain the MBP as do the other 3 immunogens and therefore the ELISA reactivity is specific to only the toxin A recombinant. The standard ELISA protocol was employed (Example 13c). From the ELISA results. all four of the anti-recombinant C.
difficile toxin A IgYs were shown to have very similar titers at greater than 1:31.250 compared to the pre-immune IgY.
c) C difficile Toxin A Neutralization Study in Hamsters Using Anti- Recombinant Toxin A IgYs The ability of the above recombinant toxin A IgYs pMA1870-2190. pMA1960- 2680 and pMA1870-2680) to provide protection against C. difficile toxin A was determined in the hamster model. Two groups of hamsters received the anti-pMA1870-2680 IgYs: therefore a total of 6 treatment groups were examined. The assay was performed as described in Example 14.
One ml of IgY was mixed and preincubated for 1 hour with 30 pg of C. difficile toxin 15 A (Tech Labs) then administered orally to 30-40 gm Golden Syrian hamsters (Charles River).
Preimmune and CTA IgY (Example 8) served as negative and positive controls, respectively.
The animals were observed for 24 hours and the number dead in each group was tallied. The results of the experiment is shown in Table 43.
Table 43 Generation of Toxin A Neutralizing Antibodies Against Different Toxin A Recombinant Fragments 25 Treatment Group Alive' Dead' Preimmune 0 CTA 5 0 pMA 1870-2190 0 pMA 1960-2680 5 0 pMA 1870-2680 a 5 0 pMA 1870-2680 b 32 Study outcome after 24 hours.
-216- As shown in Table 43. pre-treatment of C. difficile toxin A with IgY against pMA1870-2 6 80 prevented death in all 5 treated hamsters in the treatment group designated "pMAI870-2680 a" and 3 out of 5 in the treatment group designated "pMAl870-2680 b." Antibodies raised against pMA1870-2680 and the slightly smaller. carboxy-terminal polypeptide. pMA1960-2680. both prevented death in all 5 animals. In contrast, as with preimmune IgY, IgYs raised against the amino-terminal polypeptide pMA1870-2190 had no effect on the prevention of death. As expected. hamsters treated with CTA IgYs were completely protected from the enterotoxic effects of C difficile toxin A.
EXAMPLE Identification of Adjuvants that Optimally Induce Neutralizing Antibodies Against Native C difficile Toxin A In Vivo In order to compare the ability of different adjuvants to invoke neutralizing antibodies 15 against C. difficile toxin A in hens using a recombinant C. difficile toxin A protein as the immunogen. the following experiments were performed. The example involved a) immunization of hens with a recombinant C. difficile toxin A protein using four different adjuvants: b) determination of anti-recombinant C difficile toxin A IgY titers by ELISA and c) testing the neutralizing ability of the anti-recombinant C. difficile toxin A IgYs against C difficile toxin A in vivo.
a) Immunization of Hens with a Recombinant C difficile Toxin A Protein Using Four Different Adjuvants Eight groups of egg-laying Leghorn hens, each group containing 4 hens. were i 25 immunized with either 100 pg or 1 mg of recombinant toxin A protein (pMA1870-2680; Example 11) in combination with four different adjuvants. The four adjuvants tested were: Freund's (GIBCO). Fowl adjuvant LES-STM (here after referred to as the RIBI adjuvant; RIBI Immunochemical Research. Inc.), Gerbu (Biotech) and Quil A (Accurate Chemical).
Each adjuvant was tested at both concentrations of antigen. The procedures for preparation and administration for each of the adjuvants were those provided by each manufacturers' protocol. The adjuvant dose in hens was also determined according to manufacturers recommendation if specified.
For immunization with Freund's adjuvant. the standard protocol was used (Example Briefly. I volume of antigen were emulsified in 1.2 volumes of either complete Freund's -217 adjuvant for the first immunization or incomplete Freund's for the subsequent boosts. One milliliter of the antigen/Freund's mixture was administered to each hen at four sites. Since Freund's adjuvant contains an oil. the mixing of Freund's adjuvant with the immunogen required vigorous emulsification of the material until solidification using two syringes connected together by a barrel connector. The other three adjuvants (RIBI. Gerbu and Quil A) are aqueous in composition and uniform mixing with the recombinant antigen was far easier as compared to Freund's. Only gentle vortexing was required for mixing the three aqueous adjuvants. The final mixture using these aqueous adjuvants also remained a homogenous liquid allowing easier administration into the hens as compared to using Freund's.
Using the RIBI adjuvant, each hen received 500 pi of the antigen/adjuvanc-mixture at one site containing 100 pg of adjuvant. The recommended Quil-A dose for guinea pigs and rabbits was 50 pg and 100 pg. respectively. By extrapolating by weight, the hens were each given 75 pg of the Quil A adjuvant at one site in an antigen/adjuvant volume of 500 pl.
15 Using Gerbu material. each hen received 5 pg of adjuvant in 500 pl antigen mixture at one site. The hens were all immunized subcutaneously for 4 times at roughly two-week intervals.
b) Determination of Anti-Recombinant C. difficile Toxin A IgY Titers by
ELISA
Anti-recombinant toxin A antibody levels generated using the different adjuvants were compared by ELISA. About I week after the last boost, at least 3 eggs from each of the 8 groups along with pre-immune eggs were collected, yolks pooled within the group and IgYs were extracted by PEG as described in Example 1. The purified anti-recombinant toxin A IgYs were then resuspended in PBS at IX yolk volume. The protein concentration of each of the preparations. determined by absorbance at 280 nm. were all similar at about 4 to 5 mg/ml.
The IgY reactivity and liter of each of the individual antibody preparations against pMA1870- 2680 were determined by ELISA against a soluble (pPTrxAl870-2680N/C: Example 29) and an insoluble (pPA1870-2190: Example 17a) analog of the C. dficile toxin A 1870-2680 interval. These recombinant C. difficile toxin A analogs were used to coat the microtiter plates for ELISA instead of the recombinant used in the immunization (pMA1870-2680) as both pPTrxA1870-2680N/C and pPA1870-2680 were not expressed as fusions with the MBP as was the pMA1870-2680 immunogen. This was done in order to determine antibody -218 reactivity against the toxin portion of the C. difficile toxin A recombinant specifically rather than to the MBP portion of the fusion protein.
The soluble analog pPTrxA1870-2680N/C used to coat the microtiter plate was expressed as a fusion with thioredoxin which aids in solubility and the resulting fusion protein probably exists in a native conformation. The insoluble analog pPA1870-2190. which presumably contains denatured epitopes, was also used to coat microtiter plates. The ELISA reactivity of each IgY to both the soluble and insoluble analogs was tested to determine if there was any preferential reactivity to one or the other analogs when different adjuvants were used for the generation of the IgY.
The standard ELISA protocol described in Example 13c was used with the exception that 20 to 40 fold-less pPTrxAl870-2680N/C protein was used than normal-o coarthe 96well microtiter plates (Falcon. Pro-Bind Assay plates) to reduce background. One-hundred pl/well were coated overnight at 4°C with the soluble pPTrxAl870-2680N/C protein at 0.05 pg/ml or the insoluble protein pPAI870-2680 at 1 pg/ml. The results are shown in Figure S 15 In Figure 40. the results of the ELISA reactivity comparing the antibody titer of each of the adjuvant/antigen combinations to either the insoluble or the soluble C. difficile toxin A recombinant is shown. The following abbreviations were utilized: PI (pre-immune); adjuvants were designated as F. R. Q and G for Freund's. RIBI. Quil-A and Gerbu S 20 respectively at either 1 mg or 100 pg (100).
In addition, the antibody titer in each group was compared after 3 or 4 immunizations to determine if antibody response has plateaued (indicated by the use of -3 or -4 in Figure 40). All four adjuvants were able to elicit antibody responses in the hens to varying degrees.
but their antibody responses to the soluble or native antigen and insoluble or denatured antigen differed. Freund's adjuvant generated a greater antibody response to the insoluble analog as compared to the soluble one. Almost rFo reactivity was seen using Freund's adjuvant with 100 pg of antigen to the soluble analog. There was also no difference in response using Freund's to the insoluble analog at either concentration (100 pg or 1 mg) of irmnunogen while an increase in reactivity to the soluble analog was seen in the higher concentration compared to the lower concentration. In contrast. the antibody reactivity to the soluble analog was generally greater than the insoluble analog using the three other aqueous adjuvants. Antibody reactivities in the ELISA to the soluble analog were about 2-fold higher compared to the insoluble analog. The antibody response improved in the Gerbu. RIBI and -219 Quil-A groups using the increased dose of antigen (1 mg versus 100 pg, and was more pronounced against the soluble analog compared to the insoluble one. The antibody levels to both the insoluble and soluble analog in most of the groups increased after an additional boosting when comparing the 3rd and 4th immunizations.
The results shown in Figure 40 demonstrate that the immunization of chickens with Freund's adjuvant using a soluble recombinant C difficile toxin A immunogen elicits antibodies primarily against the insoluble analog. This finding is important if conformational antibodies are required to confer protection in vivo. If conformational antibodies are required.
the alternative adjuvants such as Gerbu or RIBI used here would be preferred. The soluble antigen may become denatured during the harsh emulsification process required when using Freund's adjuvant as compared with the other adjuvants. The resulting denrtured-antigen would then presumably invoke antibodies primarily against an insoluble or nonconformational analog. This effect using Freund's may be overcome by using more antigen for immunization because less of the total is being denatured and a greater amount of native 15 antigen is present. Indeed. there was an increase in soluble analog antibody reactivity at the higher immunogen concentration while there is no difference in insoluble antibody reactivity at both immunogen concentrations.
S. c) Testing the Neutralizing Ability of the Anti-Recombinant C difficile Toxin A IgYs Against C. difficile Toxin A In Vivo The ability of the antibodies raised against the pMA1870-2680 protein generated above using the different adjuvants to neutralize toxin A was compared in vivo. PEG-purified IgYs from eggs from hens immunized with each of the four adjuvants at the I mg immunogen concentration were diluted at 0.5X yolk volume in 0.1 M carbonate buffer. pH 9.5. This antibody concentration (0.5X) was chosen because it would illustrate the best differentiation in IgY neutralizing capability using the different adjuvants. Pre-immune antibodies also at concentration in carbonate were prepared as controls. The antibodies were diluted in carbonate buffer so they could be orally administered with acid less degradation in the stomach.
The IgY protein concentration by absorbance at 280 nm of all of the 0.5X preparations was 2.4-2.5 mg/ml of which 25 to 50 pg/ml was specific antibody against the C. difficile toxin A recombinant protein. An in vivo protection study of hamsters against C. difficile toxin A using the five IgY preps was preformed as described in Example 14(c). Five groups.
220 each consisting of 4 male 30-40 gms Golden Syrian hamsters (Charles River). Each hamster was given a mixture of 30 pg of C. difficile toxin A (Tech Labs) in I ml of anti-recombinant C difficile toxin A IgYs or pre-immune IgY. This mixture was first allowed to preincubate for one hour at 37 0 C prior to oral administration. The animals were then observed for 24 hours after administration for the presence of diarrhea and death. The results were tabulated and shown in Table 44.
Table 44 Generation of Toxin A Neutralizing Antibodies Using Different Adjuvants with pMA 1870-2680 Treatment Group Healthy' Diarrhea Dead Preimmune 0 1 3 Freund's 0 0 4 Gerbu 4 0 0 RIBI 4 0 0 Quil A 4 0. 0 a a a a .a a a 15 Study outcome after 24 hours.
The results shown in Table 44 demonstrate that premixture of C difficile toxin A with 20 0.5X anti-recombinant C difficile toxin A IgYs generated using the Gerbu, RIBI and Quil A adjuvants before administration prevented all oven symptoms and death from the disease in the hamsters. In contrast, all the animals treated with anti-recombinant C. difficile toxin A IgY generated by use of Freund's adjuvant (as a 0.5X antibody preparation) mixed with C difficile toxin A failed to protect and the hamsters developed diarrhea and died within 24 hours. Three out of four hamsters treated with pre-immune IgY died and the lone survivor had severe diarrhea. These results showed that the anti-recombinant C difficile toxin A IgYs generated using Gerbu. RIBI and Quil A were able to neutralize the C. difficile toxin A activity in vivo while the Freund's-generated IgY at the same concentration could not. The inability to neutralize C difficile toxin A by the Freund's-generated anti-recombinant
C
difficile toxin A IgY correlates with its low ELISA reactivity against the soluble toxin A analog. In contrast. all of the other adjuvants invoked high antibody levels to the soluble analog and were neutralizing. These results indicated that the neutralizing potential of the antibodies correlated well with their reactivity to the soluble, but not the insoluble analog.
The results also indicated that the maintenance of a soluble or conformational C difficile -221 toxin A immunogen was important in generating neutralizing antibodies. Thus, the choice of an adjuvants such as RIBI or Gerbu was important to retain the conformation of the immunogen which was important in generating anti-C. difficile toxin A antibodies which were protective in vivo.
EXAMPLE 36 In Vivo Neutralization of Toxin A Using Antibodies Against the Recombinant pPA 1870-2680 Protein To determine if the irrinunization of hens with the C difficile toxin A recombinant pPA1870-2680(N/C) induced neutralizing antibodies, the following experiment was performed. The example involved a) immunization of hens with the C. difficile toxin A recombinant pPA1870-2680(N/C) using four different adjuvants: b) purification and detection of anti-recombinant IgY; and c) in vivo neutralization study in hamsters using the anti- 15 pPA1870-26 8 0 antibodies incubated with toxin A.
a) Immunization of Hens with the C. difficile Toxin A Recombinant pPA1870- 2680 Using Four Different Adjuvants Egg-laying Leghorn hens were each immunized with the C. dfficile toxin A recombinant pPA1870-2680(N/C) (Example 29d). This recombinant protein is expressed in the pET vector and was shown to be capable of isolation in a highly pure form which contained very low levels of endotoxin as compared to the same region expressed in other vectors such as pMal-c (Example 11). These results showed that the pPA1870-2680 Srecombinant protein would be compatible for use in a vaccine. Accordingly, the ability of pPA1870-2680 to stimulate an antibody response was tested.
Four groups of hens (2 hens/group) were immunized with 100 g of pPA1870- 2680(N/C) (purified as described in Example 29d) using 4 different adjuvants. The adjuvants used were: Freund's (GIBCO). Fowl (RIBI) adjuvant (RIBI Immunochemical), Gerbu (Biotech) and Quil A (Accurate Chemical). The amount of each adjuvant used with the recombinant was described in Example 35. The hens were all immunized 4 times at 2 week intervals.
222b) Purification and Detection of Anti-Recombinant IgY The anti-recombinant pPA1870-2680(N/C) levels using the different adjuvants were compared by ELISA. About one week after the last boost, standard PEG preps were prepared from eggs from each group and resuspended at a 4X concentration (all contained about mg/ml IgY) in 0.1 M carbonate buffer, pH. 9.5. The standard ELISA protocol (Example 13c) was followed to determine specific antibody reactivity to soluble immunogen pPAl870-2680.
The ELISA results are shown in Figure 41.
In Figure 41. the absorbance at 410 nm is plotted against the logl 0 of the dilution of each antibody tested. The solid black squares represent the results of the ELISA using the pre-immune IgY: the open squares. black diamonds, open diamonds and black triangles represent the results of the ELISA using antibodies generated using pPAl870-2680(N/C) (Interval A2) and the following adjuvants: Gerbu Quil A RIBI (R-A2) and Freund's respectively.
After 4 immunizations, all the hens generated a specific IgY response against the C 15 difficile toxin A recombinant expressed in the pET vector pPA1870-2680N/C)]. The response generated by using Freund's, Fowl (RIBI) adjuvant and Quil A were comparable as shown in Figure 41. A lower antibody response was seen in the Gerbu immunized hens.
Interestingly, using the Freund's adjuvant with pPA 870-2680(N/C) gave the highest antirecombinant activity, whereas in the previous example (Example 35) using the same 20 recombinant region expressed in pMal-c (pMA1870-268 0 Freund's adjuvant generated the weakest response. The other adjuvants invoked similar antibody responses comparing both recombinants. These result indicated that the level of antibody response using Freund's adjuvant may depend on what type of antigen is used.
25 c) In Vivo Neutralization Study in Hamsters Using the Anti-pPA1870- 2680(N/C) Antibodies Incubated with C. difficile Toxin A The ability of antibodies to neutralize C. difficile toxin A in vivo was compared using antibodies raised against pPA1870-2680(N/C) protein generated using the RIBI and Freund's adjuvants. This assay was preformed as described in Example 35c with the exception that the antibodies were diluted to a 2X concentration containing 10 mg/ml of IgY protein. C difficile toxin A (Tech Labs) was mixed with antibodies generated using Freund's and Fowl (RIBI) adjuvant and orally administered to hamsters. Hamsters treated with pre-immune IgY 223 served as the control. The number of hamsters which were healthy, had diarrhea or were dead 24 hours after administration of the IgYs is shown in Table Table Generation of C difficile Toxin A Neutralizing Antibodies Using Different Adjuvants with pPA1870-2680 Treatment Group Healthy Diarrhea Dead Preimmune 0 0 4 Freund's 4 0 0 RIBI 4 0 0 10 As shown in Table 45, both the Freund's and RIBI adjuvants used in conjunction with pPA1870-2680(N/C) were able to elicit in vivo neutralizing antibodies against C. difficile 15 toxin A as compared to pre-immune IgY. The ability of the antibodies to neutralize C.
difficile toxin A shown in this example and in Example 35 appears to correlate well with their ELISA reactivity to a soluble (native) recombinant protein. These results show that the C difficile toxin A recombinant. pPAl870-2680(N/C), was immunogenic in hens and was capable of generating in vivo neutralizing antibodies: therefore, the pPA 870-2680(N/C) protein is an excellent vaccine candidate.
EXAMPLE 37 Enteric Coating of lIY Raised Against Recombinant C difficile Toxin A For Oral Delivery To determine if the avian antibodies (IgYs) raised against recombinant C. difficile toxin A could be enterically-coated and potentially retain in vivo protective abilities, the following experiment was conducted. The example involved a) enteric coating of antirecombinant C difficile toxin A antibodies, b) dissolution studies to determine the disintegration kinetics of the enteric-coated IgYs as a function of pH and c) determination of the stability of the antibody reactivity after coating and dissolution by ELISA.
a) Enteric Coating of Anti-Recombinant C. difficile Toxin A Antibodies Preliminary studies were performed to determine an effective enteric coating process.
Enterically-coated avian antibodies should be more resistant to degradation in the stomach -224compared to antibodies delivered in solution when the route of administration is oral.
Intestinal enteric coatings would remain intact at the low pH ranges found in the stomach and therefore the coated IgYs would be able to pass the through stomach undegraded but dissolve at the higher pHs (about 6.0) and release the IgYs in the intestines. An additional property of the enteric films selected for testing is that they are compatible in aqueous solutions instead of organic solvents during the coating process. This property of the enteric film should probably preserve conformation and integrity of the IgY antibody during the coating process.
Since the intestines are the site of C difficile disease. enteric coating of the anti-C difficile toxin IgY' should concentrate the amount of antibodies available at the site of infection to improve efficacy and reduce the effective dose required as compared to the use of uncoated IgYs.
The anti-C difficile toxin A antibodies were coated as follows. Sixty grams of lyophilized antibodies against the recombinant C difficile toxin A protein pMA1870-2680 (Example 11) were prepared. IgYs from eggs collected from hens immunized with the 15 recombinant protein were purified by PEG-precipitation. The IgY pellets after purification were resuspended in 0.1X PBS, pH 7.4 at about 1/4 starting yolk volume (4X) and from 200 to 250 ml volumes were transferred to 600 ml lyophilizing flasks (Labconco). The IgY solutions were flash frozen in the flasks by rotation in an reagent alcohol bath containing dry ice. The frozen antibodies were lyophilized on a Labconco Freeze Dry System/Lyph Lock 4.5 unit operated according to manufacturer's instruction. About 250 mis of the 4X IgY prep yielded about 10 grams of dry material after lyophilization.
The lyophilized IgY was sent to The Coating Place Inc. (Verona. WI) for enteric coating. The antibodies were encapsulated using a Wurster coating chamber which is wellsuited for coating materials efficiently and uniformly at a small scale in a single operation.
Encapsulated IgYs were prepared using two different coating processes. Either a single step direct process or a two-step process using a non-pariel a sugar particle of 40-60 mesh size). The lyophilized IgY was either overcoated directly with the film coatings or a two-step method was performed where first the IgY itself was used to overcoat the non-pariel. Then the IgY-coated sugar particle was then overcoated with the enteric film. The use of the sugar particle provides extra bulk necessary to maintain the antibodies in the coating chamber and can aid in a more uniform application of the enteric film.
Two different aqueous enteric films were selected and used with each coating process.
The lyophilized IgY was either overcoated with Aquateric (FMC Corp.) or Eudragit® 225 (Rohm Tech Inc). Both of these coatings are water-soluble enteric film coatings that dissolve at pH 6.5 or 5.5. respectively. Both of these enteric films were selected because they fulfill the selection criteria suitable for the needs as described above. Each of the different coating procedures using both enteric films yielded enterically-coated antibodies product. The twostep process using the sugar particle made the entire overcoating procedure in Wurster apparatus technically easier with less loss of material and subsequent greater yields of final product An enteric coating of approximately a 27-30% by weight was applied to the IgY using the direct method. About 70% of the remaining weight of this enteric-coated material was IgY. About a 32-33% by weight of the enteric coating was achieved in the IgY- 10 overcoated sugar particle. The remaining 67% by weight of the enteric particle was comprised of about 40-50% due to the sugar particle and about 20% the Ig. b) Dissolution Studies to Determine the Disintegration Kinetics of the Entericcoated IgYs as a Function of pH The performance of each of the enterically-coated IgY were tested by determining their dissolution profile. Properly coated IgY particles with intestinal enteric films should remain intact in a gastric solution of pH 1 to 2. but dissolve and release the IgYs into an intestinal solution of pH 7.5. Simulated gastric fluid at about pH 1.2 and simulated intestinal fluid at pH 7.5 were prepared according to USP guidelines except the digestive enzymes were omitted [United States Pharmacopeia. Vol. XXII (1990) United States Pharmacopeial Convention. Rockville, MD, pp. 1788-1789]. Ten milligrams of each enteric coated preparation Aquateric and Eudragit® coatings) was added per 1 ml of the simulated gastric and intestinal fluids and mixed gently for 1-2 hours at room temperature. An aliquot of the solution was taken at different time points and checked for the presence of protein released in solution. Protein amounts in solution were determined either by absorbance at 280 nm or using a BCA protein assay (Pierce).
The studies demonstrated that the IgY directly coated with both the Aquateric and Eudragit® coatings and the Aquateric-overcoated IgY sugar-particles failed to perform adequately in the dissolution studies. IgYs at both pH 1.2 and 7.5 were released in the solution within minutes after addition of these particles. The dissolution profile for the Aquateric-overcoated IgY sugar particle monitored by absorbance is shown in Figure 42. The dissolution profile for the Eudragit -overcoated IgY sugar particle is shown in Figure 43.
-226 In Figure 42 the absorbance at 280 nm is plotted against time in minutes. The release of the IgY from the Aquateric-overcoated particle in simulated gastric fluid is shown by the solid black squares; release of the IgY from the coated particle in simulated intestinal fluid is shown by the open black squares. Because the Aquateric film itself absorbs UV at a similar wavelength as protein (275-276 nm), UV absorbance at 280 nm cannot be used to accurately quantitate the amount of IgY in solution. Thus, protein at 1 hour (60 min) dissolution was quantitated using the BCA method in order to obtain an accurate determination of the protein concentration.
As shown in Figure 42. the amount of specific IgY found after dissolution of the 10 Aquateric-overcoated IgY in the two fluids were similar: 4 mg/ml at pH 1.2 and 4.9 mg/ml at pH 7.5. The difference in absorbance shown in Figure 42 between the gastftc and-intestinal solutions is due to the presence of more Aquateric film being dissolved in the intestinal solution.
In contrast to the performance of the failed coatings, the Eudragit® -overcoated IgY 15 sugar particle properly opened and released IgY into the solution in the simulated intestinal fluid in a time-dependant manner, while it remained intact in the gastric fluid. The dissolution profile in the gastric and intestinal solutions of the Eudragit® -overcoated IgY sugar particle as a function of time is shown in Figure 43.
In Figure 43, the absorbance at 280 nm is plotted against time in minutes. The release of the IgY from the Eudragit® -overcoated particle in simulated gastric fluid is shown by the solid black squares; release of the IgY from the coated particle in simulated intestinal fluid is shown by the open black squares. Since Eudragit® does not absorb UV at the amounts found in the coatings, absorbance values at 280 nm can be directly converted to protein concentration.
As shown in Figure 43. little or no protein was released in the gastric solution while protein was continually released into the intestinal solution at a linear rate reaching a maximal dissolution after about 2 hours. Ten mg/ml of Eudragit® -overcoated particles yielded from 2 to 2.5 mg/ml of IgY after dissolution. The Eudragit® -overcoated particles in the gastric solution remained intact for long periods of time, even after further incubation at 4 0 C for an additional week.
The dissolution profile Eudragit® -overcoated IgY sugar particles was determined under conditions that mimic normal physiological conditions simulated travel through the GI tract). The particle was first placed in the gastric solution for 120 minutes followed by an 227-
S..
180 minute incubation in the intestinal solution. Both of these incubations took place with gentle mixing at 370 C. Under these conditions incubation in gastric fluid followed by incubation in intestinal fluid), IgY from the Eudragit® -overcoated sugar particle was not released into the gastric solution protein as found in Figure 42 incubation in gastric fluid only), but was only released and detected in the intestinal solution at similar levels found in Figure 42 (from 2 to 2.5 mg/ml protein released after about 2 hours).
The dissolution studies discussed above demonstrated that the anti-recombinant
C.
difficile toxin A IgYs were successfully enterically-coated using Eudragit® and a non-pariel.
10 c) Determination of the Stability of the Antibody Reactivity after Coating and Dissolution by ELISA The stability of the anti-recombinant C difficile toxin A IgYs after the overcoating process was determined. This was tested by comparing the ELISA reactivity of the antibodies before coating then after the coating process followed by dissolution at pH 1.2 then pH Pre-immune IgY, lyophilized anti-recombinant toxin A IgY starting material and antirecombinant toxin A IgY obtained from the Eudragit® -overcoated IgY sugar particle after dissolution were first all quantitated for protein and normalized at 2 mg/ml in PBS (pH 7.4).
An ELISA was performed detecting antibodies against the recombinant toxin A pPTrxA1870- 2680N/C as described in Example 35b. The ELISA results are shown in Table 46.
Table 46 Comparison of Anti-Recombinant Toxin A Titers by ELISA Before and After Enteric Coating Dilution Preimmune IgY* 1:50 0.017 1:250 0.005 1:1,250 0.004 1:6.250 0.005 1:31.250 0.007 1:156.250 0.009 *Average A280 readings.
Pre-Coated Anti- Post Coated Anti- Recombinant A* Recombinant
A*
.2 1.4 0.59 0.15 0.037 0.015 0.009 1.2 0.38 0.10 0.026 0.009 0.007 228 The results shown in Table 46 demonstrate that the reactivity of the anti-recombinant C. difficile toxin A IgYs before and after Eudragit® -coating to the recombinant C. difficile toxin A protein was very similar. These results indicated that the coating process was not harmful to the IgY and that the IgY remain reactive and functional after dissolution under physiological conditions.
The results shown above demonstrate that enterically-coated IgY that remained stable and active was generated.
EXAMPLE 38 10 Vaccination of Hamsters Against C difficile Infection with Recombinant C. difficile Toxin A Proteins To determine if hamsters vaccinated with C difficile toxin A recombinant proteins Swould elicit protective antibodies against C difficile infection, the following experiment was conducted. Three different C difficile toxin A recombinants. expressed in the pMal-c or pET vectors, were compared. The example involved a) immunization of hamsters, b) detection of humoral and mucosal anti-recombinant antibody responses by ELISA. and c) challenge study of hamsters with C difficile.
20 a) Immunization of Hamsters Three groups of 90-100 gram female Golden Syrian hamsters (Charles River), each group containing 9 to 11 members, were tested as follows. Hamsters from each group were individually tagged using an ear punch for identification. The animals from each group were housed together and were given food and water ad libitum throughout the course of the experiment. Hamsters were immunized with two different recombinant C. difficile toxin A protein repeats fragments produced the in pMal-c vector and expressed with a maltose binding protein (MBP) fusion and one recombinant C difficile toxin A protein repeats fragment produced the in pET vector. The animals were immunized subcutaneously with 25 Pg of purified protein of either pPA1870-2680N/C (Example 15), pMA1870-2680, a subfragment of pMA1870-2680 called pMA1960-2680 or the MBP (pMal-c) alone as a control. All three recombinant pMal vectors were grown and protein was expressed and purified as described in Example 28c. Recombinant pPAI870-2680N/C was purified as described in Example 29f.
Mixtures comprising 200 pl of antigen and complete Freund's adjuvant (for the first injection) and incomplete Freund's adjuvant (for the subsequent injections) were given 229 subcutaneously behind the neck. The vaccination was administered using a I ml 27 gauge tuberculin syringe after the animals were lightly etherized. The animals were vaccinated five times at roughly 2 week intervals.
b) Detection of Humoral and Mucosal Anti-Recombinant Antibody Responses by ELISA The detection of humoral and mucosal anti-recombinant C difficile toxin A IgY titers in the hamsters was determined by ELISA. For the humoral response. serum from all members from each group was collected and assayed for anti-recobthinant toxin A IgG levels.
S 10 At least I week after the last boost. the hamsters were etherized. bled by cardiac puncture and serum was collected. Ninety-six well microtiter plates (Probind. Falcon) were coated overnight with the soluble C. difficile toxin A recombinant, pPTrxAI870-2680N/C (Example 29e) at 0.05 pg/ml in PBS (pH 7.4) at 100 pl per well. Standard ELISA procedure were followed as described in Example 35b. The secondary antibody used was goat anti-hamster 15 IgG-alkaline phosphatase (Southern Biotech) at a dilution of 1/2000. The average absorbance at 410 nm from duplicate test wells of each serum sample diluted at 1/250 is shown in Figure 44.
In Figure 44. the OD 41 0 of a 1:250 dilution of serum taken from hamsters immunized with either pMal-c (the pMal-c vector lacking an insert). pMA 870-2680 (Example 28c), pMA1960-2680 (Example 28b) or pPA1870-2680 (Example 15). The numerals shown on the ordinate represent the number assigned to animals within a group.
The results shown in Figure 44 demonstrate that all the hamsters immunized with the C. difficile toxin A recombinants responded by producing anti-recombinant C. difficile toxin A IgG in the serum. Some variability in the antibody response within the hamsters in a group existed although this difference was not greater than 4-fold. The average antibody response to pMA1960-2680 and pPA1870-2680 was uniformly higher than the response to pMA1870- 2680. The hamsters immunized with pMal protein did not produce an anti-serum IgG response to the C difficile toxin A recombinant protein.
Whether a mucosal IgA response was elicited after immunization was also determined by ELISA. Freshly isolated feces from 4 members of each group were collected. weighed and resuspended by vortexing at 300 pl per 100 mg of stool in PBS. pH 7.4 containing 0.05% thimerosal. The fecal suspension was centrifuged for 5 minutes at 14.000 rpm in a microcentrifuge. Microtiter plates were coated with recombinant antigen as described above.
230- Standard ELISA procedures were used with goat anti-mouse IgA-alkaline phosphatase (Southern Biotech) at 1/1000 as the secondary antibody. This conjugate was used instead of an anti-hamster IgA because the anti-hamster IgA is not commercially available and the antimouse antibody has been previously reported to cross-react with hamster IgA. In all samples of fecal extracts, mucosal IgA against recombinant toxin A was not detected by ELISA.
These results confirm previous studies [Kim and Rolfe (1989) Microbial Ecology in Health and Disease 2:47] in which IgA against toxoid A was not detected in hamsters immunized with a toxoid prepared from C. difficile toxin A.
10 c) Challenge Study of Hamsters with C difficile The vaccinated hamsters (described in section a above) were challenged viah C difficile to determine if the anti-recombinant C. difficile toxin A antibodies were protective against C difficile disease. Normal hamsters infected with a toxigenic strain of C difficile develop a fatal disease beginning with diarrhea and eventually die from severe enterocolitis of 15 the cecum (proximal colon) and ileum (as described in Example 9).
The four groups of vaccinated hamsters were first each predisposed with an intraperitoneal dose of Clindamycin-phosphate (Biomol) in 1 ml of water at I mg per 100 gm body weight. About 24 hours later, the hamsters were orally challenged with 1 x 106 C.
difficile in 1 ml of sterile saline using an 18 gauge feeding needle. The animals were lightly anethesized with ether prior to administration. The toxigenic strain of C. difficile, ATCC 43596. was used after 48-hours growth on CCFA plates (BBL). One hamster in the pMA1960-2680 immunized group died accidentally from ether overdose reducing the group number from 9 to 8. The results of the immunization study are shown in Table 47.
Table 47 Vaccination Against Lethal C. dificile Enterocolitis Using Recombinant Toxin A Fragments Vaccination Group Protection pMal-c (MBP) 10% (1/10) pMA 1960-2680 62% (5/8) pMA1870-2680 30% (3/10) pPA870-2680 19% (2/11) -231 The results shown in Table 47 demonstrate that protection against death occurred in some of the hamsters immunized with each of the recombinant toxin A proteins pMA1960-2680 and pMA1870-2680). These results were not statistically significant compared to the fusion control (pMal-c which expresses only the MBP) at a P-value of 0.05 or less using Chi-squared analysis. Ninety percent mortality occurred in the fusion control immunized group(pMal-c). The percent mortality in the pMA1960-2680 immunized group was 38%. The percent mortality in the pMA1870-2680 immunized group was 70% and in the pPA1870-2680 immunized group was 81%. The time to death in recombinant C. difficile toxin A vaccinated group was not delayed compared to the control, occurring up to 3 days 10 after infection. Necropsy of the dead hamsters revealed typical pathology such as severe megacecum.
The specific P-values of the test groups compared to the control group for pMA1960- 2680. pMA1870-2680 and pPA1870-2680 groups were less than 0.10. less than 0.75 and less than 0.90. respectively. All of the hamsters except one in the pMA1870-2680 immunized 15 group presented with diarrhea one to two days after infection. There appeared to be no correlation between anti-recombinant C difficile toxin A antibody titers and the level of .protection. For example. hamster number 6 in the pMAI960-2680 immunized group had a lower ELISA titer compared to hamster number 2 (see Figure 44) yet number 6 survived and number 2 was not protected and died. From these results, hamsters vaccinated with either of 20 the recombinant C difficile toxin A repeats proteins were not protected against C difficileinduced diarrhea and from 19 to 62% were protected from the lethal stage of the disease.
The above results correlate with previously published work [Lyerly et al. (1990) Curr.
Microbiol. 21:29] which showed that hamsters vaccinated with the smaller C dificile toxin A recombinant fragment (the 1960-2680 interval) expressed in pUC9 could also only partially protect against the lethal stage of disease and none of those hamsters were protected against diarrhea. Lyerly et al. [(1990) Curr. Microbiol. 21:29] stated that antibodies to the C. difficile toxin A recombinant protein tested did not prevent the diarrheal stage of the disease and the death in half of the hamsters was due to the varying levels of neutralizing serum antibodies to the toxin A recombinant. From the above results, differences in anti-recombinant C difficile toxin A titers seen between hamsters in a group may not explain why protection did not occur in all of the animals. The above results indicate that possibly an additional component, possibly a toxin B recombinant protein, is necessary for a more effective vaccine against C.
difficile disease.
232 EXAMPLE 39 Vaccination of Hamsters Against C. difficile Infection with C. difficile Toxin A and Toxin B Recombinant Proteins Hamsters were immunized with recombinant C. difficile toxin A or recombinant toxin B alone or in combination to test whether this would invoke a humoral response to the recombinant proteins. Furthermore, the ability of the antibodies produced by these vaccinations were tested for the ability to protect the hamsters from infection with C. difficile.
Specifically, it was determined if antibodies raised against a recombinant C difficile toxin B would provide any protection in vivo by itself or above that provided by vaccination with recombinant C difficile toxin A alone. The example involved a) the immunization of hamsters, b) determination of humoral and mucosal antibody response by ELISA and c) in vivo challenge studies in vaccinated hamsters.
15 a) Immunization of Hamsters The recombinant proteins used for vaccination were the C. difficile toxin A recombinant protein pPA1870-2680N/C (Examples 11 and 29) and the C. difficile toxin B Srecombinant protein pPB1750-2360 (Example 15b). The recombinant proteins were expressed in the pET vector instead of pMal-c vector used in Example 38 because the proteins expressed S 20 and isolated using the pET vector were found to be capable of purification at a higher level of purity with lower levels of endotoxin. Production of recombinant proteins in the pET vector is especially amenable for the potential utilization of the recombinant protein as a human vaccine which demands high purity and low levels of potentially harmful endotoxin.
For immunization, 100 pjg of pPA1870-2680, 100 pg of pPB1750-2360 or 100 jig of each in combination (200 pg total) were mixed with 2 tg of Gerbu adjuvant (Biotech). The control group were immunized with 100 pg of bovine serum albumin (BSA) with the Gerbu adjuvant Each group (four total) consisted of 9-10 members of 100 gm female Golden Syrian hamsters (Charles River). Animals were individually tagged to identify members. The hamsters were lightly anesthetized prior to injection sub-cutaneously behind the neck using 1 ml syringe with a 27 gauge needle. The hamsters were immunized 4 times at roughly 2 week intervals.
-233 b) Determination of Humoral and Mucosal Antibody Response by ELISA Serum from all individuals from each of the above groups were tested for antirecombinant protein IgG levels by ELISA. At least one week after the last boost. all of the animals from each group were bled by cardiac puncture and serum was collected. Antirecombinant C. difficile toxin A and anti-recombinant C difficile toxin B from the serum samples were determined by ELISA. Ninety-six well microtiter plates (Probind. Falcon) were coated overnight at 4"C with either pPAI870-2680 protein at 0.05 ig/ml or pPBI750-2360 protein at 1.0 pg/ml in PBS (pH 7.4) at 100 pl per well. Standard ELISA procedures were used exactly as described (Example 13c). The results are shown in Figures 45 and 46.
10 The average absorbance of each serum performed in duplicate and diluted at 1/250 is shown in Figures 45 and 46. Figure 45 shows individual antibody reactivity to the C difficile toxin A recombinant in the groups immunized with either the C. difficile toxin A recombinant (pPA 870-2680) or a mixture of recombinant C difficile toxins A and B (pPA1870-2680 and pPB1750-2360). Figure 46 shows antibody reactivity to recombinant C difficile toxin B in 15 the groups immunized with either the C difficile toxin B recombinant (pPB1750-2360) or a mixture of recombinant C. difficile toxins A and B (pPA1870-2680 and pPB1750-2360).
The results shown in Figures 45 and 46 demonstrate that in all cases each animal responded and produced a specific IgG antibody response to the immunogen. As expected.
the hamsters immunized with BSA (negative control group) did not invoke any antibody response to the recombinant antigens. The anti-recombinant C difficile toxin A or B response S. within members of the same group were similar.
The determination of a mucosal anti-recombinant C difficile toxin A or recombinant C difficile toxin B IgA response was elicited after immunization was also determined by ELISA. Freshly isolated feces from 4 members of each group were collected. weighed and processed as described in Example 21. Plates were coated with recombinant C difficile toxin A or recomnbinant C. difficile toxin B antigen as described above for determination of serum IgG levels. Standard ELISA procedures (Example 13c) were used in conjunction with goat anti-mouse IgA-alkaline phosphatase (Southern Biotech. Birmingham. AL). In all samples of fecal extracts. IgA against recombinant toxin A or B was not detected. Again this result using different recombinants confirms that found in Example 38 and with previous studies [Kim and Rolfe (1989). supra].
-234 c) In Vivo Challenge Studies in Vaccinated Hamsters The vaccinated hamsters described above in section a) above were challenged with C difficile to determine whether the serum antibody response to either recombinant C. difficile toxin A or B alone or in combination was protective against CDAD. The four groups of vaccinated hamsters were first each predisposed to CDAD with an intra-peritoneal dose of Clindamycin-phosphate (Biomol) in 1 ml of water at 1 mg per 100 gm weight. About 24 hours later. the hamsters were orally challenged with I x 106 C. difficile organisms in 1 ml of sterile saline using an 18 gauge feeding needle. The animals were lightly anethesized with ether prior to administration. The toxigenic strain ATCC 43596 was used after 48-hours 10 growth on CCFA plates (BBL). The results of the immunization study is shown in Table 48.
Table 48 Vaccination Against Lethal C difficile Enterocolitis Using Recombinant Toxin A and Toxin B Polypeptides '0 Vaccination Group' Protection BSA 0% (0/10) pPA1870-2680N/C 20% (2/10) pPB1750-2360 0%(0/10) pPA1870-2680N/C pPB1750-2360 100% (9/9) Vaccinated with 100 pg recombinant protein per hamster subcutaneously 4 times at 2 week intervals.
As-shown in Table 48. one to three days after challenge with C difficilc. all of the hamsters immunized with either pPA1870-2680 or pB1750-2360 and the BSA control group developed diarrhea. All the hamsters in those three groups except two members immunized with pPA1870-2680. died from several hours to 48 hours after the detected onset of diarrhea.
Necropsy revealed severe enterocolitis in the animals with inflamed and enlarged cecums characteristic of C. difficile disease. In contrast. hamsters immunized with the vaccine comprising the combination of pPA1870-2680 or pB1750-2360 proteins showed no signs of illness such as diarrhea and remained healthy for the entire 14-day post-infection observation period. In fact, these animals have remained healthy for a period of at least 5 months postinfection: these results demonstrate that vaccination with the combination of pPA 1870-2680 or 235 pB1750- 2 3 6 0 proteins confers complete and long term protection on hamsters inoculated with C difficile.
The protective effect seen with the combination vaccine was not due to differences in antibody titer in this group compared to the antibody titers in the hamsters vaccinated with only recombinant C difficile toxin A or C difficile toxin B. Protection of the hamsters immunized with the C. difficile toxin A/B combination pPA1870-26 80 and pB1750- 2360) was statistically significant compared to the control: the P value was determined to be less than 0.001.
The above results demonstrate that recombinant C difficile toxin A and toxin B 10 proteins are both key components for an effective vaccine against C. difficile and that *:ellictation of antibodies against recombinant C. difficile toxins A or B alone was not sufficient to confer complete protection. Antibodies generated against a recombinant C. difficile toxin B in addition to recombinant C difficile toxin A both confer protection and they both act synergistically to neutralize C d[ficile-associated diarrhea and death. While the invention is 15 not limited by any particular mechanism. the protection from the anti-C. difficile toxin serum antibodies may result from the leakage of the C difficile toxin A and B neutralizing antibodies into tissues or the intestinal lumen during the inflammation that accompanies the early stages of C. difficile enterocolitis.
The results shown above (vaccination of hamsters with recombinant C. difficile toxins A and B) and in Example 32(c)(iii) (administration of antitoxin comprising a mixture of antibodies raised against both C dificile toxins A and B) strongly support one another.
Together they demonstrate that full protection from CDAD protection from both morbidity and mortality) requires the use of recombinant proteins derived from both C difficile toxins A and B for either active or passive immunization.
EXAMPLE In Vivo Protection Against C. difficile Infection by the Parenteral Administration of Antibodies Aainst Recombinant C di ie Toxin A and Prote results shown in Example 39 demonstrated that vaccination of hamsters with recombinant C dfficile toxin A and B proteins generated neutralizing serum antibodies in the recipient animals which conferred complete protection protection from both morbidity and mortality) from the deleterious effects of infection with C. difficile. Example 38 -236demonstrated that vaccination of hamsters with recombinant C. difficile toxin A proteins produced neutralizing serum anti-toxin A antibodies (IgG) but undetectable levels of mucosal (IgA) anti-toxin A antibodies. Thus. the production of serum anti-toxin A and B antibodies is sufficient to confer protection from CDAD. In order to determine whether parenteral delivery of anti-recombinant toxin A and B IgYs is an effective way to treat C dificile infection, the following experiment is conducted.
Six groups of 80-100 gram female Golden Syrian hamsters (Charles River). each group containing 9-10 members, are infected with C difficile as described in Example 32c).
The animals are housed three per cage and are offered food and water ad libitum throughout 10 the study. At the start of the study, each hamster is predisposed to infection by the intraperitoneal administration of Clindamycin-phosphate (Biomol) at I mg/1 00 gram body weight in 1 ml of water using a 1 ml tuberculin syringe (27 gauge needle). Approximately 24 hours *o later. each animal is orally challenged. using an 18 gauge feeding needle, with 1 ml of C difficile. (strain ATCC 43596) with approximately 10' to 10 4 organisms in sterile saline. The 15 organisms are grown for 48 hours on CCFA plates (BBL) prior to infection.
,Three hours after infection (Day treatment is initiated as follows. Each hamster receives 2 mls of a solution comprising either pre-immune IgY (as an 8X PEG preparation) or a mixture of anti-recombinant toxins A and B antitoxin raised against pMA1870-2680 and pPB1750- 23 6 The 8X PEG preparations are prepared and mixed as described in 32(c)(ii) with the exception that the IgYs are resuspended in sterile saline rather than in carbonate buffer. The IgY preparations are delivered by intra-peritoneal injection. The IgY preparations are administered either once. twice or three times a day for a period of 4 days (the treatment period).
The animals are observed for the onset of diarrhea and death during and after the treatment period. The level of protection afforded by each treatment dosage is ]calculated. If the lowest dose is protective in a significant number of hamsters. then lower doses are tested in subsequent experiments using the above conditions. For example, 1.0 and 0.5 ml of IgY preparation per animal per day for 4 days would be tested to determine the lowest intraperitoneal dosage sufficient for protection. If only very small doses of IgY are needed to confer protection via intra-peritoneal injection, then the IgY would also be delivered via intravascular injection to determine whether intra-vascular delivery of the IgY PEG preparations confer protection from C. difficile infection.
-237 EXAMPLE 41 Treatment of Hamsters Infected with C difficile using Enteric-Coated IgYs Against a Recombinant C. difficile Toxin A Protein To determine whether the enterically-coated anti-recombinant toxin A IgY (Example 37a) is effective in treating C difficile infection in hamsters at a lower dose required using the same IgY without an enteric coating, the following experiment is performed.
The hamster infection model is carried out exactly as described in Example 32c with the exception that enterically-coated antitoxin (Eudragit® L30D-coated pMA1870-2680 which had first be applied to a non-pariel) is used in place of the non-coated IgY in carbonate buffer. Briefly, three groups of hamsters (Sasco) containing 7 members per group are predisposition to infection with Clindamycin-phosphate (Biomol) at t-mg/100 gram body oweight. Twenty-four later, each animal is orally challenged. using an 18 gauge feeding needle. with I ml of C. difficile (ATCC 43596) containing approximately 1 x 103 organisms in sterile saline. The organisms are grown for 48 hours on CCFA plates (BBL) prior to infection.
Three hours after infection (Day treatment is intimated by oral administration of various concentrations of Eudragit® -coated anti-toxin A IgY as follows. Each group receives 0 (the control group). 2. 20. 50. 100 or 600 mg of enterically-coated IgY once per day for a S 20 period of 4 days. The enterically-coated particles are administered orally to the hamsters by placing each dose in a microcentrifuge tube. resuspending the particles in a low pH buffer such as acetate. pH 4.0 (low pH buffers are used to prevent the release of the IgY from the enterically-coated particle prior to delivery to the hamster): the suspension is then orally administered using a 14 gauge feeding needle. The animals are observed for the onset of diarrhea and death during and after the treatment period. The percentage cumulative mortality death) and morbidity diarrhea) are calculated.
The results form the above experiment (administration of enterically-coated IgY) are compared to the results obtained in Example 32c. In Example 32c. the same infection conditions were employed but the anti-toxin A antibodies were delivered in carbonate buffer and they lacked an enteric coating. In Example 32c. 50% of the hamsters treated after infection with uncoated IgYs were protected from death from C difficile. The amount of total IgY given per day in Example 32c was about 120 mg. Of that dose. the amount of specific antibody per day necessary achieve that level of protection 50% survival) was about 1200 pg of specific IgY. In the present example, the hamsters are each given 2. 20. 238- 100 or 600 mg of enterically coated IgY. Since only 1/5 of the weight of the entericallycoated material is IgY, the actual amount of total IgY administered in the 2. 20. 50, 100 and 600 mg doses is about 0.40 mg. 4 mg, 10 mg. 20 mg and 120 mg, respectively. Of that about 1 is specific anti-recombinant toxin A IgY. The 600 mg dose of the enteric particle the Eudragit®-coated anti-recombinant C difficile toxin A IgY preparation) is roughly equivalent to the amount of antibody delivered in carbonate buffer in Example 32c which gave 50% protection. Comparison of the dose of the enteric particles required to give the same 50%) level of protection indicates the degree of increased potency afforded by enterically-coating the IgY preparation. The results of the above experiment demonstrate 10 whether enterically-coated anti-recombinant C difficile toxin A.IgY (Example 37a) is effective in treating C difficile infection in hamsters at a lower doseas compared to noncoated anti-recombinant toxin A.
Accordingly, the recombinant C difficile toxin B IgY anti-pPB1750-2360) is also enterically-coated using the methods described in Example 37a. The enterically-coated antirecombinant C. difficile toxin B IgY is tested in the hamster infection model described above alone or in combination with enterically-coated anti-recombinant C difficile toxin A the coated anti-pMA1870-2680 IgY preparation). The results of these experiments demonstrate whether enterically-coated anti-recombinant C difficile toxin A and B IgYs (Example 37a) are effective in completely protecting animals from the morbidity and mortality associated with C difficile infection at lower doses as compared to the use of non-coated anti-recombinant
C
difficile toxin A and B IgYs.
EXAMPLE 42 Determination of the Minimum Effective Dose of Avian Antibodies in Carbonate Buffer Against Recombinant C difficile Toxin A and Toxin B to Treat C dificile-lnfected Hamsters The minimum effective dose of avian antibodies (IgY) raised against recombinant toxin A protein and recombinant toxin B protein necessary to treat C difficile-associated disease (CDAD) in hamsters was determined. The experiment involved an in vivo infection study in which hamsters were treated with different concentrations of IgYs raised against recombinant toxin A and recombinant toxin B.
Antibodies were generated against recombinant C. difficile toxin A (pMA1870-2680: Interval A-6) using RIBI adjuvant and against the recombinant C difficile toxin B (pPB1750- 239 2360: Interval B-3) using Freund's adjuvant. The immunization protocol used for each adjuvant was previously described in Example 35. Antibodies were PEG-purified and resuspended at an 8X concentration (all contained about 40 mg/ml IgY) in 0.1 M carbonate buffer, pH The infection study involved the testing of three experimental groups (Group 1. Group II. and Group III). Group I animals received 2 mis pre-immune IgY. Group II animals received 1 ml of a mixture containing anti-C. difficile toxin A and anti-C. difficile toxin B IgY. Group III animals received 2 ml of the mixture given to Group II. Each group consisted of eight Golden Syrian hamsters (Sasco) weighing an average of 79 3.2 gm.
S" o: 10 The hamsters were housed two or three per cage and were given food and water ad libitum "throughout the study. The infection study was performed using the protocol described in Example 31c.
Hamsters were predisposed to infection with C difficile by I.P. administration of 1 mg/100 gm body weight of Clindamycin-phosphate (Biomol) in 1 ml of sterile water. The S 15 Clindamycin was administered I.P. using a 1 ml 27-gauge tuberculin syringe (Terumo).
About 24 hours later, the hamsters were each infected orally with approximately 1 ml of sterile saline containing 1 x 104 C. difficile (strain ATCC 43596). The C difficile were grown for about 48 hours on CCFA (cycloserine-cefoxitin-fructose-egg yolk agar. a C. difficileselective and differential medium) plates (BBL) prior to inoculation.
Eight hours after inoculation (Day treatment was initiated. The hamsters in each of the three groups orally received one of three treatments through an 18-gauge feeding needle (Popper). Group I received 2 ml of pre-immune IgY (as an 8X PEG preparation). Group II ieceived I ml of immune IgY a mixture of antibodies generated against pMA1870-2680 [Interval A-6] and pPB1750- 2 3 6 0 [Interval and Group III received 2 ml of immune IgY.
The immune IgY mixture was prepared by mixing an equal volume of an 8X concentrate of IgY raised against pMA1870-2680 and an equal volume of an 8X concentrate of IgY raised against pPB1750-2360: the resulting mixture was designated A-6/B-3 IgY. The amount of anti-toxin protein specific antibodies contained in this A-6/B-3 IgY mixture was about 1.2 mg/ml of anti-recombinant toxin A IgY and about 400 tg/ml of anti-recombinant toxin B IgY. These amounts were determined by affinity purification as previously described in Example 15c. The amounts of total IgY in the 2 ml IgY dose was about 80 mg. and about mg in the 1 ml dose.
-240 The hamsters were treated once each day for 3 days. [The dosing schedule in this treatment regimen differs from that used previous Examples Example 32), where the hamsters were treated with 2 ml three times daily for 3 days.] All hamsters were observed for the onset of diarrhea and death during and after the treatment period. The results are shown in Figure 47 and Table 49.
In Figure 47. cumulative mortality (expressed as a percentage) is displayed along the ordinate and time (expressed in days) is displayed along the abscissa. The duration of the treatment period, indicated by the horizontal bar in Figure 47. was 3 days. The administration of Clindamycin and the inoculation with C difficile ("Infection") is indicated by arrows. The solid black squares represent the Group I hamsters hamsters treated with 2 ml of preimmune IgY). The open squares represent the Group II hamsters hamsters treated with I ml of the A-6/B-3 IgY). The solid black diamonds represent the Group III hamsters hamsters treated with 2 ml of A-6/B-3 IgY).
The results shown in Figure 47 demonstrate that all the hamsters 8/8) in Group 111 were protected from death. In contrast, only 13% 1/8) of the hamsters in both Groups 1 and II survived. The degree of protection in Group III was statistically significant at P 0.005. using Chi-square analysis.
The following table shows the results observed using A-6/B-3 IgY. The dose given in this table refers to the total (not specific) IgY concentration given to the animals in a I or 2 20 ml dose.
TABLE 49 Prevention of Morbidity In Vivo Using A-6/B-3 IgY Treatment Group Animals with Diarrhea Mean Weight' Pre-immune, 80 mg 100 67.3 3.9 (Group I) A-6/B-3. 40 mg 100 69 4.2 (Group II) A-6/B-3. 80 mg 0 81.6 (Group III) Weight expressed in grams mean starting weight of hamsters was 79 3.2 gm.
For the animals that died. weight was measured at the time of death. For survivors.
the weight was measured after the end of treatment.
241 The results shown in Table 49 demonstrate that morbidity was also prevented when the hamsters were treated using 2 ml of A-6/B-3 IgY per day. Morbidity from CDAD was defined here as including diarrhea and weight loss. As shown in Table 49. none of the hamsters in Group III presented with diarrhea. In contrast. all of the hamsters treated with either 1 ml of A-6/B-3 IgY (Group II) or with 2 ml of pre-immune IgY (Group I) presented with diarrhea. Moreover, all but one of the hamsters in Groups I and II died about 48 hours after presenting with diarrhea. The prevention of diarrhea in Group III was statistically significant (P 0.001) in comparison to the other two treatment groups.
In addition, the results shown in Table 49 indicate that hamsters treated with either 2 ml of pre-immune IgY (Group I) or 1 ml of A-6/B-3 IgY (Group II) lost 14% of their mean starting weight prior to infection. In contrast the animals treated with 2 ml of the A-6/B-3 IgY (Group III) gained about 2% of their mean starting weight at the end of the treatment period.
Based on the above results. the minimal effective therapeutic dose of specific IgY to both recombinant C difficile toxins A and B (pMA1870-2680 and pPB1750-2360; Intervals A-6 and B-3. respectively) necessary to prevent mortality and morbidity of CDAD in hamsters under the conditions set forth above in carbonate buffer) is about 2.4 mg of A-6 IgY and about 800 pg of B-3 IgY per day for 3 days.
EXAMPLE 43 Determination of Specific IgY in the Cecum of a Hamster Treated with Anti-Recombinant C diffici Toxin A and Anti-Recombinant C. difficile Toxin B The amount of specific anti-C. difficile toxin IgY found in the cecum of a hamster treated with antibodies raised against the recombinant toxin A (pMA1870-2680; A-6) and the recombinant toxin B (pPB1750-2 36 0; B-3) was determined. The results of this study provide an indication of the approximate therapeutic dose of anti-recombinant toxin A and antirecombinant toxin B IgY (A-6/B-3 IgY) that must be present at the site of infection to protect an animal infected with C. difficile.
The example involved a) recovery of cecal contents from an untreated hamster and from a hamster treated with anti-recombinant C. difficile toxin A and anti-recombinant
C
difficile toxin B IgY A-6/B-3 IgY), b) direct determination by ELISA of the amount of 242 specific A-6 IgY and B-3 IgY in the cecal contents, and c) direct determination by ELISA of total IgY in the cecum and indirect determination of specific IgY.
a) Recovery of Cecal Contents from an Untreated Hamster and from a Hamster Treated with Anti-Recombinant C. difficile Toxin A and Anti-Recombinant C difficile Toxin B IgY (A- 6/B-3 IgY) A hamster was treated according to the protocol for Group 111 animals as described in 10 Example 42 a hamster that was successfully treated against CDAD using 2 ml of 8X A- 6/B-3 IgY per day for 3 days). Four hours after the last dose of IgY was administered, the hamster was sacrificed using ether. A control hamster was treated with Clindamycin to predispose the animal to infection with C. difficile. but did not receive any IgY nor C.
difficile. served as a negative control. The control hamster was sacrificed two days after administration of Clindamycin.
The ceca from both hamsters were removed and most of the cecal contents were collected in a 15 ml centrifuge tube. Each centrifuge tube contained several mis of cecal material. The contents in each tube were vortexed and a I ml aliquot from each tube was removed to a 1.5 ml microcentrifuge tube. The microcentrifuge tubes were centrifuged for 1 20 minute at 14.000 rpm. The resulting supernatant was collected and the specific IgY in the samples was quantitated by ELISA.
b) Direct Determination by ELISA of the Amount of Specific A- 6 IgY and B-3 IgY in the Cecal Contents The levels of anti-recombinant toxin A and toxin B IgY present in the cecal contents were detected by ELISA using the protocol described in Example 13c with the following modifications. The 96-well microtiter plates were coated (100 pl/well) overnight at 4 0 C with 0.05 pg/ml of recombinant C. difficile toxin A protein [pPA1870-2680 (Example 29)] or with 1 pg/ml of recombinant C difficile toxin B protein [pPB1750-2360 (Example 18b)] in PBS. pH 7.4. Cecal samples were initially diluted two-fold into PBS and placed in the wells. The diluted samples were then serially diluted 5-fold in the microtiter wells. All samples were tested in duplicate.
243 Quantitation of specific IgY levels in the cecal samples was determined by comparing the antibody reactivity directed against either recombinant toxin A or recombinant toxin B to the reactivity generated using known amounts of affinity-purified IgY in ELISA assays. IgY specific for recombinant C. difficile toxins A and B were affinity purified as described in Example 15c. Briefly, affinity-purified antibodies were isolated from PEG-purified IgYs from the eggs of hens immunized with either pMA1870-2680 (Interval A-6) or pPB1750-2360 (Interval The PEG-purified anti-pMA1870-2 6 8 0 IgY was affinity-purified using a column comprising pMA1870-2680 bound to Actigel (Sterogene). The PEG-purified antipPB1750-2360 IgY was affinity purified using an Actigel column containing pPB 850-2360 10 (Example 15c). a recombinant C difficile toxin B protein that is 100 amino acids smaller than •pPB1750-2360.
The affinity-purified anti-pMA1870-26 80 IgY and the affinity-purified antipPB1750-2360 IgY were quantitated by measuring the absorbance at 280 nm and each preparation was diluted to a concentration of 10 pg/ml in PBS. The affinity-purified IgYs were tested by ELISA starting with an initial concentration of 10 pg/ml and at serial dilutions in the respective coated-microtiter plate along side the cecal extracts. Rabbit antichicken IgY conjugated with alkaline phosphatase (Sigma) was used as the secondary antibody at a dilution of 1:750 to detect the bound IgYs in the ELISA.
Comparison of the ELISA reactivity equivalence between the cecal extracts to the 20 known concentrations of the affinity-purified IgY allowed the amount of specific antirecombinant IgY present in the cecal extracts to be estimated. From the ELISA results the amount of specific anti-pMA1870-2 6 8 0 IgY was found to be about 12 ig/ml. The amount of specific anti-pPB1750-2 3 6 0 IgY was determined to be about 800 ng/ml.
These concentrations of specific IgYs provide estimates of the effective therapeutic concentrations necessary to achieve protection at the site of infection. Since the amount of specific IgY given'orally prior to collection of the cecal fluid was about 2400-2800 pgs against the toxin A recombinant and from 400-800 jlgs against the toxin B recombinant. there was approximately a 200-233 fold reduction and 500-1000 fold reduction in the detectable amount of anti-recombinant toxin protein IgY found in the cecum directed against Interval A- 6 and Interval B-3. respectively.
These results were obtained using antibodies present in carbonate buffer. If the orally administered anti-recombinant toxin IgY were properly protected from degradation during passage through the GI tract through use of an enteric coating as described in Example 244 37) the effective therapeutic dose required for oral administration would be less than that specified in Example 42.
c) Indirect Determination of the Amount of Specific Anti-Recombinant
C.
difficile Toxin A and Toxin B IgY in the Cecal Contents by ELISA In order to confirm the values determined in Example 43(b) for the amount of antirecombinant toxin IgY levels found in the cecum of a treated hamster, the following experiment was performed. The standard ELISA format was used (described in Example 13c) unless otherwise specified.
10 The cecal extracts from an untreated hamster (serving as the negative control) and one treated with both anti-recombinant toxin A and anti-recombinant toxin B IgY [described in section a) above] were used in this experiment. The total cecal IgY. not just the amount of cecal IgY specific for the recombinant C. difficile toxin proteins, was directly determined.
Because the percent amounts of specific antibody contained within the IgY preparations was known, the determination of the total IgY level present in the cecal extracts allowed the amounts of specific antibody in the cecal extract to be calculated.
A sandwich ELISA assay was used to capture IgY in the cecal material as follows.
Rabbit anti-chicken IgG (Cappel) at 0.1 ug/ml in PBS was used to coat a microtiter plate (100 Il per well) overnight at 4C. Both of the cecal extracts were tested at an initial dilution of 20 1:500 and at serial 5-fold dilutions to a final dilution of 1:312.500. All sample dilutions were tested in duplicate. Affinity-purified antibodies directed against recombinant toxin A (pPA1870-2680. Interval A-6) were diluted to 0.1 ug/ml and then further diluted serially by five-fold to a final concentration of 0.16 ng/ml. was also tested by ELISA for allow for quantitation by comparison. After incubation and washing, rabbit anti-chicken alkaline phosphatase IgG (Sigma) was added (at 1:1000 dilution) to the plates. The plates were then washed and substrate (p-nitrophenyl phosphate) was added and the plates were evaluated as described in Example 13c.
As described above in Example 43(b), the ELISA reactivity obtained using the affinity purified anti-recombinant toxin A IgY was matched to that ELISA activity generated in dilutions of cecal extract, to quantitate the amount of total IgY found in the cecum of the treated hamster.
From the results of the ELISA assay. the amount of total IgY in the cecum of the treated hamster was estimated to be 50 tg/ml. Affinity purification studies showed that total 245 IgY preparations comprised about 7% or 3.5 pg/ml IgY specific for recombinant toxin A (anti-A-6 IgY) and about 1-2% or 500-1000 ngiml IgY specific for anti-recombinant toxin B (anti-B-3 IgY). The concentrations of both of the specific IgYs detected here correlates fairly closely with the amounts detected above in Example 43(b), namely, 3.5 pg/ml versus 12 pg/ml for anti-Interval A-6 and 800 ng/ml versus 500-1000 ng/ml for anti-Interval B-3.
The results above and in section b) of this Example, both indicate that very low levels of specific anti-Interval A-6 and B-3 IgY's were detected at the site of the C difficile infection. Since the hamster was protected from CDAD. this level of anti-recombinant toxin A and B is within the therapeutic range. These results also support the proposition that much 10 lower levels of anti-recombinant toxin IgYs would need to be orally administered if they were delivered using means to prevent degradation in the GI tract enteric coating of IgY).
EXAMPLE 44 Treatment of Diarrheic Hamsters Usine Anti-Recombinant C difficile Toxin A Protein IgY To determine whether hamsters presenting with diarrhea after infection with C. difficile could be effectively treated using the anti-recombinant C difficile toxin A IgY alone or whether a combination of anti-recombinant toxin A and B IgY is required. the following 20 experiment was performed.
Hamsters were given Clindamycin and infected with C difficile essentially as described in Example 32c. The anti-recombinant toxin A IgY and anti-recombinant toxin B IgY were produced against pMA1870-2680 (Interval A-6) and pPB1750-2360 (Interval B-3).
respectively.
Three groups of hamsters (Sasco) were predisposed to C difficile infection by I.P.
injection of 1 ml of Clindamycin-phosphate (Biomol) at 1 mg/100 g body weight. About 24hours later, each hamster was challenged, using an 18 gauge feeding needle. with a 1 ml inoculum containing approximately 1 x 104 C difficile (ATCC 43596) organisms in sterile saline. The bacteria were grown for about 48 hours on CCFA agar (BBL) plates prior to infection.
The three groups, each containing nine to ten members. were given IgY preparations using a feeding needle. Group I received pre-immune IgY; Group II received antirecombinant toxin A IgY (anti-A-6 IgY); Group III received a mixture of anti-recombinant 246 toxin A IgY and anti-recombinant toxin B IgY (anti-A-6/B-3 IgY). Each IgY preparation comprised an 8X PEG prep in 0.1 M carbonate buffer, pH 9.5 and contained about 40 mg/ml of protein. To generate the anti-A-6/B-3 IgY mixture. equal volumes of the two 8X concentrates anti-A-6 and anti-B-3) were mixed together. The amount of specific antirecombinant C difficile toxin A IgY in the anti-A-6 IgY preparation was about 2.8 mg/ml.
The amount of specific anti-recombinant C. difficile toxin A IgY in the anti-A-6/B-3 IgY prep per ml was therefore half that of the A-6 IgY prep 1.4 mg/ml). About 200-400 pg/ml of specific anti-recombinant C difficile toxin B IgY was present in the anti-A-6/B-3 IgY preparation.
After the onset of diarrhea was detected in each individual hamster, that animal was dosed with 2 ml of their respective treatment preparations either pre-immune. anti-A-6 IgY or anti-A-6/B-3 IgY). The onset of diarrhea was detected in the hamsters from 20 to 44 hours post-inoculation with C difficile. The majority of the hamsters exhibited S: diarrhea within 24 hours post-inoculation with the organisms. The majority of the animals 15 were given 3 doses of IgY per day at roughly 4 hour intervals for 2 days: however, some hamsters were only dosed once or twice on the first day of treatment due to a later onset of diarrhea.
The results of this experiment indicated that the anti-A-6 IgY was able to protect many of the hamsters from death, even if given after the onset of diarrhea. About half of the 20 hamsters treated with anti-A-6 IgY survived for approximately one month, while only 11% of the hamsters treated with preimmune IgY and 20% of the hamsters treated with anti-A-6/B-3 IgY survived.
Because it appears that the anti-A-6 IgY is the most important component for prevention of death in the hamster model Example the results obtained in 25 hamsters treated with the anti-A-6/B-3 IgY (which contains half the amount of specific anti- A-6 IgY compared to the A-6 IgY alone preparation) was not unexpected (only 20% of the animals were protected from death). In this Example. anti-A-6 IgY alone could not prevent mortality in 50% of the hamsters, and anti B-3 IgY alone did not provide protection. In addition, the results obtained in previous studies Example 32c) indicated that the anti-B- 3 antibodies are more important in preventing the onset of diarrhea rather than in preventing death due to CDAD.
All of the animals that were.successfully treated with the anti-A-6 IgY exhibited mild diarrhea before treatment was started. If diarrhea was severe and neurological symptoms were 247present before treatment was initiated, the hamsters could not be successfully treated with oral anti-A-6 IgY.
The above hamster treatment experiment was repeated. with the exception that only pre-immune or anti-A-6 IgY were administered. Ten to eleven hamsters per group were treated with 2 ml of 8X IgY concentrates after diarrhea has been detected in each individual hamster. The treatment schedule was as described above.
Diarrhea was detected in the hamsters from about 20 to 45 hours after inoculation with C. difficile. Eighty-five percent of the animals (17/20) presented with diarrhea about 28 hours after infection. The data from these two studies were combined and are shown in Figure 48.
In Figure 48. the percentage cumulative mortality is displayed along the ordinate and the time (in days) is displayed along the abscissa. The treatment period is indicated by the use of the bar between days 2 and 3. The administration of Clindamycin and the inoculation with C difficile organisms (marked as "Infection" in Fig. 48) is indicated by the arrows. The solid black squares represent hamsters which received pre-immune IgY and the open squares 15 represent hamsters which received anti-A-6 IgY.
The data shown in Figure 48 demonstrates that 45 of the hamsters treated with anti- A-6 IgY post-diarrhea survived after treatment (diarrhea resolved itself in most of these hamsters). The results obtained using anti-A-6 IgY as compared to the results obtained using pre-immune IgY was statistically significant at P 0.05. All of the hamsters treated with 20 anti-A-6 IgY survived long term month after treatment until the termination of the experiment).
These results provide the first description of a treatment regime which can be used to treat hamsters after the onset of diarrhea due to infection with C. difficile. Furthermore, these results demonstrate that the anti-recombinant C difficile toxin A IgY is an effective 25 therapeutic even when administered at the late stages of CDAD. (i.e..after the onset of diarrhea).
EXAMPLE Treatment of Established C. difficile Infection Using Anti-Recombinant C difficile Toxin A Protein and Toxin B Protein IgYs Generated Using Two Different Adiuvants The ability of recombinant toxin A and toxin B proteins produced using the pET vector to elicit neutralizing IgY in the hens capable of protecting hamsters against C. difficile 248 infection was examined. In previous studies (Example 32 or Example 44). the IgYs were generated against recombinant C. difficile toxin A proteins expressed using the pMal vector (pMAl 870-2680. Interval Because recombinant proteins expressed using the pET system could be isolated in a more highly purified form. as compared to proteins expressed using the pMal vector, production of antibodies against the toxin A recombinant produced in pET (pPA1870-2680. A-6) was preferred.
The anti-pPA1870-2680 IgYs were tested in the hamster model along with antibodies raised against the toxin B protein also expressed using the pET vector (pPB1750-2360.
Interval The use of a common expression system to produce both recombinant toxins has definite manufacturing advantages. For example, the same affinity-purification columns and protocols can be used for both recombinants and both antigens should be of comparable purity and yield.
A further objective of this example was to generate antibodies against both pET produced A-6 and B-3 toxin proteins using the same adjuvant. In previous examples 15 (Example 32 or Example 44). the IgYs tested were generated against either A-6 or B-3 recombinants using different adjuvants (the RIBI adjuvant was used for the anti-recombinant *toxin A IgY and Freund's adjuvant was used for the recombinant toxin B IgY).
The example involved a) the immunization of hens with recombinant C difficile toxin proteins expressed using the pET vector and 2 different adjuvants and the determination of anti-recombinant protein IgY titers by ELISA and b) treatment of C difficile-infected hamsters using a mixture of Gerbu or Quil A-generated anti-recombinant toxin A and toxin B IgY.
a) Immunization of Hens with Recombinant C difficile Toxin Proteins Expressed Using the pET Vector and Two Different Adjuvants and the Determination of Anti-Recombinant Protein IgY Titers by ELISA Hens were immunized with recombinant proteins expressed using the pET vector; nickel column affinity-purified recombinant toxin A (pPA1870-2680) or the recombinant toxin B (pPB1750-2360) proteins were mixed with either the Quil A (Accurate Scientific) or Gerbu (CC Biotech) adjuvants. These two adjuvants were chosen on the basis of performance (shown in Example 35) and cost. The immunization protocol followed was basically that described in Example 249 Briefly. hens were immunized with 100 jig of pPA1870-2680 for the first four immunizations followed by two immunizations using 1 mg of protein. Hens were immunized six times using 1 mg of pPB1750-2360 per immunization. Five hundred microliters of a solution containing either recombinant toxin protein and either 5 pg of Gerbu or 75 pg of Quil A was administered sub-cutaneously to each hen. About I week after the last boost. the eggs were collected and the IgYs in each group (four groups of hens. using both toxin recombinants with both adjuvants) were extracted using PEG as described in Example 1. IgY from preimmune eggs was also processed.
The IgYs were resuspended in 0.1 M carbonate buffer. pH 9.5 at 8X yolk concentration (about 40 mg/ml) and an ELISA was performed (as described in Example to determine the anti-recombinant toxin A and anti-recombinant toxin B titers. The antibody titers generated against either the pPA1870-2680 or pPB1750-2360 proteins using either the Gerbu or Quil A adjuvants was found to be 1:62.500. By affinity purification, the oamount of specific A-6 and B-3 IgY using Gerbu was 4.3% and 1.0% respectively. The 15 amount of specific IgY using Quil A was 2.2% for A-6 and 1.9% for B-3.
b) Treatment of C. difficile-Infected Hamsters Using a Mixture of Gerbu or Quil A-Generated Anti-Recombinant Toxin A and Toxin B IgY S* 20 Equal volumes of the anti-A-6 and anti-B-3 IgY PEG preps generated in section a) using the same adjuvant were mixed. These preparations were designated A-6/B-3 Gerbu and A-6/B-3 Quil A. The hamster treatment study was performed exactly as described in Example 42. Six hours after challenge with 10' C difficile organisms (ATCC strain 43596).
the hamsters were treated with 2 ml of either pre-immune IgY or an immune IgY preparation S. 25 (A-6/B-3 Gerbu and A-6/B-3 Quil The hamsters were treated with 2 ml of IgY for two more days. once per day.
The results of this hamster treatment study are shown in Figure 49. In Figure 49. the percentage cumulative mortality is displayed along the ordinate and the time (in days) is displayed along the abscissa. The treatment period is indicated by the use of the bar between days I and 3. The administration of Clindamycin and the inoculation with C. difficile organisms (marked as "Infection" in Fig. 49) is indicated by the arrows. The solid black squares represent hamsters which received pre-immune IgY; the open squares represent -250 hamsters which received anti-A-6/B-3 Gerbu IgY and the solid black diamonds represent hamsters which received anti-A-6/B-3 Quil A IgY.
The results shown in Figure 49 demonstrate that both immune IgY preparations (A- 6/B-3 Gerbu and A-6/B-3 Quil A) completely protected the hamsters from death due to CDAD. Nine out of nine of the hamsters treated with either of the immune IgY preparations survived infection with C. difficile. while all nine of the hamsters treated with pre-immune IgY died. The survival rates seen using either the A-6/B-3 Gerbu or A-6/B-3 Quil A preparations were statistically significant compared to the result obtained using pre-immune IgY (P value of <0.001 using Chi-square analysis).
Three out of nine of the animals treated with A-6/B-3 Gerbu and one out of nine of the animals treated with A-6/B-3 Quil A presented with very slight diarrhea. The slight diarrhea seen in these treated hamsters (compared to the total absence of diarrhea seen in previous Examples. such as Example 32) may be due to the lower antibody titer of the preparations used here (1:62.500 versus 1:125.000). Additional booster immunizations should 15 increase titers to the 1:125.000 range for hens immunized using either Gerbu or Quil A adjuvants. and thus increase therapeutic potency against diarrhea.
The above results indicate that the recombinant C difficile toxin A and B proteins can "be produced using the pET vector (in place of the pMal vector) without deleterious effects upon the production of neutralizing antitoxin. Furthermore. the same adjuvant can be used in conjunction with the pET-produced proteins to elicit in vivo neutralizing IgY. Moreover, the same adjuvant can be used with both recombinant toxin proteins to produce a therapeutic antirecombinant IgY response in vivo.
EXAMPLE 46 Production of Antibodies Using a Mixture Containing Recombinant C difficile Toxins A and B in Hens The ability to raise chicken antibodies directed against both a recombinant C difficile toxin A and toxin B protein using a mixture of both proteins as a combined immunogen was investigated. The example involved a) immunization of hens with a mixture of recombinant C. difficile toxin A and B proteins and b) purification and detection of anti-recombinant
C.
difficile toxin A and toxin B IgY.
251 a) Immunization of Hens With a Mixture of Recombinant C difficile Toxin A and B Proteins Egg-laying Leghorn hens were immunized with a mixture of recombinant toxin A (pPA1870-26 8 0. Interval A-6) and recombinant toxin B (pPB1750-2360. Interval B-3) proteins: both recombinant proteins were expressed using the pET vector system. Two groups of hens (each containing 4 hens) were immunized with 500 pg of each recombinant protein mixed with either Quil A (Accurate Scientific) or Gerbu (CC Biotech) adjuvant. A total volume of 1 ml containing the recombinant proteins and either 5 pg Gerbu or 75 pg Quil A adjuvant was administered to each hen. The immunization protocol followed for each adjuvant was as described in Example 35. The hens were immunized twice 2 weeks apart.
b) Purification and Detection of Anti-Recombinant C. difficile Toxin A and Toxin B IgY About I week after the last boost. 3 eggs from each group were collected and IgY was 15 extracted using PEG as described in Example 1. The IgYs were resuspended in PBS (pH 7.4) at a 4 X concentration each containing about 20 mg/ml total protein. Preimmune IgY served as a negative control.
The amount of anti-recombinant toxin A (A-6 IgY) and anti-recombinant toxin B (B-3 IgY) antibodies present in the two immune IgY preparations was determined by ELISA 20 essentially as described in Example 13c or Example 43b. Briefly. the wells of a microtiter plate were passively coated with either the toxin A recombinant (pPA1870-2680) or the toxin B recombinant (pPB1750-2360). The IgY samples were initially diluted 250-fold. then serially diluted 5-fold. All samples were tested in duplicate. Rabbit anti-chicken IgG alkaline phosphatase (Sigma) diluted 1:1000 was used to detect the specific IgY.
S. 25 The results of these ELISA assays revealed that both recombinant toxin antigens were able to elicit an IgY response in the hen. The antibody titers are expressed as the reciprocal of the highest dilution that was found to be about 3-fold higher in ELISA reactivity compared to pre-immune the negative control) at the same dilution. The titers for both the anti-A- 6 IgY and anti-B-3 IgY generated using the Gerbu adjuvant were very low at about 1:250.
The titer for the anti-A-6 IgY and anti-B-3 IgY generated using the Quil A adjuvant was from and 25 fold-higher compared to the Gerbu adjuvant. Antibody titers generated using Quil A for the anti-A-6 IgY was greater than 1:6250 and greater than 1:1250 for the anti-B-3 IgY.
252 While the IgY titers against the recombinant toxins using Quil A are lower than the levels achieved previous Examples Example 32) (mainly because these hens in this example have been immunized only twice: in comparison the hens in previous examples were immunized 5 to 10 or more times and the resulting anti-toxin protein titers reached 1:100.000 or more). the results indicated that both recombinant toxin proteins appear to be equally antigenic in the hens and the antibodies produced to each were present at comparable levels.
The results also indicated that the level of the antibody response generated depends on the adjuvant used. It was found that the Quil A adjuvant invoked a higher anti-A-6 and anti-B-3 IgY response early during the immunization process in comparison to the results obtained using the Gerbu adjuvant.
EXAMPLE 47 Affinity Purification of Native C difficile Toxin A Using Anti-Recombinant C. difficile Toxin A Antibodies Avian antibodies (IgY) raised against recombinant C difficile toxin A protein were affinity purified using interval A-6 as the affinity ligand. The resulting specific antibodies were then immobilized on a solid support to purify native toxin A from C difficile (ATCC# 43255) organisms grown in dialysis bags submerged in BHI broth. The following example 20 describes the a) affinity purification of avian antibodies directed against a recombinant fragment of toxin A and generation of a toxin A affinity column. b) growth of C difficile organisms to produce toxin A and B in dialysis bag culture supernatants. c) affinity purification of toxin A. d) in vitro characterization of affinity purified C difficile toxin A, e) investigation of an alternate strategy for coupling the anti-A-6 IgY to a solid support to affinity purify toxin A. f) affinity purification of C difficile toxin A on affinity column ,generated by periodate oxidation of A-6 IgY and g) in vitro characterization of affinity purified C. difficile toxin A.
a) Affinity Purification of Avian Antibodies Directed Against a Recombinant Fragment of Toxin A and Generation of a Toxin A Affinity Column Antibodies specific for Interval A-6 (aa 1870-2680) of C difficile toxin A were affinity purified to provide reagents for the generation of an affinity column to permit purification of C. difficile toxin A from liquid culture supernatants and to provide an 253 immunoassay reagent to permit detection of C difficile toxin A in culture supernatant and affinity purified C. difficile toxin A samples.
i) Affinity purification of A-6 IgY Hyperimmune IgY from eggs containing antibodies to A-6 recombinant protein using Freund's adjuvant was extracted using the PEG fraction method (Example The antibodycontaining supernatant was applied to a A-6 affinity column. made by covalently coupling pPA1870-2680 protein (prepared in Example 29) to Actigel A affinity resin (Sterogene Biochemicals) according to manufacturer's instructions. Approximately 10.2 mg of pPA1870- 268 protein was coupled to 5 ml Actigel affinity resin The anti-A-6 IgY was eluted with Actisep elution media (Sterogene Biochemicals) as described in Example 15c. and dialyzed against PBS for 24-48 hours at 2-8°C.
ii) Coupling of Affinity-Purified Anti-A-6 IgY to an Activated Affinity Resin 15 to Make a C difficile Toxin A Affinity Column An initial toxin A affinity column was prepared as described in Example 48a below.
by coupling the anti-A-6 IgY to Actigel A affinity resin. By comparing the pre- and postcoupling absorbance values of the IgY at 280 nm. it was estimated that 58%. or about 7.6 mg, of the anti-A-6 IgY was coupled to the affinity resin.
b) Growth of C difficile Organisms to Produce Toxins A and B in Dialysis S: Bag Culture C. difficile strain #43255 was grown as described in Example 49b. sections iv and v.
below. SDS-PAGE/Western blot analysis was conducted to evaluate the C difficile dialysis bag culture supernatants for the presence of toxin A as follows.
The dialysis bag culture supernatant samples and a known toxin A sample purchased commercially were analyzed as described in Example 49b. section iv. with the exception that affinity purified A-6 IgY was used as the primary antibody for the western blot.
Following SDS-PAGE polyacrylamide gel) the proteins were transferred to nitrocellulose using a Milliblot transfer apparatus (Millipore) according to the manufacturer's instructions. The blot was temporarily stained with 10% Ponceau S. and blocked overnight in PBS containing I mg/ml dry milk. The preimmune and anti-A-6 IgY primary antibodies were diluted to I tg/ml in PBS containing I mg/ml BSA. and the appropriate antibody was 254incubated with the corresponding blot for 2 hours at room temperature with gentle agitation.
The strips were washed with PBS (10 mM sodium phosphate. 150 mM NaCI. pH BBS- Tween (0.1 M boric acid. 0.025 M sodium borate. 1 M NaC1. 0.1% v/v Tween 20) and PBS to remove unbound primary antibody, and incubated with Rabbit anti-chicken IgG alkaline phosphatase conjugated secondary antibody (Sigma Chemical Co). diluted 1:2000 in PBS/BSA. The blots were washed to remove unbound secondary antibody, and the strips were developed in BCIP/NBT substrate solution (as described in Example 48 below).
Both culture supernatant samples analyzed appeared to contain immunoreactive
C.
difficile toxin A when analyzed by Western blot. This protein co-migrated with the commercial toxin A and was recognized by the affinity-purified anti-A-6 IgY. The culture supernatant samples were pooled prior to affinity purification of toxin A. The pooled culture supernatants were not concentrated prior to loading on the affinity column.
c) Affinity Purification of C difficile Toxin A 15 The C difficile toxin culture supernatant samples were affinity purified as described in Example 48c. The volume of the Actisep fraction following elution and dialysis was 42 ml, 15 ml of which were removed and concentrated to 3 ml prior to analysis. A Centricon concentrator (Amicon) was used to concentrate the sample.
d) Analysis of C difficile Culture Supernatant, Actisep-Eluted Fraction and SColumn Effluent for the Presence of Toxin A In order to determine the presence or absence of toxin A in the Actisep eluted sample and effluent from the affinity column, these samples were analyzed by SDS-PAGE and o Western blot along with the culture supernatant starting material. These analyses were performed as described in section b above to evaluate the relative amount of toxin A in the samples and the efficiency of the affinity purification.
The resulting Western blot is shown in Figure 50. In Figure 50. lanes 1-3 were incubated with pre-immune IgY as the primary antibody and lanes 4-6 were incubated with anti-A-6 IgY as the primary antibody. Lanes I and 4 contain culture supernatant starting material; lanes 2 and 5 contain column flow-through and lanes 3 and 6 contain affinity purified toxin A.
The results shown in Figure 50 demonstrated that immunoreactive toxin A was detected in the culture supernatant starting material and the Actisep fraction. Furthermore. no 255 toxin A was observed in the column effluent sample. indicating most of the toxin was bound by the affinity column. There appeared to be significantly more toxin A in the starting material than in the Actisep fraction. Since the column effluent apparently contains no toxin A. the difference in toxin A amounts between the starting material and Actisep fraction suggested a significant amount of the toxin was still bound to column. even after Actisep elution. One possible explanation for the inability of the Actisep to elute all of the toxin A is the tendency for toxin A to bind nonspecifically to the carbohydrate region of molecules such as immunoglobulins. This is possible because the anti-A-6 IgY on the column is coupled via primary amines. which would allow for a subpopulation of the IgY to couple via the Fab region. leaving the carbohydrate-containing Fc region accessible for binding to toxin A.
e) Investigation of an Alternate Strategy for Coupling the Anti-A-6 IgY to a Solid Support to Affinity Purify Toxin A The possibility of coupling IgY to a solid support by periodate oxidation of the 15 carbohydrate region was next examined. This method of coupling was predicted to accomplish the following: 1) couple the IgY to the support via the Fc region. leaving the Fab regions accessible for binding to C. dificile toxin, and 2) alter the carbohydrates enough to eliminate or reduce the nonspecific binding of C. difficile toxin A.
20 i) Oxidation of Anti-A-6 IgY with Sodium Periodate The anti-A-6 IgY was oxidized using sodium periodate (Sigma Chemical Co) as follows. A sodium periodate stock solution was made by dissolving 25 mg of sodium periodate in 1.2 ml of distilled. deionized water. To six ml of A-6 IgY (at 2.7 mg/ml) in a 15 ml polystyrene tube 600 pl (0.1 volume) of the sodium periodate stock solution was added. The tube was then covered with aluminum foil and mixed gently at room temperature for 1 hour and 20 minutes. Glycerol was then added to a final concentration of 20 mM. and the tube was inverted for 10 more minutes. The solution was then dialyzed against 100 mM sodium acetate. 150 mM NaCI. pH 5.5 to remove the sodium periodate.
ii) Coupling of Oxidized IgG to Affi-Gel Hz Hydrazide Gel (BioRad) Six ml of Affi-Gel resin was washed with coupling buffer (100 mM sodium acetate, pH 5.5 and 150 mM sodium chloride). The oxidized anti-A-6 IgY was filtered through a glass fiber syringe filter to remove the precipitate which had formed during the oxidation 256 procedure. and a 100 pl aliquot was removed for A 0 ,o analysis. The washed Affi-Gel resin was added to the oxidized IgY in a 15 ml polystyrene tube. and the tube was inverted overnight at room temperature (Total volume 12 ml).
iii) Determination of Coupling Efficiency The anti-A-6 IgY-Affi-Gel affinity resin was poured into a BioRad Econo column and the unbound antibody was washed through the resin and saved for A, analysis. The resin was then washed with I bed volume of PBS (10 mM sodium phosphate. 0.5 M NaCI. pH This wash was also collected and saved for analysis. The resin was then washed with several more volumes of PBS. and treated with the Actisep elution buffer (Sterogene Bioseparations) to ensure no unbound antibody remained in the resin. By comparing the preand post-coupling values of the A-6 IgY. it was estimated that 95%. or 8.4 mg, of the IgY was coupled to the resin.
15 f) Affinity Purification of C. difficile toxin A on Affinity Column Generated by Periodate Oxidation of Anti-A-6 IgY STwo dialysis bag culture supematants. grown as described in Example 48b. sections iv and v. were pooled and concentrated to about 10.5 ml using an Amicon centriprep concentrator. The pooled, concentrated supernatants were then applied to the anti-A-6 IgY 20 Affi Gel affinity column and the column effluent was collected and reloaded several times to bind as much toxin as possible. The unbound protein was then removed by washing the column with several bed volumes of PBS and the bound toxin A was eluted with 2 bed 'i volumes of Actisep elution media. The column effluent was saved for analysis to evaluate the efficiency of the affinity purification. The Actisep-eluted toxin was then dialyzed against TBS for 24-48 hours at 2-8 0 C. and concentrated from 53 to 3 ml using a Centriprep concentrator (Amicon).
g) In Vitro Characterization of Affinity Purified C. difficile Toxin A i) Protein Assay The purified toxin concentration was determined using a BCA protein assay (Pierce) and was found to be 70 pg/ml. or about 210 pg total from 37 ml of culture supernatant.
indicating there was about 5.7 pg of toxin/ml of culture supernatant.
257 ii) Comparison of Toxin Purity and Retention Times by HPLC HPLC analysis was used to compare both the purity and retention times of the affinity purified toxin A samples. Commercial and affinity purified toxin A samples were applied to a Shodex KW 803 HPLC column and eluted with PBS. using a Waters HPLC system. The toxin A retention times were approximately 7 minutes for both toxin samples. suggesting the toxins are identical. Furthermore, the purities of both toxins were similar.
iii) Western Blot Analysis of Culture Supernatant Starting Material, Affinity Purified Toxin A and Column Effluent (flow through) 10 In order to evaluate the efficiency of the affinity purification and immunochemically identify the affinity purified toxin A. the culture supernatant affinity purified toxin A. and column effluent samples were electrophoresed by SDS-PAGE on a 5% gel under reducing conditions and transferred to nitrocellulose using standard methods. The blot was temporarily stained with 10% Ponceau S to allow the lanes to be marked and the remaining protein 15 binding sites were blocked overnight at 2-8"C with a PBS solution containing 1 mg/ml dry milk. The blot was cut into two halves, one of which was incubated with anti-A-6 IgY primary antibody. diluted to 1 gg/ml in PBS containing 1 mg/ml BSA. and the second half incubated with preimmune IgY diluted to 1 g/ml in PBS/BSA. After a two hour incubation in the presence of the primary antibody (with gentle agitation), the unbound primary antibody 20 was removed with successive washes of PBS. BBS-Tween and PBS. Rabbit anti-chicken IgY .alkaline phosphatase conjugated secondary antibody. diluted 1:2000 in PBS containing I mgiml BSA was then added to each blot. After two hours. the blots were washed to remove unbound secondary antibody and developed with BLIP/NBT (Kirkegaard and Perry) substrate solution. Color development was stopped by flooding the blots with water. The resulting Western blot is shown in Figure 51.
In Figure 51. lanes 1-7 were incubated with anti-A-6 IgY as the primary antibody and lanes 8-15 were incubated with pre-immune IgY as the primary antibody. Lanes 1 and 9 contain broad range molecular weight markers (BioRad). Lanes 2 and 10 contain C. difficile culture supernatant Lanes 3 and 11 contain C difficile culture supernatant Lanes 4 and 12 contain C. difficile culture supernatants #1 and #2 (pooled). Lanes 5 and 13 contain column flow-through. Lanes 6 and 14 contain affinity purified Toxin A (high load; 2X the load shown in lanes 7 and 15). Lanes 7 and 15 contain affinity purified Toxin A (low load). Lane 8 does not contain any sample material (blank).
258 The affinity purified toxin A sample (lane 7) was 3.5 fold more concentrated than the pooled starting material sample (lane however. 1/3 the volume (5 Pl vs 15 pl) of the affinity purified sample was loaded compared to the pooled starting material sample.
Consequently, if most of the toxin A was recovered from the column.the toxin A levels detected on the Western blot should be similar. As shown in Figure 51. the signals corresponding to the main high molecular weight bands are comparable. Therefore. the recovery of toxin A from the affinity column appeared to be quantitative.
EXAMPLE 48 10 Affinity Purification of Native C difficile Toxin B Usine Anti-Recombinant C difficile Toxin B Antibodies Avian antibodies (IgY) raised against recombinant C difficile toxin B protein (pPB 1750-2360: Interval B-3) were affinity purified using Interval B-3 aa 1750-2360 of C.
15 difficile toxin B) as the affinity ligand. The resulting purified anti-Interval B-3 specific antibodies were then immobilized on a solid support to facilitate purification of native toxin B derived from C difficile organisms (ATCC #43255) grown under conditions favorable for S* toxin production.
The example involved a) affinity purification of avian antibodies directed against a recombinant fragment of C difficile toxin B and generation of a C difficile toxin B affinity column. b) growth of C difficile organisms to produce toxins A and B in liquid culture and dialysis bag culture supematants. c) affinity purification of C difficile toxin B. and d) in vitro and in vivo characterization of affinity purified toxin B from C. difficile.
a) Affinity Purification of Avian Antibodies Directed Against a Recombinant Fragment of C. difficile Toxin B and Generation of a C. difficile Toxin B Affinity Column Antibodies specific for Interval B-3 of C difficile toxin B protein were affinity purified to provide reagents for the generation of an affinity column to permit purification of C difficile toxin B from liquid culture supernatants and to provide an immunoassay reagent to permit detection of C difficile toxin B in culture supernatants and affinity-purified C difficile toxin B samples.
259i) Affinity Purification of Anti-Interval B-3 IgY Hyperimmune IgY was extracted from eggs containing antibodies to the Interval B-3 recombinant protein (pPB 1750-2360) generated using Gerbu adjuvant (see Example 45) using the PEG fractionation method (Example The antibody-containing supernatant was applied to an Interval B-3 affinity column. made by covalently coupling pPB 1750-2360 protein (prepared in Example 29) to Actigel A affinity resin (Sterogene) as described in Example This fragment was chosen because it contains the C difficile toxin B repeat region and does not contain regions of homology with the C difficile toxin A protein, therefore the resulting purified antibody should not cross-react with C difficile toxin A. The anti-Interval B-3 10 antibodies (anti-B-3 IgY) were eluted from the column with 4 M guanidine HCI. pH 8.0 and dialyzed against PBS for 24 to 48 hours at 2-8 0
C.
ii) Coupling of Affinity-Purified Anti-B-3 IgY to an Activated Affinity Resin to Make a C. difficile Toxin B Affinity Column.
15 AC. difficile toxin B affinity column was made by coupling 11 mg of the affinity purified avian anti-B-3 antibodies prepared above to 5 ml of Actigel affinity resin (Sterogene).
A coupling time of 30 minutes was used rather than the minimum 2 hours recommended by the manufacturer in order to minimize the number of sites where each antibody molecule is coupled to the resin, thereby making the antibody more accessible to the toxin. In addition, the column was only exposed to high salt buffers or the Actisep elution buffer (Sterogene): no guanidine solutions were utilized during the preparation of the affinity column in order to minimize denaturation of the anti-B-3 antibodies. Comparison of the pre- and post-coupling absorbance values of the IgY at 289 nm. revealed that approximately estimate 62%. or about 6.8 mg. of the anti-B-3 IgY was coupled to the resin.
b) Growth of C difficile Organisms to Produce Toxins A and B in Liquid Culture and Dialysis Bag Culture Supernatants i) Liquid Culture of C. difficile in BHI Broth A frozen stock vial of C. difficile (ATCC #43255) was thawed and plated on CCFA plates (BBL) and grown for 36 to 48 hours at 37 0 C in an anaerobic chamber. Colonies were harvested from the CCFA plates using a sterile swab. The harvested colonies were used to inoculate a 20 ml liquid culture of BHI broth (BBL). This culture was grown for 260 approximately 24 hours in an anaerobic jar at 37°C. Ten milliliters of the overnight culture were used to inoculate a 500 ml liquid culture of BHI broth, and the culture was grown for approximately 72 hours at 37CC in an anaerobic chamber.
ii) Harvest of Liquid Culture Supernatant The 72 hour culture was centrifuged at 5000 rpm (4420 x g) for 10 minutes in a Beckman J2-21 centrifuge to pellet the C. difficile organisms and the supernatant was filtered through a 0.45 p filter (Nalgene) and saved for toxin purification and analysis at 2-8 0
C.
S 10 iii) In Vitro Analysis of Culture Supernatant to Detect Presence of C difficile Toxin B In order to determine whether C. difficile toxin B was present in the culture supernatant. the supernatant was analyzed by native PAGE and Western blotting as follows.
The harvested culture supernatant was concentrated about 10-fold using a Centricon concentrator (Amicon) prior to electrophoresis on native PAGE gels. The concentrated sample was then mixed with an equal volume of native gel sample buffer (50% sucrose. 0.1% bromophenol blue-) and loaded on a 4-15% Tris-glycine gradient gel (Bio-Rad). along with a known sample of C. difficile toxin B. purchased from Techlab. The samples were Selectrophoresed for 3 hours at 150 volts, constant voltage, using a Hoefer power supply.
Following electrophoresis. the gel was cut in half and one half was stained with Coomassie blue and destained with a solution comprising 10% glacial acetic acid/40% methanol to visualize any protein bands. The other half of the gel was blotted and probed using affinity purified anti-B-3 antibody (section i above).
The Coomassie blue stained gel is shown in Figure 52. In Figure 52. lane I contains broad range molecular weight markers. Lane 2 contains the commercial toxin B (Techlab); lane 3 contains BHI broth; lane 4 contains culture supernatant and lane 5 contains concentrated culture supernatant.
As shown in Figure 52. the commercial toxin B was detectable on the Coomassie stained gel. The concentrated culture supernatant showed several relatively faint bands.
however no detectable proteins in the supernatant samples co-migrated with the known C difficile toxin B sample. Furthermore, western blot analysis did not detect any proteins in the culture supernatant that were recognized by the affinity purified anti-B-3 antibody. These results demonstrated that the above growth conditions did not appear to be optimal for toxin production.
261 Miv) Production of C. difficile Toxin B in Dialysis Bag Culture In order to identify growth conditions optimal for the production of C. difficile toxin B. the following experiment was conducted. A liquid culture of C difficile 43255 was grown overnight as described above. The overnight culture was used to inoculate large scale cultures grown in dialysis bags containing PBS and submerged in BHI broth, as described by Meador and Tweten [Infection and Immunity. 56:7 (1988)]. Briefly. two 500 ml wide-mouth media bottles were filled with 400 ml of BHI broth. A dialysis bag having a molecular weight cut-off of 12-14.000 (Spectrapor) containing 25 ml of PBS was tied off at both ends and submerged in the BHI media in each 500 ml bottle. The bottles and dialysis bags were 10 then autoclaved for 30 minutes to sterilize the broth and PBS. The bottles were then incubated for 1 hour at 37 0 C under anaerobic conditions to cool the liquids and remove oxygen from the media.
The PBS in each dialysis bag was then inoculated with 10 ml of the overnight culture using a 10 cc syringe fitted with a 27 gauge needle to puncture the bags above the broth 15 level. The bottles were then incubated for 48 to 72 hours at 37 0 C under anaerobic conditions.
Both bottles showed heavy growth inside the dialysis bags. however one of the bottles also showed turbidity in the BHI broth outside of the dialysis bag. suggesting the BHI was contaminated. Therefore. the contents of the two dialysis bags were kept separate until the contents could be analyzed separately. It was thought that the BHI growth in the contaminated bag was due to C. difficile which may have fallen from the needle during inoculation of the dialysis bag.
v) Harvest of Dialysis Bag Culture Supernatants The dialysis bag contents were removed and centrifuged at 5000 rpm (3440 x g) for 10 minutes to pellet the cells and the dialysis bag culture supernatants from each bag were handled separately: each was filtered through a 0.45 p syringe filter and concentrated using a Centriprep 30 concentrator (Amicon). The two samples were concentrated from 35 ml to 3 ml. and stored at 2-8 0
C.
The culture supernatants from the dialysis bag liquid cultures were analyzed by native PAGE and western blot analysis to evaluate the amount of toxin B produced in the dialysis bag culture supernatants.
-262 vi) Native PAGE/Western Blot Analysis of C difficile Dialysis Bag Culture Supernatants Aliquots of the concentrated dialysis bag culture supernatants and a known C. difficile toxin B sample were electrophoresed on a 4-15% Tris-glycine gel (Bio-Rad) as described above in section iii). One half of the gel was stained with Coomassie blue and the other half was transferred to a nitrocellulose membrane for western blot analysis using a semi-dry transfer apparatus (Millipore)and standard transfer conditions (12 volts, constant voltage, for minutes). The remaining protein binding sites on the membrane were blocked overnight in PBS containing 1 mg/ml dry milk.
10 Figure 53 shows the resulting Coomassie stained gel and Western blot. In Figure 53.
lanes 1-4 represent the Coomassie stained gel and lanes 6-9 represent the Western blot. Lanes I and 6 contain broad range molecular weight markers: lanes 2 and 7 contain commercial toin B (Techlab): lanes 3 and 8 contain dialysis bag culture supernatant from the culture having sterile BHI broth: lanes 4 and 9 contain dialysis bag culture supernatant from the culture 15 having "contaminated" BHI broth: lane 5 is blank.
A shown in Figure 53. the presence of C. difficile toxin B was detected by incubating the blot strips with affinity purified anti-B-3 IgY. After washing the blots to remove unbound anti-B-3 antibodies, bound anti-B-3 antibodies were detected by incubating the strips with a secondary antibody comprising rabbit anti-chicken Ig conjugated to alkaline phosphatase (Sigma). The blots were washed again to remove any unbound secondary antibody and the blots were developed in freshly prepared BLIP/NBT substrate solution. Development was stopped by flooding the blots with water once an adequate signal was obtained.
The results of the PAGE and Western blot analysis showed that the amount of toxin B present in the dialysis bag supernatant samples was too dilute to be detected by staining with Coomassie. However. both culture supernatant samples (one from the bottle with sterile BHI broth and one from the bottle with contaminated BHI broth) contained immunoreactive toxin B when analyzed by Western blotting. The only difference seen between the two culture supernatant samples appeared to be in the amount of toxin B produced. The sterile broth sample appeared to contain more toxin B than did the contaminated broth sample.
Comparison of the commercial toxin B sample to the toxin B produced in the dialysis bag culture supernatant samples revealed that the culture supernatant sample contained a higher percentage of intact toxin B protein there was much less evidence of degradation in the form of minor immunoreactive bands present in the culture supernatant samples).
263 Because both culture supernatant samples contained toxin B (although at different concentrations). they were pooled prior to affinity purification.
c) Affinity Purification of C. difficile Toxin B The dialysis bag culture supernatant samples were pooled and applied to the toxin B affinity column [prepared in section Nonspecific proteins were removed by washing the column with PBS until the baseline OD was achieved. The bound protein was eluted using Actisep elution media (Sterogene) and was then dialyzed against Tris-buffered saline. pH (50 mM Tris. 150 mM NaCI). Following dialysis. the affinity purified protein was 10 concentrated from 40 ml to 4.5 ml using a Centricon 30 concentrator (Amicon).
o d) In Vitro and In Vivo Characterization of Affinity Purified Toxin B From C difficile In order to determine the presence or absence of C difficile toxin B in the Actisep- 15 eluted sample and effluent from the affinity column the flow-through). these samples were analyzed by native PAGE and Western blotting along with the culture supernatant starting material. These analyses were performed to evaluate the relative amount of C.
difficile toxin B in the culture supernatant and the efficiency of the affinity purification.
The affinity purified, culture supernatant. flow-through, and commercial C difficile toxin B samples were each mixed with an equal volume native sample buffer and loaded on a 4-15% native Tris-glycine gradient gel (Bio-Rad). The sample were electrophoresed for approximately 2.5 hours at 200 volts, constant voltage, using a Hoefer power supply, and transferred to nitrocellulose using a semi-dry blotting apparatus (Millipore) according to manufacturer's instructions. The blot was blocked ovemight using a solution containing 1% powdered milk in PBS. The blot was then incubated with affinity purified anti-B-3 IgY as the primary antibody and rabbit anti-chicken conjugated to alkaline phosphatase as the secondary antibody. The blots were handled as described in section b(vi) to permit visualization of the C difficile toxin B protein.
Figure 54 shows the Coomassie stained gel and corresponding Western blots. In Figure 54. lanes 1-3 were stained with Coomassie blue: lanes 5-10 were probed with anti-B-3 IgY and lanes 8-10 were probed with pre-immune IgY. Lanes 1. 5 and 8 contain affinity purified toxin B; lanes 2. 6 and 9 contain the column flow through; lanes 3, 7 and 10 contain commercial toxin B (Techlab). Lane 4 does not contain any protein (blank).
264 The following results were obtained upon western blot analysis. All three samples (culture supernatant. eluted protein and flow-through) contained immunoreactive toxin B.
These results indicated that the affinity purification protocol was successful in purifying some toxin B. However, as the flow-through fraction was found to contain significant amounts of toxin B the following modifications would be examined for the ability to further optimize the purification process coupling of B-3 IgY to Affigel hydrazide support (BioRad) via periodate oxidation of IgY).
ii) Yield of Affinity Purified C difficile Toxin B 10 The yield of affinity purified C. difficile toxin B was determined by BCA protein assay (Pierce), using BSA as the protein standard. This assay showed that the toxin B concentration was 73 ag/ml x 4.5 ml (volume of affinity purified material) 365 pg of toxin B. Approximately 70 ml of dialysis bag culture supematant was used as the starting material: therefore, about 5 pg toxin B was recovered per milliliter of culture. This yield was 15 consistent with previously reported yields using this method of culturing C difficile [7.8 pg toxin B/ml of culture supematant: Meador and Tweten (1988). supra].
iii) Measurement of the In Vivo Activity of the Affinity Purified C.
difficile Toxin B The in vivo activity of the affinity purified C difficile toxin B was determined by injecting various amounts of the purified toxin B preparation (described below) into 30 to gram female syrian hamsters. Another group of hamsters was injected with various amounts of a commercial toxin B preparation (TechLabs) for comparison with results previously obtained. The LD,o of the TechLabs preparation of C difficile toxin B was found to be about 5 pg for 30-40 g hamsters when administered I.P. (Example 19). At this concentration pg/30-40 g hamster). the hamsters died within about 3 hours post-I.P. injection.
The LD,o concentration of the affinity purified toxin B was determined by I.P.
injection of 1 ml of a solution containing either 5 or 50 pg of affinity purified toxin B diluted in saline. Two 30-40 gram hamsters were injected with each concentration of affinity purified toxin B. The hamsters injected with 50 pg of affinity purified material hamster died within 2 hours: the hamsters injected with 5 pg of affinity purified toxin B died within 4 hours. These results demonstrated that the toxicity of the affinity purified C. difficile toxin B preparation was comparable to the commercially available C difficile toxin B.
265 EXAMPLE 49 Diagnostic Agglutination Assay for the Detection of C. difficile Toxin A and Toxin B In this example, a rapid agglutination assay designed to detect C. difficile toxin A and toxin B in either culture supernatants or biological specimens such as feces was developed.
Affinity purified antibodies against recombinant C. difficile toxin A and toxin B from hens were used to passively coat small polystyrene particles. In principle, the particles coated with the specific avian antibodies (IgY) to toxin A and toxin B should form visible aggregates when they are mixed with a sample containing the toxins. This format should produce a specific, sensitive and rapid assay. Affinity purified IgY in this case confers specificity and sensitivity to C difficile toxin, while ease of use and speed of the assay is conferred using an agglutination assay format. This example describes: a) initial development of the agglutination assay for the detection of C difficile toxin A and toxin B: and b) evaluation and optimization of the agglutination assay.
a) Development of an Agglutination Assay for Detection of C. difficile Toxin A and Toxin B Antibodies were generated in hens using the toxin A recombinant (pMAL 1870-2680) S. and the toxin B recombinant (pPB1750-2360) using Freund's adjuvant as described in previous Examples. The recombinant toxin A antibodies (A-6 IgY) and the recombinant toxin B antibodies(B-3 IgY) were PEG fractionated the then affinity purified as described in Example 15c. The A-6 IgY was affinity purified against pPA1870-268 0 and the B-3 IgY was affinity purified against pB1750-236 9 The affinity-purified antibodies were then passively coated onto the polystyrene particles.
25 For each IgY preparation to be coated. 100 pl of a 5% bead suspension of 1 p beads (Spherotech Inc..Libertyville. IL) was removed and centrifuged for 2 minutes at 14,000 x g in a Beckman microfuge to pellet the particles. The particles were then washed with TBS mM Tris. 150 mM NaCI, pH 8) PBS-Tween (10 mM sodium phosphate. 150 mM NaCI, pH 7.2 0.05% Tween 20) and TBS. The particles were centrifuged for 2 minutes following each wash and the wash buffer was discarded. Following the last TBS wash. the particles were resuspended in I ml of the antibody coating solution: affinity purified avian A-6 or B-3 IgY at 100 pg/ml in TBS. PEG-fractionated preimmune IgY was also coated in the same manner to serve as a negative control in the agglutination assays. The particle suspensions 266 were then inverted at room temperature for 18 to 24 hours to allow the IgY to coat the particles.
To remove the unbound antibody. the suspensions were centrifuged for 2 minutes, the antibody solution was discarded, and the particles were washed as before (TBS. PBS-Tween.
TBS). After the last TBS wash. the IgY-coated particles were resuspended in 200 pl of TBS.
giving a 2.5% particle suspension.
In order to demonstrate that the particles were coated with IgY. 10 l of the particles were incubated with 5 pl of undiluted goat anti-chicken IgG (Fisher Biotech) in depression wells. Samples were then evaluated for macroscopic agglutination. Particles that had not been coated with IgY showed no agglutination in this procedure.
In order to demonstrate the feasibility of using affinity purified polyclonal avian IgY in this type of assay. the ability of A-6 IgY coated particles to agglutinate in the presence of various concentrations of toxin A was evaluated.
Commercial toxin A (Tech labs) was diluted 10-fold serially from a starting concentration of 0.29 mg/ml. using PBS containing BSA at 1 mg/ml as the diluent. Ten tl of S each dilution was mixed with 10 ll of the coated beads in a depression-well slide, and the mixture was incubated for 20 minutes at 37' C. The slides were then analyzed macroscopically for evidence of agglutination.
Strong agglutination was observed with the 1:10 and 1:100 dilutions. and weak agglutination was observed in the 1:1000 dilution. The dilutions greater than 1:1000 showed no agglutination. The pre-immune coated particles did not agglutinate at any dilution tested.
The 1:100 dilution of toxin A had a concentration of 2.9 tg/ml. Ten pl of the 2.9 lg/ml dilution contains 29 ng of toxin A. therefore the assay is sensitive to 29 ng of toxin A. or 2.9 pg/ml. The agglutination assay format appears to be suitable for detecting C difficile toxins 25 A and B.
Affinity purified polyclonal avian antibodies were most commonly used to coat the panicles, however the use of PEG-fractionated and water-diluted IgY preparations was also investigated, in order to determine if it was possible to increase the sensitivity of the agglutination assay by using polyclonal antibodies which might contain a population of high affinity antibodies lost during affinity purification.
PEG-fractionated polyclonal A2 IgY was used to coat I p polystyrene particles under conditions identical to those described above, and the particles were evaluated for sensitivity 267 in the C difficile toxin A agglutination assay. These particles were less sensitive than particles ccated with affinity purified IgY.
To investigate the possibility that residual PEG in the PEG-fractionated IgY may inhibit particle agglutination in the assay, A-2 IgY was extracted by the acidified water dilution method described by Akita and Nakai of Food Science. 57:629 (1992)].
Polystyrene particles were then coated with water diluted IgY under conditions identical to those described above, and the particles were evaluated for sensitivity in the C difficile toxin A agglutination assay.
It was determined that particles coated with water-diluted IgY preparations were less sensitive than particles coated with affinity purified IgY. Affinity purified IgY therefore appears superior to batch-fraciionated IgY preparations in this assay format. In order to increase the sensitivity and maintain the specificity of the agglutination assays, we then evaluated the effect of several other variables on the assay performance.
15 b) Evaluation and Optimization of the C. difficile Toxin A and Toxin B Agglutination Assay A-6 and B-3 IgY-coated beads were evaluated for their agglutinability with lowest amount of toxin sensitivity) and specificity. Instead of using PEG-fractionated preimmune IgY, affinity-purified IgY against an irrelevant antigen. C atrox snake venom.
was used to coat the particles as a negative control. Toxin A and toxin B were serially diluted in PBS from 1 pg/ml to 0.1 ng/ml. Ten pi of bead suspension was mixed with 20 p I sample in wells of glass agglutination plates. mixed well and rotated on nutator (Lab Quake) or manually for two minutes. Agglutination was read after two minutes. A completely uniform suspension was rated as a slightly gritty appearance was rated as and distinct 25 agglutination was rated as or according to the size of the aggregates.
Various parameters which affect the sensitivity and/or specificity of the assay such as bead size. concentration of coating antibody, temperature of reaction, pH of coating buffer.
antibodies generated using different adjuvants, final density of the beads w/v) and sample diluents were evaluated. Four different bead sizes 0.39 p. 0.81 pt, 1 p. and 1.2 t were initially evaluated. The 1 p bead agglutinated very rapidly. resulting in large aggregates with little or no non-specific agglutination. Hence. 1 p bead size was chosen for further optimization studies. Samples were initially diluted in PBS. If the beads autoagglutinated in PBS, PBS with I mg/ml BSA or PBS with 0.01% Tween-20 was substituted. Both diluents 268 prevented autoagglutination. but PBS with Tween-20 also inhibited the specific signal.
Various other blocking agents such as sucrose. BSA at higher concentration and gelatin were evaluated as diluents. PBS containing 1 mg/ml BSA was found to be optimal at preventing autoagglutination without inhibiting specific signal.
The density of the beads in final suspension was also evaluated. In order to improve the sensitivity and specificity. A-6 or B-3 IgY-coated latex particles were tested for their agglutinability at 1.25%. and 0.25% suspensions. All the bead suspensions except 2.5% resulted in no or low signal.. Antibodies generated using different adjuvants have different avidities and affinities, and hence agglutinate differently. It was known that antibodies with higher avidity and affinity form large and distinct aggregates. A-6 and B-3 IgY generated using Freund's and Gerbu as adjuvants were evaluated for their agglutinability at lowest concentration of toxin. Antibodies generated using Gerbu adjuvant were found to be better in giving distinct and large aggregates at 10-times lower concentrations of toxin A or toxin B. compared to antibodies generated with Freund's adjuvant.
15 The effect of antibody concentration/mg of beads with lower or higher incubation temperature was also tested. The polystyrene particles were coated with 20 Pg IgY or 50 pg IgY/mg of beads. and incubated at room temperature. 37 0 C. or 56 0 C. There was a direct correlation between higher concentration of coating antibody and higher temperature with respect to increase in sensitivity. However, it was found that coating the particles at higher temperature also resulted in increased non-specific signal. High pH and low pH coating buffers were evaluated in order to optimize maximum sensitivity and specificity. The polystyrene particles coated in 50 mM sodium acetate. 150 mM sodium chloride. pH 5.5 (low pH buffer) agglutinated non-specifically. while beads coated using 50 mM sodium carbonate.
pH 9.5 (high pH buffer) buffer increased sensitivity and specificity with A-6 IgY coated 25 particles, but not for B-3 IgY coated particles. The sensitivity and specificity of A-6 or B-3 IgY sensitized particles using various methods is summarized ir. Table -269 i Tabl Sunr of Results 0 A-6 IgY-sensitized beads B-3 IgY-sensitized beads Sensitivity Specificity Sensitivity Specificity conditions 100 50 neg Fees E vh 100 10 1 n Fees E.Coli 1g/m 1gm gm ng/rnI control ng/mI ng/nil nglml control A. 100 Pg/mI, lp, I-1S. 4 pH 7.5 (F/RT) Be 100 pg/mI. liu, TBS, PH 7.5 (G/RT) C. 100 pg/mI, Ip, Ills, nia PH- 7.5 (G/37TC) n/ D. -250 pg/mI, Ip, TBS, pH 7.5 (GRT) -250 2 pg~mI, Ip, TBS. PH 7.5, (G/37*C) Fg/mg of beads 0.81 p (Yamamoto, et+ at procedure 4 C. 30 pg/mg of beads 0.39 p (Yamamoto, et al procedure) II. IS50 p.g/mi.I +4 PHt 7.5 blocking proceduire+ 1. 150 Pg/ml, 0.81 P, TBS, pH 7.5, (G/RT) blocking procedure 1. 100 pg/mI, I, l-BS. PH 7.5, (FRT) n/ K. 100 pg/mi, lp. TBS, PH 7.5 9 9..
9 9 9. 9 .9.
999 99 9 9 9 9..
9 99 99 9 9 99 99 99 999 9 9 999 99 .9 9 *9 9 9 9 *9 9 9 9 9 9 *99 9 99 999 9 9 9. 9 99 50 Otg/ml, Ip li BS. n/a pH 7.5 (F/RT) n/ M. 250 jig/mI, hi, +4 Acetate. pH 5.5 (FRT) N. 250 ptg/mI, Ili,CO) buffer, pH 9.5, (F/RT) 0. -2S0pg/ml, IA. CO) buffer, pH1 9.5, (G!RT)I -250 jug/mi. lip, Glycine 44 44 n/a saline buffer, pli 8.2 (FIS6*C)+ 4+ Q. 250 jig/nil. 1.2p, pi I 4 44 +4 4 4 +41 (F/111) 7+ R. -same as Qexcept after 44 +4 4 4 overcoating w/BSAI Ex~planation of conditions: coating concentration of IgY. head size, coating solution. ph I, adjuant used. temlperature during coating.
No autoagglutinatioi wvith sensitivity of I ng/nil in presence of PI3S4BSA40.01% TW2O. however nonspecific agglutination did occur wvith E. co/i and Mv feces even with this diluent.
F IgY generated using Freund's adjuvant G IgY generated using Gerbu adjuvant E. co/i E coli colony was picked from agar plate and resuspended in 500 PI diluent Fees 50 mg mouse feces wvas suspended in 500 gal diluent vortexed and spun at 14,000 x g for 2 min. supemnatant was used in the assay rl1 20 Based on the information from various parameters tested, the following bead coating protocols with A-6 and B-3 IgY were established: i) A-6 IgY coated particles to detect toxin A directly from feces of human patients.
Five mg polystyrene particles (1 Spherotech Inc., Libertyville. IL) were added to a tube and washed with I ml of TBS. PBS-T. and TBS followed by another wash with 50 mM Na,CO 3 pH 9.5 buffer. The beads were resuspended in the latter buffer to a total volume of 1 ml. A-6 IgY (affinity-purified. Gerbu-generated) was added to beads to a final concentration of 250 g/ml and incubated at room temperature on a nutator overnight. The next day, IgY-sensitized particles were washed with TBS. PBS-T. and TBS and resuspended in TBS to a final concentration of These IgY-sensitized particles were stored at 4"C until use.
ii) B-3 coated latex particles to detect toxin B directly from feces of human patients.
Five mg polystyrene particles (I g. Spherotech Inc.. Libertyville. IL) were o..
added to a tube and washed with 1 ml of TBS. PBS-T. and TBS followed by another wash S. with 50 mM NaCO3. pH 9.5 buffer. The beads were resuspended in the latter buffer to a 0 20 total volume of I ml. B-3 IgY (affinity-purified. Gerbu-generated) was added to beads to a final concentration of 100 pg/ml and incubated at room temperature on a nutator overnight.
The next day, IgY-sensitized latex particles were washed with TBS. PBS-T. and TBS and resuspended in TBS to a final concentration of These IgY-sensitizcd latex particles were stored at 4*C until use.
25 The agglutination assay to detect toxin A and B from feces was compared with commercially available assays to detect toxin A and toxin B from human stool specimens.
CyctocloneTM A+B EIA (Cambridge Biotech). which detects both toxins A and B, and ****PremierTM C difficile Toxin A Test (Meridian Diagnostics Inc.). which detects only toxin A, were used for comparison. Normal human stool specimens were processed according to each of the manufacturer's instructions. Stool samples were spiked with 1 pg/ml of toxin A or toxin B and serially diluted 10-fold. to 0.01 ng/ml toxin A and 0.1 ng/ml toxin B. For agglutination assays, stool was diluted 5-fold with PBS containing 1 mg/ml BSA and centrifuged at 2500 xg for 3 minutes. The supematant was then used in the assay.
272 EIA's were performed according to the manufacturer's instructions, and results were read spectrophotometrically. Interpretation of results was made based on optical density values and the manufacturer's recommendations. For the agglutination assay, 10 pl suspension of A-6. B-3 IgY- or non-specific IgY-sensitized particles were placed in the wells of glass agglutination plates. Twenty pl samples were added to each well. mixed well, and rotated on a nutator. Agglutination was read visually after 2 minutes of rotation.
Interpretation of results was made as described earlier. The summary of the results is presented in Table 51. The agglutination assay of the present invention detected toxin A at 1 ng/ml. while both the CytocloneTM A+B EIA. and PremierTM C difficile toxin A test detected toxins at 10-fold lower levels. Toxin B was detected at I ng/ml. using both agglutination and Cvtoclone A+B EIA. The results show that agglutination assay of the invention, is simple, easy to perform. and very rapid, as the results can be obtained in 5 minutes.
273 TABLE SI Comparison of Results of Three Different Methods to Detect Toxin A and Toxin B Spiked in Normal Human Stool Specimens C vtoclone' Premier' C d~iJile Agglutination assay for K.B EIA toxin Atest Toxin A& B Cambridge Biotech Meridian Diagnostics Ophidian Pharmaceuticals -Stool spiked with Toxin A 100ng/ml ng/mI I ng/nal 0.1 ng/mI 0.01 ng/mI ND
ND
20 Stool spiked with Toxin B 100 ng/mI N/A 25 10 ng/ml 1 ng/ml++ 0.1 ng/mI 30 1 I otal i ime 150 nun 150 min 3 min
S
S. S
S
*5
*SS*
S
*5 S S S
*S
*SSSS.
*5 S
S
S.
S
*5*5 ND Not determined N/A Not applicable EXAMPLE Characterization of Hamsters After Successful Treatment with Avian Antibodies Directed against Recombinant C difficile Toxin A and Toxin B Proteins In order to investigate why hamsters treated with IgYs directed against recombinant toxin A (A-6 IgY) and recombinant toxin B (B-3 IgY) before or after challenge with C difficile do not relapse and contract C diffictle associated disease CDAD) after the withdrawal of treatment, the following experiment was performed.
274 Relapse is commonly seen in hamsters (as demonstrated in Example 33) and in humans treated with drugs, such as vancomycin or metronidazole, to combat CDAD once drug treatment is terminated. In contrast, data presented in Examples 16 and 32 show that IgYs directed against recombinant C difficile toxin A alone (given prophylactically) or a mixture containing IgYs directed against recombinant toxin A and B proteins (given therapeutically) can be used to successfully prevent or treat CDAD and also prevent relapse.
The example involved a) the detection of C. difficile organisms and toxins in the feces of hamsters treated with anti-A-6/B-3 IgY, b) the detection of anti-C difficile toxin A and anti-C difficile toxin B IgG in the serum of treated hamsters by ELISA, c) the detection of anti-C. difficile toxin A and anti-C. difficile toxin B IgA in the saliva of treated hamsters by ELISA and d) re-exposure of A-6/B-3 treated hamsters with antibiotics.
a) Detection of C difficile Organisms and Toxins in the Feces of Hamsters 15 Treated with A-6/B-3 IgY The 7 hamsters that were successfully treated with 2 ml per day of A-6/B-3 IgY and the lone surviving hamster treated with 1 ml per day of A-6/B-3 IgY (Example 42) were tested for the presence of C difficle and toxin A and toxin B in fecal material after treatment was withdrawn. This determination was performed to investigate whether hamsters treated A-6/B-3 IgY were protected from relapse because the IgY treatment either reduced or completely eliminated C difficile organisms and toxins from the GI tract of the treated hamsters.
Stools were collected from the 7 individual hamsters 4 days after termination of treatment with A-6/B-3 IgY. A suspension was made from the stool samples as follows.
25 Fifty milligrams of feces were added to 100 pl of PBS (pH 7.4) and the mixture was suspended by vortexing the sample. An aliquot (50 pl) of each suspension was streaked unto a C difficile selective agar plate (CCFA plates;BBL) and the plates were incubated for 48 hours under anaerobic conditions. The remaining suspension was tested for the presence of C difficile toxin A and toxin B using the toxin agglutination assay described in Example 49.
The results obtained by culturing stool suspensions on the CCFA plates demonstrated that all of the hamsters successfully treated with A-6/B-3 IgY still harbored C difficile organisms 4 days after treatment (ranging from approximately 6-100 colonies).
275 Furthermore, C difficile toxin A was detected in the feces from all nine treated hamsters using the agglutination assay (Example 49). Surprisingly, C. difficile toxin B was not detected in the feces of any of the hamsters.
Stool samples were collected from the same 7 hamsters again about 5 weeks after the termination of antibody treatment. Suspensions were prepared and plated onto CCFA plates as described above. After this prolonged period, the presence of C. difficile was only detected in the stool from one of the hamsters. Interestingly, the organisms were detected in the hamster that was treated with the lower (1 ml) dose of A-6/B-3 IgY. Only a low number of colonies (5 colonies) was detected in the stool of that animal. In control animals there were no organisms detected, as normally, only a very low percentage of hamsters have detectable levels of organisms..
These results indicate that although the A-6/B-3 treated hamsters have been successfully treated for CDAD and the disease does not relapse, they still shed C difficile organisms and contain toxin A in their feces early after treatment. The anti-recombinant 15 C difficile toxin A and B antibodies A-6/B-3 IgY) apparently eliminate disease 0* symptoms without completely eliminating C. difficile organisms or toxin A from the GI tract of the treated hamsters. While not limiting the invention to a particular theory of action, the avian IgYs may exert their therapeutic effects by lowering the level of toxin present and thus possibly reducing organism number enough to not only prevent CDAD but also prevent CDAD from re-occurring, as it is possible that toxin A may aid in the colonization of C. difficile.
Five weeks after treatment with the avian antitoxin preparation, C difficile organisms were not detected in the feces of most of the treated hamsters. These i results indicate that long-term colonization of the GI tract by C difficile does not occur 25 following treatment with A-6/B-3 IgY.
b) Detection of Anti-C dfficile Toxin A and Anti-C dificile Toxin B IgG in the Serum of Treated Hamsters by ELISA Serum was collected from hamsters following treatment with anti-A-6/B-3 IgY to determine if an endogenous serum IgG response directed against C. difficile toxins was elicited in the treated hamsters. The generation of an anti-toxin IgG response could account for the prevention of subsequent relapse in the animals.
276 Blood was collected by cardiac puncture in the seven hamsters that were still available after five weeks (described above) four days after termination of treatment. The blood was allowed to clot and serum was prepared by centrifugation of the clotted sample.
Serum was also collected from an uninfected hamster and from a hamster vaccinated with a mixture of recombinant toxin A and toxin B proteins (Example 39) to serve as negative and positive controls, respectively. The ELISA was conducted using the protocol described in Example 1. Briefly, the wells of the microtiter plates were coated with 0.05 Ug/ml of the toxin A recombinant pPA1870-2680 or 1.0 gg/ml of toxin B recombinant pPB 1750- 2360 at 100 pl per well. Serum samples were tested at a starting dilution of 1:50 followed by 5-fold serial dilutions. Goat-anti-hamster IgG alkaline phosphatase (Southern Biotechnology Assoc.) was used at a dilution of 1:1000 as the secondary antibody. All antibody incubations were carried out at 37 0 C for 2 hours. The plates were developed for minutes using para-nitrophenyl phosphate (Sigma).
The results of the ELISA demonstrated that all of the serum samples from the test 15 hamsters contained significantly lower levels of anti-toxin A and anti-toxin B IgG as compared to the positive control serum (serum from a hamster that generated a protective *ooo. IgG response after active immunization with recombinant toxin A and B proteins).
Antibody titers present in the serum from the 7 treated hamsters were comparable to those present in the negative control. These results demonstrated that the protection from CDAD relapse achieved by treatment of hamsters with the A-6/B-3 IgY is probably not due to the generation of an active serum IgG response in the hamsters following infection with C difficile organisms.
c) Detection of an Anti-C difficie Toxin A and Anti-C difficile Toxin B IgA 25 Response in the Saliva by ELISA in Treated Hamsters To investigate whether the protection from relapse from CDAD seen in the anti-A- S' 6/B-3 IgY treated hamsters was due to the production of a mucosal IgA response in the animals, the following experiment was performed. Saliva was collected from 6 hamsters previously treatment with anti-A-6/B-3 IgY (Example 42; 2 ml) using pilocarpine (Sigma) which causes hyper-secretion of saliva. Hamsters were injected I.P. with a solution containing pilocarpine (1 mg/ml) in sterile water; 1 to 3 mgs pilocarpine was administered to the animals. Saliva was collected from 6 hamsters using a pipettor. As a negative control, saliva was collected from an mouse given 200 pg of pilocarpine. A mouse 277 specimen was used as a negative control because the only anti-IgA conjugate commercially available is a goat anti-mouse IgA (this reagent has been reported to cross-react with hamster IgA).
The ELISA was performed as described above with the following modifications.
The saliva samples were tested at an initial 1:10 dilution followed by serial five-fold dilutions. Goat anti-mouse IgA (Southern Biotechnology Assoc.) was used as the secondary antibody at a 1:1000 dilution.
The results of the ELISA showed that saliva from only 2 out of the 6 treated hamsters contained levels of anti-toxin A and anti-toxin B IgA higher than that seen in the mouse negative control. The saliva of the two anti-toxin IgA-positive hamsters had fairly low titers (between 1:250 and 1:1250). Typical hyper-immune IgA titers normally range about 1:10,000 or greater. The remaining 4 hamsters did not have a significant anti-toxin IgA response to either toxin A or toxin B as compared to the negative control. Since all six of the hamsters were successfully treated against CDAD relapse and the majority (4/6) 15 of the treated hamsters did not produce a significant anti-toxin IgA response, it is unlikely that the prevention of relapse was due to the generation of an anti-toxin A or anti-toxin B IgA response by the hamster.
The results shown in sections b) and c) indicate that the protection against relapse seen in hamsters successfully treated with IgY directed against C difficile Intervals A-6 and B-3 is not due to the production of a anti-C difficile toxin humoral response by the host.
Thus, prevention of relapse is a function of the administration of the IgY preparations.
This indicates that the host's immune status may not be relevant in terms of prediction of disease outcome survival) or whether relapse will occur. This is important as many of the patients who would most benefit most from treatment with an A-6/B-3 IgY therapeutic are immunocompromised.
d) Re-exposure of A-6/B-3 Treated Hamsters with Antibiotics As shown in section a) above, the treated hamsters still possess detectable levels of C. difficile organisms in their feces (4 days after termination of IgY treatment) and thus have the potential for developing CDAD. An experiment was performed to investigate whether re-exposure of these treated hamsters to Clindamycin would initiate onset of
CDAD.
-278- Four of the same hamsters used in the above experiments in Example 42) that were successfully treated with A-6/B-3 IgY were again predisposed to C. difficile infection by administration of Clindamycin-phosphate. Seven days after termination of the initial antibody treatment, the hamsters were given another I.P. injection of Clindamycinphosphate (Biomol) at 1 mg/100 g body weight. Twelve days post-Clindamycin predisposition the second application of Clindamycin), none of the hamsters developed any signs of CDAD.
These results demonstrated that once the hamsters are successfully treated with the anti-A-6/B-3 IgY, they were resistant to developing CDAD even after another exposure to Clindamycin. To take this result another step further, the same hamsters were given another antibiotic, namely Cefoxitin (Sigma) which is also known to predispose the hamsters to C. difficile infection. This was done as it was possible that the prevention of CDAD in the hamsters after the Clindamycin re-treatment was due to the generation of Clindamycin-resistant normal flora which may have prevented colonization of the GI tract 15 with C difficile. The 4 hamsters were each given a subcutaneous injection of 10 mg of Cefoxitin in saline 11 days after (18 days post treatment with A-6/B-3 IgY). This dosage of 0* Cefoxitin is known to predispose hamsters to CDAD.
Seven days post-Cefoxitin treatment, 1 of the 4 hamsters developed diarrhea and died. The remaining 3 hamsters remained healthy and have survived long-term at least one month). The results obtained after treatment with Cefoxitin indicate that protection from the re-occurrence of CDAD in the treated hamsters is probably not due to the development of specific antibiotic resistance resistance to Clindamycin) in the hamster flora.
Together the above results showed that hamsters treated for CDAD using anti-A- 25 6/B-3 IgY contain viable C difficile organisms and C difficile toxin A in their GI tract early after the withdrawal of treatment and yet the hamsters do not relapse. Even more.
surprising was the finding that while the hamsters still harbor C. difficile in the gut, they were resistant to a subsequent challenge using antibiotics capable of predisposing hamsters to CDAD. As was shown above, 5 weeks after withdrawal of the avian antitoxin, the hamsters no longer shed organisms into feces and thus were probably no longer colonized by C difficile. The results further indicate that oral administration of A-6/B-3 IgY to hamsters not only successfully treated CDAD, but also conferred resistance to relapse.
279 Moreover, the A-6/B-3 IgY protected the hamsters against CDAD from repeated antibiotic predisposition using two different antibiotics.
From the above it is clear that the present invention provides antitoxins and vaccines for the treatment and prevention of C. difficile disease. Furthermore, these antitoxins prevent the relapse of C. difficile disease which is commonly seen using conventional treatment protocols. Additionally, the invention provides a rapid agglutination assay for the detection of C difficile toxins A and B in samples.
EXAMPLE 51 Formation and Enteric Overcoating of Tablets Containing Avian Antitoxin Directed Against Clostrdial Toxin Proteins For Oral Delivery This example describes the formation of tablets containing chicken IgY directed against Clostridial toxin proteins suitable for oral delivery for the effective treatment for C.
difficile disease. As shown in Example 43, when IgYs are administered orally to hamsters in a carbonate buffer, only a very small amount of the delivered antibody is detected at the cecum, the relevant site where infection occurs. Most of the IgY is probably hydrolyzed in the acid environment or degraded by proteases in the stomach. Furthermore, much of the remaining functional IgY passing through the stomach would then be digested by the 20 various enzymes found in the small intestine. The low levels of IgY found in the cecums of treated hamsters is supported by the work of Losch ct al. who has found in in vitro experiments that chicken IgYs are very sensitive to the effects of low pH and intestinal proteases. In order to maximize the effectiveness of a given dose of an oral antibody therapeutic, experiments were performed to determine if the PEG-fractionated IgYs could 25 be tableted for easy administration and enterically overcoated to prevent degradation in the GI tract. The Example involved the formation of tablets containing PEG-purified anti- A-6 IgY, overcoating the IgY tablets with a pH sensitive enteric film, testing the dissolution profile of the overcoated IgY tablets, determination of the stability of the IgY reactivity after tableting, enteric overcoating and dissolution by ELISA and (e) demonstration of the retention of the ability of the tableted IgY to neutralize C difficile toxin A in vivo.
280 a) The Formation of Tablets Containing PEG-Purified Anti-A-6 IgY Chicken IgY from hens immunized with the recombinant C difficile toxin A protein pMA 1870-2680 (A-6 region) was fractionated by PEG as described in Example 37(a) IgYs from eggs collected from hens immunized with the recombinant protein were purified by PEG-precipitation and after purification the IgY pellets were resuspended in 0.1X PBS, pH This protocol differs slightly from the protocol described in Example 1 in that 0.1X PBS is used here (and Ex. 37a) rather than IX PBS (Ex. The change to 0.1X PBS was done primarily to reduce the final proportion of salt in relation to IgY in the final dried preparation.
One-hundred thirteen eggs were fractionated and the final IgY pellet after the 12% PEG step was resuspended in process water at 1/4 the yolk volume. The resuspended IgY in water was transferred to lyophilization vessels and quick-frozen with on dry-ice in reagent alcohol. The vessels were rotated in the dry-ice bath to allow for even freezing of the IgY solution by layering on the walls of the vessel. The frozen IgY solution was 15 placed on a Labconco Freeze-Dry System/Lyph Lock 4.5 apparatus and lyophilized for about 18 hours until dry. The final lyophilized IgY weighed 13.56 grams or about 120 mgs of dried material per egg.
Twelve grams of the lyophilized IgY were processed into forty, 250 milligram tablets using a Stokes B2 tablet press. Conventional flat-face 1/4 inch die tooling was used.
The tablets were prepared by double compression using 4500 pounds of pressure and were hard and flat-faced with an average weight of 256 mgs 6 mgs.
a.
b) Overcoating of the IgY Tablets with a pH Sensitive Enteric Film The tablets were coated with Eudragit S-100 (Rohm Tech. Inc. Maiden, MA), an 25 enteric film coating (methacrylic acid copolymer Type B, USP/NF) that is soluble in solutions from pH 7.00 and above. A stock solution of Eudragit S-100 was prepared according to the manufacturer's instructions. Briefly, Eudragit S-100 was dissolved by mixing 13 parts (by weight) of Eudragit S-100 (12.5% of dry polymer substance) in a mixture of 82 parts (by weight) of isopropyl alcohol and 5 parts (by weight) of process water. The enteric coating film was prepared by weight in grams as follows: 480 g of the Eudragit S-100 12.5% solution, 6 g of triethyl citrate as the plasticizer, 30 g of talc as the anti-adhesive, 50 g of process water and 434 g of isopropyl alcohol. The solid content of the final suspension was 9.6% and the content of the dry polymer substance was -281 The enteric coating mixture was placed in a beaker and individual tablets were dunked into the solution for about 1 second using fine tipped tweezers. The coated tablets were then allowed to dry at room temperature on a sheet of parafilm. Some tablets were dipped again in the enteric coating an additional two times with room temperature drying between coatings. This was done to ensure a more complete overcoating of the tablets.
The tablets dipped once and three times in the enteric solution were designated as Ix and 3x tablets, respectively.
c) Testing the Dissolution Profile of the Overcoated IgY Tablets Dissolution studies were conducted to determine the disintegration kinetics of the enteric tablets using the methods described in Example 37(b). The dissolution of the tablets was conducted under two conditions: either in simulated gastric solution, pH 1.2 or simulated intestinal solution at pH 7.5, both prepared using USP guidelines. Each tablet was weighed and placed in a beaker containing the respective solutions at 10 mg tablet per 15 ml of solution. The following forms of tablets were tested: 1) uncoated IgY tablets, 2) Ix coated IgY tablets and 3) 3x coated IgY tablets.
The tablets were allowed to dissolve by gentle mixing using a stir bar at room temperature. At various times, aliquots of each were taken and the absorbance at 280 nm was measured to quantitate the amount of IgY in solution. The dissolution profile of the Eudragit overcoated IgY tablets are shown in Figure In Figure 55, the absorbance at 280 nm is plotted against time in minutes. The release of IgY from the uncoated IgY tablets in gastric or intestinal fluid is shown by the solid black squares and the open diamonds, respectively. As shown in Figure 55, the rate .i of dissolution of the uncoated IgY tablet was slower in gastric solution compared to 25 intestinal solution. This inherent physical property of the uncoated tablet is an unexpected advantage of the tablet, making it naturally more resistant to gastric dissolution.
The dissolution profile of the Ix and 3x coated IgY tablets placed in the intestinal fluid is shown by the black triangles and the open triangles in Figure 55. As shown in Figure 55, both the lx and 3x coated tablets dissolved at similar rates with complete dissolution occurring after about 1 hour. Moreover, the dissolution rates of the enteric coated IgY tablets was slightly faster than the uncoated IgY tablet in the intestinal fluid. In contrast, the Ix or 3x coated IgY tablets in gastric fluid (shown by the open boxes and the black triangles, respectively in Figure 55) dissolved very slowly and only a small fraction of -282 the total IgY contained in the tablets was released into the solution. The difference in the dissolution profile of the lx or 3x coated IgY tablets was minimal. From these results, the Eudragit over-coated IgY tablets properly opened into the solution of the simulated intestinal fluid at pH 7.5 in a time dependent manner, but largely remained intact in the gastric fluid at pH 1.2.
The dissolution studies described above demonstrate that the IgY, formulated as a tablet, can be successfully enterically coated.
d) Determination of the Stability of the IgY Reactivity after Tableting, Enteric Over-Coating and Dissolution by ELISA The stability of the anti-recombinant C. difficile toxin A IgY (anti-A-6 IgY) after the tableting formation into a tablet) and enteric overcoating process was determined by comparing the ELISA reactivity of the lyophilized A-6 starting material (not tableted) with either the Eudragit S-100 enterically-coated or uncoated anti-A-6 IgY tablets. The enterically coated (3x tablet) or uncoated IgY tablets were allowed to dissolve in the simulated intestinal fluid at pH 7.5 at a concentration of 10 mg per ml. The starting untableted IgY (designated as bulk antibody) was also dissolved in the intestinal fluid at the same concentration. A standard ELISA was performed detecting the presence of antibodies Sdirected against the toxin A recombinant pPA1870-2680 N/C protein as described in Example 35. Pre-immune IgY at the same normalized concentration (designation as the placebo; shown by solid squares in Figure 56)) was also tested as a negative control. The ELISA results are shown in Figure 56.
In Figure 56 the absorbance at 410 nm is plotted against the reciprocal of the antibody dilution tested. The results shown in Figure 56 demonstrate that the reactivity of 25 the anti-A-6 IgY after tableting (intestinal uncoated; shown by the black diamonds) and the anti-A-6 IgY after tableting followed by enteric over-ceating with the Eudragit S-100 mixture (intestinal 3x; shown by the open diamonds) were very similar to the starting bulk o anti-A-6 IgY (bulk antibody; shown by the open squares). The results indicated that the tableting and the enteric over-coating processes were not harmful to the IgY preparation and that the anti-A-6 IgY remains active and functional after dissolution under physiological conditions.
The above results demonstrate that enterically-coated IgY tablets were generated that were stable and reactive.
283 e) Demonstration of the Retention of the Ability of the Tableted IgY to Neutralize C difficile Toxin A In Vivo The ability of the A-6 IgY tablet after dissolution to neutralize C. difficile toxin A was determined in vivo. Mice were used instead of hamsters to test toxin A neutralization because mice require less toxin for an LD (lethal dose) 100.
The LD,00 of C difficile toxin A for mice was determined by I.P. injection of gram Balb/c mice (Charles River) with 1 ml of PBS containing either 50 ng, 500 ng, or 5000 ng of C difficle toxin A. The C difficile toxin A used in this study was purchased from Dade International, Inc., Bartells Division Seattle, WA. C difficile toxin A at 50 ng was found to be the minimum lethal dose tested and all mice died within 24-hours posttreatment. All mice died within 3 and 4 hours when 500 ng and 5000 ng of C difficile toxin A were administered, respectively.
The effect of injecting C difficile toxin A in the presence of preimmune IgY was also tested to determine if preimmune IgY would reduce toxicity in vivo. Fifty nanograms 15 of toxin A was preincubated for 1 hour at 37 0 C with up to 50,000 ng (100-fold more) of preimmune IgY before administering to the mice. Even in the presence of preimmune IgY, 50 ng of C difficile toxin A was found to be lethal to the mice within 24 hours.
The ability of the anti-A-6 IgY tablet to neutralize C difficile toxin A in vivo was compared to the ability of anti-A-6 bulk IgY (before tableting) to neutralize the toxin.
One 250 mg anti-A-6 IgY tablet (the tablet had been stored for 3 weeks at room temperature) was dissolved in simulated intestinal fluid and the amount of IgY in solution was quantitated by absorbance at 280 nm. Lyophilized A-6 IgY before tableting (bulk) and preimmune (PI) IgY were also prepared in intestinal fluid and quantitated by absorbance at 280 nm. Five-thousand nanograms, 25,000 ng and 50,000 ng per ml of preimmune, anti-A- 25 6 bulk or dissolved anti-A-6 tablet IgY were each preincubated for 1 hour at 37 0 C with ng per ml of C difficile toxin A. This represents, respeetively, 100, 500, and 1000-fold more IgY compared to the amount of toxin (in terms of weight).
After incubation for 1 hour, the mixtures (9 total) were administered intraperitoneally to 20-25 gram Balb/c mice. Three mice were tested per group, with nine groups total. At the end of the 27 hour observation period, the number of mice surviving in each group were ascertained. The results are summarized below in Table 52.
284- TABLE 52 Treatment Group (ng of IgY mixed with 50 ng of Toxin A) 1 5000 ng anti-A-6 tablet 2 250,000 ng anti-A-6 tablet 3 500,000 ng anti-A-6 tablet 4 5000 ng anti-A-6 bulk 250,000 ng anti-A-6 bulk 6 500,000 ng anti-A-6 bulk 7 5000 ng PI 8 250,000 ng PI 9 500,000 ng PI Survivors 100 (3/3) 100 (3/3) 100 (3/3) 100 (3/3) 100 (3/3)
I
66 (2/3) 0 (0/3) 0 (0/3) 33 (1/3) The results shown above demonstrate that the anti-A-6 IgY tablet, after dissolution at pH 7.5, is able to neutralize the lethal effects of C. difficile toxin A in a manner 15 comparable to that observed when the starting bulk or uncoated) material was used.
These results show that the tableting process does not have an adverse effect on the capaciyt of the anti-A-6 IgY to neutralize C difficile toxin A. In contrast to the results obtaining using preimmune IgY, both the bulk and tableted anti-A-6 IgY preparations were able to neutralize C difficile toxin A at a comparable 100-fold excess of protein (5000 ng of A-b IgY neutralized 50 ng of toxin).
These results demonstrate the ability to formulate anti-clostridial toxin IgYs in a solid dosage form a tablet) form that is enterically coated for delivery in the large intestine. Moreover, the anti-clostridial toxin IgY present in the tablet remains stable, active and functional.
EXAMPLE 52 Prophylactic Treatment of C Difficile Disease in Hamsters by Antibodies Against Toxin A Recombinant Interval 6 The experiment involved infection of hamsters with C difficile and treatment of C difficile-induced disease with antibodies raised against recombinant toxin A (IgY). IgY antibodies were generated against the C difficile toxin A recombinant pMA1870-2 6 8 0 -285 (Interval Control animals were infected hamsters treated with preimmunization immunoglobulin fraction Crude PI and IgY were fractionated with polyethylene glycol and resuspended at an 8 X concentration (40 mg/ml) in 0.1 M carbonate buffer pH The two groups of hamsters each consisted of nine or ten female Golden Syrian hamsters (Sasco; outbred LVG) weighing about 80 g. The hamsters were housed at three per cage and were given food and water ad libitum throughout the study.
Hamsters were predisposed to C difficile infection using the method of Lyerly et al.' Clindamycin-HCl (Sigma) was intragastrically administered to each hamster at 3 mg/1 0 0 g 10 body weight in 1 ml of sterile water using a gavage needle. Two and four hours after clindamycin treatment, the hamsters were orally dosed with 2 mis of the 8X PI or 8X IgY.
Twenty-four hours after the administration of clindamycin-HCl the hamsters were challenged with 1 ml of phosphate buffered saline (PBS) pH 7.2 containing approximately 10' organisms of C difficile VPI 7698. The challenge dose was prepared from brain heart 15 infusion (BHI) broth cultures grown anaerobically in a Gaspak anaerobic chamber at 37°C overnight (growth conditions followed those of Lyerly et One hour prior to challenge and 8 hours after challenge the hamsters were dosed with 8X PI or 8X IgY. Treatments were continued twice a day at approximately 8 hour intervals for 2 more days (4 days total).
The results are shown in Figure 57. The duration of treatment is indicated by the horizontal bar below the abscissa. The administration of clindamycin and C difficile (infection) is indicated by arrows. Black squares represent PI-treated hamsters and open squares represent A-6 IgY-treated hamsters.
PI-treated hamsters developed diarrhea within 24-48 hours and died within 24 hours after the onset of diarrhea. All nine of the PI-treated hamsters were dead 48 hours after challenge. In contrast, sixty percent (6/10) of the hamsters receiving anti-A-6 IgY survived through 19 days post-challenge, when monitoring of the hamsters ended. The onset of death in the other four anti-A-6-IgY-treated hamsters was delayed from one to three days compared to the PI-treated hamsters. The level of long-term protection by anti-A- 6 IgY was statistically significant (chi-square analysis with P< 0.025). Although most of the anti- A-6 IgY-treated hamsters developed diarrhea (8 of 10), only half of those with diarrhea died Lverly et al. Infect. Immun.. 59 2215-2218 (1991).
286from C. difficile infection. Notwithstanding the onset of diarrhea, the anti-A-6 IgY promoted survival of the diseased hamsters.
These results indicate that low prophylactic doses of anti-A-6 IgY protects hamsters long-term from the severest effects of C difficile-induced disease. This protection was achieved on a dosage regimen of eight administrations over 4 days, of which only 3 doses were before infection. This minimal treatment contrasts with the Lyerly et al. regimen wherein hamsters were prophylatically treated a total of 39 times: bovine antitoxin was administered three times daily starting three days before challenge, and continuing ten days after challenge. Lyerly's bovine antitoxin dosage was also larger than that in the present *10 experiment 300 mg bovine antibody/dose versus 80 mg avian antibody/dose).
Moreover, in contrast to our findings, Lyerly reported that the animals showed no long term protection after termination of treatment (all died 72 hour after treatment) or once S* diarrhea developed.
In contrast to the Lyerly study, the present results demonstrate that avian antirecombinant toxin A antibodies are an effective prophylactic whose protective effects last well after treatment ends.
EXAMPLE 53 Therapeutic Treatment of C Difficile Disease in 20 Hamsters by Antibodies Against Toxin A Recombinant Interval 6 In another experiment, hamsters were treated therapeutically (post-infection) with anti-A-6 IgY. This is a more difficult treatment approach and has not been reported by Lyerly.
The C. difficile strain, growth media, challenge dose and infection procedure were the same as in Example 52. Two groups each containing nine female hamsters (Sasco) were challenged with approximately 10' organisms of C. diffcile strain VPI 7698. Four hours after challenge, treatment was initiated with 2 mis of either PI or anti-A-6 IgY at the same concentration as in Example 52. The hamsters were dosed again about 4 hours later, followed by two more daily treatments twice a day at 8-hour intervals. The hamsters were treated a total of six times over 3 days.
The results are shown in Figure 58. The duration of treatment is indicated by the horizontal bar below the abscissa and the clindamycin administration and C. difficile challenge are indicated by arrows.
287 The hamsters that were therapeutically treated with anti-A-6 IgY (open squares) had a lower cumulative mortality rate than the control group (closed squares). Additionally, the onset of diarrhea was delayed at least 24 hours in the anti-A-6 IgY-treated hamsters compared to the PI-treated hamsters. All hamsters treated with PI developed diarrhea and died within 2 days, demonstrating the lack of protection against the disease using control immune fraction. Fifty-five percent of the anti-A-6 treated hamsters survived longterm (observation period was 20 days). The protection provided by anti-A-6 IgY (survival) was statistically significant as compared to the PI-treated group (chi-square value P 0.05).
These results demonstrate that hamsters infected with C. difficile under infection 10 conditions identical to those of Lyerly received effective therapeutic protection with the avian anti-A-6 IgY and exhibited long-term survival.
288 SEQUENCE LISTING INFORMATION FOR SEQ ID NO:1: SEQUENCE CHARACTERISTICS: LENGTH: 24 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic) (xi) SEQUENCE DESCRIPTION: SEQ ID NO:1: GGAAATTTAG CTGCAGCATC
TGAC
24 INFORMATION FOR SEQ ID NO:2: S(i) SEQUENCE CHARACTERISTICS: LENGTH: 24 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic) S(xi) SEQUENCE DESCRIPTION: SEQ ID NO:2: TCTAGCAAAT TCGCTTGTGT TGAA 24 INFORMATION FOR SEQ ID NO:3: SEQUENCE CHARACTERISTICS: LENGTH: 20 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic) (xi) SEQUENCE DESCRIPTION: SEQ ID NO:3: CTCGCATATA GCATTAGACC INFORMATION FOR SEQ ID NO:4: SEQUENCE
CHARACTERISTICS:
LENGTH: 19 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic) (xi) SEQUENCE DESCRIPTION: SEQ ID NO:4: CTATCTAGGC
CTAAAGTAT
19 INFORMATION FOR SEQ ID SEQUENCE
CHARACTERISTICS:
LENGTH: 8133 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear -289 (ii) MOLECULE TYPE: DNA (genornic) (ix) FEATURE: NAME/KEY: CDS LOCATION: 1. .8130 (xi) SEQUENCE DESCRIPTION: SEQ ID ATG TCT TTA ATA TCT AAA GAA GAG TTA ATA AAA CTC GCA TAT AGC ATT 48 Met Ser Leu Ile Ser Lys Giu Glu Leu Ile Lys Leu Ala Tyr Ser Ile 1 5 10 AGA CCA AGA GAA AAT GAG TAT AAA ACT ATA CTA ACT AAT TTA GAC GAA 96 Arg Pro Arg Giu Asn Glu Tyr Lys Thr 25 GAA AAT AAA TAT TTG CAA TTA .4 a.
a. a a.
TAT AAT 144 Tyr Asn AAA AAA 192 Lys Lys 50 TCA AGC 240 Ser Ser GAA GTA 288 Glu Val AAG TTA ACT ACA AAC AAT AAT Lys Leu Thr Thr Asn Asn Asn 40 Glu Asn Lys CTA AAT GAA TCA ATT GAT Leu Asn Glu Ser Ile Asp AGA AAT AGA GCA CTC TCT Arg Asn Arg Ala Leu Ser ~70 GTT TTT ATG Val Phe Met AAT CTA AAA Asn Leu Lys 75 AAT ACA AGC Asn Thr Ser 90 AAA TAT AAA ACT Lys Tyr Lys Thr GAT ATA TTA AAA Asp Ile Leu Lys GTA OAA AAA AAT Val Glu Lys Asn
ATT
Ile CTT ATT Leu Ile 85 AAA AAT TCC Lys Asn Ser
CCT
Pro TTA CAT TTT GTA 336 Leu His Phe Val 100 TAC ATA AAA CAA 384 Tyr Ile Lys Gin TGG ATA GOT OGA GAA Trp Ile Gly Giy Giu 105 TOG GCT GAT ATT AAT Trp Ala Asp Ile Asn 120 AGT GAT ATT Ser Asp Ile GCT CTT GAA Ala Leu Giu 110 GCA GAA TAT AAT ATT AAA CTG Ala Glu Tyr Asn Ile Lys Leu 125 AAT ACA CTA AAA AAG OCT ATA Asn Thr Leu Lys Lys Ala Ile TOG TAT 432 Trp Tyr 130 OTT GAA 480 Val Glu AGT GA.A OCA TTC TTA OTA Ser Giu Ala Phe Leu Val 135 TCT ACC ACT GAA GCA TTA Ser Thr Thr Giu Ala Leu
TCT
Ser
CAG
Gin CTA CTA GAO Leu Leu Giu GAA GAG ATT Oiu Glu Ile 160 AGO ATO GAA Arg Met Oiu 175 AAT CCT CAA TTT OAT AAT ATO AAA TTT Asn Pro Gin Phe Asp Asn Met Lys Phe 165 110 AAA AAA Lys Lys TTT ATA TAT OAT AGA CAA AAA AGO TTT ATA AAT TAT TAT AAA TCT CAA 576 Phe Ile Tyr Asp Arg Gin Lys Arg Phe Ile Asn Tyr Tyr Lys Ser Gin .180 185 190 ATC AAT .AAA CCT ACA GTA CCT ACA ATA OAT OAT ATT ATA AAG TCT CAT 624 290 lie Asn Lys Pro Thr Val Pro Thr Ile Asp Asp 195 200 CTA GTA TCT GAA TAT AAT AGA GAT GAA ACT GTA 672 Leu Val Ser Glu Tyr Asn Arg Asp Giu Thr Val 210 215 Ile lie Lys Ser His 205 TTA GAA TCA TAT AGA Leu Glu Ser Tyr Arg 220 000 ATA GAT ATC AGG Gly Ile Asp Ile Arg 240 ACA AAT 720 Thr Asn 225 OCT AAT 768B Ala Asn TCT TTG AGA AAA ATA Ser Leu Arg Lys Ile 230 AGT TTG TTT ACA GAA Ser Leu Phe Thr Giu 245 AAT AGT AAT CAT Asn Ser Asn His 235 CAA GAG TTA TTA Gin Giu Leu Leu 250 TTA OCT GCA GCA Leu Ala Ala Aia Asn Ile Tyr Ser Gin 255 ATA OTA AGA Ile Val Arg 270 0.0.
0.
O*o GAG rro TTA AAT 816 Giu Leu Leu Asn 260 TTA TTA 0CC CTA 864 Leu Leu Ala Leu 275 CTT CCA GOT ATT 912 Leu Pro Giy Ile 290 TCT ATT OGA CTA 960 Ser Ile Gly Leu 305 AAO TAT AAA AAA 1008 Lys Tyr Lys Lys COT GGA AAT Arg Oly Asn AAA AAT TTT Lys Asn Phe TCT GAC Ser Asp 265 GOC OGA Oly Oly OTA TAT Val Tyr Leu Asp Val Asp Met 285 ATA TCT AGA CCT AGC Ilie Ser Arg Pro Ser 300 CAC TCT OAT TTA His Ser Asp Leu 295 GAC CGT TOO GAA Asp Arg Trp Giu TTT AAA ACA Phe Lys Thr ATG ATA AAA TTA Met Ile Lys Leu 315 TAT ACA TCA GAA Tyr Thr Ser 01u 330
GAG
Giu OCT ATT ATG Ala Ile Met 320 AAT AAT Asn Asn Asn Phe Asp Lys 335 ATA OAA AGT AAA Ile Giu Ser Lys CTT OAT 1056 Leu Asp AGT GAA 1104 Ser Giu GAT CTT 1152 Asp Leu 370 0CC TTG 1200 Ala Leu 365 CAA OTA 1248 Gin Val CAA CA.A TTA AAA OAT AAT TTT AAA CTC Gin Gin Leu Lys Asp Asn Phe Lys Leu 340 345
ATT
Ile AAA TCT Lys Ser 355 GAA ATT Giu Ile GAG ATA TTT Glu Ile Phe TCT AAA TTA GAA Ser Lys Leu Giu 360 TTC OCT TTA GGC Phe Ala Leu Gly AAT TTA AAT OTA TCT Asn Leu Asn Val Ser 365 AGT OTT ATA AAT CAA Ser Val Ile Asn Gin AAA ATA Lys Ile
OCT
Ala 375 380 ATA TCA AAA CAA Ilie Ser Lys Gin 390 AAA AAT AGA TAT Lys Asn Arg Tyr 405 GOT TCA TAT CTT ACT AAC CTA GTA ATA Gly Ser Tyr Leu Thr Asn Leu Val Ile 395 CAA TTT TTA AAC CAA CAC CTT AAC CCA Gin Phe Leu Asfl Gin His Leu Asn Pro 410 415
GAA
Giu 400 0CC Al a ATA GAG TCT GAT AAT AAC TTC ACA OAT ACT ACT AAA ATT TTT CAT OAT 1296 291 Ile Giu Ser Asp Asn Asn Phe Thr Asp Thr Thr Lys Ile Phe His Asp 420 425 430 TCA T TTT AAT TCA GCT ACC GCA GAA AAC TCT ATG TTT TTA ACA AAA 1344 Ser Leu Phe Asn Ser Ala Thr Ala Giu Asn Ser Met Phe Leu Thr Lys 435 440 445 ATA GCA CCA TAC TTA CAA OTA GGT TTT ATG CCA GAA GCT CGC TCC ACA 1392 Ile Ala Pro Tyr Leu Gin Val Gly Phe Met Pro Giu Ala Arg Ser Thr 450 455 460 ATA AGT TTA AGT GOT CCA GGA GCT TAT 1440 GCO TCA GCT TAC TAT GAT TTC Ala Ser Ala Tyr Tyr Asp Phe 475 480 a a.
a a.
a a.
a a a .a a. a a a a.
a. a Ile Ser 465 ATA AAT 1488 Ile Asn TTA ATA 1536 Leu Ile TTA CAA GAA Leu Gin Giu 485 470 AAT ACT ATA GAA Asn Thr Ile Giu AAA ACT TTA Lys Thr Leu 490 A.AT CTA TCT Asn Leu Ser AAA GCA TCA OAT Lys Ala Ser Asp 495 CAA TTO ACA GAA Gin Leu Thr Giu 510 Leu Ser Giy Pro Gly Ala Tyr CAA OAA 1584 Gin Oiu CAA TTT 1632 Gin Phe- 530 GAC AAT 1680 Asp Asn 545 TTA TTA 1728 Leu Leu OAA TTT AAA TTC CCA OAA AAT Oiu Phe Lys Phe Pro Giu Asn 500 505 ATA AAT AOT CTA TOO AOC TTT Ile Asn Ser Leu Trp Ser Phe 515 520 GAO AAA TAT OTA AGA OAT TAT Oiu Lys Tyr Vai Arg Asp Tyr Asp Gin Ala Ser Ala Lys Tyr 525 ACT GOT OGA TCT CTT TCT OAA Thr Gly Glv Ser Leu Ser Glu 000 GTA GAC TTT Oly Vai Asp Phe 550 AAT AAT AAA ATT Asn Asn Lys Ilie 565 AAA AAT ACT 0CC Lys Asn Thr Aia 555 TCA AAC AAT OTA Ser Asn Asn Val 540
CTC
Leu GAC AAA AAC TAT Asp Lys Asn Tyr 560 OAA OCT OGA AOT Giu Ala Giy Ser
CCA
Pro
GAA
Glu AAA AAT 1776 Lys Asn TAT OTT Tyr Val 580 CAT TAT ATC ATA His Tyr Ile Ile
TTA
Leu
AAA
Lys 575 TAT OAA OCA 1824 Tyr Giu Ala 595 ATT ATA CAA 1872 Ile Ile Gin ACA TOC AAT TTA Thr Cys Asn Leu COA AAT ATO AAT Arg Asn Met Asn 615 GAA TCT ATT TTA Glii Ser Ile Leu CAA GGA GAT GAT ATA AOT Gin Gly Asp Asp Ile Ser 590 A.AT CCT AAA AAT AGT ATT Asn Pro Lys Asn Ser Ile 605 AAA AOC TAC TTT TTA AOT Lys Ser Tyr Phe Leu Ser OAA AOT OCA Giu Ser Ala 610 OAT OAT 1920 Asp Asp 625
GOA
GAA TTA AAT AAA TAT Oiu Leu Asfl Lys Tyr 635 AGO ATA CCT Arg Ile Pro 7 630 AGA TTA AAA AAT AAG GAA AAA 1968 GTA AAA GTA ACC TTT ATT GOA CAT GOT 2 92 Arg Leu Lys Asn Lys Glu Lys Val Lys Val Thr Phe Ile Gly His Gly 645 650 655 AAA GAT GAA TTC AAC ACA AGC GAA Trr GCT AGA TTA ACT GTA GAT TCA 2016 Lys Asp Giu Phe Asn Thr Ser Glu Phe Ala Arg Leu Ser Vai Asp Ser 660 665 670 TTTCC AAT GAG ATA ACT TCA TTT TTA GAT ACC ATA AAA TTA GAT ATA 2064 Leu Ser Asn Giu Ile Ser Ser Phe Leu Asp Thr 675 680 TCA. CCT AAA AAT GTA GAA GTA AAC TTA CTT GGA 2112 Ser Pro Lys Asn Val Glu Val Asn Leu Leu Gly 690 695 TAT GAT TTT AAT OTT OAA GAA ACT TAT CCT G 2160 Tyr Asp Phe Asn Val Glu Glu Thr Tyr Pro Gly 705 710 715 ATT ATO GAC AAA ATT ACT TCC ACT TTA CCT GAT 2208 Ile Met Asp Lys Ile Thr Ser Thr Leu Pro Asp 725 730 ATT ACT ATA OGA OCA AAT CAA TAT GAA GTA AGA 2256 ile Thr Ilie Gly Ala Asn Gin Tyr Giu Val Arg '740 745 AGA AAA OAA CTT CTG OCT CAC TCA GOT AAA TOO 2304 Arg Lys Glu Leu Leu Ala His Ser Oly Lys Trp 755 760 685 TGT AAT ATO TTT ACT Cys Asn Met Phe Ser 700 AAG TTG CTA TTA AGT Lys Leu Leu Leu Ser 720 GTA AAT AAA AAT TCT Val Asn Lys Asn Ser 735 ATT AAT AGT GAG OGA Ile Asn Ser Oiu Oly 750 ATA AAT AAA OAA GAA Ile Asn Lys Oiu Oiu 765 a.
a OCT ATT ATO AOC OAT TTA TCT ACT AAA GAA TAC ATI' TTT TTT GAT TCT 2352 Ala Ile Met Ser Asp Leu Ser Ser Lys Oiu Tyr Ile Phe Phe Asp Ser 770 775 780 ATA OAT AAT AAG CTA AAA OCA AAG TCC AAO AAT ATr CCA OGA TTA OCA 2400 Ile Asp Asn Lys Leu Lys Ala Lys Ser Lys Asn Ile Pro Oiy Leu Ala 785 790 795 800 TCA ATA 2448 Ser Ilie CCT OAT 2496 Pro Asp TCT ATT 2544 Ser Ile ATA ATT 2592 Ile Ile 850 TCA OAA OAT ATA Ser Oiu Asp Ile 805 AAA ACA TTA TTA Lys Thr Leu Leu 810 TTA AAT AATCT Leu Asfl Asn Leu Leu Asp Ala Ser Val 815 AAT ATI' OAA TCT Asn Ile Giu Ser ACA AAA TTr Thr Lys Phe 820 GG0 OAT TAC Oly Asp Tyr 835 CAC AAT TCT His Asn Ser Ile
AAG
Lys crr Leu 825 830 Ile
ATA
Ile TAT TAT OAA AAA TTA GAG CCT OTT AA.A PAAT Tyr Tyr Giu Lys Leu Oiu Pro Val Lys Asfl 840 845 OAT OAT TTA ATA GAT GAG TTC AAT CIA CTT Asp Asp Leu Ile Asp Oiu Phe Asn Leu Leu 855 860 OAA AAT OTA TCT GAT GAA TTA TAT OAA TTA AAA AAA TTA AAT AAT CTA 2640 293 Giu Asn Val Ser Aso Giu Leu Tyr Glu Leu Lys Lys Leu Asn Asn Leu 865 870 875 880 GAT GAG AAG TAT TI'A ATA TCT TTT GAA GAT ATC TCA AAA AAT AAT TCA 2688 Asp Giu Lys Tyr Leu Ile Ser Phe Giu Asp 885 890 ACT TAC TCT GTA AGA TTT ATT AAC AAA AGT 2736 Thr Tyr Ser Val Arg Phe Ilie Asn Lys Ser 900 905 GTA GAA ACA GAA AAA GAA ATT TTr TCA A 2784 Val Giu Thr Giu Lys Giu Ile Phe Ser Lys 895 A.AT GGT GAG TCA GTT TAT Asn Gly Glu Ser Val Tyr 910 TAT AGC GMA CAT ATT ACA Tyr Ser Giu His Ile Thr 925 ATT ACA GAT GTT MAT GGT Ile Thr Asp Val Asn Gly 915 6* *O 0 ~S*
S
*0*S S. OS
S
.5 S o
S.
0 *5*9
S
5* 6 S. S
S.
0* S. S .3 5* *5 *55* 5 *55e AAA GMA ATA 2832 Lys Giu Ile 930 MAT TTA TTG 2880 Asn Leu Leu 945 TTA MAC GCA 2928 Leu Asn Ala AGT ACT ATA MAG Ser Thr Ile Lys 935 GAT MAT ATA CAG Aso An Ile Gin MAT AGT ATA Asn Ser Ile TTA GAT Leu Asp 950 ACT TCT Thr Ser 955 ATA GAT Ile Asp CMA GTT Gin Val MAT ACA Asn Thr 960 AMA GAT GTA 2976 Lys Asp Val GCT CMA CTA 3024 Ala Gin Leu 995 TTA GTA MAT 3072 Leu Vai Asn 1010 CCT ACA ATA 3120 Pro Thr Ile 1025 ATA MAC TTA 3168 Ile Asn Leu GCA TTC TTT ATT CMA TCA TTA Ala Phe Phe Ile Gin Ser Leu 965 970 CTG MAT GAT TTA AGT ACC TCA Leu Asn Asp Leu Ser Thr Ser 980 985 TTT AGT ACA GGT TTA MAT ACT Phe Ser Thr Gly Leu Asn Thr 1000 TTA ATA TCA MAT GCA GTA AAT Leu Ile Ser Asn Ala Val Asn 1015 ACA GAG GGG ATA CCT ATT GTA Thr Glu Giy Ile Pro Ile Val 1030 CGT GCA GCA ATT MAG GMA TTA TAT AGT AGC MAT Tvr Ser Ser Asn 975 Val Lys Val Gin Leu Tyr 990 ATA TAT GAC TCT ATC CMA Ile Tyr Asp Ser Ile Gin 1005 GAT ACT ATA MAT GTA CTA Asp Thr Ile Asn Val Leu 1020 TCT ACT ATA TTA GAC GGA Ser Thr Ile Leu Asp Gly 1035 1040 CTA GAC GMA CAT GAC CCA Gly Ala Aia Ile Lys Giu Leu Leu Asp 1045 TTA CTA AMA MAA GMA 3216 Leu LeU Lys Lys Giu 1060 ATG TCA TTA TCT ATA 3264 Met Ser Leu Ser Ile 1075 TTA GMA Leu Glu GCT GCA Ala Ala GCT MAG GTG Ala Lys Vai 1065 ACT GTA GCT Thr Vai Aia 1080 GGT GTT Gly Val TCA ATT Ser Ile TTA GCA ATA MAT Leu Ala Ile Asn 1070 GT1T GGA ATA GGT Val Giy Ile Giy 1085 GCT GMA GTT ACT ATT TTC TTA TTA CCT ATA GCT GGT ATA TCT GCA GGA 3312 294 Ala Glu Val Thr Ile Phe Leu Leu 1090 1095 ATA CCT TCA TTA GTT AAT AAT GAA 3360 Ile Pro Ser Leu Val Asn Asn Glu 1105 1110 TCA GTG GTA AAC TAT TTT AAT CAT 3406 Ser Val Val Asn Tyr Phe Asn His Pro Ile Ala TTA ATA TTG Leu Ile Leu 12115 TTG TCT GAA Leu Ser Glu 1130 1125 Gly Ile Ser Ala Gly 1100 CAT GAT AAG GCA ACT His Asp Lys Ala Thr 1120 TCT AAA AAA TAT GGC Ser Lys Lys Tyr Gly 1135 CCT ATT GAT GAT TTA Pro Ile Asp Asp Leu 1150 ATA AAA CTA GGA ACA Ile Lys Leu Gly Thr 1165 CAC ACA GTG ACT GGT CCT CT1' AAA ACA GAA GAT 3456 Pro Leu Lys Thr Glu Asp 1140 GTA ATA TCA GAA ATA GAT 3504 Val Ile Ser Glu Ile Asp 1155 TGT AAT ATA TTA GCA ATO 3552 Cys Asn Ile Leu Ala Met 1170 AAT ATA GAT CAC TTT TTC 3600 Asn Ilie Asp His Phe Phe 1185 119 TCA TTA TCA ATT TAT TCT 3648 Ser Leu Ser Ile Tyr Ser 1205 GAT AAA ATT TTA GTT Asp Lys Ile Leu Val 1145 TTT AAT AAT AAT TCG a a a a. a. a a.
2a* a.
a 1160 GAG GGG GGA Glu Gly Oly 1175 TCA TCT CCA Ser Ser Pro 0 GCA ATA GGT Ala Ile Gly Asn Ser TCA GGA 1180 TCT ATA ACT Ser Ile Ser 1195 ATA GAA ACA Ile Glu Thr 1210 Thr Val Thr Gly TCT CAT ATT CCT Ser His Ile Pro 1200 GAA AAT CTA OAT Giu Asn Leu Asp 1215 TTT TCA AAA AAA ATA ATG ATO TTA CCT AAT GCT CCT TCA AGA GTO TTT 3696 Phe Ser Lys Lys Ilie Met Met Leu Pro Asn Ala Pro Ser Arg Val Phe 1220 1225 1230 TGG TOG GAA ACT GGA OCA GTT CCA GOT TTA AGA TCA TTG GAA AAT GAC 3744 Trp, Trp Olu Thr Gly Ala Val Pro Oly Leu Arg Ser Leu Olu Asn Asp 1235 1240 1245 OGA ACT AGA TTA CTT GAT TCA ATA AGA GAT TTA TAC CCA GOT AAA TTT 3792 Gly Thr Arg Leu Leu Asp Ser Ile Arg Asp Leu Tyr Pro Gly Lys Phe 1250 1255 1260 TAC TGO AGA TTC TAT GCT TTT TTC OAT TAT GCA ATA ACT ACA TTA AAA 3840 Tyr Trp, Arg Phe Tyr Ala Phe Phe Asp Tyr Ala Ile Thr Thr Leu Lys 1265 1270 1275 1280 CCA OTT TAT OAA GAC ACT AAT ATT AAA ATT AAA CTA GAT AAA OAT ACT 3888 Pro Val Tyr Glu Asp Thr Asn Ile Lys Ile Lys Leu Asp Lys Asp Thr 1285 1290 1295 AGA AAC TTC ATA ATG CCA ACT ATA ACT ACT AAC GAA ATT AGA AAC AAA 3936 Arg Asn Phe Ile met Pro Thr Ile Thr Thr Asn Glu Ile Arg Asn Lys 1300 1305 1310 TTA TCT TAT TCA TTT OAT OGA OCA GGA OGA ACT TAC TCT TTA TTA TTA 3984 295 Leu Ser T-yr Ser Phe Asp Gly Ala Gly Gly Thr Tyr Ser Leu Leu Leu 1315 1320 1325 TCT TCA TAT CCA ATA TCA ACG AAT ATA AAT TTA TCT AAA GAT GAT TTA 4032 Ser Ser Tyr Pro Ile Ser Thr Asn Ile Asn Leu Ser Lys Asp Asp Leu 1330 1335 1340 TGG ATA TTT AAT ATT GAT AAT GAA GTA AGA GAA ATA 4080 Trp Ile Phe Asn Ile Asp Asn Glu Val Arg Giu Ile 1345 1350 1355 GOT ACT ATT AAA AAA OGA AAG TTA ATA AAA OAT OTT 4128 Gly Thr Ile Lys Lys Gly 1365 GAT ATA AAT AAA AAT AAA 4176 Asp Ile Asn Lys Asn Lys 1380 TCA GGC GAT ATA GAT AAT 4224 Ser Gly Asp Ile Asp Asn 1395 TTA GAT GAT AAA ATT AGT 4272 Leu Asp Asp Lys Ile Ser 1410 TCT TAT AGT TTG TTA TTG 1370 CTT ATT ATA GGC AAT Leu Ile Ile Gly Asn 1385 Val
CAA
Gin
TTC
Ser Ile Giu Asn 1360 TTA AGT AAA ATT Leu Ser Lys Ile 1375 ACA ATA GAT TTT Thr Ile Asp Phe 1390 TTG ACT TGT GAG AAA GAT AGA TAT Lys Asp Arg Tyr 1400 TTA ATA ATA GAA lieu Ile Ile Glu 1415 TCT GGG GAT AAA
ATA
1405 ATA AAT CTT GTT GCA AAA Ile Asn Leu Val Ala Lys 1420 AAT TAT TTG ATA TCC AAT Asn Tyr Leu Ile Ser Asn 1435 144 TTA 0CC CTA GAT ACT AAA 4320 Ser Tyr 1425 MT TCZT Ser Leu Leu Leu Ser Gly Asp Lys 1430 AAT ACT ATT GAO AA6A ATC AAT ACT 0 Leu Ser Asn Thr Ile Giu Lys Ile Asn Thr Leu Oly Leu Asp Ser Lys 1445 1450 1455 AAT ATA GCG TAC AAT TAC ACT GAT GAA TCT AAT AAT AAA TAT TTT GGA 4416 Asn Ile Ala Tyr Asn Tyr Thr Asp Ciu Ser Asn Asn Lys Tyr Phe Gly 1460 1465 1470 GCT ATA TCT AAA ACA AGT CAA AAA AGC ATA ATA CAT TAT AAA AAA GAC 4464 Ala Ile Ser Lys Thr Ser Gin Lys Ser Ile Ile His Tyr Lys Lys Asp 1475 1480 1485 AGT AAA AAT ATA TTA GAA TTT TAT AAT GAC AGT ACA TTA GAA TTT AAC 4512 Ser Lvs Asn Ile Leu Glu Phe Tyr Asn Asp Ser Thr Leu Giu Phe Asn 1490 1495 1500 AGT AAA GAT TTT ATT GCT GAA GAT ATA AAT CTA TTT ATG AAA CAT CAT 4560 Ser Lys Asp Phe Ile Ala Glu Asp Ile Asn Val Phe Met Lys Asp Asp 1505 1510 1515 1520 ATT AAT ACT ATA ACA CCA AAA TAC TAT GTT OAT MAT MAT ACT OAT A 4608 lie Asn Thr Ile Thr Cly Lys Tyr Tyr Val Asp Asn Asn Thr Asp Lys 1525 1530 1535 ACT ATA CAT TTC TCT ATT TCT TTA GTT ACT AMA MT CMA GTA AMA GTA 4656 296 Ser Ile Asp Phe Ser Ile Ser Leu Val Ser Lys Asn Gin Val Lys Val 1540 1545 1550 AAT GGA TTA TAT TTA AAT GAA TCC GTA TAC TCA TCT TAC CTT GAT TTT 4704 Asn Gly Leu Tyr Leu Asn GiU Ser Val Tyr Ser Ser Tyr Leu Asp Phe 1555 1560 1565 GTG AAA AAT TCA OAT OGA CAC CAT AAT ACT TCT AAT TTT ATG AAT TTA 4752 Val Lys Asn Ser Asp Gly His His Asn Thr Ser Asn Phe Met Asn Leu 1570 1575 1580 Trr TI'G GAC AAT ATA AGT TTC TOG AAA TTG TTT 000 ITTT OAA AAT ATA 4800 Phe Leu Asp Asn Ile Ser Phe Trp Lys Leu Phe Gly Phe Giu Asn Ile 1585 1590 1595 1600 AAT TTT GTA ATC OAT AAA TAC TTT ACC CT!' OTT GOT AMA ACT AAT CT' 4848 Asn Phe Val Ile Asp Lys Tyr Phe Thr Leu Val Giy Lys Thr Asn Leu 1605 1610 1615 GGA TAT OTA OAA TTT ATT TOT GAC AAT AAT AA.A MAT ATA OAT ATA TAT 4896 Oly Tyr Val Giu Phe Ile Cys Asp Asn Asn Lys Asn Ile Asp Ile Tyr 1620 1625 1630 *TTT GOT GMA TOG AAA ACA TCO TCA TCT AAA AGC ACT ATA TTT AGC OGA 4944 Phe Gly Oiu Trp Lys Thr, Ser Ser Ser Lys Ser Thr Ilie Phe Ser Oly 1635 1640 1645 MT GOT AGA MAT OTT OTA OTA GAG CCT ATA TAT MAT CCT OAT ACO GOT As 4992 Arg Asn Val Val Val Giu Pro Ile Tyr Asn Pro Asp Thr Oly 1650 1655 1660 GMA OAT ATA TCT ACT TCA CTA OAT TTT TCC TAT GMA CCT CTC TAT GGA 5040 Giu Asp Ile Ser Thr Ser Leu Asp Phe Ser Tyr Oiu Pro Leu Tyr Gly *1665 1670 1675 1680 ATA OAT AGA TAT ATA MAT AMA OTA TTO ATA OCA CCT OAT TTA TAT ACA 5088 Sle Asp Arg Tyr Ile Asn Lys Val Leu Ile Ala Pro Asp Leu Tyr Thr 1685 1690 1695 AGT TTA ATA MAT ATT MAT ACC MAT TAT TAT TCA MAT GAO TAC TAC CCT 5136 *Ser Leu Ile Asn Ile Asn Thr Asn Tyr Tyr Ser Asn Oiu Tyr Tyr Pro *1700 1705 1710 GAO ATT ATA OTT CTT MAC CCA MAT ACA TTC CAC AMA AMA OTA MAT ATA 5184 Glu Ile Ile Val Leu Asn Pro Asn Thr Phe His Lys Lys Val Asn Ile 1715 1720 1725 MAT -TTA OAT AGT TCT TCT TTT GAG TAT AMA TOG TCT ACA GMA OGA AOT 5232 Asn Leu Asp Ser Ser Ser Phe Oiu Tyr Lys Trp Ser Thr Giu Oly Ser 1730 1735 1740 GAC TTT ATT TTA OTT AGA TAC TTA GAM GMA AGT MAT AMA AAA ATA TTA 5280 Asp Phe Ilie Leu Val Arg Tyr Leu Giu Giu Ser Asn Lys Lys Ile Leu 1745 *1750 1755 1760 CMA AMA ATA AGA ATC AMA GOT ATC ITA TCT MAT ACT CMA TCA TTT MAT 5328 297 Gin Lys Ile Arg Ile Lys Gly Ile Leu Ser Asn Thr Gin Ser Phe Asn 1765 1770 1775 AAA ATG AGT ATA GAT TTT AAA GAT ATT AAA AAA CTA TCA TTA GGA TAT 5376 Lys Met Ser Ile Asp Phe Lys Asp Ile Lys Lys Leu Ser Leu Gly Tyr 1780 1785 1790 ATA ATG AGT AAT TTT AAA TCA TTT AAT TCT GAA AAT GAA TTA GAT AGA 5424 Ile Met Ser Asn Phe Lys Ser Phe Asn Ser Glu Asn Glu Leu Asp Arg 1795 1800 1805 GAT CAT TTA GGA TTT AAA ATA ATA GAT AAT AAA ACT TAT TAC TAT GAT 5472 Asp His Leu Gly Phe Lys Ile Ile Asp Asn Lys Thr Tyr Tyr Tyr Asp 1810 1815 1820 GAA GAT AGT AAA TTA GTT AAA GGA TTA ATC MAT ATA AAT AAT TCA TTA 5520 Glu Asp Ser Lys Leu Val Lys Gly Leu Ile Asn Ile Asn Asn Ser Leu 1825 1830 1835 1840 TTC TAT TTT GAT CCT ATA GAA TTT AAC TTA GTA ACT GGA TGG CAA ACT 5568 Phe Tyr Phe Asp Pro Ile Glu Phe Asn Leu Val Thr Gly Trp Gin Thr 1845 1850 1855 ATC AAT GGT AAA AAA TAT TAT TTT GAT ATA AAT ACT GGA GCA GCT TTA 5616 le Asn Gly Lys Lys Tyr Tyr Phe Asp Ile Asn Thr Gly Ala Ala Leu 1860 1865 1870 .ACT AGT TAT AAA ATT ATT AAT GGT AAA CAC TTI TAT TTT AAT AAT GAT 5664 Thr Ser Tyr Lys Ile Ile Asn Gly Lys His Phe Tyr Phe Asn Asn Asp S1875 1880 1885 GGT GTG ATG CAG TTG GGA GTA TTT AAA GGA CCT GAT GGA TTT GAA TAT 5712 Gly Val Met Gin Leu Gly Val Phe Lys Gly Pro Asp Gly Phe Glu Tyr 1890 1895 1900 TTT GCA CCT GCC AAT ACT CAA AAT AAT AAC ATA GAA GGT CAG GCT ATA 5760 **Phe Ala Pro Ala Asn Thr Gin Asn Asn Asn le Glu Gly Gin Ala lie 1905 1910 1915 1920 GTT TAT CAA AGT AAA TTC TTA ACT TTG AAT GGC AAA AAA TAT TAT TTT 5808 Val Tyr Gin Ser Lys Phe Leu Thr Leu Asn Gly Lys Lys Tyr Tyr Phe 1925 1930 1935 LGAT AAT AAC TCA AAA GCA GTC ACT GGA TGG AGA ATT ATT AAC AAT GAG 5856 Asp Asn Asn Ser Lys Ala Val Thr Gly Trp Arg Ile Ile Asn Asn Glu 1940 1945 1950 AAA TAT TAC TTT AAT CCT AAT AAT GCT ATT GCT GCA GTC GGA TTG CAA 5904 4Lys Tyr Tyr Phe Asn Pro Asn Asn Ala Ile Ala Ala Val Gly Leu Gin 1955 1960 1965 GTA ATT GAC AAT AAT AAG TAT TAT TTC AAT CCT GAC ACT GCT ATC ATC 5952 2Val Ile Asp Asn Asn Lys Tyr Tyr Phe Asn Pro Asp Thr Ala Ile Ile 1970 1975 1980 TCA AAA GGT TGG CAG ACT GTT AAT GGT AGT AGA TAC TAC TTT GAT ACT 6000 -298- Ser Lys Gly 1985 GAT ACC GCT 6048 Asp Thr Ala Trp Gin Thr Val 1990 ATT GCC TTT AAT Ile Ala Phe Asn 2005 Asn Gly Ser Arg Tyr 1995 GGT TAT AAA ACT ATT Gly Tyr Lys Thr Ile 2010 CTA GTG AAA ATA GGT Val Val Lys Ile Gly 2025 Tyr Phe Asp Thr 2000 GAT GGT AAA CAC Asp Gly Lys His 2015 GTG TTT AGT ACC Val Phe Ser Thr 2030 TTT TAT 6096 Phe Tyr TCT AAT TTT GAT AGT Phe Asp Ser 2020 CAT TGT Asp Cys GGA TTT GAA TAT TTT GCA CCT GCT AAT ACT TAT AAT AAT AAC 6144 Ser Asn Gly Phe 2035 ATA CAA CT CAC 6192 Ile Glu Cly Cmn 2050 CGT AAA AAA TAT 6240 Cly Lys Lys Tyr 2065 CAA ACT ATT CAT 6288 Gin Thr Ile Asp GCA GCT ACT CGA Glu Tyr Phe Ala 2040 GCT ATA CTT TAT Ala Ile Val Tyr 2055 TAC TTT CAT AAT Tyr Phe Asp Asn Pro Ala Asn Thr Tyr Asn Asn Asn 2045 CAA AGT AAA TTC TTA ACT TTC AAT Gin Ser Lys Phe Leu Thr Leu Asn 2060 AAC TCA AAA CCA GTI ACC GGA TTG Asn Ser Lys Ala Val Thr Cly Leu 2075 2080 TAC TTI AAT ACT AAC ACT GCT CAA Tyr Phe Asn Thr Asn Thr Aia Glu 2090 2095 CAT CGT AAA AAA TAT TAC TT AAT
S
S
2070 AGT AAA AAA TAT Ser Lys Lys Tyr 2085 TGG CAA ACT ATT 6336 Ala Ala Thr C 2 ACT AAC ACT C 6384 Thr Asn Thr 2115 AAA TAT TAC 6432 Lys Tyr Tyr 2130 ATT ATT AAT 6480 Ile Ile Asn 2145 ATA GGA CTG 6528 Ile Cly Vai ;iy Trp Gin Thr 1i00 CT CAA CCA CCT kla Glu Ala Ala TT AAT ACT AAC Phe Asn Thr Asn 2135 GGT AAA CAT TTT Cly Lys His Phe 2150 TTT AAA GGA CCT Phe Lys Cly Pro 2165 Ile Asp 2105 ACT GGA rhr Gly 2120 TGG CAA Trp Gin ACT ATT GAT GGT AAA Thr Ile Asp Gly Lys 2125 Gly Lys Lys Tyr Tyr Phe Asn 2110 ACT CCT ATA CCT TCA ACT Thr Ala Ile Ala Ser Thr 2140 TAT TIT AAT ACT CAT GGT Tyr Phe Asn Thr Asp Gly 2155 AAT OGA T1T GAA TAT TIT Asn Gly phe Giu Tyr Phe 2170 GAA GGT CAA GCT ATA CTT Glu Gly Gin Ala Ile Leu 2185 AAA AAA TAT TAC TTT GGT Lys Lys Tyr Tyr Phe Gly 2200 220 GGT TAT ACA Cly Tyr Thr ATT ATG CAC Ile Met Cln 2160 GCA CCT GCT Ala Pro Ala 2175 TAC CAA AAT Tyr Gin Asn 2190 AGT CAC TCA Ser Asp Ser '5 AAT -ACC 6576 Asn Thr GAA TTC 6624 Glu Phe AAA CCA 6672 CAT GCT AAC Asp Ala Asn 2180 TTA ACT TTC Leu Thr Leu 2195 AAC ATA Asn Ile AAT GGT Asn Cly GTT ACT CGA TGG AGA ATT ATT AAC AAT AAG AAA TAT TAC TTT -299 Lys Ala Val Thr 2210 AAT CCT AAT AAT 6720 Asn Pro Asn Asn 2225 GAC AAG TAT TAC 6768 Asp Lys Tyr Tyr ACT ATT GAA AGA 6816 Thr Ile Glu Arg 226 Gly Trp Arg Ile Ile 2215 GCT ATT OCT GCA ATT Asn Asn Lys Lys 2220 CAT CTA TGC ACT Tyr Tyr Phe ATA AAT AAT Al a Ile Ala 2230 Phe Ser Tyr 2245 AAT AAT- TTC Asn Asn Phe 0 Ala Ile His GAT GGA ATT Asp Gly Ile 2250 TAT TTT GAT Tyr Phe Asp 2265 Leu Cys CTT CA.A AAT GGA TAT ATT Leu Gin Asn Gly Tyr Ile 2255 GCT AAT AAT GAA TCT AAA Ala Asn Asn Giu Ser Lys 2270 OGA TTT GAG TAT TTT GCA Gly Phe Giu Tyr Phe Ala 2285 GOT CAG OCT ATA OTT TAC Giy Gin Ala Ile Val Tyr 2300 Thr Ile Asn Asn ATO GTA 6864 Met Val CCT OCT 6912 Pro Ala 229' CAG AAC 6960 Gin Asn 2305 GAC TCA 7008 Asp Ser ACA OGA4 Thr Gly 2275 AAT ACT Asn Thr AAA TTC Lys Phe AAA GCA Lys Aia GTA TTT AAA OGA CCT AAT VJal Phe Lys Gly Pro Asn 2280 CAC AAT AAT AAC ATA GAA His Asn Asn Asn Ile Oiu 2295 TTA ACT TTG AAT GGC AAA Leu Thr Leu Asn Gly Lys 2310 OTT ACT OGA TOG CAA ACC Val Thr Gly Trp Gin Thr 2325 233
AAA'
LYS
2315
ATT
Ile 0 TAT TAT TTT OAT AAT Tyr Tyr Phe Asp Asn 2320 GAT GGT AAA AAA TAT Asp Oly Lys Lys Tyr 2335 GGA TGO CAA ACT ATT Oly Trp Gin Thr Ile 2350 OCT GAA OCA OCT ACT Ala Giu Ala Ala Thr 2365 TAC TTT AAT CTT AAC ACT OCT GAA OCA OCT ACT 7056 Tyr Phe Asn Leu Asn Thr Ala Oiu Ala Ala Thr 2340 2345 OAT GOT AAA AAA TAT TAC TTT AAT CTT AAC ACT 7104 Asp Gly Lys Lys Tyr Tyr Phe Asn Leu Asn Thr 2355 2360 OGA TG 7152 Oly Trp 2370 TTC ATA 7200 Phe Ile 2385 TT-- AAT 7248 Phe Asn OGA TTT 7296 Gly Phe CAA ACT ATT GAT GOT AAA AAA Gin Thr Ile Asp Gly Lys Lys 2375 0CC TCA ACT GOT TAT ACA ACT Ala Ser Thr Giy Tyr Thr Ser 2390 ACT OAT GOT ATT ATO CAG ATA Thr Asp Oly Ilie Met Gin Ilie 2405 GAA TAC TTT OCA CCT OCT AAT Giu Tyr Phe Ala Pro Aia Asn 2420 242 TAT TAC Tyr Tyr TTT AAT Phe Asn 2380 ACT AAC ACT Thr Asn Thr AT? AAT GOT AAA CAT TTT TAT Ile Asn Oly Lys His Phe Tyr 2395 2400 OGA OTG TTT AAA OGA CCT AAT Oly Val Phe Lys Gly Pro Asfl 2410 2415 ACO OAT OCT AAC AAC ATA OAA Thr Asp Ala Asn Asn Ile Giu 5 2430 00? CAA OCT ATA CT? TAC CAA AAT AAA TTC TTA ACT TTO AAT GOT AAA 7344 00 Gly Gin Ala Ile Leu Tyr Gin Asn Lys Phe Leu Thr Leu Asn Gly Lys 2435 2440 2445 AAA TAT TAC TTT GGT AGT GAC TCA AAA GCA GTT ACC GGA CTG CGA ACT 7392 Lys Tyr Tyr Phe Gly Ser Asp Ser Lys Ala Val Thr Gly Leu Arg Thr 2450 2455 2460 ATT GAT GGT AAA AAA TAT TAC TTT AAT ACT AAC ACT GCT GTT GCA GTT 7440 Ile Asp Gly Lys Lys Tyr Tyr Phe Asn Thr Asn Thr Ala Val Ala Val 2465 2470 2475 2480 ACT GGA TGG CAA ACT ATT AAT GGT AAA AAA TAC TAC TTT AAT ACT AAC 7488 Thr Gly Trp Gin Thr Ile Asn Gly Lys Lys Tyr Tyr Phe Asn Thr Asn 2485 2490 2495 ACT TCT ATA GCT TCA ACT 7536 Thr Ser Ile Ala Ser Tnr 2500 TAT TTT AAT ACT GAT GGT 7584 Tyr Phe Asn Thr Asp Gly 2515 GAT GGA TTT GAA TAC TTT 7632 Asp Gly Phe Glu Tyr Phe 2530 GAA GGT CAA GCT ATA CGT 7680 Glu Gly Gin Ala Ile Arg GGT TAT ACA ATT ATT Gly Tyr Thr Ile Ile 2505 ATT ATG CAG ATA GGA Ile Met Gin Ile Gly AGT GGT AAA CAT TTT Ser Gly Lys His Phe 2510 GTG TTT AAA GGA CCT Val Phe Lys Gly Pro 2525 GAT GCT AAC AAT ATA ASp Ala Asn Asn Ile 2520 GCA CCT Ala Pro 2535 TAT CAA Tyr Gin AAT ACA Asn Thr 2 AAT AGA TTC Asn Arg Phe 2555 540
TA
Leu TAT TTA CAT GAC Tyr Leu His Asp 2560 ACT GGT TGG GTA Thr Gly Tro Val 2575 2545 2550 AAT ATA TAT 7728 Asn Ile Tyr TAT TTT GGT AAT Tyr Phe Gly Asn 2565 AAT TCA AAA GCG GCT Asn Ser Lys Ala Ala 2570 ACT ATT GAT GGT 7776 Thr Ile Asp Gly 2580 GCG AAT GGT TAT 7824 Ala Asn Gly Tyr 2595 GGT TTA CCT CAG 7872 Gly Leu Pro Gin 2610 TTT GCA CCT GCT 7920 Phe Ala Pro Ala 2625 CGT TAT CAA AAT 7968 Arg Tyr Gin Asn LAT AGA TAT TAC TTC sn Arg Tyr Tyr Phe 2585 AAA ACT ATT GAT AAT Lys Thr Ile Asp Asn 2600 ATA GGA GTG TTT AAA Ile Gly Val Phe Lys 2615 AAT ACG GAT GCT AAC GAG CCT AAT ACA GCT ATG GGT Glu Pro Asn Thr Ala Met Gly 2590 AAA AAT TTT TAC TTT AGA AAT Lys Asn Phe Tyr Phe Arg Asn 2605 GGG TCT AAT GGA TTT GAA TAC Gly Ser Asn Gly Phe Glu Tyr 2620 AAT ATA GAA GGT CAA GCT ATA Asn Ile Glu Gly Gin Ala Ile 2635 '2640 CTT GGA AAA ATA TAT TAC TTT Leu Gly Lys lie Tyr Tyr Phe 2650 2655 Asn Thr Asp Ala Asn 2630 AGA TTC CTA CAT TTA Arg Phe Leu His Leu 2645 2645 GGT AAT AAT TCA AAA GCA GTT ACT GGA TGG CAA ACT ATT AAT GGT AAA 8016 301 Glv Asn Asn Ser Lys Ala Val Thr Gly Ti-v Gin Thr Ile Asn Gly Lys 2660 2665 2670 GTA TAT TAC TTT ATG CCT GAT ACT GCT ATG GCT GCA GCT GGT GGA CTT 8064 Val Tyr Tyr Phe Met Pro Asp Thi- Ala Met Ala Ala Ala Gly Gly Leu 2675 2680 2685 TTC GAG ATT GAT GGT GTT ATA TAT TTC TTT GGT GTT GAT GGA GTA AAA 8112 Phe Glu Ile Asp Gly Val Ile Tyr Phe Phe Gly Val Asp Gly Val Lys 2690 2695 2700 GCC CCT GGG ATA TAT GGC TAA 6133 Ala Pro Gly Ile Tyr Gly 2705 2710 INFORMATION FOR SEQ ID NO:6: SEQUENCE CHARACTERISTICS: LENGTH: 2710 amino acids TYPE: amino acid TOPOLOGY: linear (ii) MOLECULE TYPE: protein 9 SEQUENCE DESCRIPTION: SEQ ID N4O:6: Met Ser Leu Ile Ser Lys Giu Giu Leu Ile Lys Leu Ala Tyr Ser Ile I5 10 ***Arg Pro Arg Giu Asn Giu Tyr Lys Thr Ile Leu Thr Asn Leu Asp Giu .20 25 *Tyr Asn Lys Leu Thr Thi- Asn Asn Asn Giu Asn. Lys Tyr Leu Gin Leu 35 40 Ly s Lys Leu Asn Giu Ser Ile Asp Val Phe Met Asn Lys Tyr Lys Thr s0 55 *Ser Ser Arg As n Arg Ala Leu Ser Asn Leu Lys Lys Asp Ile Leu Lys 70 75 *Giu Val Ile Leu Ilie Lys Asn Ser Asn Thr Ser Pro Val Giu Lys Asn 90 Leu His Phe Val Trp Ile Gly Gly Glu Val Ser Asp Ile Ala Leu Giu 100 105 110 ***Tyr Ilie Lys Gin Trp Ala Asp Ile Asn Ala Giu Tyr Asn Ile Lys Leu 15 120 125 T-p, Tyi- Asp Ser Giu Ala Phe Leu Val Asn Thr Leu Lys Lys Ala Ile 130 135 140 'Val Giu Ser Ser Thr Thr Giu Ala Leu Gin Leu Leu Giu Giu Giu Ile 145 150 155 160 Gin Asn Pro Gin Phe Asp Asn Met Lys Phe Tyr Lys Lys Arg Met Giu 165 170 175 Phe Ile Tyr Asp Arg Gin Lys Arg Phe Ile Asn Tyr Tyr Lys Ser Gin 180 185 190 Ile Asn Lys Pro Thr Val Pro Thr Ile Asp Asp Ile Ile Lys Ser His 195 200 .205 Leu Val Ser Giu Tyr Asn Arg Asp Giu Thr Val Leu Giu Ser Tyr Arg 210 215 220 302 Thr Asri Ser Leu Arg Lys Ile Asn Ser A-sn 225 230 His Gly Ile Asp Ile Arg 235 240 A-1a Giu Leu Leu Ser 305 Lys Leu Ser Asp Ala 385 Gin Ile Ser Asn Leu Leu Pro 290 Ile Tyr Asp Giu Leu 370 Leu Val1 Giu Leu Ser Leu Phe Thr Giu Gin Giu Leu Leu Asn Ile Tvr Leu Ala 275 Gly Giy Lys Gin Lys 355 Glu Ile Lys Ser Phe 435 Asn 260 Leu Ile Leu Lys Gin 340 Ser Ile Se r Asn Asp 420 Asr 245 Arg Lys His Asp Tyr 325 Leu Glu Lys Lys Arg 405 Asn iSer Gly Asn Ser Arg 310 Ile Lys Ile Ile Gin 390 Tyr Asn Ala Asn I Phe Asp 295 Trp Asn Asp Phe Aila 375 Giy Gin Phe Thr ~eu 1 fly ~80 .eu PAsn Asn Ser 360 Phe Ser Phe Thr Aia 440 ~65 Phe Met Tyr Phe 345 Lys Ala Tyr Leu Asp 425 Gl.
Ala Val' Lys Ile Thr 330 Lys Leu Leu Leu Asn 410 Thr IAsn Al a Thr Lys 315 Se r Leu Giu Gly Thr 395 Gin Thr Ser Ser Leu Ile 300 Leu Giu Ile Asn Ser 380 Asn His Lys Met Asp A~sp 285 Ser Giu Asn Ile Leu 365 Val1 Leu Leu Ile Phe 445 Ile 270 Val Arg Ala Phe Giu 350 Asn Ile Val Asn Phe 430 Leu Ser Gin 255 Val Arg Asp Met Pro Ser Ile Met 320 Asp Lys 335 Ser Lys Vai Ser Asn Gin Ile Glu 400 Pro Ala 415 His Asp Thr Lys *e0.
a *aa.
a.
a. a a a. a a a.
a Leu Gin Vai Gly Phe Met Pro Giu Ala Arg Ser Thr Ile Ala Pro Tyr 450 455 460 Ile 465 Ile Leu Gin Gin Asp 545 Leu Lys Tyr Ser Asn Ile G iu TPhe 530 Asn Leu Asn Giu Leu Leu Glu Ile 515 Giu Gly Asn Tyr Ala Ser Gin Phe 500 Asn Lys Val1 Asn Val 580 Thr Gly Giu 485 Lys Ser Tyr Asp Lys 565 His Cys Pro Gly Ala Tyr Ala Ser 470 475 Asn Thr Ilie Giu Lys Thr 490 Phe Pro Giu Asn Asn Leu 50E- Leu Trp Ser Phe A.sp Gin 520 Val Arg Asp Tyr Thr Gly 535 Phe Asn Lys Asn Thr Ala 550 555 Ile Pro Ser Asn Asn Val 570 Tyr Ile Ile Gin Leu Gin 565 Asn Leu Phe Ser Lys Asn 303- Al a Leu Ser Ala Gly 540 Leu Glu Gly Prc Tyr Lys Gin Ser 525 Ser Asp Giu Asp Lys Tyr Asp Ala Ser 495 Leu Thr 510 Ala Lys Leu Ser Lys Asfl Aia Giy 575 Asp Ile 590 Asn Ser Phe 480 A'sp Giu Tyr Giu Tyr 560 Ser Ser Ile 595 Ile Ile Gin Arg Asn 610 600 Met Asn Giu 615 Ser Ala Lys Ser 620 Phe Leu Ser Asp 625 Arg Lys Leu Ser Tyr 705 Ile Ile Arg Ala Asp Gly Glu Ser Ile Leu Giu Leu Asn Leu Asp Ser Pro 690 Asp Met Thr Lys Ile 770 Lys Giu Asn 675 Lys Phe Asp Ile Giu 755 Met *q Giu Lys Vai L Thr Ser Giu P Ser Ser Phe I 680 Glu Val Asfl 1 695 Giu Giu Thr 710 Thr er Thr Asn Gin Tyr Ala His Ser 760 Leu Ser Ser 775 Lys Ala Lys 790 Ile Lys Thr Ile Leu Asn Ile Tyr Tyr 840 Slie Asp Asp 855 Giu Leu Tyr 870 SIle Ser Phe g Phe Ile Asn S Gu Ile Phe 920 ~ys 1% ~he 65 ~eu 3 ~euI L'yr Leu ilu 745 Gly Lys Se r Leu Asn 825 Giu Leu Giu Giu Lys 905 Ser Pal ~50 aeu Pro Pro 730 VJal Lys Glu Lys Leu 810 Leu Lys Ile Leu Asp 8 90 Ser Lyv Lys 635 Thr P Arg I Thr I Gly( GlyI 715 Asp Arg Tr-p Tyr Asn 795 Leu Lys Leu
ASP
Lys 875 Ile As n sTyr ~he ~eu :le :ys 100 -,ys la 1 Ile Ile 11ie_ 780 Ile Asp Leu Giu Giu 860 Lys Ser Gly Ser Ile Ser Lys 685 Asn Leu Asn Asn Asn 765 Phe Pro Ala Asn Pro 845 Phe Leu Lys Giu Giu 925 iy lal 670 Leu Met Leu Lys Ser 750 Lys Phe Giy Ser Ile 830 Val Asn Asn Asn Ser 910 His 'yr Arg Ile Pro Giu 640 His Gly 655 Asp Ser Asp Ile Phe Ser Leu Ser 720 Asn Ser 735 Giu Gly Giu Giu Asp Ser Leu Ala 800 Val Ser 815 Giu Ser Lys Asn Leu Leu Asn Leu 880 Asn' Ser 895 Val Tyr Ile Thr Ile Asp Asn Lys Leu 785 Ser Pro Ser Ile Giu 865 Asp Th~r Val1 Ile Asp Ile Ile 850 Asn Giu Tyr Glu Ser Thr Gly 835 Hi s Vai Lys Ser Thr 915 Glu Lys 820 Asp Asn Ser Tyr Val 900 Giu Asp 805 Phe Tyr Sex
AST
Le 88~ Arc Ly Lys Giu Ile Ser Thr Ile 930 Asn Ser Ile Ile Thr Asp Val Asn Giy 940 Asn Leu Leu Asp Asn Ile Gin Leu Asp His Thr 945 950 955 Ser Gin Val Asn 3 304 Leu Asn Ala Ala Phe Phe Ile Gin Ser Leu Ile Asp Tyr Ser Ser Asn 965 910 975 Lys Asp Vai Leu Asn Asp Leu Ser Thr Ser Val Lys Vai Gin Leu Tyr 980 985 990 Ala Gin Leu Phe Ser Thr Giy Leu Asn Thr Ile Tyr Asp Ser Ile Gin 995 1000 1005 Leu Val Asn Leu Ile Ser Asn Ala Val Asn Asp Thr Ile Asn Val- Leu 1010 lois 1020 Pro Thr Ile Thr Giu Gly Ile Pro Ile Val Ser Thr Ile Leu Asp Giy 1025 1030 1035 1040 Ile Asn Leu Giy Aia Ala Ile Lys Glu Leu Leu Asp Giu His Asp Pro 1045 1050 1055 Leu Leu Lys Lys Giu Leu Glu Ala Lys Val Gly Val Leu Ala Ile Asn 1060 1065 1070 Met Ser Leu Ser Ile Ala Ala Thr.Vai Ala Ser Ile Val Gly Ile Gly 1075 1080 1085 Ala Giu Val Thr Ile Phe Leu Leu Pro Ile Ala Gl Ile Ser Ala Gly 1090 1095 1100 Ile Pro Ser Leu Val Asn Asn Glu Leu Ile Leu His Asp Lys Ala Thr .1105 1110 1115 1120 Ser Val Val Asn Tyr Phe Asn His Leu Ser Giu Ser Lys Lys Tyr Gly 1125 1130 1135 *Pro Leu Lys Thr Glu Asp Asp Lys Ile Leu Va1,Pro Ile Asp Asp Leu *1140 1145 1150 **.Val Ile Ser Glu Ile Asp Phe Asn Asn Asn Ser Ile Lys Leu Gly Thr 1155 1160 1165 Cys Asn Ile Leu Ala Met Glu Gly Gly Ser Gly His Thr Val Thr Gly *1170 1175 1180 *Asn Ile Asp His Phe Phe Ser Ser Pro Ser Ile Ser Ser His Ile Pro 1185 1190 1195 1200 *Ser Leu Ser Ilie Tyr Ser Ala Ile Gly Ile Glu Thr Giu Asn Leu Asp :1205 1210 1215 Phe Ser Lys Lys Ile Met Met Leu Pro Asn Ala Pro Ser Arg Val Phe 1220 1225 1230 *Trp Trp Giu Thr Gly Ala Val Pro Gly Leu Arg Ser Leu Giu Asn Asp *1235 1240 1245 Thr Arg Leu Leu Asp Ser Ile Arg Asp Leu Tyr Pro Gly Lys Phe 1250 1255 1260 Tyr Trp Arg Phe Tyr Ala Phe Phe Asp Tyr Ala Ile Thr Thr Leu Lys 1265 1270 1275 1280 Pro Val Tyr G1u Asp Thr Asn Ile Lys Ile Lys Leu Asp Lys Asp Thr 1285 1290 1295 305 Arg Asn Phe Ile Met Pro Thr Ile Thr Thr Asn Giu Ile Arg Asn Lys 1300 1305 1310 Leu Ser Tyr Ser Phe Asp Gly Ala Gly Gly Th-r Tyr Ser Leu Leu Leu 1315 1320 1325 Ser Ser Tyr Pro Ilie Ser Thr Asn Ile Asn Leu Ser Lys Asp Asp Leu 1330 1335 1340 Trp Ile Phe Asn Ile Asp Asn Glu Val Arg Giu Ile Ser Ile Giu Asn 1345 1350 1355 1360 Gly Thr Ile Lys Lys Gly Lys Leu Ile Lys Asp Val Leu Ser Lys Ile 1365 1370 1375 Asp Ile Asn Lys Asn Lys Leu Ile Ile Gly Asn Gin Thr Ile Asp Phe 1380 1385 1390 Ser Gly Asp Ile Asp Asn Lys Asp Arg Tyr Ile Phe Leu Thr Cys Glu 1395 1400 1405 Leu Asp Asp Lys Ile Ser Leu Ile Ile Giu Ile Asn Leu Val Ala Lys 1410 1415 1420 Ser Tyr Ser Leu Leu Leu Ser Gly Asp Lys Asn Tyr Leu Ile Ser Asn 1425 1430 1435 1440 ***Leu Ser Asn Thr Ile Giu Lys Ilie Asn Thr Leu Gly Les Asp Ser Lys *1445 1450 1455 Asn Ile Ala Tyr Asn Tyr Thr Asp Glu Ser Asn Asn Lys Tyr Phe Gly .to. 1460 1465 1470 **Ala Ile Ser Lys Thr Ser Gin Lys Ser Ile Ile His Tyr Lys Lys Asp S1475 1480 1485 Ser Lys Asn Ilie Leu Glu Phe Tyr Asn Asp Ser Thr Leu Giu Phe Asn 1490 1495 1500 Ser Lys Asp Phe Ilie Ala Glu Asp Ile Asn Val Phe Met Lys Asp Asp 1505 1510 1515 1520 *lie Asn Thr Ile Thr Gly Lys Tyr Tyr Val Asp Asn Asn Thr Asp Lys 1525 1530 1535 *Ser Ile Asp Phe Ser Ile Ser Leu Vai Ser Lys Asn Gin Val Lys Vai *1540 1545 1550 *Asn Gly Leu Tyr Leu Asn Glu Ser Val Tyr Ser Ser Tyr Leu Asp Phe 1555 1560 1565 **:Vai Lys Asn Ser Asp Gly His His Asn Thr Ser Asn Phe Met Asn Leu 1570 1575 1580 ea*se Phe Leu Asp Asn Ile Ser Phe Trp Lys Leu Phe Giy Phe Giu Asn Ile 1585 1590 1595 1600 Asn Phe Val Ile Asp Lys Tyr Phe Thr Leu Val Gly Lys Thr Asn Leu -1605 1610 1615 Gly Tyr Val Giu Phe Ile Cys Asp ASn Asn Lys Asn Ile Asp Ile Tyr 1620 1625 1630 306 Phe Gly Glu Trp Lys Thr Ser Ser Ser Lys Ser Thr le Phe Ser Gly 1635 1640 1645 Asn Gly Arg Asn Val Val Val Glu Pro Ile Tyr Asn Pro Asp Thr Gly 1650 1655 1660 Glu Asp Ile Ser Thr Ser Leu Asp Phe Ser Tyr Glu Pro Leu Tyr Gly 1665 1670 1675 1680 Ile Asp Arg Tyr Ile Asn Lys Val Leu Ile Ala Pro Asp Leu Tyr Thr 1685 1690 1695 Ser Leu Ile Asn Ile Asn Thr Asn Tyr Tyr Ser Asn Glu Tyr Tyr Pro 1700 1705 1710 Glu Ile Ile Val Leu Asn Pro Asn Thr Phe His Lys Lys Val Asn Ile 1715 1720 1725 Asn Leu Asp Ser Ser Ser Phe Glu Tyr Lys Trp Ser Thr Glu Gly Ser 1730 1735 1740 Asp Phe Ile Leu Val Arg Tyr Leu Glu Glu Ser Asn Lys Lys Ile Leu 1745 1750 1755 1760 Gin Lvs Ile Arg Ile Lys Gly Ile Leu Ser Asn Thr Gin Ser Phe Asn 1765 1770 1775 Lys Met Ser Ile Asp Phe Lys Asp Ile Lys Lys Leu Ser Leu Gly Tyr S1780 1785 1790 *000 Ile Met Ser Asn Phe Lys Ser Phe Asn Ser Glu Asn Glu Leu Asp Arg 1795 1800 1805 Asp His Leu Gly Phe Lys Ile Ile Asp Asn Lys Thr Tyr Tyr Tyr Asp 1810 1815 1820 Glu Asp Ser Lys Leu Val Lys Gly Leu Ile Asn Ile Asn Asn Ser Leu 1825 1830 1835 1840 Phe Tyr Phe Asp Pro Ile Glu Phe Asn Leu Val Thr Gly Trp Gin Thr 1845 1850 1855 lie Asn Gly Lys Lys Tyr Tyr Phe Asp Ile Asn Thr Gly Ala Ala Leu 1860 1865 1870 SThr Ser Tyr Lys Ile Ile Asn Gly Lys His Phe Tyr Phe Asn Asn Asp 1875 1880 1885 o Gly Val Met Gin Leu Gly Val Phe Lys Gly Pro Asp Gly Phe Glu Tyr 1890 1895 1900 SPhe Ala Pro Ala Asn Thr Gin Asn Asn Asn Ile Glu Gly Gin Ala Ile 0 0 1905 1910 1915 1920 *oo Val Tyr Gin Ser Lys Phe Leu Thr Leu Asn Gly Lys Lys Tyr Tyr Phe 1925 1930 1935 Asp Asn Asn Ser Lys Ala Val Thr Gly Trp Arg Ile Ile Asn Asn Glu 1940 1945 1950 Lys Tyr Tyr Phe Asn Pro Asn Asn Ala Ile Ala Ala Val Gly Leu Gin 1955 1960 1965 307- Val Ilie Asp Asn Asn Lys Tyr Tyr Phe Asn Pro Asp Thr Ala Ile Ile 1970 1975 1980 Ser Lys Gly Trp Gin Thr Val Asn Gly Ser Arg Tyr Tyr Phe Asp Thr 1985 1990 1995 2000 Asp Thr Ala Ile Ala Phe Asn Gly Tyr Lys Thr Ile Asp Gly Lys His 2005 2010 2015 Phe Tyr Phe Asp Ser Asp Cys Val Val Lys Ile Gly Val Phe Ser Thr 2020 2025 2030 Ser Asn Gly Phe Glu Tyr Phe Ala Pro Ala Asn Thr Tyr Asn Asn Asn 2035 2040 2045 Ilie Giu Gly Gin Ala Ile Val Tyr Gin Ser Lys Phe Leu Thr Leu Asn 2050 2055 2060 Gly Lys Lys Tyr Tyr Phe Asp Asn Asn Ser Lys Ala Val Thr Gly Leu 2065 2070 2075 2080 Gin Thr Ile Asp Ser Lys Lys Tyr Tyr Phe Asn Thr Asn Thr Aia Giu 2085 2090 2095 Ala Ala Thr Gly Trp Gin Thr Ile Asp Gly Lys Lys Tyr Tyr Phe Asn 2100 2105 2110 *Thr Asn Thr Ala Giu Ala Ala Thr Gly Trp Gin Thr Ile Asp Gly Lys 2115 2120 2,125 Lys Tyr Tyr Phe Asn Thr Asn Thr Ala Ile Ala Ser Thr Gly Tyr Thr *2130 2135 2140 *le Ilie Asn Gly Lys His Phe Tyr Phe Asn Thr Asp Gly Ile Met Gin **.2145 2150 2155 2160 Ilie Gly Val Phe Lys Gly Pro Asn Gly Phe Giu Tyr Phe Ala Pro Ala *2165 2170 2175 Asn Thr Asp Ala Asn Asn Ile Giu Gly Gin Ala Ile Leu Tyr Gin Asn .2180 2185 2190 Giu Phe Leu Thr Leu Asn Gly Lys 'Lys Tyr Tyr Phe Gly Ser Asp Ser *2195 2200 2205 *Lys Ala Val Thr Gly Trp, Arg Ile Ile Asn Asn Lys Lys Tvr Tyr Phe 2210 2215 2220 Asn Pro Asn Asn Ala Ile Ala Ala Ile His Leu Cys Thr Ile Asn Asn 2225 2230 2235 2240 Asp Lys Tyr Tyr Phe Ser Tyr Asp Gly Ile Leu Gin Asn Gly Tyr Ile 2245 -250 2255 Thr Ile Glu Arg Asn Asn Phe Tyr Phe Asp Ala Asn Asn Giu Ser Lys 2260 2265 2270 Met Val Thr Gly Val Phe Lys Gly Pro Asn Gly Phe Giu Tyr Phe Ala 2275 2280 2285 Pro Ala Asn Thr His Asn Asn Asn Ile Giu Gly Gin Ala Ile Val Tyr 2290 2295 2300 308 Gin Asn Lys Phe Leu Thr Leu Asn Gly Lys Lys Tyr Tyr Phe Asp Asn 2305 2310 2315 2320 Asp Ser Lys Ala Val Thr Gly Trp Gin Thr Ile Asp Gly Lys Lys Tyr 2325 2330 2335 Tyr Phe Asn Leu Asn Thr Ala Glu Ala Ala Thr Gly Trp Gin Thr Ile 2340 2345 2350 Asp Gly Lys Lys Tyr Tyr Phe Asn Leu Asn Thr Ala Glu Ala Ala Thr 2355 2360 2365 Gly Trp Gin Thr Ile Asp Gly Lys Lys Tyr Tyr Phe Asn Thr Asn Thr 2370 2375 2380 Phe Ile Ala Ser Thr Gly Tyr Thr Ser Ile Asn Gly Lys His Phe Tyr 2385 2390 2395 2400 Phe Asn Thr Asp Gly Ile Met Gin Ile Gly Val Phe Lys Gly Pro Asn 2405 2410 2415 Gly Phe Glu Tyr Phe Ala Pro Ala Asn Thr Asp Ala Asn Asn Ile Glu S* 2420 2425 2430 Gly Gin Ala Ile Leu Tyr Gin Asn Lys Phe Leu Thr Leu Asn Gly Lys 2435 2440 2445 Lys Tyr Tyr Phe Gly Ser Asp Ser Lys Ala Val Thr Gly Leu Arg Thr S*2450 2455 2460 Ile Asp Gly Lys Lys Tyr Tyr Phe Asn Thr Asn Thr Ala Val Ala Val 2465 2470 2475 2480 Thr Gly Trp Gin Thr Ile Asn Gly Lys Lys Tyr Tyr Phe Asn Thr Asn S2485 2490 2495 Thr Ser Ile Ala Ser Thr Gly Tyr Thr Ile Ile Ser Gly Lys His Phe 2500 2505 2510 STyr Phe Asn'Thr Asp Gly Ile Met Gin Ile Gly Val Phe Lys Gly Pro 2515 2520 2525 Asp Gly Phe Glu Tyr Phe Ala Pro Ala Asn Thr Asp Ala Asn Asn Ile 2530 2535 2540 Glu Gly Gin Ala Ile Arg Tyr Gin Asn Arg Phe Leu Tyr Leu His Asp 2545 2550 2555 2560 Asn Ile Tyr Tyr Phe Gly Asn Asn Ser Lys Ala Ala Thr Gly Trp Val 2565 2570 2575 Thr Ile Asp Gly Asn Arg Tyr Tyr Phe Glu Pro Asn Thr Ala Met Gly 2580 2585 2590 Ala Asn Gly Tyr Lys Thr Ile Asp Asn Lys Asn Phe Tyr Phe Arg Asn 2595 2600 2605 Gly Leu Pro Gin Ile Gly Val Phe Lys Gly Ser Asn Gly Phe Glu Tyr -2610 2615 2620 Phe Ala Pro Ala Asn Thr Asp Ala Asn Asn Ile Glu Gly Gin Ala Ile 2625 2630 2635 2640 309 lw Arg Tyr Gin Asn Arg Phe Leu His Leu Leu Gly Lys Ile Tyr Tyr Phe 2645 2650 2655 Giy Asn Asn Ser Lys Ala, Val Thr Gly Trp Gin Thr Ile Asn Gly Lys 2660 2665 2670 Val Tyr Tyr Phe Met Pro Asp Thr Ala Met Ala Ala Ala Gly Gly Leu 2675 2680 2685 Phe Glu Ilie Asp Gly Val Ile Tyr Phe Phe Gly Val Asp Gly Val Lys 2690 2695 2700 Ala Pro Gly Ile Tyr Gly 2705 2710 INFORMATION FOR SEQ ID NO:7: Ci) SEQUENCE CHARACTERISTICS: LENGTH: 811 amino acids TYPE: amino acid STR.ANDEDNESS: unknown TOPOLOGY: unknown (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:7: Ser Tyr Lys Ilie Ile Asn Giy Lys His Phe Tyr Phe Asn Asn Asp Gly 10 is **.Val Met Gin Leu Gly Val Phe Lys Gly Pro Asp Giy Phe Giu Tyr Phe 20 25 Ala Pro Ala Asn Thr Gin Asn Asn Asn Ile Giu Gly Gin Ala Ile Val 40 *Tyr Gin Ser Lys Phe Leu Thr Leu Asn Gly Lys Lys Tyr Tyr Phe Asp 50 .55 **Asn Asn Ser Lys Ala Val Thr Gly Trp Arg Ile Ile Asn Asn Glu Lys 70 75 Tyr Tyr Phe Asn Pro Asn Asn Ala Ilie Ala Ala Val Gly Leu Gin Val 90 I**le Asp Asn Asn Lys Tyr Tyr Phe Asn Pro Asp Thr Ala Ile Ile Ser *.100 105 110 Lys Gly Trp Gin Thr Val Asn Gly Ser Arg Tyr Tyr Phe Asp Thr Asp 115 120 125 Thr Ala Ile Ala Phe Asn Gly Tyr Lys Thr Ile Asp Gly Lys His Phe 130 135 140 Tyr Phe Asp Ser Asp Cys Val Val Lys Ile Gly Val Phe Ser Thr Ser 145 iSO 155 160 Asn Gly Phe Glu Tyr Phe Ala Pro Ala Asn Thr Tyr Asn Asn Asn Ile 165 170 175 Glu Gly Gin Ala Ile Val Tyr Gin Ser Lys Phe Leu Thr Leu Asn Gly 180 185 190 310- Lys Lys Tvr Tyr 195 Phe Asp Asf Asn Ser Lys Ala Val 200 Thr Gly Leu Gin 205 Thr Ala 225 Asn Tyr Ile Gly Thr 305 Phe Ala Asp Gly Ala Phe Gly 275 Phe Ala Thr Thr Ser Trp Glu Asn 260 Lys Lys Asn Leu Gly 340 Lys Gin Ala 245 Thr His Gly Asn Asn 325 Trp Lys Tyr Tyr Phe 1 215 Ile Thr Thr Tyr Asn 295 Glu Lys Ile Asp Gly Gly Trp Ala Ile 265 Phe Asn 280 Gly Phe Gly Gin Lys Tyr Ile Asn 345 Ile HiS 360 Gly Ile Asn Lys Gin 250 A1a Thr.
Glu Ala Tyr 330 Asn Leu Leu Thr Lys 235 Thr Ser Asp Tyr Ile 315 Phe Lys Cys Gin Asn 395 Asn 220 Tyr Ile Thr Gly Phe 300 Leu Gly Lys Thr Asn 380 Thr Tyr Asp Gly Ile 285 Ala Tyr Ser Tyr Ile 365 Gly kla Phe Gly Tyr 270 Met Pro Gin Asp Tyr 350 Asn Tyr Glu Asn Lys 255 Thr Gin Ala Asn Ser 335 Phe Asn Ile Pro Asn Asn Ala Ile Ala Ala 355 Lys Tyr Tyr Phe Ser Tyr Asp 370 375 Ala Thr 240 Lys Ile Ile Asn Glu 320 Lys Asn Asp Thr Met 400 Pro Gin 1 Asp Tyr Asp 480 r Gly
S
r Phe r Phe Ile Glu Arg Asn 385 Asn Phe Tyr Phe Asp Ala Asn Glu Ser Lys 390 Val Ala Asn Ser Phe 465 Gly Trp Ile Thr Gly Val Phe Lys Gly Pro 405 Asn Thr His Asn Asn Asn Ile 420 Lys Phe Leu Thr Leu Asn Gly 435 440 Lys Ala Val Thr Gly Trp Gin 450 455 Asn Leu Asn Thr Ala Giu Ala 470 Lys Lys Tyr Tyr Phe Asn Leu 485 Gin Thr Ile Asp Gly Lys Lys 500 Ala Ser Thr Gly Tyr Thr Ser 515 520 Asn Glu 425 Lys Thr Ala Asn Tyr 505 Ile Gly 410 Gly Lys Ile Thr Thr 490 Tyr Asn Phe Glu Tyr Phe Gin Ala Ile Val 430 Tyr Tyr Phe Asp 445 Asp Gly Lys Lys 460 Gly Trp Gin Thr 475 Ala Giu Ala Ala Phe Asn Thr Asn 510 Gly Lys His Phe 525 Ala 415 Tyr Asr Iie Th: 49 Th Ty .311 Asn Phe 545 Gin Tyr Asp Gly Ser 625 Phe Gly Gly Thr 530 Giu Al a Tyr Gly Trp 610 Ile Asn Phe Gin Asp Tyr Ile Phe Lys 595 Gin Aia Thr Giu Al a 675 Gly Ile Met Phe Ala Pro 550 Leu Tyr Gin 565 Gly Ser Asp 580 Lys Tyr Tyr Thr Ile Asn Ser Thr Giy 630 Asp Giy Ile 645 Tyr Phe Ala 660 Ile Arg Tyr Phe Giy Asn Gin Ilie 535 Ala Asn Val Phe Lys Gly Pro Asn Giy 540 Asp Ala Asn Asn Ile Giu Giy Asn Lys Phe Leu Ser Lys Ala 585 Phe Asn Thr 600 Gly Lys Lys 615 Tyr Thr Ile Met Gin Ilie Pro Ala Asn 665 Gin Asn Arg 680 Asn Ser Lys 695 Val Asn Tyr Ile Gly 650 Thr Phe Ala Pro Thr Leu Thr Giy Thr Ala Tyr Phe 620 Ser Gly 635 Val Phe Asp Ala Leu Tyr Ala Thr 700 Asn Thr 715 Gly Arg 590 Ala Thr His Gly Asn 670 His Trp Met Lys 575 Thr Val Asn Phe Pro 655 Ile Asp Val1 Gly Lys le Thr Thr Tyr 640 Asp Giu Asn Thr Ala 720 Ile Tyr Tyr 690 Ile Asp Gly Asn Arg Tyr Tyr Phe Giu 705 710 Asn Gly Tyr Lys Thr Ile Asp Asn Lys Asn Phe Tyr Phe Arg 725 730 Asn Gly 735 Leu Pro Gin Ile Gly Val Phe Lys Gly Ser Asn Giy Phe Giu Tyr Phe 740 745 750 Ala Pro Ala Asn Thr Asp Ala Asn Asn Ilie Giu Gly Gin Ala Ile Arg 755 '760 765 Tyr Gin Asn Arg Phe Leu His Leu Leu Gly Lys Ile Tyr Tyr Phe Gly 770 775 780 Asn Asn Ser Lys Ala Val Thr Giy Tr-p Gin Thr Ile Asn Gly Lys Val 785 790 795 800 Tyr Tyr Phe Met Pro Asp Thr Ala Met Ala Ala 1805 810 INFORMATION FOR SEQ ID NO:8: SEQUENCE CHARACTERISTICS: LENGTH: 91 amino acids TYPE: amino acid STRANDEDNESS: unknown TOPOLOGY: unknown (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:B: Ser Tyr Lys Ile Ile Asn Gly Lys His Phe Tyr Phe Asn Asn Asp Gly 1 5 10 Val Met Gin Leu Gly Val Phe Lys Gly Pro Asp Gly Phe Giu Tyr Phe 2530 Ala Pro Ala Asn Thr Gin Asn Asn Asn Ile Glu Gly Gin Ala Ile Val 40 Tyr Gin Ser Lys Phe Leu Thr Leu Asn Gly Lys Lys Tyr Tyr Phe Asp 55 Asn Asn Ser Lys Ala Val Thr Gly Trp Ag Ile Ile Asn Asfl Giu Lys 10 75 Tyr Tyr Phe Asn Pro Asn Asn Ala Ile Ala Ala 85 INFORMATION FOR SEQ ID NO:9: SEQUENCE CHARACTERISTICS: LENGTH: 7101 base pairs TYPE: nucleic acid STRANDEDNESS: singie TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic) (ix) FEATURE: NAME/KEY:
CDS
LOCATION: 1. .7098 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:9: ATG AGT TTA GTT AAT AGA AAA CAG TTA GAA AAA ATG GCA AAT GTA AGA 48 Met Ser Leu Val Asn Arg Lys Gin Leu Giu Lys Met Ala Asn Val Arg *1 5 10 *TTT CGT ACT CAR GAA GAT GAA TAT GTT GCA ATA TTG GAT GCT TTA GAA 96 Phe Arg Thr Gin Glu Asp Glu Tyr Val Ala Ile Leu Asp Ala Leu Giu 20 25 *GAA TAT CAT ART ATG TCA GAG ART ACT GTA GTC GAA AAA TAT TTA AAA 144 *Giu Tyr His Asn Met Ser Glu Asn Thr Val Val Glu Lys Tyr Leu Lys 35 40 **.TTA AAA GAT ATA ART AGT TTA ACA GAT ATT TAT ATA GAT ACA TAT AAA 192 Leu Lys Asp Ile Asn Ser Leu Thr Asp Ile Tyr Ile Asp Thr Tyr Lys 55 AAA TCT GGT AGA ART AAA GCC T1'A AAA AMA TTT AAG GAA TAT CTA GTT 240 Lys Ser Gly Arg Asn Lys Ala Leu Lys Lys Phe Lys Giu Tyi- Leu Val 70 75 ACA GMA GTA TTA GAG CTA MAG ART AMT AT TTA ACT CCA GTT GAG AMA 288 Thr Glu Val Leu Giu Leu Lys Asn Asn Asn Leu Thr Pro Val Glu Lys 85 90 MAT TTA CAT TTT GTT TGG ATT GGA GGT CMA ATA ART GAC ACT GCT ATT 336 Asn Leu His Phe Val Trp Ile Gly Gly Gin Ile Asn Asp Thr Ala Ile 100 105 110 MAT TAT ATA MAT CMA TGG AMA GAT GTA AAT AGT GAT TAT MAT GTT AAT 384 Asfl Tyr Ile Asn Gin Trp Lys Asp Val Asn Ser Asp Tyr Asn Val Asn 115 120 125 313 GTT TTT TAT GAT AGT A.AT GCA TTT TTG ATA AAC ACA TTG AAA AAA ACT 432 Val Phe Tyr Asp Ser Asn Ala Phe Leu Ile Asn Thr Leu Lys Lys Thr 130 135 140 GTA GTA GAA TCA GCA ATA AAT GAT ACA CTT GAA TCA TTT AGA GAA AAC 480 Val Vai Giu Ser Ala Ile Asn Asp Thr Leu Giu Ser Phe Arg Giu Asn 150 AAT GAC CCT AGA TTT GAC TAT AAT Asn Asp Pro Arg Phe Asp Tyr Asn 165 AAA TTC TTC AGA AAA CGT Lys Phe Phe Arg Lys Arg 170 175 GAA ATA ATT TAT GAT AAA CAG AAA 576 Gu Ile Ile Ty~r Asp Lys Gin Lys 160 AAT TTC ATA AAC TAC Asn Phe Ile Asfl Tyr 185 ATA ATT GAT GAT ATT Ile Ile Asp Asp Ile 205 TAT AAA GCT Tyr Lys Ala 190 GTA AAG ACA Val Lys Thr S.
S
CAA AGA GAA 624 Gin Arg Giu 195 TAT CTT TCA 672 Tyr Leu Ser 210 ATT GAA GAA 720 Ile Glu Glu GAA AAT Giu Asn AAT GAG Asn Giu CCT GAA CTT Pro Giu Leu 200 TAT TCA AAG Tyr Ser Lys GAG ATA GAT Giu Ile Asp 225 AGA AAC 768 Arg Asn TCC TTA AAT Ser Leu Asn 230 GAA GAA TTT Giu Giu Phe 245
AAA
Lys
AAA
Lys GAA CTT AAT Giu Leu Asn 220 AGT GGA AAT Ser Gly Asn mT Phe ACC TAT Thr Tyr ATT ACA CAG AAT Ile Thr Gin Asn 235 AAT GGA GAG TCA Asn Gly Giu Ser 250 AAT TTA GCT OCT Asn Leu Ala Ala TTC AAC Phe Asn TTA TAT GAA Leu Tyr Giu 255 CAA GAG TTG GTA GAA 816 Gin Giu Leu Val Glu 260 AGA ATA TCT GCA TTA 864 Arcx Ie Ser Ala Leu AGG TGG Arg Trp AAA GAA Lys Glu Ala Ser Asp Ile Leu 270 275 ATG TTA CCA GGA 912 Met Leu Pro Gly 290 ATT GGT Ile Giy 280 GAC TTA Asp Leu Oly Met Tyr Leu Asp Val Asp 285 TT' GAG TCT ATA GAG AAA CCT Phe Giu Ser Ile Oiu Lys Pro ATA CAA CCA Ile Gin Pro 295 AGT TCA 960 Ser Ser 305.
ATG AAA, loos8 Met Lys GTA ACA GTG GAT Val Thr Val Asp 310 TAC AAA GAA TAT Tyr Lys Giu Tyr 325 GAC GAA GAA. OTT Asp Giu Glu Val 340 Phe TGG GAA ATG ACA Trp, Giu Met Thr 315 TTA GAA GCT Leu Giu Ala ATA CCA GAA Ile Pro Giu CAA AGT AGT Gin Ser Ser 345 ACC TCA GAA CAT TTT GAC Thr Ser Giu His Phe Asp 335 GAA TCT GTT CTA OCT TCT Giu Ser Val Leu Ala Ser 350 ATO TTA 1056 Met Leu -314 AAG TCA GAT AAA TCA GAA ATA TTC TCA. TCA CTT GGT GAT ATG GAG GCA 1104 Lys Ser Asp Lys Ser Giu Ile Phe Ser Ser Leu Gly Asp Met Giu Ala 355 360 365 TCA CCA CTA GAA GTT AAA ATT GCA TTT AAT AGT AAG GGT ATT ATA AAT 1152 Ser Pro Leu Giu Val Lys Ile Ala Phe Asn Ser Lys Gly Ile Ile Asn 3'70 375 380 CAA GGG CTA ATT TCT GTG AAA GAC TCA TAT TGT AGC AAT TTA ATA GTA 1200 Gin Gly Leu Ile Ser Val Lys Asp Ser Tyr Cys Ser Asn Leu Ile Val 385 390 395 400 AAA CAA ATC GAG AAT AGA TAT AAA ATA TTG AAT AAT AGT TTA AAT CCA 1248 Lys Gin Ile Giu Asn Arg Tyr Lys Ile Leu Asn Asn Ser Leu Asn Pro 405 410 415 GCT ATT AGC GAG GAT AAT GAT TTT AAT ACT ACA ACG AAT ACC TTT ATT 1296 *Ala Ilie Ser Giu Asp Asn Asp Phe Asn Thr Thr Thr Asn Thr Phe Ile 420 425 430 *GAT AGT ATA ATG GCT GAA GCT AAT GCA GAT AAT GGT AGA TTT ATG ATG 1344 .Asp Ser Ile Met Ala Giu Ala Asn Ala Asp Asn Gly Arg Phe Met Met *435 440 445 GAA CTA GGA AAG TAT TTA AGA GTT GGT 'ITC TTC CCA GAT GTT AAA ACT 1392 Giu Leu Giy Lys Tyr Leu Arg Val Gly Phe Phe Pro Asp Val Lys Thr *450 455 460 *ACT ATT AAC TTA AGT GGC CCT GAA GCA TAT GCG GCA GCT TAT CAA GAT 1440 Thr Ile Asn Leu Ser Gly Pro Giu Ala Tyr Ala Ala Ala Tyr Gin Asp **9465 470 475 480 TTA TTA ATG TTT AAA GAA GGC AGT ATG AAT ATC CAT TTG ATA GAA GCT 1488 *9**Leu Leu Met Phe Lys Giu Gly Ser t4et Asn Ile His Leu Ile Giu Ala 485 490 495 *GAT TTA AGA AAC rrr GAA ATC TCT AAA ACT AAT ATT TCT CAA TCA ACT 1536 Asp Leu Arg Asn Phe Giu Ile Ser Lys Thr Asn Ile Ser Gin Ser Thr 500 505 1 GAA CAA GAA ATG GCT AGC TTA TGG TCA TTT GAC GAT GCA AGA GCT AAA 1584 Giu Gin Giu Met Ala Ser Leu Trp Ser Phe Asp Asp Ala Arg Aia Lys 515 520 525 GCT CAA ITT GAA GAA TAT AAA AGG AAT TAT TTT GAA GGT TCT CIT GGT 1632 Ala Gin Phe Giu Giu Tyr Lys Arg Asn Tyr Phe Glu Giy Ser Leu Gly 530 535 540 GAA GAT GAT AAT CTT GAT TTT TCT CAA AAT ATA GTA GTT GAC AAG GAG 1680 Giu Asp Asp Asn Leu ASP Phe Ser Gin Asn Ile Val Val Asp Lys G1u 545 550 555 560 TAT CIT ITA GAA AAA ATA TCT TCA TTA GCA AGA AGT TCA GAG AGA GGA 1128 Tyr Leu Leu Giu Lys Ile Ser Ser Leu Ala Arg Ser 5er Giu Arg Gly 565 570 575 -315 TAT ATA CAC TAT AT? GTT CAG TTA CAA GGA GAT AAA ATT AGT TAT GAA 1776 Tyr Ile His Tyr Ile Val Gin Leu Gin Gly Asp Lys Ile Ser Tyr Giu 560 585 590 GCA GCA TGT AAC TTA TTT GCA AAG ACT CCT TAT GAT AGT GTA CTG TTT 1824 Ala Ala Cys Asn Leu Phe Ala Lys Thr Pro Tyr Asp Ser Val Leo Phe 595 600 605 CAG AAA AAT ATA GAA GAT TCA GAA ATT GCA TAT TAT TAT AAT CCT GGA 1872 Gin Lys Asn Ile Giu Asp Ser Giu Ile Ala Tyr 610 615 GAT GOT GAA ATA CAA GAA ATA GAC AAG TAT AAA Tyr Tyr Asn Pro Gly 620 ATT CCA AGT ATA ATT le Pro Ser Ile Ile *4
S
*4
S
*5 S S S *55* 4*S* 1920 Asp Giy 625 TCT GAT 1968 Ser Asp GAA TTT 2016 Gio Phe Giu Ile Gin AGA CCT AAG Arg Pro Lys 645 AAT ACT GAT Asn Thr Asp AAA TTA ACA Lys Leu Thr GOT CAT Giy His GGT AAA GAT Giy Lys Asp 655 Ile Asp Lys Tyr ATA TTT GCA GGT Ile Phe Ala Gly 665 ACA GAA ATA 2064 Thr Giu Ile 675 A.AG TCA ATA 2112 Lys Ser Ile 690 ATC AAC OTA 2160 Ile Asn Vai 705 GAT AAA ATA 2208 Asp Lys Ile 660
GAA
Giu
GAA
Giu GCA GCA ATA GAT TTA GCT Ala Aia Ilie Asp Leu Ala 680 ATA AAT TTA TTA GGA TGT Ile Asn Leu Leu Gly Cys 695 OAT GTA GAT TCA TTA TCC Asp Val Asp Ser Leo Ser 670 AAA GAG GAT ATT TCT CCT Lys Glu Asp Ile Ser Pro 685 AAT ATG Ti'? AGC TAC TCT Asn Met Phe Ser Tyr Ser GAG GAG G10 Glu ACT TAT CCT GGA Thr Tyr Pro Gly 710 AAA TTA Lys Leu '715 ATA AGT Ilie Ser CTT AAA OTT Leo Lys Val TCA GAA TTA ATO Ser Giu Leo Met 725 Pro Ser GAC TCT ATT ATA Asp Ser Ile Ile 735 GTA AGT 2256 Val Ser
GCA
Ala
AAT
Asn 740
CAA
Gin TAT GAA OTT AGA Tyr Giu Val Arg 745 TCT GOT GAA TG Ser Gly Glu Trp 760 ATA AAT AGT GAA OGA AGA AGA Ile Asn Ser 010 Oly Arg Arg 750 ATA AAT AAA GAA GAA AGT ATT Ilie Asn Lys Oiu Oiu Ser Ile 765 OAA TTA TTG OAT CAT 2304 Glu Leu Leo Asp His 755 ATA AAG OAT ATT TCA 2352 Ile Lys Asp Ile Ser 770 TCA AAA GAA TAT ATA TCA TTT AAT CCT AAA GAA Ser Lys Glu Tyr Ile Ser Phe Asn Pro Lys OiU 775 780 AAT AAA ATT ACA GTA AAA TCT AMA AAT TTA CCT GAG CTA TCT ACA TTA 2400 Asn Lys Ile Thr Val Lys Ser Lys Asn Leo Pro Glu Leu Ser Thr Leu 785 790 795 800 316 TTA CAA 2448 Leu Gin GAA AAA 2496 Glu Lys GAT ACG 2544 Asp Thr TCT GAC 2592 Ser Asp 850 ATT TCT 2640 Ile Ser 865 TCT CAT 2688 Ser His AGT ATA 2736 Ser Ile GAA ATT Glu Ile GTA ATG Val Met 820 CAA ATT Gin Ile AAT AAT TCT AAT Asn Asn Ser Asn ACA GAA TGT GAG Thr Giu Cys Glu 825 TCA AGT GAT Ser Ser Asp 83.0 ATA AAT GT? Ile Asn Vai GAA C-A GAA Giu Leu Giu 815 TCA AAT ATA Ser Asn Ile GT? GAG GAA AGG AT? Val Giu Giu Arg Ile 840 4* 0**S *4 6.
S
OSS*
S.
S
S.
S *5
S.
LB S TCT AT? AAT TAT ATA Ser Ile Msn Tyr Ile 855 GA? GCA CTA TGT GAC Asp Ala Leu Cys Asp 870 ?T ATA TCT TTT GAG Phe Ile Ser Phe Giu 885 Lys Asp TTA AAA Leu Lys GAA GAA Giu Giu GAA TTT Giu Phe CAA CAG Gin Gin 875 GAA TTA Glu Leu GAA GAT Glu Asp 880 GCT AAG AAT TA ACT Ala Lys Msn Leu Thx 845 AAA CTA ATA GAA TCT Lys Leu Ile Giu Ser GAC ATA TCA Asp Ile Ser 890 Giu
GAA
Glu ACT GAT Thr Asp TCT ATA Ser Ile Giu Gly Phe 895 TTT GTA GAA Phe Val Giu 910 AGA TT Arg Phe 900 AAA ACA Lys Thr AAT APLA GAA ACT Msn Lys Giu Thr 905 TTC TCT GAA TAT Phe Ser Giu Tyr Gly ACT GAA 2784 ?hr Giu AT? TCT 2832 lle, Ser 930 OTA AAA 2880 Val Lys 945 GCT GCA 2928 Ala Ala
ATA
Ile GCT AAT Ala Asfl GAT ACT Asp Thr His Ile Thr Glu Glu 925 GTA AAT GGT AAG TTA Val Asn Gly Lys Leu 940 06.
**as AAG ATA AAA GGT ACT ATA TT? Lys Ile Lys Gly Thr Ile Phe 935 AAA GTA AAT TTA GAT ACT ACA Lys Val Asn Leu Asp Thr Thr 950 TTT TTT ATA CAA TCA TTA ATA Phe. Phe Ie Gin Ser Leu Ile CAC GAA GTA AAT His Giu Val Msn ACT TTA AAT Thr Leu Aen 960 TCT AAA GAA Ser Lys Giu 975 ?CT CTT 2976 Ser Leu TTA TIT 3024 Leu Phe 965 AGT AAT TTA Ser Msn Leu GAA TAT Glu Tyr 970 AAA GTC Lys Val AGT GTA GCA ATG Ser Val Ala Met 985 TTA AAT ACT AT? Leu Asn Thr Ile 1000 CAA GT? TAC GCT CAA Gin Val Tyr Ala Gin 990 AAT AGT Asn Ser
GGT
Gly ACA GAT GCA GCC AAA GTT CT? Thr Asp Ala Ala Lys Val Val 1005 ACT ATA GAC TTA CT? CCT ACA Thr Ile Asp Leu Leu Pro Thr 1020 GAA TA GA TCA ACT GCA TA GAT GAA 3072 Giu Leu Val Ser Thr Ala Leu Asp Glu 1010 1015 -317 ±TIA TC-T GAA GGA TTA CCT ATA 3120 Leu Ser Glu Gly Leu Pro Ile 1025 1030~ TWA GGT GCA GCA ATC AAA GAG 3168 Leu Gly Ala Ala Ile Lys Glu 1045 ATT GCA ACT ATW ATA GAT GGT GTA AGT Ile Ala Thr Ile Ilie Asp Gly Val Ser 1035 1040 CTA AGT GAA ACG AGT GAC CCA TWA TWA Leu Ser Glu Thr Ser Asp Pro Leu Leu 1050 1055 ATA GGT ATA ATG GCA GTA AAT TWA ACA Ile Gly Ile Met Ala Val Asn Leu Thr 1065 1070 ACT TCA TCT TTG 000 ATA GCT AGT GGA Thr Ser Ser Leu Gly Ile Ala Ser Gly 1080 1085 AGA CAA GAA ATA GAA 3216 Arg Gin Glu Ile Oiu 1060 ACA GCT ACA ACT GCA 3264 Thr Ala Thr Thr Ala 1015 TTT AGT ATA CTT TWA 3312 Phe Ser Ile Leu Leu 1090 AGC TWA GTA AAC AAT 3360 Ser Leu Val Asn Asn 1105 GCT AAG Ala Lys ATC ATW Ile Ile GTT CCT Val Pro 1095 GAA CTW Glu Leu 1110
TA
Leu
GTA
Val GCA GGA ATW TCA OCA Ala Gly Ile Ser Ala 1100 CTT CGA GAT AAG GCA Leu Arg Asp Lys Ala 1115 GOT ATA CCA Gly Ile Pro 0.
.0.
0 ACA AAG Thr Lys
GTT
Val 1120 GTA GAT 3408 Val Asp ACT TWA 3456 Thr Leu TAT TW AAA Tyr Phe Lys 1125 TWA GAT GAT Leu Asp Asp 1140 CAT GTT TCA TWA GTT GAA ACT His Val Ser Leu Val Giu Thr 1130 AAA AWA ATG ATG CCA CAA GAT Lys Ile Met Met Pro Gin Asp 1145 AAT AAW AAT TCA ATA OTT TWA Asn Asn Asn Ser Ile Val Leu 1160 GOT GGT TCA 0GW CAT ACT GTA Oly Gly Ser Gly His Thr Val 1175 119' GCA CCA TCA ATA ACA TAT AGA OAA GGA GWA TT Glu Gly Val Phe 1135 OAT TWA OWO AWA Asp Leu Val Ile 1150 GOT AAA TOT GAA Gly Lys Cys Glu 1165 ACT OAT GAT AWA Thr Asp Asp Ile 0 GAG CCA CAC TWA 3504 Ser Oiu Ile Asp Phe 1155 ATC TOO AGA ATO GAA 3552 Ile Trp Arg Met Giu 1170 GAT CAC T-TC TTT WCA 3600 Asp His Phe Phe Ser 1185 Ala Pro 1190 Ser Ile Thr -Tyr Arg Giu Pro His 1195 Leu 1200 TCT ATA TAT GAC OWA TWG OAA GWA CAA AAA GAA GAA CTW OAT TWO WCA 3648 Ser Ile Tyr Asp Val Leu Glu Vai Gin Lys Giu Giu Leu Asp Let' Ser -1205 1210 1215 AAA OAT TWA AWO OWA TWA CCT AAW OCT CCA AAT AGA OTA TTW OCT TG 3696 Lys Asp Leu Met Val Let' Pro Asn Ala Pro Asn Arg Val Phe Ala Wrp 1220 1225 1230 OAA ACA OGA TOG ACA CCA GOT TWA AGA AGC TWA GAA AAT OAT GGC ACA 3744 Olt' Thr Gly Trp Whr Pro Gly Leu Arg Ser Let' Git' Asn Asp Oly Whr 1235 1240 1245 318 AAA CTG TTA GAC CGT ATA AGA GAT AAC TAT GAA GGT GAG TTT TAT TGG 3792 Lys Leu 1250 AGA TAT 3840 Arg Tyr 1265 AGA TAT 3888 Arg Tyr ACT TTT 3936 Ser Phe Leu Asp Arg Ile Arg Asp 1255 TTT GCT TTT ATA GCT CAT Phe Ala Phe Ile Ala Asp 1270 GAA GAT ACT AAT ATA AGA Giu Asp Thr Asn Ilie Arg 1285 ATA GTT CCA ATA ATA ACT Ile Val Pro Ile Ile Thr 1300 1260 GCT TTA ATA ACA ACA TTA AAA CCA Ala Leu Ile Thr Thr Leu Lys Pro 1275 1280 ATA AAT TTA Ile Asn Leu 1290 ACA GAA TAT Thr Giu Tyr 1305 CAT ACT AAT ACT AGA Asp Ser Asn Thr Arg 1295 ATA AGA CAA AAA TTA Ile Arg Giu Lys Leu 1310 GCA TI'G TCT CTT TCT Ala Leu Ser Leu Ser TCA TAT TCT TTC TAT CGT TCA GGA GGA ACT TAT 3984 Ser Tyr Ser Phe Tyr Cly Ser Gly Cly Thr Tyr 1315 1320 CAA TAT AAT ATC CGT ATA AAT ATA GAA TTA ACT 4032 Gin Tyr Asn Met Gly Ile Asn Ile Glu Leu Ser 1330 1335 ATT ATA GAT GTT CAT AAT CTT CTG AGA CAT CTA 4080 Ile Ile Asp Val Asp Asn Vai Val Arg Asp Val 1345 1350 135! AAA ATT AAA AAA GGT CAT TTA ATA GAA GGT ATT 4128 Lys Ilie Lys Lys Gly Asp LeuIle Giu Cly Ile 1365 1370 1325 GAA ACT Ciu Ser 1340 ACT ATA CAT GTT TGG Asp Vai Trp GAA TCT CAT Thr
TTA
Leu 1360 TCT ACA CTA ACT Ser Thr Leu Ser 1375 ATT GAA GAG AAT AAA ATT ATC TTA MAT AGC CAT GAG ATT AAT Tfl TCT 4176 Ile Ciu Ciu Asn Lys Ile Ile Leu Asn Ser His Giu Ile Asn Phe Ser 1380 1385 1390 CGT GAG GTA AAT GCA ACT AAT GGA ITT 4224 Gly Ciu Val Asn Gly Ser Asn Gly Phe 1395 1400 TTA CAA CCA ATA MAT GCA ATT ATA GMA 4272 Leu Glu Cly Ile Asn Ala Ile Ile Giu 1410 1415 TAT AMA TTA CTT ATT TCT CCC GAA TTA 4320 Tyr Lys Leu Leu Ile Ser Cly Clu Leu 1425 1430 MAT CAT ATT CAA CAG AMA ATA CAT TAT 4368 Asn His Ile Gin Gin Lys Ile Asp Tyr 1445 GTT TCT TTA ACA TTT TCA ATT Val Ser Leu Thr Phe Ser Ile 1405 GTT CAT TTA TTA TCT AMA TCA Val Asp Leu Leu Ser Lys Ser 1420 AAA ATA TTC ATC TTA MAT TCA Lys Ile Leu Met Leu Asn Ser 1435 1440 ATA GGA TTC MAT AGC GMA TTA Ile Cly Phe Asn Ser Glu Leu 1450 1455 CAG AMA MT 4416 Gin Lys Asn ATA CCA TAT AGC TT Ile Pro Tyr Ser Phe 1460 GTA GAT ACT GMA GCA AMA GAG MAT Val Asp Ser Giu Cly Lys Giu Asfl 1465 1470 3119- GOT TT-4 ATT AAT GOT TCA ACA AAA GAA GGT TTA TTT GTA TCT GAA TTA 4464 Gly Phe Ile Asn Gly Ser Thr Lys Giu Gly Leu Phe Val Ser Glu Leu 1475 1480 1485 CCT GAT GTA OTT CTT ATA AGT AAG OTT TAT ATG GAT OAT ACT AAG CCT 4512 Pro Asp Val Val 1490 TCA TTT GGA TAT 4560 Ser Phe Gly Tyr 1505 AAA GAT AAT GTT 4608 Lys Asp Asn Val AAA ATC TCT CTT 4656 Lys Ile Ser Leu 1540 AAT AGT GTG CAT 4704 Asn Ser Val His 1555 ATG AAT AGA AAA 4752 Met Asn Arg Lys 1570 TTA GAA AGT ATG 4800 Leu Giu Ser Met 1585 1495 FAT AGT AAT Tyr Ser Asn 1510 AAT ATA TTA As n Ile Leu 1525 AAT TTG AAA OAT Asn Leu Lys Asp 1515 ACA GOT TAT TAT Thr Oly Tyr Tyr 1530 CTA CAA GAT OAA Leu Gin Asp Oiu 1545 ACT GGA GTA OCT Ser Giy Val Ala .500 Ser Lys Pro
TCT
Ser TTO ACT Leu Thr TC AAA OTT ATA ACT fai Lys Val Ile Thr 1520 CTT AAG OAT GAT ATA Leu Lys Asp Asp Ile 1535 A.AA ACT ATA AAO TTA Lys Thr Ile Lys Leu 1550 GAG ATT TTG AAG TTC Giu Ile Leu Lys Phe 1565 TCT TTA ATO AGC TTT Ser Leu Met Ser Phe 1580 AAT TTC TTA CAA TCT Asn Phe Leu Gin Ser TTA GAT GAA Leu Asp Glu GOT AAT ACA AAT Gly Asn Thr Asn 1.575 AAT ATA AAA AGT Asn Ile Lys Ser 1.590 A.CT TCA OAT rhr Ser Asp ATT TTC Ile Phe
OTT
Val 1595 AAT ATT AAO TTT ATA TTA OAT OCT AAT TT ATA.
4848 Asn Ile Lys Phe Ile Leu Asp Ala Asn Phe Ile 1605 1610 TCT ATT GOC CAA TTT GAG TIT ATT TOT OAT GAA ATA AGT OGT ACT ACT Ile Ser Gly Thr Thr 1615 AAT OAT AAT ATA CAA Asn Asp Asn Ile Gin 1630 AAT TAT ACT TTA TAT Asn Tyr Thr Leu Tyr 1645 4896 Giu Ser le.Oly Gin Phe 1620 CCA TAT TTC ATT AAO 4944 Pro Tyr Phe Ile LYS 1635 OTA OOA AAT AGA CAA 4992 Val Oly Asfl Arg Gin 1650 OAT TCT GGA OAT ATA 5040 Asp Ser Oly Asp Ilie 1665 CTT TAT OGA ATA GAC
TTF
Phe Phe Ile A.AT ACA Asn Thr 1640 1625 CTA OAA ACT Leu Oiu Thr AAT ATG'ATA OTO GAA CCA AAT TAT OAT TTA GAT Asfl Met Ile Val Oiu Pro Asn Tyr Asp Leu Asp 1655 1660 TCT TCA ACT OTT ATC AAT TTC TCT CAA AAO TAT Ser Ser Thr Val Ile Asn Phe Ser Gin Lys Tyr 1670 1675 1680 AGT TOT GTT AAT AAA OTT OTA ATT TCA CCA AAT 5088 Leu Tyr Oiy Ile Asp Ser Cys Val Asn Lys Val Vai Ile Ser Pro Asn 1685 1690 1695 320 AITT TAT ACA GAT GAA ATA AAT ATA ACG CCT GTA TAT GAA ACA AAT AAT 5136 Ile Tyr Thr Asp Giu Ile Asn Ile Thr Pro Val Tyr Giu Thr Asn Asn 1700 1105 1710 ACT TAT CCA GAA GTT ATT GTA TTA GAT GCA AAT TAT ATA AAT GAA AAA 5184 Thr Tyr Pro Glu Val lie Val Leu Asp Ala Asn Tyr Ile Asn Glu Lys 1715 1720 1725 ATA AAT 4 5232 Ile Asn 1730 GAT GGT 5280 Asp Gly 1745 TCA CAA 5328 Ser Gin GCA AAT 5376 Ala Asn AGT GAA 5424 Ser Glu 3TT AAT ATC AAT GAT CTA Vai Asn Ile Asn Asp Leu 1735 AAT GAT TTT ATT CTT ATG Asn Asp Phe Ile Leu Met 1750 GTT AAA ATA AGA TTC GTT Val Lys Ile Arg Phe Vai 1765 AAG CTA TCT TTT AAC TTT Lys Leu Ser Phe Asn Phe 1780 TCT ATA Ser Ile TCA ACT Ser Thr AAT OTT Asn Vai 177( CGA TAT GTA TOG AGT AAT Arg Tyr Vai Trp Ser Asn 1740 AGT GAA GAA AAT AAG GTO Ser Giu Giu Asn Lys Val 1755 1760 TTT AAA OAT AAO ACT TTG Phe Lys Asp Lys Thr Leu 0 ATA ATC TTA TCA Ile Ile Leu Ser 1795 TTT ACA Phe Thr 180 AGT OAT AAA Ser Asp Lys 1785 CCT TCA TAT Pro Ser Tyr 0 1775 CAA OAT OTA COT OTA Gin Asp Val Pro Val 1790 TAT GAG GAT OGA TTO Tyr Giu Asp Gly Leu 1805 ATT GOC TAT OAT TTG GOT CTA GTT TCT TTA TAT AAT GAG AAA TTT TAT 5472 Ile Gly Tyr Asp Leu Giy Leu Val Ser Leu Tyr Asn Glu Lys Phe Tyr 1810 1815 1820 ATT AAT AAC TTT GGA ATO ATG GTA TOT GGA TTA ATA TAT ATT AAT GAT 5520 Ile Asn Asn Phe Gly Met Met Val Ser Gly Leu Ile Tyr le Asn Asp 1825 1830 1835 184 TCA TTA TAT TAT TTT AAA CCA CCA GTA AAT AAT TTO ATA ACT GGA TTT 5568 Ser Les Tyr Tyr Phe Lys Pro Pro Val Asn Asn Leu Ile Thr Gly Phe 1845 1850 1855 OTO ACT GTA GGC OAT OAT AAA TAC TAC TTT AAT CCA ATT AAT GOT OGA 5616 Val Thr Val Oly Asp Asp Lys Tyr Tyr Phe Asn Pro Ile Asn Gly Gly 1860 1865 1870 GCT GCT TCA ATT GGA GAG ACA ATA ATT GAT GAC AAA AAT TAT TAT TTC 5664 Ala Ala Ser Ile Gly Glu Thr Ile Ile Asp Asp Lys Asn Tyr Tyr Phe .875 1880 1885 AAC CAA AGT OGA GTG TA CAA ACA GGT GTA TTT AOT ACA GAA OAT GGA 5712 Asf Gln Ser Gly Val Leu Gin Thr Gly Val Phe Ser Thr Giu Asp Gl) 1890 1895 1900 TTT AAA TAT TTT 0CC CCA GCT AAT ACA CTT GAT OAA AAC OTA OAA GO) 5760 Phe Lys Tyr Phe Ala Pro Ala Asn Thr Leu Asp Glu Asn Leu Giu Gil 1905 1910 1915 19: 0 2( 321 GAA GCA ATT GAT TTT ACT GGA AAA TTA ATT ATT GAC GAA AAT ATT TAT 5808 Glu Ala Ile Asp Phe Thr Gly Lys Leu Ile Ile Asp Giu Asn Ile Tyr 1925 1930 1935 TAT TTT GAT GAT AAT TAT AGA GGA GCT GTA GAA TOO AAA GAA TTA GAT 5856 Tyr Phe Asp Asp Asn Tyr Arg Gly Ala Val Giu Ti-p Lys Giu Leu Asp 1940 1945 1950 GOT GAA ATG CAC TAT TTT AGC CCA GAA ACA GOT AAA GCT TTT AAA GOT 5904 Oly Giu Met His Tyr Phe Ser Pro Glu Thr Gly Lys Ala Phe Lys Gly 195.5 cTA AAT CAA 5952 Leu Asn Gin 1970 ATG CAA. AAA 6000 P.TA GGT GAT TAT AAA TAC TAT Ile Gly Asp Tyr Lys Tyr Tyr 1975 GGA TTT GTT AGT ATA AAT GAT TTC AAT Phe Asri 198( AAT AAA Met Gin Lys Gly Phe Val Ser 1985 1990 GAT TCT GGT GTT ATG AAA GTA 6048 Asp Ser Oly Vai Met Lys Val 2005 TTC TAC TTT GCT GAP. AAC GGA 6096 Phe Tyr Phe Ala Giu Asn Gly 2020 GAP. GAT GGA TTT AA.A TAT TTT 6144 Giu Asp Gly Phe Lys Tyr Phe 2035 GAP. GAP. GGT GAA GAA ATC TCA Ile Asn Asp Asn Lys 1995 GGT TAC ACT GAP. ATA Gly Tyr Th- Gli Ile 2010 GAA ATG CAP. ATA GGA TCT GAT GOP. GTT Ser Asp Gly Val CAC TAT TTT OAT His Tyr Phe Asp 2000 GAT GGC AP.G CAT Asp Gly Lys His 2015 GTA TTT AAT ACA Val Phe Asn Thi- 2030 GAT TTA GGA AP.T Asp Leu Gly Asn 2045 2025 OCT CAT CAT Ala His His 2040 TAT TCT GGT Ile Gly A.T GAP.
Asn Giu 6192 Giu Glu Gly Oiu Oiu Ile Ser Tyr 'Ser Gly 2050 2055 AAA ATT TAC TAT TTT OAT OAT TCA TTT ACA 6240 Lys Ile Tyr Tyr Phe Asp Asp Ser Phe Thi- 2065 2070 OAT TTA GAG OAT GOT TCA AP.G TAT TAT TTT ATA TTA AP.T TTC A.AT AP.T Ile Leu Asn Phe Asn Asn 2060 OCT GTA OTT GOP. TOG AAA Ala Val Val Oly Ti-p Lys 2075 2080 OAT GAP. OAT ACA OCA GA.
Asp Glu Asp Thr Ala Giu 0 2095 GOT CA. TAT TAT TI? MAT Oly Gin Tyr Tyr Phe Asn 2110 ACT ATA AP.T OAT AAAP GTC Thr Ile Asn Asp Lys Val 6288 Asp Leic- Giu Asp Oly 2085 OCA TAT ATA GOT TTO 6336 Ala Tyr Ile Gly Leu 2100 OAT OAT GOP. ATT ATO 6384 Asp Asp Oly Ile Met 2115 TTC TAC TTC TCT GAC Ser Lys Tyr Tyr TCA TI'A ATA AAT Ser Leu Ile Asn 210 209'
OAT
Asp 5 CAP. OTT GOP. TTT OTC Gin Val Gly Phe Val 2120 TCT GGA ATI' ATA OAA TCT GOP. OTA CAP. AAC ATA 6432 Phe Tyr Phe Ser Asp Ser Gly Ile Ile Oiu Ser Gly Val Gin Asn Ile 2130 2135 2140 322 GAT GAC AAT TAT TTC TAT ATA GAT GAT AAT GGT ATA GTT CAA ATT GGT 6480 Asp Asp Asn Tyr Phe Tyr Ile Asp Asp Asn Giy Ile Val Gin Ile Gly 2145 2150 2155 2160 GTA TTT GAT ACT TCA GAT GCA TAT AAA TAT TTT GCA CCT GCT AAT ACT 6528 Vai Phe Asp Thr Ser Asp Giy Tyr Lys Tyr Phe Ala Pro Ala Asn Thr 2165 2170 2175 GTA AAT GAT AAT ATT TAC GGA CAA GCA GTT GAA TAT AGT GGT TTA GTT 6576 Val Asn Asp Asn Ile Tyr Giy Gin Ala Val Glu Tyr Ser Cly Leu Vai 2180 2185 2190 AGA GTT GGG GAA CAT GTA TAT TAT TTT GGA GAA ACA TAT ACA ATI' GAG 6624 Arg Val Gly Ciii Asp Val Tyr Tyr Phe Giy Ciu Thr Tyr Thr Ile Clu 2195 2200 2205 ACT CGA TGC ATA TAT CAT ATG GMA AAT GAA ACT CAT AAA TAT TAT TTC 6672 Thr Cly Trp Ile Tyr Asp Met Giu Asn Giu Ser Asp Lys Tyr Tyr Phe 2210 2215 2220 MAT CCA GMA ACT AMA AAA GCA TGC AAA CGT 6720 Asn Pro Giu Thr Lys Lys Ala Cys Lys ClyI 2225 22302 ATA AAA TAT TAT TTT CAT GAG AAG GGC ATA t6768 Ile Lys Tyr Tyr Phe Asp Giu Lys Gly Ile 2245 2250 TCA TTT GMA MT MAT MAT TAT TAC TTT MAT 6816 Ser Phe Glu As n Asn Asn Tyr Tyr Phe Asn 2260 2265 TTT GGT TAT ATA MAT ATA CMA CAT AAG ATG 6864 Phe Cly Tyr Ile Asn Ile Giu Asp' Lys Met 2275 2280 CCT CTC ATC CAG ATT GCA GTA TTT AAT ACA 6912 Gly Val Met Gin Ile Gly Val Phe Asn Thr 2290 2295 TTT GCA CAT CMA MT ACT TTG CAT GAG MAT 6960 Phe Ala His Gin Asn Thr Leu Asp GJlu Asn 2305 2310 =T MAT TTA lie Asn Leu ~235 Ile Asp Asp 2240 'let Arg Thr Gly Leu Ile 2255 GAG MAT CGT GMA ATG CMA Giu Asn Gly Giu Met Gin 2270 TTC TAT TTT CGT CMA CAT Phe Tyr Phe Cly Glu Asp 2285 CC.A CAT GGA TTT AMA TAC Pro Asp Gly Phe Lys Tyr 2300 TTT GAG GGA CMA TCA ATA Phe Clu Gly Clu ger Ile 2315 232 0 MAC TAT 7008 Asn Tyr CAT GMA 7056 Asp Ciu TAT TAT 7098 Tyr Tyr ACT GGT TGG TTA CAT Thr Gly Trp Leu Asp 2325 TAT ATT CCA GCA ACT Tyr Ile Ala Ala Thr 2340 TTT- CAT CCT CAT ACA Phe Asp Pro Asp Thr 2355 TTA GAT GMA MG AGA TAT TAT TTT ACA LeU Asp Clii Lys Arg Tyr Tyr Phe Thr 2330 2335 GGT TCA CTT ATT ATT CAT GCT GAG GAG Gly Set Val Ile Ile Asp Gly Giu Ciu 2345 2350 CCT CMA TTA GTG ATT ACT GMA Ala Gin Leu Val Ile Ser Giu 2360 2365 323
TAG
7101 INFORMATION FOR SEQ, ID NO:1O: SEQUENCE CHARACTERISTICS: LENGTH: 2366 amino acids TYPE: amino acid TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ, ID 140:10: Met Ser Leu Val Asn Arg Lys Gin Leu Glu LYS Met 1 5 10 Phe Arg Thr Gin Giu Asp Giu Tyr Val Ala Ile Leu .20 25 Glu Tvr His Asn Met Ser Giu Asn Thr Val Val Giu Asn Val Arg 15 Ala Leu Giu Tyr Leu Lys Leu Lys 65 Thr Asn Asn Val Val1 145 Leu Giu Lys Ser Glu Leu Tyr Phe 130 Val1 Asn Ile Gly Val His Ile 115 Tyr Glu
ASP
Ile Arg Leu Phe 100 Asn Asp Se r Pro Tyr I80 Asn Giu 85 Val Gin Ser Al a Arg 165 Asp Lys 70 Leu Trp, TrP Asn Ile 150 Phe Lys Ila LeuI .sys Asn lie Gly Lys Asp 120 kla Phe 135 hsn Asp Asp Tyr Gin Lys Lys As n Gin Asn Ile Leu Lys 170 Phe Phe Leu Ile Ser As n Giu 155 Phe Ile Lys Thr Asn
ASP
Thr 140 Ser Phe Asn Glu Pro Asp Tyr 125 Leu Phe Arg Tyr Tyr Val Thr 110 Asn Lys Arg Lys Tyr 190 Asp Ilie Asn Ser Leu Thr Asp Ile Tyr Ilie Asp Thr Tyr Lys Gin Arg Giu Giu Asn Pro Glu Leu 195 200 Ilie Ile Asp Asp Ile Val Lys Thr 205 Tyr Leu Ser 210 Ile Giu Giu 225 Arg Asfl Phe Gin Giu Leu Arg Ile Ser 275 Met Leu Pro 290 Asn Giu Ser Leu Glu G1u 245 Val G1u 260 Ala Leu Gly Ile Tyr Ser 215 Asn Lys 230 Phe Lys Arg TrP Lys Giu Gin Pro 295 Lys Ile Asn Asn Ile 280 Asp Glu Ile Asp Giu 220 Thr Gin Asn Ser 235 Gly Giu Ser Phe 250 Leu Ala Ala Ala 265 Gly Gly Met Tyr Leu Phe Giu Ser 300 Leu Gly Asn Ser Leu 285 Ile Asn Thr Tyr Asn Asp Val 240 Leu Tyr G1u 255 Asp Ile Leu 270 Asp Val ASP Giu Lys Pro 324 Ser Ser Val Thr Val Asp Pk 305 310 Met Lys Tyr Lys Giu Tyr I 325 Met Leu Asp Giu Giu Val G 340 Lys Ser Asp Lys Ser Glu I.
355 Ser Pro Leu Giu Val Lys I 370 3 Gin Gly Leu Ile Ser Val L 385 390 Lys Gin Ile Giu Asn Arg T 405 Ala Ile Ser Giu Asp Asn A 420 Asp Ser Ile Met Ala Giu A 435 Glu Leu Gly Lys Tyr Leu A 450 4 Thr Ile Asn Leu Ser Gly I 465 470 Leu Leu Met Phe Lys Glu 485 Asp Leu Arg Asn Phe Glu 500 Giu Gin Giu Met Ala Ser 515 Ala Gin Phe Giu Giu Tyr 530 Glu Asp Asp Asn Leu Asp 545 550 Tyr Leu Leu Giu Lys Ile 565 Tyr Ile His Tyr Ile Vai 580 Ala Ala Cys Asn Leu Phe 595 Gin Lys Asn Ile Giu Asp 610 Asp Gly Giu Ile Gin Glu 625 630 Ser Asp Arg Pro Lys Ile 645 ie TI Le P1 In Si le PI 3 le A 75 ys A yr L sp P ia A 4 rg 155 ,ro C 31y lie Leu Lys 535 Phe Ser Gin Ala Ser 615 Ile Lys rp ro er he 60 la sp ys 'he 1sn 40 ral flu Ser Ser TrT.
52C ArS Sei Se: Le Ly 60 Gi As Le Glu Met Thr Lys Li 315 Glu Tyr Thr Ser G 330 Ser Phe Giu Ser V 345 Ser Ser Leu Gly A 3 Phe Asn Ser Lys G 380 Ser Tyr CYs Ser A 395 Ile Leu Asn Asn S 410 Asn Thr Thr Thr A 425 Ala Asp Asn Gly 4 Gly Phe Phe Pro 460 Ala Tyr Ala Aia 475 Met Asn Ile His I 490 Lys Thr Asn Ile 505 Ser Phe Asp Asp Asn Tyr Phe Glu 540 r Gin Asn Ile Val 555 r Leu Ala Arg Ser 570 u Gin Gly Asp Lys 585 s Thr Pro Tyr Asp 0 u Ile Ala Tyr Tyr 620 ;p Lys T'yr Lys Ile 635 :u Thr Phe Ile Gly 650 eu Giu Ala Ile 320 lu His Phe Asp 335 al Leu Ala Ser 350 sp Met Giu Ala ly Ile Ile Asn sn Leu Ile Val 400 er Leu Asn Pro 415 sn Thr Phe lie 430 .rg Phe Met Met sp Val Lys Thr la Tyr Gin Asp 480 Leu Ile Giu Ala 495 Ser Gin Ser Thr 510 Ala Arg Ala Lys 525 Gly Ser Leu Gly Val Asp Lys Glu 560 Ser Giu Arg Gly 575 Ile Ser Tyr Glu 590 Ser Val Leu Phe 605 Tyr Asn Pro Gly Pro Ser Ile Ile 640 His Gly Lys Asp 655 325 Glu P1 Thr G Lys S 6 Ile A 705 Asp L Val S Glu L lie 1 7 Asn I 785 Leu( Glu] Asp
S.
S e r.
Sex Ile 865 Ser .Ser Thr Ile Val 945 Ala Ser he As iu 11 67 er I 90 Sn V; ys Ii er A ,eu L 7 ,ys A r70 .vs I 3In G Lys V Thr C 8 Asp 850 Ser His Ile Glu Ser 930 Lys Ala Leu n5 Le 55 ie al Le la eu 55 sP le r 91% 1 Ph Ar Ly 91
L)
Sc Thr As 660 Glu Al Glu IJ Glu G Ser G 7 Asn G 740 Asp H Ile S Thr V Ile A 8 L Met 1 820 le r Ile p Ala e Ile g Phe 900 's Thr .5 rs Ile (s Val he Phe er As 980
PP
aa le Lu lu 25 in is er 'ai Lrg 105 ,eu lal ksr Let Se: 88 Iie Ii Ly As 11 96 Le Ile Ph Ala Il Asn L( 65 Thr T' 710 Leu M Tyr G Ser G Ser L 7 Lys S 790 Asn A Thr G Glu C Tyr I Cys 870 r Phe 5 e Asn e Phe s Gly ,n Leu 950 .e Gin 5e :u Ser ee ee 95 et lu ly ys 75 er isn lu lu le 855 AsI Gl Ly Se Th 93 As Se Va Ala Giy Phe Asp Vai 665 Asp Leu Aia Lys Glu 680 Leu Gly Cys Asn Met 700 Pro Gly Lys Leu Leu 715 Pro Ser Ile Ser Gin 730 Val Arg Ile Asn Ser 745 Glu Trp Ile Asn Lys 760 Glu Tyr Ile Ser Phe 780 Lys Asn Leu Pro Glu 795 Ser Asn Ser Ser Asp 810 Cys Giu Ile Asn Vai 825 Arg Ile Giu Giu Ala 840 Lys Asp Giu Phe Lys 860 Leu Lys Gin Gin Asr 875 Asp Ile Ser Giu Th2 890 s Giu Thr Gly GIu Sei 905 Giu Tyr Ala Asn Hi: 920 r Ile Phe Asp Thr Va 5 94 p Thr Thr His Giu Va 955 r Leu Ile Giu Tyr As 970 i1 Aia Met Lys Vai GI 985 Asp Ser Leu Ser 670 Asp Ile Ser Pro 685 Phe Ser Tyr Ser Leu Lys Vai Lys 720 Asp Ser Ile Ile 735 Glu Gly Arg Arg 750 Glu Giu Ser Ile 765 Asn Pro Lys Giu Leu Ser Thr Leu 800 Ile Giu Leu Glu 815 Ile Ser Asn Ile 830 Lys Asn Leu Thr 845 Leu Ile Giu Ser Glu Leu Giu Asp 880 Asp Giu Giy Phe 895 r Ile Phe Vai Glu 910 s Ile Thr Giu Glu 925 I Asn Gly Lys Leu 0 1 Asn Thr Leu As 960 n Ser Ser Lys Glu 975 n Val Tyr Aia Gin 990 326- Leu Phe Ser Thr Gly Leu Asn Thr Ile Thr Asp Ala Ala Lys Val Val 995 1000 1005 Glu Leu Val Ser Thr Ala Leu Asp Glu Thr Ile Asp Leu Leu Pro Thr 1010 1015 1020 Leu Ser Glu Gly Leu Pro Ile Ile Ala Thr Ile Ile Asp Gly Val Ser 1025 1030 1035 1040 Leu Gly Ala Ala Ile Lys Glu Leu Ser Glu Thr Ser Asp Pro Leu Leu 1045 1050 1055 Arg Gin Glu Ile Glu Ala Lys Ile Gly Ile Met Ala Val Asn Leu Thr 1060 1065 1070 Thr Ala Thr Thr Ala Ile Ile Thr Ser Ser Leu Gly Ile Ala Ser Gly 1075 1080 1085 Phe Ser Ile Leu Leu Val Pro Leu Ala Gly Ile Ser Ala Gly Ile Pro 1090 1095 1100 Ser Leu Val Asn Asn Glu Leu Val Leu Arg Asp Lys Ala Thr Lys Val 1105 1110 1115 1120 Val Asp Tyr Phe Lys His Val Ser Leu Val Glu Thr Glu Gly Val Phe .o 1125 1130 1135 Thr Leu Leu Asp Asp Lys Ile Met Met Pro Gin Asp Asp Leu Val Ile 1140 1145 1150 Ser Glu Ile Asp Phe Asn Asn Asn Ser Ile Val Leu Gly Lys Cys Glu 1155 1160 1165 S.Ile Trp Arg Met Glu Gly Gly Ser Gly His Thr Val Thr Asp Asp Ile S1170 1175 1180 Asp His Phe Phe Ser Ala Pro Ser Ile Thr Tyr Arg Glu Pro His Leu 1185 1190 1195 1200 Ser Ile Tyr Asp Val Leu Glu Val Gin Lys Glu Glu Leu Asp Leu Ser 1205 1210 1215 Lys Asp Leu Met Val Leu Pro AsnAla Pro Asn Arg Val Phe Ala Trp 1220 1225 1230 s Glu Thr Gly Trp Thr Pro Gly Leu Arg Ser Leu Glu Asn Asp Gly Thr 1235 1240 1245 Lys Leu Leu Asp Arg Ile Arg Asp Asn Tyr Glu Gly Glu Phe Tyr Trp 1250 1255 1260 Arg Tyr Phe Ala Phe Ile Ala Asp Ala Leu Ile Thr Thr Leu Lys Pro 1265 1270 1275 1280 Arg Tyr Glu Asp Thr Asn Ile Arg Ile Asn Leu Asp Ser Asn Thr Arg 1285 1290 1295 Ser Phe Ile Val Pro Ile Ile Thr Thr Glu Tyr Ile Arg Glu Lys Leu 1300 1305 1310 Ser Tyr Ser Phe Tyr Gly Ser Gly Gly Thr Tyr Ala Leu Ser Leu Ser 1315 1320 1325 327 Gin Tyr Asn met Gly Ile Asn Ile Glu Leu Ser Glu Ser Asp Val Trp 1330 1335 1340 Ile Ile Asp Val Asp Asn Val Val Arg Asp Val Thr Ile Giu Ser Asp 1345 1350 1355 1360 Lys Ile Lys Lys Gly Asp Leu Ile Giu Gly Ile Leu Ser Thr Leu Ser 1365 1370 1375 ile Giu Glu Asn Lys Ile Ilie Leu Asri Ser His Giu Ile Asn Phe Ser 1380 1365 1390 Giy Glu Val Asn Giy Ser Asn Giy Phe Val Ser Leu Thr Phe Ser Ile .1395 1400 1405 Leu Giu Giy Ile Asn Ala Ilie Ile Giu Val Asp Leu Leu Ser Lys Ser 1410 1415 1420 Tyr Lys Leu Leu Ile Ser Giy Giu Leu Lys Ile Leu Met Leu Asn Ser 1425 1430 1435 1440 Asn His Ile Gin Gin Lys Ilie Asp Tyr Ile Giy Phe Asn Ser Giu Leu .v1445 1450 1455 Gin Lys Asn Ilie Pro Tyr Ser Phe Val Asp Ser Giu Gly Lys Giu Asn **1460 1465 1470 Phe Ile Asn Gly Ser Thr Lys Glu Gly Leu Phe Vai Ser Giu Leu 1475 1480 1485 Pro Asp Vai Vai Leu Ile Ser Lys Val Tyr Met Asp Asp Ser Lys Pro *1490 1495 1500 *Ser Phe Gly Tyr Tyr Ser Asn Asn Leu Lys Asp Val Lys Val Ile Thr 1505 1510 1515 1520 LYE Asp Asn Val Asn Ile Leu Thr Gly Tyr Tyr Leu Lys Asp Asp Ile *1525 1530 1535 Lys Ile Ser Leu Ser Leu Thr Leu Gin Asp Giu Lys Thr Ile Lys Leu .1540 1545 1550 Asn Ser Val His Leu Asp Giu Ser Gly Val Ala Glu Ile Leu Lys Phe *.:1555 1560 1565 Met Asn Arg Lys Giy ASn Thr Asn Thr Ser A-sp Ser Leu Met Ser Phe Ooo*1570 1575 1580 Leu Giu Ser Met Asn Ile Lys Ser Ilie Phe Val Asn Phe Leu Gin Ser 1585 1590 1595 1600 Asn Ile Lys Phe Ile Leu Asp Aia Asn Phe Ile Ile Ser Giy Thr Thr 1605 1610 -~1615 Ser Ile Gly Gin Phe Giu Phe Ile Cys Asp Giu Asn Asp Asn Ile Gin 1620 1625 1630 Pro Tyr Phe Ile Lys Phe Asn Thr Leu Giu Thr Asn Tyr Thr Leu Tyr 1635 1640 1645 Val Giy Asn Arg Gin Asfl Met Ile Val Giu Pro Asn Tyr Asp Leu Asp 1650 1655 1660 328 Asp Ser Gly Asp Ile Ser Ser Thr Val Ile Asn Phe Ser Gin Lys Tyr 1665 1670 1675 1680 Leu Tyr Gly Ile Asp Ser Cys Val Asn Lys Val Val Ile Ser Pro Asn 1685 1690 1695 Ile Tyr Thr Asp Glu Ile Asn Ile Thr Pro Val Tyr Glu Thr Asn Asn 1700 1705 1710 Thr Tyr Pro Glu Val Ile Val Leu Asp Ala Asn Tyr Ile Asn Glu Lys 1715 1720 1725 Ile Asn Val Asn Ile Asn Asp Leu Ser Ile Arg Tyr Val Trp Ser Asn 1730 1735 1740 Asp Gly Asn Asp Phe Ile Leu Met Ser Thr Ser Glu Glu Asn Lys Val 1745 1750 1755 1760 Ser Gin Val Lys Ile Arg Phe Val Asn Val Phe Lys Asp Lys Thr Leu S1765 1770 1775 Ala Asn Lys Leu Ser Phe Asn Phe Ser Asp Lys Gin Asp Val Pro Val o* 1780 1785 1790 Ser Glu Ile Ile Leu Ser Phe Thr Pro Ser Tyr Tyr Glu Asp Gly Leu 1795 1800 1805 Ile Gly Tyr Asp Leu Gly Leu Val Ser Leu Tyr Asn Glu Lys Phe Tyr S.1810 1815 1820 Ile Asn Asn Phe Gly Met Met Val Ser Gly Leu Ile Tyr Ile Asn Asp 1825 1830 1835 1840 Ser Leu Tyr Tyr Phe Lys Pro Pro Val Asn Asn Leu Ile Thr Gly Phe 1845 1850 1855 Val Thr Val Gly Asp Asp Lys Tyr Tyr Phe Asn Pro Ile Asn Gly Gly 1860 1865 1870 S" Ala Ala Ser Ile Gly Glu Thr Ile Ile Asp Asp Lys Asn Tyr Tyr Phe 1875 1880 1885 Asn Gin Ser Gly Val Leu Gin Thr Gly Val Phe Ser Thr Glu Asp Gly 1890 1895 1900 Phe Lys Tyr Phe Ala Pro Ala Asn Thr Leu Asp Glu Asn Leu Glu Gly 1905 1910 1915 1920 Glu Ala Ile Asp Phe Thr Gly Lys Leu Ile Ile Asp Glu Asn Ile Tyr 1925 1930 1935 Tyr Phe Asp Asp Asn Tyr Arg Gly Ala Val Glu Trp Lys Glu Leu Asp 1940 1945 1950 Gly Glu Met His Tyr Phe Ser Pro Glu Thr Gly Lys Ala Phe Lys Gly 1955 1960 1965 Leu Asn Gin Ile Gly Asp Tyr Lys Tyr Tyr Phe Asn Ser Asp Gly Val -1970 1975 1980 Met Gin Lys Gly Phe Val Ser Ile Asn Asp Asn Lys His Tyr Phe Asp 1985 1990 1995 2000 329 Asp Ser Gly Val Met Lys Val Gly Tyr Thr Giu Ile Asp Gly Lys His 2005 2010 2015 Phe Tyr Phe Ala Glu Asn Gly Giu Met Gin Ile Gly Val Phe Asn Thr 2020 2025 2030 Giu Asp Gly Phe Lys Tyr Phe Ala His His Asn Giu Asp Leu Gly Asn 2035 2040 2045 Glu Giu Gly Glu Glu Ile Ser Tyr Ser Gly Ile Leu Asn Phe Asn Asn 2050 2055 2060 Lys Ile Tyr Tyr Phe Asp Asp Ser Phe Thr Ala Val Val Gly Trp Lys 2065 2070 2075 2080 Asp Leu Giu Asp Gly Ser Lys Tyr Tyr Phe Asp Giu Asp Thr Ala Giu 2085 2090 2095 Ala Tyr Ile Gly Leu Ser Leu Ile Asn Asp Gly Gin Tyr Tyr Phe Asn 2100 2105 2110 Asp Asp Giy Ile Met Gin Val Gly Phe Val Thr Ile Asn Asp Lys Val 2115 2120 2125 Phe Tyr Phe Ser Asp Ser Gly Ile Ilie Giu Ser Gly Val Gin Asn Ile *2130 2135 2140 Asp Asp Asn Tyr Phe Tyr Ilie Asp Asp Asn Gly Ile Val Gin Ile Gly 2145 2150 2155 2160 Val Phe Asp Thr Ser Asp Gly Tyr Lys Tyr Phe Ala Pro Ala Asn Thr *2165 2170 2175 *Val Asn Asp Asn Ile Tyr Giy Gin Ala Val Giu Tyr Ser Giy Leu Val .*.2180 2185 2190 Arg Val Giy Giu Asp Val Tyr Tyr Phe Gly Giu Thr Tyr Thr Ile Giu ***2195 2200 2205 Thr Gly Trp Ile Tyr Asp Met Glu Asn Glu Ser Asp Lys Tyr Tyr Phe 2210 2215 2220 Asn Pro Giu Thr Lys Lys Ala Cys- Lys Gly Ile Asn Leu Ilie Asp Asp *.*2225 2230 2235 2240 Ile Lys Tyr Tyr Phe Asp Glu Lys Gly Ile Met Arg Thr Giy Leu Ile 2245 2250 2255 Ser Phe Giu Asn Asn Asn Tyr Tyr Phe Asn Glu Asn Gly G1u Met Gin 2260 2265 2270 Phe Gly Tyr Ile Asl Ile Glu Asp Lys Met Phe Tyr Phe Gly Giu Asp 2275 2280 2285 Gly Val Met Gin Ile Gly Vai Phe Asn Thr Pro Asp Gly Phe Lys Tyr, 2290 2295 2300 Phe Ala His Gin Asn Thr Leu Asp Giu Asn Phe Giu Gly Giu Ser Ile 2305 2310 2315 2320 Asn Tyr Thr Gly Trp Leu Asp Leu Asp Giu Lys Arg Tyr Tyr Phe Thr 2325 2330 2335 330 Asp Glu Tyr lie Ala Ala Thr Gly Ser Val Ile Ile Asp Gly Glu Glu 2340 2345 2350 Tyr Tyr Phe Asp Pro Asp Thr Ala Gln Leu Val Ile Ser Glu 2355 2360 2365 INFORMATION FOR SEQ ID NO:11: SEQUENCE
CHARACTERISTICS:
LENGTH: 19 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic) (xi) SEQUENCE DESCRIPTION: SEQ ID NO:11: TAGAAAAAAT
GGCAAATGT
19 INFORMATION FOR SEQ ID NO:12: SEQUENCE CHARACTERISTICS: LENGTH: 21 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic) (xi) SEQUENCE DESCRIPTION: SEQ ID NO:12: TTTCATCTTG TAGAGTCAAA G •21 INFORMATION FOR SEQ ID NO:13: SEQUENCE CHARACTERISTICS: LENGTH: 22 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic) (xi) SEQUENCE DESCRIPTION: SEQ ID NO:13: GATGCCACAA GATGATTTAG
TG
22 INFORMATION FOR SEQ ID NO:14: SEQUENCE
CHARACTERISTICS:
LENGTH: 22 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic) (xi) SEQUENCE DESCRIPTION: SEQ ID NO:14: CTAATTGAGC TGTATCAGGA
TC
22 331 INFORMATION FOR SEQ ID SEQUENCE CHARACTERISTICS: LENGTH: 27 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic) (xi) SEQUENCE DESCRIPTION: SEQ ID CGGAATTCCT AGAAAAAATG GCAAATG 27 INFORMATION FOR SEQ ID NO:16: ii) SEQUENCE CHARACTERISTICS: LENGTH: 26 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic) (xi) SEQUENCE DESCRIPTION: SEQ ID NO:16: GCTCTAGAAT GACCATAAGC TAGCCA 26 INFORMATION FOR SEQ ID NO:17: SEQUENCE CHARACTERISTICS: LENGTH: 27 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic) (xi) SEQUENCE DESCRIPTION: SEQ ID NO:17: CGGAATTCGA GTTGGTAGAA AGGTGGA S1-27 INFORMATION FOR SEQ ID NO:18: SEQUENCE
CHARACTERISTICS:
LENGTH: 27 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic) (xi) SEQUENCE DESCRIPTION: SEQ ID NO:18: CGGAATTCGG TTATTATCTT
AAGGATG
27 INFORMATION FOR SEQ ID NO:19: SEQUENCE CHARACTERISTICS: LENGTH: 28 base pairs TYPE: nucleic acid STRANDEDNESS: single TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic) 332 *4 S 4 4.e.
.9.4 .9 .9 9 .5 9 4 *4 5S*S 4 4.
49 4 9. 9
S
4.
S
.4 (xi) SEQUENCE DESCRIPTION: SEQ ID NO: 19: CGGAATTCTT GATAACTGGA
TTTGTGAC
28 INFORMATION FOR SEQ ID SEQUENCE
CHARACTERISTICS:
LENGTH: 511 amino acids TYPE: amino acid STRANDEDNESS: unknown TOPOLOGY: unknown (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID Leu Ile Thr Gly Phe Val Thr Vai Gly Asp A 1 5 10 Pro Ilie Asn Gly Gly Ala Ala Ser Ile Gly G 20 25 Lys Asn Tyr Tyr Phe Asn Gin Ser Gly Val L 35 40 Ser Thr Glu Asp Gly Phe Lys Tyr Phe Ala P 50 55 Giu Asn Leu Glu Gly Glu Ala Ile Asp Phe 1 65 Asp Giu Asn Ile Tyr Tyr Phe Asp Asp Asn 85 90 Trp Lys Giu Leu Asp Gly Glu Met His Tyr 100 105 Lys Ala Phe Lys Gly Leu Asn Gin Ile Giy 115 120 Asn Ser Asp Gly Val Met Gin Lys Gly Phe 130 135 Lys His Tyr Phe Asp Asp Ser Gly Val Met 145 150 Il~e Asp Gly Lys His Phe Tyr Phe Ala Glu 165 170 Gly Val Phe Asn Thr Giu Asp Gly Phe Lys 180 185 Giu Asp Leu Gly Asfl Giu Giu Gly Giu Giu 195 200 Leu Asn Phe Asn Asfl LYS Ile Tyr Tyr Phe 210 215 Val Val Gly Trp Lys Asp Leu Glu Asp Gly 225 230 sp lu ,eu ~ro 'hr ryr Phe Asp Vtal Lys 155 Asr Ty2 11i As Se 235 Lys Tyr Tyr Phe 1 15 Thr Ile Ile Asp A~ Gin Thr Giy Val P Ala Asn Thr Leu Gly Lys Leu Ile Arg Gly Ala Val Ser Pro Glu Thr 110 Tyr Lys Tyr Tyr 125 Ser Ile Asn Asp 140 Val Gly Tyr Thr Gly Giu Met Gin .175 Phe Ala His His 190 e Ser Tyr Set Gly 205 p Asp Ser Phe Thr 220 r Lys Tyr Tyr Phe -sn ~he Sp Ele s0 3iu G1y Phe Asn Giu 160 Ile Asn Ile Ala Asp 240 333 Glu Asp Thr Ala Glu Ala Tyr Ile Gil 245 Gin Tyr Tyr Phe Asn Asp Asp Gly Iil 260 26! Ilie Asn Asp Lys Val Phe Tyr Phe Se: 275 280 Gly Vai Gin Asn Ile Asp Asp A-sn Ty 290 295 Ilie Vai Gin Ile Gly Val Phe Asp Th 305 310 Ala Pro Ala Asn Thr Val Asn Asp AS 325 Tyr Ser Giy Leu Vai Arg Vai Gly Gi 340 34 Thr Tyr Thr Ile Giu Thr Giy Trp Il 355 360 Asp Lys Tyr Tyr Phe Asn Pro Giu Tit 370 375 Asn Leu Ile Asp Asp Ile Lys Tyr T) 385 390 *Arg Thr Gly Leu Ile Ser Phe Giu A~ 405 Asn Giy Giu Met Gin Phe Gly Tyr I 420 4.
*Tyr Phe Gly Giu Asp Giy Vai Met G *435 440 Asp Gly Phe Lys Tyr Phe Aia His G 450 455 Giu Giy Glu Ser Ile Asn Tyr Thr G 465 470 Arg Tyr Tyr Phe Thr Asp Giu Tyr I 485 Ile Asp Giy Giu Giu Tyr Tyr P5he 500 INFORMATION FOR SEQ ID NO:21: Wi SEQUENCE
CHARACTERISTICS:
LENGTH: 608 amino acids TYPE: amino acid STRANDEDNESS: unknown TOPOLOGY: unknown (ii) MOLECULE TYPE: protein Leu Ser L 250 e Met Gin V Asp Ser G Phe Tyr I 3 r Ser Asp G 315 n Ile Tyr C 330 u Asp Val
S
e Tyr Asp ~r Lys Lys rr Phe Asp 395 sn Asn Asn 410 le Asn Ile 25 in Ile Gly in Asn Thr iy Trp Leu 475 le Ala Ala 490 Lsp Pro ASP 05s eu Ile al Gly iy Ile 285 lie Asp 00 Ily Tyr ;iy Gin ryr Tyr 4et Giu 365 klia Cys 360 Giu Lys Tyr Tyr Giu Asp Vai Phe 44 c Leu ASY 460 Asp Lei Thr Gi', Thr AL Asn Asp Gly 25 Phe Vai Thr 270 Ile Giu Ser Asp Asn Giy Lys Tyr Phe 320 Ala Val Giu 335 Phe Gly Giu 350 Asn Giu Ser Lys Gly Ile Gly Ile Met 400 Phe Asn Giu 415 Lys Met Phe 430 Asn Thr Pro Giu Asn Phe Asp Giu Lys 480 SSer Val Ile 495 a Gin Leu 510 334 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:21: Ser Glu Glu Asn Lys Val Ser Gin Val Lys Ile Ar 1 5 10 Phe Lys Asp Lys Thr Leu Ala Asn Lys Leu Ser Ph 20 25 Lys Gin Asp Val Pro Val Ser Glu Ile Ile Leu Se 40 Tyr Tyr Glu Asp Gly Leu Ile Gly Tyr Asp Leu Gi 55 Tyr Asn Glu Lys Phe Tyr Ile Asn Asn Phe Gly Me 70 75 Leu Ile Tyr Ile Asn Asp Ser Leu Tyr Tyr Phe Ly 85 90 Asn Leu Ile Thr Gly Phe Val Thr Val Gly Asp As 100 105 Asn Pro Ile Asn Gly Gly Ala Ala Ser Ile Gly G 115 120 Asp Lys Asn Tyr Tyr Phe Asn Gin Ser Gly Val L 130 135 1 Phe Ser Thr Glu Asp Gly Phe Lys Tyr Phe Ala P 145 150 155 Asp Glu Asn Leu Glu Gly Glu Ala Ile Asp Phe T 165 170 Ile Asp Glu Asn Ile Tyr Tyr Phe Asp Asp Asn T 180 185 Glu Trp Lys Glu Leu Asp Gly Glu Met His Tyr P 195 200 Gly Lys Ala Phe Lys Gly Leu Asn Gin Ile Gly A 210 215 2 Phe Asn Ser Asp Gly Val Met Gin Lys Gly Phe N 225 230 235 Asn Lys His Tyr Phe Asp Asp Ser Gly Val Met I 245 250 Glu Ile Asp Gly Lys His Phe Tyr Phe Ala Glu 260 265 Ile Gly Val Phe Asn Thr Glu Asp Gly Phe Lys 275 280 Asn Glu Asp Leu Gly Asn Glu Glu Gly Glu Glu 290 295 Ile Leu Asn Phe Asn Asn Lys Ile Tyr Tyr Phe 305 310 315 Ala Val Val Gly Trp Lys Asp Leu Glu Asp Gly 325 330 Asp Glu Asp Thr Ala Glu Ala Tyr Ile Gly Leu 340 345 Gly Gin Tyr Tyr Phe Asn Asp Asp Gly Ile Met 355 360 335 e r y ;t 's sp lu eu ro hr yr 'he spF i2( Val -Ly Asi Ty Ii 30 As Se Se
G
Phe Val Asn Val Asn Phe Ser Asp Phe Thr Pro Ser Leu Val Ser Leu Met Val Ser Gly Pro Pro Val Asn Lys Tyr Tyr Phe 110 Thr Ile Ile Asp 125 Gin Thr Gly Val Ala Asn Thr Leu 160 Gly Lys Leu Ile 175 Arg Gly Ala Val 190 Ser Pro Glu Thr 205 STyr Lys Tyr Tyr l Ser Ile Asn Asp 240 s Val Gly Tyr Thr 255 n Gly Glu Met Gin 270 r Phe Ala His His 285 e Ser Tyr Ser Gly 0 p Asp Ser Phe Thr 320 er Lys Tyr Tyr Phe 335 er Leu Ile Asn Asp 350 In Val Gly Phe Val 365 Thr Ilie 370 Ser Gly 385 Asn Aso Lys Val Phe 375 Val Gin Asn Ile Asp 390 Giy Ile Vai Gin Ile Giy Val Phe Giu Giu Ser 465 Ilie Met Giu Phe Pro Aia 420 Ser Gly 435 Tyr Thr Lys Tyr Leu Ile Thr Glyv 500 Giy Giu 515 Phe Giy Asn Leu Ile Tyr Asp 485 Leu Met Thr Val Giu Phe 470 Asp Ile Gin Val1 Arg Thr 455 Asn Ile Se r Phe Tyr Phe Ser Asp Ser 380 Asp Asfl Tyr Phe Tyr 395 Phe Asp Thr Ser Asp 410 Asn Asp Asn Ile Tyr 425 Val Gly Giu Asp Val 440 Giy Ti-p Ile Tyr Asp 460 Pro Giu Thr Lys Lys 475 Lys Tyr Tyr Phe A-sp 490 Phe Glu Asn Asn Asn 505 Giy Tyr Ilie Asn Ile 520 31y Ile Ile Giu Ilie Asp Asp Asn 400 31ly Tyr Lys Tyr 415 Gly Gin Ala Vai 430 Tyr Tyr Phe Gly 445 Met Glu Asn Glu Ala Cys Lys Gly 480 Glu Lys Gly Ile 495 Tyr Tyr Phe Asn 510 Giu Asp Lys Met 525 Val Phe Asn Thr
S
S
S S *5 Giu Asp Giy 535 Val met Gin Ile Pro Asp Gly Phe Lys Tyr Phe Ala His Gin 545 550 Asn Thr Leu Asp Glu Asn 555 560 Phe Giu Gly Glu Ser Ile Asn Tyr Thr Giy Trp Leu Asp Leu A-sp Glu 565 570 575 Lys Arg Tyr Tyr Phe-Thr Asp Glu Tyr Ile Ala Ala Thr Gly Ser Val 580 585 590 Ile Ile Asp Giy Glu Glu Tyr Tvr Phe Asp Pro Asp Thr Ala Gin Leu 595 600 605 INFORMATION FOR SEQ ID NO:22: SEQUENCE CHARACTERISTICS: LENGTH: 1330 base pairs TYPE: nucleic acid STRANDEDNESS: double TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genomic) (ix) FEATURE: NAME/KEY:
CDS
LOCATION: 1. .1314 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:22: ATG GCT CGT CTG CTG TCT ACC TTC ACT GAA TAC ATC AAG AAC ATC ATC 48 Met Ala Arg Leu Leu Ser Thr Phe Thr Giu Tyr Ile Lys Asn Ile Ile 1 5 10 AAT ACC TCC ATC CTG AAC CTG CGC TAC GAA TCC AAT CAC CTG ATC GAC 96 Asn Thr Ser Ile Leu Asn Leu Arg Tyr Giu Ser Asn His Leu Ile Asp 20 25 336 CTG TCT CGC TAC GCT TCC AAA ATC MAC ATC GGT TCT AAA GTT AAC TTC 144 Leu Ser Arg Tyr Ala Ser Lys Ile Asn Ile Giy Ser Lys 40 Val Asn Phe GAT CCG ATC GAC MAG AAT CAG ATC CAG 2.92 Asp Pro Ile Asp Lys Asn Gin Ile Gin 55 AAA ATC GMA GTT ATC CTG MAG MAT GCT CTG TTC MAT CTG GMA Leu Phe Asn Leu Giu TCT TCC Ser Ser 240 Lys Ile Glu GMA AAC TTC 288 Giu Asn Phe ATC GTA TAC Ile Val Tyr 75 70 TCC ACC TCC TTC TGG ATC -Ser Thr Ser Phe Trp Ile 85 CGT ATC Arg Ilie 90 ATC ATC MAC TCT ATG TAC Asn Ser Met Tyr MAA TAC TTC MAC Lys Tyr Phe Asn TGC ATG GMA MC Cvs Met Glu Asn TCC ATC TCT CTG MAC 336 Ser Ile Ser Leu Asn 100 MAT TCT GGT TGG AMA 384 Asn Ser Gly Trp Lys MAT GMA TAC ACC Asn Giu Tyr Thr Ile 105 GTA TCT CTG MAC TAC Val Ser Leu Asn Tyr 120 GMA ATC AMA CAG CGT Giu Ile Lys Gin Arg Ile 110 CTG CAG 432 Leu Gin 130 115 GAC ACT Asp Thr GGT GMA ATC ATC TGG ACT Gly Giu Ile Ile Trp Thr 125 GTT GTA TTC AMA TAC TCT Val Val Phe Lys Tyr Ser 135
CAG
480 Gin 145
ATC
528 Ile
CTG
576 Leu
MAT
624 Asn
ATG
Met ATC MAC ATC TCT GAC Ile Asn Ilie Ser Asp 150 ACC MAC M.T CGT Thr Asn Asn Arg 165 ATC GAC CAG MA Ile Asp Gin Lys 180 MAC ATC ATG T-rC Asn Ile Met Phe Leu Asn CCG ATC Pro Ile AMA CTG Lys Leu TAC ATC Tyr Ile AAC TCC Asn'Ser TCC MAT Ser Asn 185 GAC GGT Asp Gly 200 CTG TTC Leu Phe Lvs Ile Tyr CTG GGT MAC Leu Gly Asn M.T CGC TGG Asn Arg Trp 155 ATC TTC GTT ACC Ile Phe Val Thr 160 ATC MAC GGC CGT Ile Asn Gly Arg
ATC
Ile CAC GCT TCT His Ala Ser 190 TGT CGT GAC ACT CAC CGC TAC Cys Arg Asp Thr His Arg Tyr 205 GAC AMA GMA CTG MAC GM A Asp Lys Giu Leu Asn Giu Lys 220
I
ATC TGG ATC AMA TAC TTC MAT 672 Ile Trp Ile Lys Tyr Phe Asn 210 215 GMA ATC MAA GAC CTG TAC GAC AAC CAG TCC M.T TCT GGT ATC CTG MA 720 Glu Ile Lys Asp Leu Tyr Asp Asfl Gin Ser Asn Ser Gly Ile Leu Lys 225 230 235 240 GAC TTC TGG GGT GAC TAC CTG CAG TAC GAC MAA CCG TAC TAC ATG CTG 768 Asp Phe Trp, Gly Asp Tyr Leu Gin Tyr Asp Lys Pro Tyr Tyr Met Leu 245 250 255 337 AAT GTG 916 Asn Leu CGC GGT 864 Arg Gly AAC ATC TAC GAT CCG AAC AAA Tyr Asp Pro Asn Lys 260 TAC ATG TAC GTG AAA Tyr Met Tyr Leu Lys TAC GTT GAG Tyr Val Asp 265 GTC AAC Val Asn
AAT
Asn
GGT
Gly 280 275 TAC GTG AAC TGT TCC GTG CCG CGT GGT TGT GTT Pro Arg Gly Ser Val 265 TAC CGT GGT ACC AAA Tyr Arg Gly Thr Lys -300 GAC AAT ATC OTT CG Asp Asn Ile Vai Arg GTA GGT ATC Val Gly Ile 270 ATG ACT ACC Met Thr Thr TTC ATC ATG Phe Ile Ile 912 Asn Ile 290 AAG AAA 960 Lys Lys 305 GGT GTA 1008 Arg Val ACC AAT 1056 Thr Asn ATC CCG 1104 Ile Pro Tyr Leu Asn Ser Ser Leu 295 TAC GCG TGT Tyr Ala Ser TAC ATG AAT Tyr Ile Asn 325 GCT TCT GAG Ala Ser Gin GOT AAC Gly Asn 310 OTT OTA Val Val AAGC AAT Asn Asn OTT AAG AAC AAA Val Lys Asn Lys 330 OAA TAG Giu Tyr GT CTG OCT Arg Leu Ala 335 GOT GTA Gly Val 9* GOT AAT GTG TCT Gly Asn Leu Ser 360 GAA AAG Glu Lys 345 GAG OTA Gin Val TOG AAA Gys Lys
OTT
Val Val Leu Ser Ala Leu Giu 350 AAG GAG GAG GOT ATC ACT 1152 Asn Asp Gin Oly Ile Thr 370 AAT GOT AACGOAT ATG GOT 1200 Asn Gly Asn Asp Ile Gly 385 390 Asn Lys
AT(
Me.
CA
Hi 39 ATG GOT TTG Ile Gly Phe G AAT t Asn 380
CGAG
s Gin ,T CG g Gin C GCG .e Pro Met Lys Ser Lys 365 CTG GAG GAG AAC Leu Gin Asp Asn TTG AAG AAT ATG Phe Asn Asn Ile 400 ATG GAA GT TGG Ile Giu Arg Ser 415 OTT OAT GAG GOT Val Asp Asp Gly 430 OCT AAA CTO OTT OCT TGC AAC TOG TAG AAT GO 1248 Aia Lys Leu Val Ala Ser Asn Trp, Tyr Asn Ar 405 410 TCT GG ACT CTG GOT TG TCT TOG GAO TTG AT 1296 Ser Arg Thr Leu Gly Gys Ser Trp Glu Phe 13 420 425 TO GOT GAA GT CG CTG TAACL UL- 1330 Trp Gly Giu Arg Pro Leu 435 INFORMATION FOR SEQ ID NO: 23: SEQUENCE
CHARACTERISTICS:
LENGTH: 438 amino acids TYPE: amino acid TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:23: -338 Met Ala 1 Asn Thr Axg Leu Leu Ser Thr Phe 5 Ser Ile Leu Asn Leu Arg Gilu Tyr Ile Giu Ser Asri Ile Ile Ile Asp Leu Ser Arg Tyr Asp Pro Ile Asp Lys Ile Giu Vai Giu Asn Phe Ser Ser Ile Ser Leu 100 Asn Ser Gly Trp, 115 Leu Gin Asp Thr 130 Gin Met Ilie Asn 145 Ile Thr Asn Asn Leu Ilie Asp Gin 180 Ala Ser Lys Ilie Asn Ile Gly Ser Lys Val Asn Phe 40 Lys Ile Thr 85 As n Lys Gin Ile Arg 165 Asn Gin Ile Gin Leu Phe Asn Leu Giu Ser Ser Leu 70 Ser Asn Val Glu Ser 150 Leu Lys Phe Giu Ser Ile 135 Asp Asn Ala Ile Ile Vai Arg Ile Tyr Asn Ser Met Pro Lys Tyr Phe Asn Thr Ile Ile 105 Asn Tyr Gly Gin Arg Vai Ile Asfl Arg 155 Ser Lys Ile 170 Asn Leu Gly 185 Met 110 Ile Lys Phe Asn His 190 His As n Giu Trp, Tyr Val1 Gly Al a Arg Giu Asn Thr Ser Thr 160 Arg Ser Tyr Lys Lys 240 Lys Pro Ile Ser Asn Asn Ile Met Phe Lys Leu Asp Gly Cys Arg Asp 195 200 Ile Trp Ile Lys Tyr Phe Asn Leu Phe Asp Lys Giu 210 215 220
S
Giu Ile Lys Asp Leu Tyr 225 230 Asp Asn Gin Ser Asn Ser Gly Ilie Leu 235 Asp Asn Arg Asn Lys 305 Arg Thr Ile Asn Phe Leu Gly Ile 290 Lys Val As n Pro Asp Trp Tyr Tyr 275 Tyr Tyr Tyr Ala Asp 355 Gin Gly Asp Tyr 245 Asp Pro Asn 260 Met Tyr Leu Leu Asn Ser Ala Ser Gly 310 Ile Asn Val 325 Ser Gin Aia 340 Val Gly Asn Gly Ile Thr Leu Lys Lys Ser 295 Asn Val Gly Leu Asn Gln Tyr Gly 280 Leu Lys Val Val Ser 360 Lys Tyr ASP 250 Val Asp 265 Pro Arg Tyr Arg Asp Asn Lys Asn 330 Giu Lys 345 Gin Val Cys Lys 339 Lys Pro Val Asn Gly Ser Giy Thr 300 Ile Val 315 Lys Giu Ile Leu Val Val Met Asn Tyr PAsn Val 285 Lys Arg Tiyr Ser Met 365 Leu Tyr Val1 270 Met Phe Asn Arg Ala 350 Lys Gir Met Leu 255 Gly Ile Thr Thr Ile Ile Asn Asp 320 Leu Aia 335 Leu G1U Ser Lys Asp 370 375 380 Asn Gly Asn Asp Ilie Gly Phe Ile Gly Phe His Gin Phe Asn Asn Ile 385 390 395 400 Ala Lys Leu Val Ala Ser Asn Trp Tyr Asn Arg Gin Ile Glu Arg Ser 405 410 415 Ser Arg Thr Leu Gly Cys Ser Trp, Giu Phe Ilie Pro Val Asp Asp Gly 420 425 430 Trp Gly Glu Arg Pro Leu 435 INFORMATION FOR SEQ ID NO:24: SEQUENCE CHARACTERISTICS: LENGTH: 23 amino acids TYPE: amino acid STRANDEDNESS: unknown TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:24: Met Gly His His His His His His His His His His Ser Ser Giy His 1 5 10 Sle Glu Gly Arg His Met Ala INFORMATION FOR SEQ ID SEQUENCE CHARACTERISTICS: LENGTH: 1402 base pairs TYPE: nucleic acid STRANDEDNESS: double TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genornic) (ix) FEATURE: NAME/KEY: CDS LOCATION: 1. .1386 (xi) SEQUENCE DESCRIPTION: SEQ ID ATG GGC CAT CAT CAT CAT CAT CAT CAT CAT CAT CAC AGC AGC GGC CAT 48 met Gly His His His His His His His His His His Ser Ser Gly His 5 10 *ATC GAA GGT CGT CAT ATG GCT AGC ATG GCT CGT CTG CTG TCT ACC TTC Ile Giu Gly Arg His Met Ala Ser Met Ala Arg Leu Leu Ser Thr Phe 20 25 ACT GAA TAC ATC AAG ARC ATC ATC AAT ACC TCC ATC CTG ARC CTG CGC 144 Thr GiU Tyr Ile Lys Asn Ile Ile Asn Thr Ser Ile Leu Asn Leu Arg 40 TAC GAA TCC ART CAC CTG ATC GAC CTG TCT CGC TAC GCT TCC AAA ATC 192 Tyr Glu Ser Asfn His Leu Ile Asp Leu Ser Arg Tyr Ala Ser Lys Ile 55 ARC ATC GGT TCT AAA GTT AAC TTC GAT CCG ATC GAC AAG ART CAG ATC 240 340 Asn Ile Gly Ser Lys Val Asn Phe Asp Pro Ilie Asp Lys Asn Gin Ile 70 75 CAG CTG TTC AAT CTG GAA TCT TCC AAA ATC GAA GTX' ATC CTG AAG AAT 288 Gin Leu Phe Asn Leu Giu Ser Ser Lys Ile Giu Val Ile Leu Lys Asn 85 90 GCT ATC GTA TAC AAC TCT ATG TAC GAA AAC TTC TCC ACC TCC TTC TGG Ala Ile Val Tyr 100 ATC CGT ATC CCG 384 Ile Arg Ile Pro 115 ACC ATC ATC AAC 432 Thr Ile Ile Asn Asn Ser AAA TAC Lys Tyr TGC ATG Cys Met Met Tyr TTC AAC Phe Asn 120 GAA AAC Giu Asn 135 TGG ACT Trp Thr Giu 105 Asn Phe Ser Thr 110 TCC ATC TCT CTG AAC AAT GAA TAC Ser Ile Ser Leu Asfl Asn Giu Tyr 125 AAT TCT GGT TGG AAA OTA TCT CTG Asn Ser Gly Trp Lys Val Ser Leu 130 AAC TAC GGT 480 Asn Tyr Gly GAA ATC Giu Ile CTG CAG Leu Gin CAG GAA ATC Gin Giu Ile CGT GTT GTA rrC TAC TCT CAG ATG AAC ATC TCT GAC Gin Arg Vai Val Phe Lys Tyr Ser Gin Met Ile Asn Ile Ser Asp Tyr 165 170 175 ATC AAT CGC TGG ATC TTC GTT ACC ATC ACC AAC AAT CGT CTG AAT AAC 576 Ile Asn Arg Tr-p Ile Phe Val Thr Ile Thr Asn Asn Arg Leu Asn Asn 180 185 190 a
S
TCC AAA ATC 624 Ser Lys Ile 195 AAT CTG GGT 672 Asn Leu Gly 210 GGT TGT CGT 720 Gly Cys Arg 225 TTC GAC AAA 768 Phe Asp Lys TAC ATC MAC GGC CGT CTG Tyr Ile Asn Giy Arg Leu 200 AAC ATC CAC OCT TCT MAT Asn Ile His Aia Ser Asn 215 GAC ACT CAC CGC TAC ATC Asp Thr His Arg Tyr Ile Asn Ile Met Phe 220 TGG ATC AAA TAC Trp Ile Lys Tyr 235 ATC M- A GAC CTG Ilie Lys Asp Leu 250 Lys Leu ASP ATC GAC CAG MAA CCG ATC 7CC le Asp Gin Lys Pro Ile Ser 205 TTC MAT Phe Asn
GMA
Glu Leu 245 AAA GMA Lys Giu TAC GAC MAC Tyr Asp Asn 255 CAG TCC MAT TCT GOT ATC CTG MAA GAC TTC 816 Gin Ser Asn Ser Gly Ile Leu Lys Asp Phe 260 265 TAC GAC MAA CCG TAC TAC ATG CTG MAT CTO TGG GGT GAC TAC CTG CAL, Trp Giy ASP Tyr Leu Gin 270 TAC GAT CCG MAC MAA TAC Tyr Asp Lys Pro T1yr Tyr Met Leu Asn Leu Tyr Asp Pro 275 280 285 OTT GAC GTC MAC MAT GTA GGT ATC CGC GGT TAC ATG TAC 912 341 CTG MAA GGT a. a.
a a a a a a a. a a a.
a Val Asp Val Asn Asn Val Gly Ile Arg Gly Tyr met Tyr Leu Lys Gly 290 295 300 CCG CC'! GGT TCT GTT ATG ACT ACC AAC ATC TAC CTG AAC TCT 'rCC CTG 960 Pro Arg Gly Ser Val Met Thr Thr Asn Ilie Tiyr Leu Asn Ser Ser Leu 305 310 315 320 TAC CGT GGT ACC AAA TTC ATC ATC AAG AAA TAC GCG TC'r GGT AAC AAG 1008 Tyr Arg Gly Thr Lys Phe Ile Ile Lys Lys Tyr Ala Ser Gly Asn Lys 325 330 335 CAC AAT ATC GTT CCC AAC AAT GAT CGT GTA TAC ATC AAT GTT GTA GTT 1056 Asp Asn Ile Val Arg Asn Asn Asp Arg Val Tyr Ile Asn Val Val Val 340 345 350 AAG AAC AAA GAA TAC CGT CTG GCT ACC AAT GCT TCT CAG GCT GGT GTA 1104 Lys Asn Lys Giu Tyr Arg Leu Ala Thr Asn Ala Ser Gin Ala Gly Vai 355 360 365 GAA AAG ATC TTG TCT GCT CTG GAA ATC CCG GAC GTT GGT AAT CTG TCT 1152 Ciu Lys Ile Leu Ser Ala Leu Giu Ile Pro Asp Val Gly Asfl Leu Ser 37 0 375 380 CAG GTA GTT GTA ATG AAA TCC AAG AAC GAC CAG GGT ATC ACT AAC AAA 1200 Gin Val Val Val Met Lys Ser Lys Asn Asp Gin Gly Ile Thr Asn Lys 385 390 395 400 TGC AAA ATG AAT CTG CAG GAC AAC AAT GGT AAC GAT ATC GGT TTC ATC 1248 Cys Lys Met Asn Leu Gin Asp Asn Asn Cly Asn Asp Ile Gly Phe Ile 405 410 415 GGT TTC CAC CAG TTC AAC AAT ATC GCT AAA CTG OTT GCT TCC AAC TGG 1296 Gly Phe His Gin Phe Asn Asn Ile Ala Lys Leu Val Ala Ser Asn Tr-p 420 425 430 TAC AAT CGT CAG ATC GAA CGT TCC TCT CCC ACT CTG GGT TGC TCT TGG 1344 Tyr Asn Arg Gin Ile Giu Arg Ser Ser Arg Thr Leu Gly Cys Ser Tr-p 435 440 445 GAG TTC ATC CCG GTT GAT GAC GGT TGG GGT GAA CGT CCC CTG 1386 Ciu Phe Ile Pro Val Asp Asp Gly Trp Gly Giu Arg Pro Leu 450 455 460 TAACCCGGGA
AAGCTT
1402 INFORMATION FOR SEQ ID NO:26: SEQUENCE
CHAACTERISTICS:
LENGTH: 462 amino acids TYPE: amino acid TOPOLOGY: linear (ii) MOLECULE TYPE: protein (xi) SEQUENCE DESCRIPTION: SEQ ID NO:26: Met Gly His His His His His His His His His His Ser Ser Gly His 1 5 10 342 Ile G1 Thr G] Tyr G1 Asn I: Gin L Ala I.
Ile A Thr I 1 Asn T 145 Gin A S Ile P Ser I .5 AsnI Gly I 225 Phe Glr Tyr Val *SS Pro 305 Tyr Asp Lys uu uu uu Le eu le rg le 30 yr rg sn S f .ys ,eu 21C AsI Se: As As 29 Ar Ar As Ai Gly Arg His Met Al Tyr Ile Lys Asn Ii Ser Asn His Leu II 5 Gly Ser Lys Vai As 70 Phe Asn Leu Giu Se 85 Val Tyr Asn Ser ME 100 ile Pro Lys Tyr Pk 115 Ile Asn Cys Met G Gly Giu Ile Ile T 150 Val Vai Phe Lys T 165 Arg Trp Ile Phe V 180 Ile Tyr Ile Asn G 195 Gly Asn Ile His A 2 Arg Asp Thr His I 230 Lys Giu Leu As 245 r Asn Ser Gly Ile 260 p Lys Pro Tyr Tyr 275 p Vai Asn Asn Val 0 g Giy Ser Val Met 310 g Giy Thr Lys Phe 325 ;n Ile Vai Arg Asn 340 ;n Lys Giu Tyr Arg 355 a e e 35 rp yr al ly la 15\ Le' Me G1 29 Th I1 As Li Ser Met Ala Arg Leu Leu Se 25 Ile Asn Thr Ser Ile Leu As 40 Asp Leu Ser Arg Tyr Ala SE Phe Asp Pro Ile Asp Lys A- 75 Ser Lys Ile Giu Val Ile LI 90 Tyr Giu Asn Phe Ser Thr S 105 1 Asf Ser Ile Ser Leu Asn A 120 125 As Asn Ser Giy Trp Lys V 140 Thr Leu Gin Asp Thr Gin G 155 Ser Gin Met Ile Asn Ile S 170 Thr Ile Thr As Asn Arg L 185 1 Arg Leu Ile Asp Gin Lys 1 200 205 Ser Asn Asn Ile Met Phe I 220 Tyr Ile Trp Ile Lys Tyr I 235 u Lys Giu Ile Lys Asp Leu 250 L Lys Asp Phe Trp Giy Asp 265 t Leu Asn Leu Tyr Asp Pro 280 285 y Ile Arg Gly Tyr Met Tyr 5 300 .r Thr Asn Ile Tyr Leu Asn 315 .e Ile Lys Lys Tyr Ala Ser 330 ;n Asp Arg Vai Tyr Ile Asn 345 su Ala Thr Asn Aa Ser Gin 360 365 !r Thr P1 ;n Leu Ai ir Lys I: sn Gin I: -u Lys X, er Phe T sn Giu T al Ser L iu Ile L 1 er Asp T 175 .eu Asn I ,ro Ile S ,ys Leu Phe Asn Tyr Asp 255 Tyr Leu 270 Asn Lys Leu Lys Ser Ser Gly Asn 335 Vai Vai 350 Ala Gly Xe Le Le
BO
sn rp yr eu ,ys yr Lsn er ksp Leu 240 Asn Gin Tyr Gly Leu 320 Lys Val Val 343 Giu Lys Ile Leu Ser Ala Leu Giu Ile Pro Aso Val Gly Asn Leu Ser 370 375 380 Girn Val Val Val Met Lys Ser Lys Asn Asp Gin Gly Ile Thr Asn Lys 385 390 395 400 Cys Lys met Asn Leu Gin Asp Asn Asn Gly Asn Asp Ile Gly Phe Ile 405 410 415 Gly Phe His Gin Phe Asn Asn Ile Ala Lys Leu Val Ala Ser Asn Trp 420 425 430 Tyr Asn Ax-g Gin Ile Giu Arg Ser Ser Arg Thr Leu Gly Cys Ser Trp 435 440 445 Giu Phe Ile Pro Vai Asp Asp Gly Trp Gly Giu Arg Pro Leu 450 455 460 INFORMATION FOR SEQ ID 140:27: SEQUENCE
CHARACTERISTICS:
LENGTH: 3691 base pairs TYPE: nucleic acid STRANDEDNESS: double ID) TOPOLOGY: linear (ii) MOLECULE TYPE: DNA (genornic) (ix) FEATURE: NAME/KEY:
CDS
LOCATION: 1. .3888 (xi) SEQUENCE DESCRIPTION: SEQ ID NO:27: .ATG CAA TTT GTT ALAT AAA CAA TTT AAT TAT AAA GAT CCT GTA AAT GGT 48 Met Gin Phe Val Asn Lys Gin Phe Asn Tyr Lys Asp Pro Val Asn Gly 1 5 10 is GTT GAT ATT GCT TAT ATA AAA ATT CCA AAT GTA GGA CAR ATO CAR CCA 96 *Val Asp Ile Ala Tyr Ile Lys Ile Pro Asn Val Gly Gin Met Gin Pro 20 .25 OTA AAA OCT TTT AAA ATT CAT ART AAA ATA TOO GTT ATT CCA OAR AGA 144 *Vai Lys Ala Phe Lys Ile His Asn Lys Ile Trp Val Ile Pro Olu Arg 35 40 OAT ACA TTT ACA ART CCT OAA OAA OGA GAT TTA ART CCA CCA CCA GAA 192 ***Asp Thr Phe Thr Asn Pro Giu Oiu Oly Asp Leu Asn Pro Pro Pro 01".
:50 55 OCA AAA CAR OTT CCA OTT TCA TAT TAT GAT TCA ACA TAT TTA AOT ACA 240 Ala Lys Gin Val Pro Vai Ser Tyr Tyr Asp Ser Thr Tyr Leu Ser Thr 70 75 GAT ART GAR AAA GAT ART TAT TTA AAG GOA OTT ACA AAA TTA TTT GAG 288 Asp Asn Oiu Lys Asp Asn Tyr Leu Lys Gly Val Thr Lys Leu Phe G1u 85 90 AGA ATT TAT TCA ACT OAT CTT OGA AGA ATG TTG TTA ACA TCA ATA GTA 336 Arg Ile Tyr Ser Thr Asp Leu Gly Arg Met Leu Leu Thr Ser Ile Val 100 105 110 3,44 AGG GGA ATA C-C-A TTT TGG 384 Arg Gly Ile Pro Phe Tr-p 115 GTT ATT GAT ACT AAT TGT 432 Val Ile Asp Thr Asn Cys 130 GGT GGA AGT ACA ATA GAT ACA GAA TTA AAA Gly Gly Ser Thr Ile Asp Thr Giu Leu Lys 120 125 ATT AAT GTG ATA C-AA CCA GAT GGT AGT TAT Ile Asn Val Ile Gin Pro Asp Gly Ser Tyr 135 140 AGA TCA GAA GAA C-TT AAT C-TA GTA ATA ATA GGA CCC TCA GCT GAT ATT 480 Arg Ser Giu Giu Leu Asn Leu Vai Ile Ile Gly Pro Ser Ala Asp Ile 145 150 155 160 ATA C-AG TTT GAA TGT AAA AGC TTT GGA C-AT GAA GTI' TTG AAT C-TT ACG 528 Ile Gin Phe Giu C-ys Lys Scr Phe Gly His Giu Vai Leu Asn Leu Thr 165 170 175 C-GA AAT GGT TAT GGC TCT ACT CAA TAC AT!' AGA TTT AGC C-C-A GAT TT!' 576 Arg Asn Gly Tyr Gly Ser Thr Gin Tyr Ile Arg Phe Ser Pro Asp Phe 180 185 190 AC-A TTT GGT TTT GAG GAG TCA CTT GAA GTT GAT AC-A AAT C-CT C-TT TTA 624 Thr Phe Giy Phe Giu Giu Ser Leu Giu Val Asp Thr Asn Pro Leu Leu 195 200 205 GGT GCA GGC AAA TTT GCT AC-A GAT C-CA CA GTA AC-A TTA GC-A CAT GAA 67 2 Gly Ala Giy Lys Phe A-1a Thr Asp Pro Ala Val Thr Leu Ala His Giu 210 215 220 CTT ATA C-AT GCT GGA C-AT AGA TTA TAT GGA ATA GCA ATT AAT C-CA A.AT 720 Leu Ile His Ala Giy His Arg Leu Tyr Gly Ile Ala Ile Asfl Pro Asfl 225 230 235 240 AGG GTT TTT AAA GTA AAT ACT AAT CC TAT TAT GAA ATG AGT GGG TTA 768 Arg Val Phe Lys Val Asn Thr Asn*Ala Tyr Tyr Giu Met Scr Gly Leu 245 250 255 CAA GTA AGC TT!' GAG GAA CT!' AGA ACA TTI' GOG GGA C-AT GAT GCA AAG 816 Ciu Val Ser Phe Ciu Giu Leu Arg Thr Phe Cly Gly His Asp Ala Lys 260 265 270 TTT ATA GAT AGT TTA C-AG GA.A AAC GAA TTT C-CT C-TA TAT TAT TAT AAT 864 Phe Ile Asp Set Leu Gin Giu Asn Giu Phe Arg Leu Tyr Tyr Tyr Asn 275 280 285 AAG TTT AAA CAT ATA GC-A AGT AC-A C-TT AAT AAA GCT AAA TCA ATA GTA 912 Lys Phe Lys Asp Ile Ala Ser Thr Leu Asn Lys Ala Lys Ser Ile Vai 290 295 300 GGT ACT ACT GCT TC-A TTA C-AG TAT ATG AAA AAT CTT TTT AAA GAG AAA 960 Cly Thr Thr Ala Ser Leu Gin Tyr Met Lys Asn Val Phe Lys Giu Lys 305 310 315 320 TAT C-TC C-TA TCT GAA GAT ACA. TCT GGA AAA TI'T TCG GTA GAT AAA TTA 1008 Tyr Leu Leu Scr Ciu Asp Thr Ser Cly Lys Phe 5cr Val Asp Lys Leu 325 330 335 345 AAA TTT GAT AAG TTA TAC AAA ATG TTA ACA GAG ATT TAC ACA GAG GAT 1056 Lys Phe Asp Lys Leu Tyr Lys Met Leu Thr Giu Ile Tyr Thr Glu Asp 340 345 350 AAT TTT GTT AAG TTT TTT AAA GTA CTT MAC AGA AAA ACA TAT TTG AAT 1104 Asfl Phe Val Lys Phe Phe Lys Val Leu Asn Arg Lys Thr Tyr Leu Asn 355 360 365 T1'T GAT AAA GCC GTA TTT MAG ATA AAT ATA GTA CCT MAG GTA AAT TAC 1152 Phe Asp Lys Ala Val Phe Lys Ile Asn Ile Val Pro Lys Val Ann Tyr 370 375 380 ACA ATA TAT GAT GGA TTT MAT TTA AGA MAT ACA MAT TTA GCA GCA MAC 1200 Thr Ile Tyr Asp Gly Phe Asn Leu Arg Ann Thr Asn Leu Ala Ala Asn 385 390 395 400 TTT MAT GGT CAA M.T ACA GMA ATT MAT MAT ATG MAT TTr ACT AMA CTA 1248 Phe Asn AA MT 1296 Lys Asn Gly Gin Asn Thr Glu Ile An MtT. Asn Phe. Th 415 GTA AGA
TTT
Phe ACT GGA TTG Thr Gly Leu 420 ACT TCT AMA Thr Ser Lys TTT GMA Phe Giu ACT AMA Thr Lys 440 TTT TAT MAG Phe Tyr L.ys 425 TCA TTA GAT Ser Leu Asp Leu Leu CyT Vl r 9* s.d
S
5*
S
S S GGG ATA ATA *:1344 Gly Ile Ile 435 GCA TTA MAT 1392 Ala Leu Asn 450 AGT CCT TCA 430 AMA GGA TAC MAT MAG Lys Gly Tyr Asn Lys 445 TGG GAC TTG TTT TTT Tr-p Asp Leu Phe Phe GAT TTA TGT ATC Asp Leu Cys Ile 455 AMA GTT Lys Val MAT MAT Ann An (Thh GAA GAT 1440 Ser Pro 465 Ser Giu Asp MAT TTT ACT MAT GAT CTA Ann Phe Thr Ann Asp Leu 470 475 AMA GGA GMA GMA Lys Gly Giu Giu 480
S
S S
S.
S
S. S S S
S.
S
55.5 ATT ACA 1488 Ile Thr GAT TTA 1536 Asp Leu
TCT
Ser
ATA
Ile GAT ACT MAT ATA GMA OCA OCA GMA GMA MT ATT AGT TTA Asp Thr Ann Ile Giu Ala Ala Giu Giu Ann Ile Ser Leu 485 490 495 CAA CMA TAT TAT TTA ACC TI'T MT iT GAT MAT GMA CCT Gin Gin Tyr Tyr Leu Thr Phe Ann Phe Asp Ann Giu Pro 500 505 510 TCA ATA GMA AT CTT TCA AGT GAC ATT ATA GGC CMA TTA Ser Ile Giu Asn Leu Ser Ser Asp Ile Ile Gly Gin Leu 520 525 CCT MAT ATA GAA AGA TTT CCT MAT GGA AMA MG TAT GAG Pro Asn Ile Giu Arg Phe Pro Ann Gly Lys Lys Tyr G1u 535 540 GMA MT ATT 1584 Giu Asn Ile 515 GMA CTT ATG 1632 Glu Leu Met 530 TTA GAT 1680 Leu Asp 545 AMA TAT ACT ATG TTC CAT TAT CTT CGT GCT CMA GMA TTT GMA Lys Tyr Thr Met Phe His Tyr Leu ALrg Ala Gin Glu Phe GiU 550 555 560 346 CAT GGT AAA TCT AGG ATT GCT TTA ACA AAT TCT GTT MAC GAA GCA TTA 1726 His Gly Lys Ser Arg lie Ala Leu Thr Asn Ser Val Asn Glu Ala Leu 565 570 575 TTA AAT CCT AGT CGT GTT TAT ACA TTT TTT TCT TCA GAC TAT GTA AAG 1776 Leu Asn Pro Ser Arg Val Tyr Thr Phe Phe Ser Ser Asp Tyr Val Lys 580 585 590 AAA GTT MAT AAA GCT ACG GAG GCA GCT ATG TTT TTA GGC TGG GTA GAA 1824 Lys Val Asn Lys Ala Thr Giu Ala Ala Met Phe Leu Gly Trp Val Glu 595 600 605 CAA TTA GTA TAT GAT TTT ACC GAT GAA ACT AGC GAA GTA AGT ACT ACG 1872 Gin Leu Val Tyr Asp Phe Thr Asp Glu Thr Ser Giu Val Ser Thr Thr 610 615 620 GAT AMA ATT GCG GAT ATA ACT ATA ATT ATT CCA TAT ATA GGA CCT OCT 1920 Asp Lys Ile Ala Asp Ile Thr Ile Ile Ile Pro Tyr Ile Gly Pro Ala 625 630 635 640 TTA AAT ATA GGT AAT ATG TTA TAT AAA GAT GAT TTT GTA GGT GCT TTA 1968 Leu Asn Ile Gly Asn Met Leu Tyr Lys Asp Asp Phe Val Gly Ala Leu 645 650 655 ATA TTT TCA GGA GCT GTT ATT CTG TTA GMA TTT ATA CCA GAG ATT GCA 2016 em.. Ile Phe Ser Gly Ala Val Ile Leu Leu Giu Phe Ile Pro Giu Ile Ala 660 665 670 ATA CCT GTA TTA GOT ACT TTT GCA CTT GTA TCA TAT ATT GCG AAT AAG 00 2064 Ile Pro Vai Leu Gly Thr Phe Ala Leu Val Ser Tyr Ile Ala Asn Lys 675 680 685 GTT CTA ACC GTT CMA ACA ATA OAT MAT GCT TTA AGT AAA AGA AAT GAA 0211 Val Leu Thr Val Gin Thr Ile Asp"Asn Ala Leu Ser Lys Arg Asn Glu 690 695 700 AA TOG GAT GAG GTC TAT AAA TAT ATA OTA ACA AT TGG TTA GCA AAG 00 2160 Lys Trp, Asp Glu Val Tyr Lys Tyr Ile Val Thr Asn Trp Leu Ala Lys 705 710 715 720 OTT AAT ACA CAG ATT GAT CTA ATA AGA AAA MAA ATG AAA GAA GCT TTA 2208 Val Asn Thr Gin Ile Asp Leu Ile Arg Lys Lys Met Lys Giu Ala Leu 725 730 GAA AAT CAA GCA GAA GCA ACA AAG GCT ATA ATA AAC TAT CAG TAT AAT 0e 2256 Giu Asn Gin Ala Giu Ala Thr Lys Ala lie Ile Asn Tyr Gin Tyr Asn 740 745 750 CAA TAT ACT GAG GAA GAG AAA AAT MT ATT MAT TTT AAT ATT GAT GAT 2304 Gin Tyr Thr Giu Giu Giu Lys Asn ASn Ile Asn Phe Asn Ile Asp Asp 755 760 765 TTA AGT TCG AAA CTT AAT GAG TCT ATA AAT AAA GCT ATG ATT AAT ATA 2352 Leu Ser Ser Lys Leu Asf Glu Ser Ile Asn Lys Ala Met lie Asn Ile 770 775 780 347 AAT AAA TTr TTG AAT CAA TGC TCT GTT TCA TAT TTA ATG AAT TCT ATG 2400 Asn Lys Phe Leu Asn Gin Cys Ser Vai Ser Tyr Leu Met Asn Ser Met 785 790 795 S00 ATC CCT TAT GGT GTT AAA CGG TTA GAA GAT TTT GAT GCT AGT CTT AAA 2448 Ile Pro Tyr Giy Val Lys Arg Leu Giu Asp Phe Asp Ala Ser Leu Lys 805 810 815 GAT GCA TTA Tl'A AAG TAT ATA TAT GAT AAT AGA GGA ACT TTA ATT GGT 2496 Asp Ala Leu Leu Lys Tyr Ile Tyr Asp Asn Arg Giy Thr Leu Ile Gly 820 825 830 CAA GTA GAT AGA TTA AAA GAT AAA CTT AAT AAT ACA CTT AGT ACA GAT 2544 Gin Val Asp Arg Leu Lys Asp Lys Val Asn Asn Thr Leu Ser Thr Asp 835 840 845 0 ATA CCT TTT CAG CTT TCC AAA TAC GTA GAT AAT CAA AGA TTA TTA TCT 2592 Ile Pro Phe Gin Leu Ser Lys Tyr Val Asp Asn Gin Arg Leu Leu Ser o o850 855 860 *ACA TTT ACT GAA TAT ATT AAG AAT ATT ATT AAT ACT TCT ATA TTG AAT 2640 0 Thr Phe Thr Giu Tyr Ile Lys Asn Ile Ile Asn Thr Ser Ile Leu Asn o865 870 875 880 TTA AGA TAT GAA AGT AAT CAT TTA ATA GAC TTA TCT AGG TAT GCA TCA o 2688 ::000Leu Arg Tyr Giu Ser Asn His Leu Ile Asp Leu Ser Arg Tyr Ala Ser 885 890 895 AA.A ATA AAT ATT CGT AGT AAA GTA AAT TT1' GAT CCA ATA GAT AAA AAT 2736 *..Lys Ile Asn Ile Giy Ser Lys Val Asn Phe Asp Pro Ile Asp Lys Asn 900 905 910 *CAA ATT CAA TTA TTT AAT TTA GAA AGT AGT AAA ATT GAG GTA ATT TTA 2784 **.Gin Ile Gin Leu Phe Asn Leu Giu Ser Ser Lys Ile Giu Val Ile Leu 00. 915 920 925 o :.0::AAA AAT GCT ATT GTA TAT AAT ACT ATG TAT GAA AAT TTT AGT ACT AGC 2832 Lys Asn Ala Ile Val Tyr Asn Ser Met Tyr Giu Asn Phe Ser Thr Ser 930 935 940 TTT TGG ATA AGA ATT CCT AAG TAT TTT AAC ACT ATA AGT CTA AAT AAT 2860 Phe Trp Ile Arg Ilie Pro Lys Tyr Phe Asn Ser Ile Ser Leu Asn Asn 945 950 955 960 GAA TAT ACA ATA ATA AAT TCT ATG GAA AAT AAT TCA GCA TGG AAA GTA 2928 Giu Tyr Thr Ile Ile Asn Cys Met Giu Asn Asn Ser Gly Trp Lys Val 965 970 975 TCA cTT AAT TAT GGT CAA ATA ATC TCG ACT TTA CAG GAT ACT CAG CAA 2976 Ser Leu Asn Tyr Gly Gu Ile Ile Trp Thr Leu Gin Asp Thr Gin Giu 980 985 990 ATA AAA CAA AGA GTA CTT TTT AAA TAC ACT CAA ATG ATi' AAT ATA TCA 3024 Ile Lys Gin Arg Vai Val Phe Lys Tyr Ser Gin Met Ile Asn Ile Ser 995 1000 1005 348 GAT TAT ATA AAC AGA TGG ATT TTT GTA ACT ATC ACT MAT AAT AGA TTA 3072 Asp Tyr Ile Asn Arg Trp Ile Phe Val Thr Ile Thr Asn Asn Arg Leu 1010 1015 1020 AAT AAC 3120 Asn Asn 1025 ATT TCA 3168 lle Ser TTA GAT 3216 Leu ASP AAT CTT 3264 Asn Leu TCT AAA ATT TAT ATA AAT GGA AGA TTA ATA Ser Lys Ile Tyr Ile Asn Gly Arg Leu Ile 1030 1035 AAT TTA GOT AAT ATT CAT GCT AGT AAT AAT As n Leu Gly Asn Ile His Ala Ser Asn Msn 1045 1050 GOT TGT AGA GAT ACA CAT AGA TAT ATT TGG Gly Cys Arg Asp Thr His Arg Tyr Ile Trp 1060 1065 TTT GAT AAG OAA TTA AAT GAA AAA GAA ATC Phe Asp Lys Glu Leu Asn Glu Lys Giu Ile 1075 1080 Asp Gin Lys Pro 1040 Ile Met Phe Lys 1055 ATA AAA TAT TTT Ile Lys Tyr Phe 1070 AAA OAT TTA TAT Lys Asp Leu Tyr 1085 C OAT A.AT CAA TCA AAT TCA GGT ATT TTA AAA GAC TTT TOG GOT OAT TAT 3312 Asp Asn Gin Ser Asn Ser Oly Ile Leu Lys Asp Phe Trp Oly Asp Tyr 1090 1095 1100 TTA CAA TAT OAT AAA CCA TAC TAT ATO TTA AAT TTA TAT OAT CCA AAT 3360 Leu Gin Tyr Asp Lys Pro Tyr Tyr Met Leu Msn Leu Tyr Asp Pro Asn 1105 1110 1115 1120 AAA TAT GTC OAT OTA AAT AAT OTA GOT ATT AGA GOT TAT ATO TAT CTT 3408 Lys Tyr Val Asp Val Asn Asn Val Oly Ile Arg Oly Tyr Met Tyr Leu 1125 1130 1135 AAA CGOG CCT AGA GOT AOC GTA ATO ACT ACA AAC ATT TAT TTA AAT TCA 3456 Lys Gly Pro Arg Oly Ser Vai Met Thr Thr Asn Ile Tyr Leu Asn Ser 1140 1145 1150 AOT TTG TAT AGO GOG ACA AAA TTT ATT ATA MAA AMA TAT OCT TCT GGA 3504 Ser Leu.Tyr Arg Oly Thr Lys Phe Ile Ile Lys Lys Tyr Ala Ser Oly 1155 1160 1165 MAT AMA OAT MAT ATT OTT AGA MAT MAT OAT COT GTA TAT ATT MAT OTA 3552 Msn Lys Asp Asn Ile Val Arg Asn Asn Asp Arg Val Tyr Ile Msn Val 1170 1175 1180 OTA OTT AMA MT AMA GMA TAT AGO TTA OCT ACT MAT GCA TCA CAG OCA 3600 Val Val Lys' Mn Lys O1u Tyr Arg Leu Ala Thr Asn Ala Ser Gin Ala 1185- 1190 1195 1200 GOC OTA GMA AMA ATA CTA AGT OCA TTA GMA ATA CCT OAT OTA OGA MAT 3648 Oly Val Olu Lys Ile Leu Ser Ala Leu Gu Ile Pro Asp Val Oly Msn 1205 1210 1215 CTA AGT CMA GTA OTA GTA ATG MAG TCA AMA MT OAT CMA OGA ATA ACA 3696 Leu Ser Gin Val Val Val Met Lys Ser Lys Msn Asp Gin Oly Ile Thr 1220 1225 1230 349 AAT ;AA TGC AAA 374~4 Asn :Lys Cys Lys 1235 rrr ;ATA GGA TIT 3792 Phe -le Gly Phe 1259 AAT TGG TAT AAT 3840 Asn Trp Tyr Asn 1265 TCA -IrGG GAA TTT 388B Ser TIrp Giu Phe ATG AAT TTA CAA GAT Met Asn Leu Gin Asp 1240 CAT CAG TiT AAT AAT His Gin Phe Asn Asn 1255 AGA CAA ATA GAA AGA Arg Gin Ile Glu Arg 1270 ATT CCT GTA GAT GAT Ile Pro Val Asp Asp 1285 AAT A.AT GGG AAT GAT Asn Asn Gly Asn Asp 1245 ATA GCT AAA CTA GTA Ile Ala Lys Leu Val 1260 TCT AGT AGG ACT TTG Ser Ser Arg Thr Leu 1275 GGA TGG GGA GAA AGG Gly Trp Gly Glu Arg 1290 ATA GGC Ile Gly GCA AGT Ala Ser GGT TGC Gly Cys 1280 CCA CTG Pro Leu 1295
TAA
3851.
INMRMATION FOR SEQ ID NO:28: SEQUENCE CHARACTERISTICS: LENGTH: 1296 amino acids TYPE: amino acid TOPOLOGY: linear Iii) MOLECULE TYPE: protein Axi) SEQUENCE DESCRIPTION: SEQ ID NO:28: met CIn Phe Val Asn Lys Gin Phe Asn Tyr Lys Asp Pro 1 5 Val Asp Ile Ala Tyr Ile Lys Ile Pro Asn Val Gly Gin 20 Val Asn Gly 15 Met Gin Pro Val1 Asp Ala Asp Arg Arg Val Arg 145 Ala Phe Gin Giu Tiyr Ile 115 Asp Glu Phe Thr Val Lys Se r 100 Pro Thr Glu Lys Asri Pro Asp 85 Thr Phe Asn Leu Ile Pro Val 70 Asn Asp Trp Cys Asn 150 His Asn Lys Ilie Trp Val Ile Pro Glu Axg Giu Ser Leu Gly Ile 135 Leu Glu Tyr Leu Gly Gly 120 Asfl Val1 Gly Tyr Lys Arg 105 Ser Val1 Ile Asp Asp Gly 90 Met Thr Ile Ile Leu Ser 75 Val Leu Ile Gin Gly 155 Pro Tyr Lys Thr Thr 125 Asp Ser Pro Leu Leu Ser 110 Glu Gly Ala Pro Glu Ser Thr Phe Glu Ile Val Leu Lys Ser Tyr Asp Ile 160 Leu Thr 175 Ile Gln Phe Giu Cys Lys Ser 165 Phe Gly His Giu Val Leu A-sn 170 Arg Asn Giy Gly Ser Thr Gin Tyr Ile ArS Phe Ser Pro Asp Phe 185 190 350 Thr Phe Gly 195 Gly Ala Gl' 210 Leu Ile Hi~ 225 Arg Val Ph4 Glu Val Se Phe Ile AS 27 Lys Phe Ly 290 Gly Thr Th 305 Tyr Leu Le Lys Phe Al Asn Phe V 3, Phe Asp L 370 Thr Ile T 385 Phe Asn G Lys Asn P Gly Ile 1 Ala Leu 450 Ser Pro 465 Ile Thr Asp- Leu Glu Asn Glu Leu 530 Leu ASP 545 His Gly r
P
5 y
S
S
I
Phe Giu Glu Ser Leu Glu Val Asp Thr Asn Pro Leu Leu 200 205 Lys Phe Ala Thr Asp Pro Ala Val Thr Leu Ala His Glu 215 220 Ala Gly His Arg Leu Tyr Gly Ile Ala Ile Asn Pro Asn 230 235 240 Lys Val Asn Thr Asn Ala Tyr Tyr Giu Met Ser Gly Leu 245 250 255 Phe Giu Giu Leu Arg Thr Phe Gly Gly His Asp Ala Lys 260 265 270 Ser Leu Gin Glu Asn Giu Phe Arg Leu Tyr Tyr Tyr ASn 280 285 Asp lie Ala Ser Thr Leu Asn Lys Ala Lys Ser Ile Val 295 300 Ala Ser Leu Gin Tyr Met Lys As Val Phe Lys Giu Lys 310 315 320 u Ser Giu Asp Thr Ser Gly Lys Phe Ser Val Asp Lys Leu 325 330 335 p Lys Leu Tyr Lys Met Leu Thr Giu Ile Tyr Thr Giu Asp 340 345 350 1 Lys Phe Phe Lys Val Leu Asn Arg Lys Thr Tyr Leu Asn 5 360 365 s Ala Val Phe Lys Ile Asn Ile Val Pro Lys Val Asn Tyr 375 380 r Asp Giy Phe Asn Leu Arg Asn Thr Asn Leu Ala Ala Asn 390 395 400 .y Gin Asn Thr Giu Ile Asn Asn Met Asn Phe Thr Lys Leu 405 410 415 %e Thr Gly Leu Phe Glu'Phe Tyr Lys Leu Leu Cys Val Arg 420 425 430 le Thr Ser Lys Thr Lys Ser Leu Asp Lys Giy Tyr Asn Lys 35 440 445 sn Asp Leu Cys Ile Lys Val Asn ASn Trp Asp Leu Phe Phe 455 460 er Giu Asp Asn Phe Thr Asn Asp Leu ASn Lys Giy Glu Giu 470 475 480 er Asp Thr Asn Ile Giu Ala Aia Glu Glu Asn lie Ser Leu 485 490 495 :le Gin Gin Tyr Tyr Leu Thr Phe Asn Phe Asp Asn Giu Pro 500 505 510 lie Ser Ile Giu Asn Leu Ser Ser Asp Ile Ile Giy Gin Leu 515 520 525 4et Pro Asn lie Giu Arg Phe Pro Asn Gly Lys Lys Tyr GiU 535 540 Lys Tyr Thr Met Phe His Tyr Leu Arg Aia Gin Giu Phe Giu 550 555 560 Lys Ser Arg Ile Ala Leu Thr Asn Ser Vai Asn Giu Ala Leu -351- 565 570 Leu Asn Pro Ser Arg Val Tyr Thr Phe Phe Ser Ser ASI 580 585 Lys Val Asn Lys Ala Thr Giu Ala Ala Met Phe Leu GI~ 595 600 Gin Leu Val Tyr Asp Phe Thr Asp Giu Thr Ser Giu Va 610 615 620 Asp Lys Ile Ala Asp Ile Thr Ile Ile Ile Pro Tyr Ii.
625 630 635 Leu Asn Ile Gly Asn Met Leu Tyr Lys Asp Asp Phe Va 645 650 Ile Phe Ser Gly Ala Val Ile Leu Leu Giu Phe Ile Pr 660 665 Ile Pro Val Leu Gly Thr Phe Ala Leu Val Ser Tyr 11 675 680 68 Val Leo Thr Val Gin Thr Ile Asp Asn Ala Leu Ser Ly 690 695 700 Lys Trp Asp Giu Val Tyr Lys Tyr Ile Val Thr Asn T2 705 110 715 Val Asn Thr Gin Ile Asp Leu Ile Arg Lys Lys Met L~ 725 730 Giu Asn Gin Ala Giu Ala Thr Lys Ala Ile Ile Msn 740 745 Gin Tyr Thr Giu Giu Giu Lys Asn Asn Ile Asn Phe A 755 760 7 Leu Ser Ser Lys Leo Asn Giu Ser Ile Msn Lys Ala M 770 775 780 Asn Lys Phe Leo Asn Gin Cys Ser Vai Ser Tyr Leu M 785 790 795 Ile Pro T.yr Gly Val Lys Arg Leu Giu Asp Phe Asp 805 810 Asp Ala .Leu Leu Lys Tyr Ile Tyr Asp Msn Arg Gly 1 820 825 Gin Val Asp Arg Leu Lys Asp Lys Val Asn Asn Thr 835 840 Ile Pro Phe Gin Leu Ser Lys Tyr Val Asp Asn Gin 850 855 860 Thr Phe Thr Giu Tyr Ile Lys Mn Ile Ile Msn Thr 865 670 875 Leo Arg Tyr Giu Ser Asn His Leu Ile Asp Leo Ser 885 890 Lys Ilie Asn Ile Giy Ser Lys Vai Msn Phe Asp Pro 900 905 Gin Ile Gin Leu Phe Msn Leu Glu Ser Ser Lys Ile 915 920 Lys Msn Ala Ile Vai Tyr Msn Ser Met Tyr GlU Msn 930 935 940 -352- 575 Tyr Vai Lys 590 ~Trp, Val Giu 1 Ser Thr Thr e Gly Pro Ala 640 i Gly Ala Leu 655 Gu Ile Ala 670 e Ala Mn Lys 's Arg Asn Giu -p Leo Ala Lys 720 Glu Ala Leo 735 yr Gin Tyr Asn 750 sn Ile Asp Asp et. Ile Mn Ile et Asn Ser Met 800 lia Ser Leo Lys 815 rhr Leo Ile Gly 830 Leu Ser Thr Asp 845 Arg Leo Leo Ser Ser Ile Leu Msn 880 Arg Tyr Ala Ser 895 Ile Asp Lys Asn 910 Giu Val Ile Leo 925 Phe Ser Thr Ser Phe Trp Ile Arg Ile Pro Lys Tyr Phe Asn Ser Ile Ser Leu Asn Asn 945 950 955 960 Giu Tyr Thr Ile Ile Asn Cys Met Giu Asfl As n Ser Gly Trp Lys Val 965 970 975 Ser Leu Asn Tyr Gly Giu Ile Ile Trp Thr Leu Gin Asp Thr Gin Giu 980 985 990 Ile Lys Gin Arg Val Vai Phe Lys Tyr Ser Gin Met Ile Asn Ile Ser 995 1000 1005 Asp Tyr Ile Asn Arg Trp Ile Phe Val Thr Ile Thr Asn Asn Arg Leu 1010 10is 1020 Asn Asn Ser Lys Ile Tyr Ile Asn Gly Arg Leu Ile Asp Gin Lys Pro 1025 1030 1035 1040 Ile Ser Asn Leu Gly Asn Ile His Ala Ser Asn Asn Ile Met Phe Lys 1045 1050 1055 Leu Asp Gly Cys Arg Asp Thr His Arg Tyr Ile Trp Ile Lys Tyr Phe 1060 1065 1070 Asn Leu Phe Asp Lys Glu Leu Asn Giu Lys Giu Ile Lys Asp Leu Tyr *1075 1060 1085 Asp Asn Gir. Ser Asn Ser Gly Ile Leu Lys Asp Phe Trp Gly Asp Tyr 1090 1095 1100 *Leu Gin Tyr Asp Lys Pro Tyr Tyr Met Leu Asn Leu Tyr Asp Pro Asn *1105 1110 1115 1120 *Lys Tyr Val Asp Val Asn Asn Val Gly Ile Arg Gly Tyr Met Tyr Leu 1125 1130 1135 *Lys Gly Pro Arg Gly Ser Vai Met Thr Thr Asn Ile Tyr Leu Asn Ser *1140 1145 1150 Ser Leu Tyr Arg Gly Thr Lys Phe Ile Ilie Lys Lys Tyr Ala Ser Gly 1155 1160 1165 ***Asn Lys Asp Asn Ilie Val Arg Asn Asn Asp Arg Val Tyr Ile Asn Val *1170 1175 1180 Val Val Lys Asn Lys Glu Tyr Arg Leu Ala Thr Asn Ala Ser Gin Ala 1185 1190 1195 1200 Gly Val Giu Lys Ile Leu Ser Ala Leu Giu Ile Pro Asp Val Gly Asn 1205 1210 1215 Leu Ser Gin Val Val Val Met Lys Ser Lys Asn Asp Gin Gly Ile Thr 1220 1225 11230 Asn Lys Cys LYS Met Asn Leu Gin Asp Asn Asn Gly Asn Asp Ile Gly 1235 1240 1245 Phe Ile Gly Phe His Gin Phe Asn Asn Ile Ala Lys Leu Val Ala Ser 1250 1255 1260 Asn Trp Tyr Asfl Arg Gin Ilie Glu Arg Ser Ser Arg Thr Leu Gly Cys 1265 1270 1275 1280 Ser Tr-p Glu Phe Ile Pro Val Asp Asp Gly Trp Gly Giu Arg Pro Leu 1285 1290 1295 INFORMATION FOR SEQ ID NO:29: SEQUENCE
CHARACTEISTICS:
LENGTH: 812 amino acids -353- (ii) (xi) Thr 1 TYPE: amino acid STRANDEDNESS: unknown TOPOLOGY: linear MOLECULE TYPE: protein SEQUENCE DESCRIPTION: SEQ ID NO:29: Ser Tyr Lys Ile Ile Asn Gly Lys His Phe Tyr Phe Asn 5 10 Asn Asp is Gly Val Met Phe Ala Pro Val Tyr Gln s0 Asp Asn Asn Lys Tryr Tyr Val Ile Asp Ser Lys Gly 115 Asp Thr Ala 130 Phe Tyr Phe 145 Gin3 Ala Ser Ser Phe As n 100 Tr-p Ile Asp Thr Phe Ala 70 Pro Lys Thr Phe Asp 150 Asn Asn Leu Asn Gly Trp Ala Ile 90 Phe Asn 105 Gly Ser Tyr Lys Vai Lys Glu Lys Ile Al1a Asp Tyr Ile 140 Gly Thr Ala Ile Tyr Phe Asn Glu Leu Gin Ile Ile Asp Thr Lys His Ser Thr 160 Asn Asn 175 Leu Gly Val Phe Lys Gly Pro Asp Gly Phe Giu Tyr Ser Asn Gly Phe Giu Tyr Phe Ala Pro Ala A-sn 170 Ile Gly Gin Ala 225 Thr Lys Ile Ile Asr 305 Gli Giu Gly Gin Ala Ile Val Tyr 180 Lys Lys Tyr Tyr Phe Asp Asn 195 200 Thr Ile Asp Ser Lys Lys Tyr 210 215 Ala Thr Gly Trp Gin Thr Ilie 230 Asn Thr Ala Giu Ala Ala Thr 245 Tyr Tyr Phe Asn Thr Asn Thr 260 Ilie Asn Gly Lys His Phe Tyr 275 280 Gly Val Phe Lys Gly Pro Asn 290 295 Thr Asp Ala A-sn Asn Ile Giu 310 Phe Leu Thr Leu Asn Gly Lys 325 Gln 185 A-sn Tyr Asp Gly Ala 265 Phe Gly Gly Lys Ser Lys Phe Ser Lys Ala Phe Asn Thr 220 Gly Lys Lys 235 Trp Gin Thr 250 Ile Ala Ser Asn Thr Asp Phe Giu Tyr 300 *Gin Ala Ile 315 Tyr Tyr Phe 330 Leu Val 205 A-sn Tyr Ile Thr Gly 285 Phe Leu Gly Thr 190 Thr Thr Tyr Asp Giy 270 Ile Ala Tyr Ser Gly Ala Phe Gly 255 Tyr Met Pro *Gin *Asp 335 Leu Giu Asn 240 Lys Thr Gin Ala Asn 320 Ser 354 Lys Ala Val Thr Gly Trp Arg Ile Ile Asn Asn Lys Lys Tyr Tyr Phe 340 345 350 Asn Pro Asn Asn Ala Ile Ala Ala Ile His Leu Cys Asp Thr 365 Met Pro Gin Asp Tyr 465 Asp 355 Lys Tyr 370 Ile Giu Vai Thr Ala Asn Asn Lys 435 Ser Lys 450 Phe Asn Gly Lys Arg Gly Thr 420 Phe Al a Leu Lys Ser Asn 390 Phe Asn Thr Thr Thr 470 Tyr Asp Tyr Gly Asn Asn 440 Trp Giu Asn Gly Ile Phe Asp Pro Asn 410 Ile Giu 425 Gly Lys Gin Thr Ala Ala Leu Asn 490 Gin 380 Asn Phe Gin Tyr Asp 460 Gly Al a Tyr Ile Ser Lys 400 Phe Ala 415 Val Tyr Asp Asn Lys Tyr Thr Ile 480 Ala Thr 495 Gly Trp Gin Thr Ilie Asp Gly Lys Lys Tyr Tyr Phe Asn Thr Asn Thr 500 505 510 Phe Ile Ala Ser Thr Gly 515 Phe Asn Thr Asp Gly Ilie 530 Gly Phe Giu Tyr Phe Ala 545 550 Gly Gin Ala Ile Leu Tyr 565 Lys Tyr Tyr Phe Gly Ser 580 Ile Asp Gly Lys Lys Tyr 595 Thr Gly Trp Gin Thr Ile 610 Thr Ser Ile Ala Ser Thi 625 63( Tyr Phe Asn Thr Asp Glj 645 Asp Gly Phe Giu Tyr Phi 660 Glu Gly Gin Ala Ile Ari, 675 Asn Ilie Tyr Tyr Phe GI' 690 Thr Ile Asp Gly Asn Ar Tyr Thr S 520 Met Gin I Pro Ala Gin'Asfl I Asp Ser Tyr Phe2 600 Asn Gly 615 Gly Tyr Sle Met Ala Pro ;Tyr Gin 680 y Asfl Asn 695 g Tyr Tyr 355 le Gly Lsn Thr ~ys Phe 570 L.ys Ala 585 Asn Thr Lys Lys Thr Ile Gin Ile 650 Ala Asn 665 Asn Ar5 Ser Lyl Phe GIl 525 Val Phe Lys Gly Pro Asn 540 Asp Ala Asn Asn Ile Giu 555 560 Leu Thr Leu Asn Giy Lys 575 Val Thr Gly Leu Arg Thr 590 Asn Thr Ala Val Ala Val 605 Tyr Tyr Phe Asn Thr Asn 620 Ile Ser Giy Lys His Phe 635 640 Gly Val Phe Lys Gly Pro 655 Thr Asp Ala Asn Asn Ile 670 Phe Leu Tyr Leu His Asp 685 Ala Ala Thr Gly Trp Val 700 u Pro Asn Thr Ala Met Gly 705 710 715 720 Ala Asn Gly Tyr Lys Thr Ile Asp Asn Lys Asn Phe Tyr Phe Arg Asn 725 730 735 Gly Leu Pro Gin Ile Gly Val Phe Lys Gly Ser Asn Gly Phe Glu Tyr 740 745 750 Phe Ala Pro Ala Asn Thr Asp Ala Asn Asn Ile Glu Gly Gin Ala Ile 755 760 765 Arg Tyr Gin Asn Arg Phe Leu His Leu Leu Gly Lys Ile Tyr Tlyr Phe 770 775 780 Gly Asn Asn Ser Lys Ala Vai Thr Gly Trp Gin Thr Ile Asn Gly Lys 785 790 795 B00 Val Tyr Tyr Phe Met Pro Asp Thr Ala Met Ala Ala ~*805 810 INFORMATION FOR SEQ ID SEQUENCE
CHARACTERISTICS:
LENGTH: 609 amino acids TYPE: amino acid STRANDEDNESS: unknown TOPOLOGY: linear (ii) MOLECULE TYPE: protein SEQUENCE DESCRIPTION: SEQ ID Thr Ser Giu Glu Asn Lys Val Ser Gin Val Lys Ile Arg Phe Val Asn 1 5 10 Val Phe Lys Asp Lys Thr Leu Ala Asn Lys Leu Ser Phe Asn Phe Ser 25 Asp Lys Gin Asp Val Pro Val Ser Giu Ile Ile Leu Ser Phe Thr Pro 40 Ser Tyr Tyr Giu Asp Gly Leu Ile Gly Tyr Asp Leu Gly Leu Val Ser 55' *Leu Tyr Asn Glu Lys Phe Tyr Ile Asn Asn Phe Gly Met Met Val Ser 65 70 75 Gly Leu Ile Tyr Ilie Asn Asp Ser LeU Tyr Tyr Phe Lys Pro Pro Val 85 90 Asn Asm Leu Ile Thr Gly Phe Val Thr Val Gly Asp Asp Lys Tyr Tyr 100 105 110 Phe Asn Pro Ile Asn Gly Gly Ala Ala Ser Ile Gly Giu Thr Ile Ile 115 120 125 Asp Asp Lys Asn Tyr Tyr Phe Asn Gin Ser Gly Val Leu Gin Thr Giy 130 135 140 Val Phe Ser Thr Giu Asp Gly Phe Lys Tyr Phe Ala Pro Ala Asn Thr 145 150 155 160 Leu Asp Giu Asn Leu Glu Gly Giu Ala Ile Asp Phe Thr Gly Lys Leu 165 170 175 3,56
C
a a C a Ile Ile Asp Glu Asn Ile Tyr Tyr 180 Val Glu Trp Lys Glu Leu Asp Gly 195 200 Thr Gly Lys Ala Phe Lys Gly Leu 210 215 Tyr Phe Asn Ser Asp Gly Val Met 225 230 Asp Asn Lys His Tyr Phe Asp Asp 245 Thr Glu Ile Asp Gly Lys His Phe 260 Gin Ile Gly Val Phe Asn Thr Glu 275 280 His Asn Glu Asp Leu Gly Asn Glu 290 295 Gly Ile Leu Asn Phe Asn Asn Lys 305 310 Thr Ala Val Val Gly Trp Lys Asp 325 Phe Asp Glu Asp Thr Ala Glu Ala 340 Asp Gly Gin Tyr Tyr Phe Asn Asp 355 360 Val Thr Ile Asn Asp Lys Val Phe 370 375 Glu Ser Gly Val Gin Asn Ile Asp 385 390 Asn Gly Ile Val Gin Ile Gly Val 405 Tyr Phe Ala Pro Ala Asn Thr Val 420 Val Glu Tyr Ser Gly Leu Val Ar 435 441 Gly Glu Thr Tyr Thr Ile Glu Th 450 455 Glu Ser Asp Lys Tyr Tyr Phe As 465 470 Gly Ile Asn Leu Ile Asp Asp Ii 485 Ile Met Arg Thr Gly Leu Ile Se 500 Phe Asp Asp Asn Tyr Arg Gly Ala 185 190 Glu Met His Tyr Phe Ser Pro Glu 205 Asn Gin Ile Gly Asp Tyr Lys Tyr 220 Gin Lys Gly Phe Val Ser Ile Asn 235 240 Ser Gly Val Met Lys Val Gly Tyr 250 255 Tyr Phe Ala Glu Asn Gly Glu Met 265 270 Asp Gly Phe Lys Tyr Phe Ala His 285 Glu Gly Glu Glu Ile Ser Tyr Ser 300 Ile Tyr Tyr Phe Asp Asp Ser Phe 315 320 Leu Glu Asp Gly Ser Lys Tyr Tyr 330 335 Tyr Ile Gly Leu Ser Leu Ile Asn 345 350 Asp Gly Ile Met Gin Val Gly Phe 365 Tyr Phe Ser Asp Ser Gly Ile Ile 380 Asp Asn Tyr Phe Tyr Ile Asp Asp 395 400 Phe Asp Thr Ser Asp Gly Tyr Lys 410 415 L Asn Asp Asn Ile Tyr Gly Gin Ala 425 430 3 Val Gly Glu Asp Val Tyr Tyr Phe 3 445 r Gly Trp Ile Tyr Asp Met Glu Asn 460 n Pro Glu Thr Lys Lys Ala Cys Lys 475 480 e Lys Tyr Tyr Phe Asp Glu Lys Gly 490 495 r Phe Glu Asn Asn Asn Tyr Tyr Phe 505 510 357 Asn Giu Asn Gly Giu Met Gin Phe Gly Tyr Ile Asn 515 520 Met Phe Tyr Phe Gly Glu Asp Gly Val Met Gin Ile 530 535 540 Thr Pro Asp Gly Phe Lys Tyr Phe Ala His Gin Asn 545 550 555 Asn Phe Giu Gly Giu Ser Ile Asn Tyr Thr Gly Trp, 565 570 Giu Lys Arg Tyr Tyr Phe Thr Asp Giu Tyr Ile Ala 580 585 Val Ile Ile Asp Giy Giu Giu Tyr Tyr Phe Asp Pro 595 600 .Leu Ile Giu Asp Lys 525 Gly Val Phe Asn Thr Leu Asp Giu 560 Leu Asp Leu Asp 575 Ala Thr Giy Ser 590 Asp Thr Ala Gin 605 358

Claims (9)

1. A recombinant Clostridium botulinum toxin derived from the cleavage of a soluble, recombinant Clostridium botulinum toxin fusion protein.
2. A recombinant Clostridium botulinum toxin protein derived from the cleavage of a soluble, recombinant Clostridium botulinum toxin protein or portion thereof, capable of eliciting an immune response, fused to a non-toxin protein sequence.
3. A soluble fusion protein comprising: a portion of a Clostridium botulinum toxin capable of eliciting an immune response, and; a non-toxin protein.
4. A soluble fusion protein comprising: a Clostridium botulinum toxin, and; a non-toxin protein.
5. The soluble fusion protein of claim 4 comprising SEQ ID NO. 26.
6. The soluble fusion protein of claim 4 wherein the Clostridium botulinum toxin comprises SEQ ID NO. 28.
7. The soluble fusion protein of claim 3 wherein the portion of the Clostridium botulinum toxin comprises SEQ ID NO. 23.
8. A recombinant Clostridium botulinum toxin derived from the cleavage of a soluble, recombinant Clostridium botulinum toxin fusion protein as claimed in any one of claims 3 to 7.
9. A soluble, recombinant Clostridium botulinum toxin substantially as hereinbefore described with reference to any one of Examples 22 to 27. A soluble, recombinant protein comprising a portion of a Clostridium S 25 botulinum toxin substantially as hereinbefore described with reference to any one of Examples 22 to 27. Dated 20 January, 2003 Allergan, Inc. Allergan Botox Limited Patent Attorneys for the Applicants/Nominated Persons SPRUSON FERGUSON [I:\DayLib\LIBVV]02862specidoc:sxc
AU63043/99A 1994-10-24 1999-12-02 Soluble recombinant botulinum toxin proteins Ceased AU758820B2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
AU63043/99A AU758820B2 (en) 1994-10-24 1999-12-02 Soluble recombinant botulinum toxin proteins
AU2002317523A AU2002317523B2 (en) 1994-10-24 2002-12-09 Vaccine for Clostridium Botulinum Neurotoxin

Applications Claiming Priority (6)

Application Number Priority Date Filing Date Title
US08/329154 1994-10-24
US08/405496 1995-03-16
US08/422711 1995-04-14
US08/480604 1995-06-07
AU39683/95A AU709586B2 (en) 1994-10-24 1995-10-23 Vaccine and antitoxin for treatment and prevention of C. difficile disease
AU63043/99A AU758820B2 (en) 1994-10-24 1999-12-02 Soluble recombinant botulinum toxin proteins

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
AU39683/95A Division AU709586B2 (en) 1994-10-24 1995-10-23 Vaccine and antitoxin for treatment and prevention of C. difficile disease

Related Child Applications (1)

Application Number Title Priority Date Filing Date
AU2002317523A Division AU2002317523B2 (en) 1994-10-24 2002-12-09 Vaccine for Clostridium Botulinum Neurotoxin

Publications (2)

Publication Number Publication Date
AU6304399A AU6304399A (en) 2000-05-11
AU758820B2 true AU758820B2 (en) 2003-04-03

Family

ID=3726762

Family Applications (2)

Application Number Title Priority Date Filing Date
AU48763/99A Ceased AU747841B2 (en) 1994-10-24 1999-09-16 Vaccine and antitoxin for treatment and prevention of C. difficile disease
AU63043/99A Ceased AU758820B2 (en) 1994-10-24 1999-12-02 Soluble recombinant botulinum toxin proteins

Family Applications Before (1)

Application Number Title Priority Date Filing Date
AU48763/99A Ceased AU747841B2 (en) 1994-10-24 1999-09-16 Vaccine and antitoxin for treatment and prevention of C. difficile disease

Country Status (1)

Country Link
AU (2) AU747841B2 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11175293B1 (en) 2021-01-04 2021-11-16 University Of Utah Research Foundation Rapid assay for detection of SARS-CoV-2 antibodies

Also Published As

Publication number Publication date
AU747841B2 (en) 2002-05-23
AU6304399A (en) 2000-05-11
AU4876399A (en) 1999-11-25

Similar Documents

Publication Publication Date Title
AU709586B2 (en) Vaccine and antitoxin for treatment and prevention of C. difficile disease
US6613329B1 (en) Vaccine and antitoxin for treatment and prevention of C. difficile disease
US6967088B1 (en) Soluble recombinant botulinum toxin proteins
US5919665A (en) Vaccine for clostridium botulinum neurotoxin
US5814477A (en) Recombinant clostridial toxin protein
EP1105153A1 (en) Multivalent vaccine for (clostridium botulinum) neurotoxin
AU758820B2 (en) Soluble recombinant botulinum toxin proteins
AU2002317523B2 (en) Vaccine for Clostridium Botulinum Neurotoxin
KR100497700B1 (en) Soluble Recombinant Botulinum Toxin Proteins
CA2416318A1 (en) Soluble, recombinant botulinum toxin proteins
AU2521600A (en) Treatment for verotoxin-producing escherichia coli

Legal Events

Date Code Title Description
PC1 Assignment before grant (sect. 113)

Owner name: ALLERGAN, INC.

Free format text: THE FORMER OWNER WAS: OPHIDIAN PHARMACEUTICALS, INC.

TH Corrigenda

Free format text: IN VOL 16, NO 12, PAGE(S) 2713-2713 UNDER THE HEADING ASSIGNMENTS BEFORE GRANT, SECTION 113 THE NAME OF THE ASSIGNEE IN REGARD TO PATENT APPLICATION NO. 63043/99 SHOULD READ: ALLERGAN INC. AND ALLERGAN BOTOX LIMITED AND THEIR ADDRESSES TO READ: 2525 DUPONT DRIVE, IRVINE, CA 92612,USA

FGA Letters patent sealed or granted (standard patent)
DA3 Amendments made section 104

Free format text: THE NATURE OF THE AMENDMENT IS: AMEND THE TITLE OF THE INVENTION TO READ SOLUBLE RECOMBINANT BOTULINUM TOXIN PROTEINS