US20080107673A1 - Mutants of clostridium difficile toxin B and methods of use - Google Patents

Mutants of clostridium difficile toxin B and methods of use Download PDF

Info

Publication number
US20080107673A1
US20080107673A1 US11/809,970 US80997007A US2008107673A1 US 20080107673 A1 US20080107673 A1 US 20080107673A1 US 80997007 A US80997007 A US 80997007A US 2008107673 A1 US2008107673 A1 US 2008107673A1
Authority
US
United States
Prior art keywords
tcdb
toxin
mutant
antibody
clostridium difficile
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US11/809,970
Inventor
Jimmy D. Ballard
Lea M. Spyres
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.)
Individual
Original Assignee
Individual
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Individual filed Critical Individual
Priority to US11/809,970 priority Critical patent/US20080107673A1/en
Publication of US20080107673A1 publication Critical patent/US20080107673A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/569Immunoassay; Biospecific binding assay; Materials therefor for microorganisms, e.g. protozoa, bacteria, viruses
    • G01N33/56911Bacteria
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/33Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Clostridium (G)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies

Definitions

  • Intracellular bacterial toxins enter cells, modify targets, and in many cases ultimately destroy the targeted cells thereby contributing to the disease process.
  • TcdA and TcdB are two large clostridial toxins (LCTs) produced by C. difficile and are involved in development of pseudomembranous colitis.
  • TcdB (SEQ ID NO: 1), the focus of this study, glucosylates isoforms of small GTPases Rho, Rac and Cdc42 within the effector binding region at residues Threonine-37 (Rho) or Threonine-35 (Rac and Cdc42).
  • the physiological impact of TcdB's activity includes disruption of tight junctions, increased epithelial permeability, as well as actin condensation and cell death.
  • TcdB can be divided into enzymatic, translocation and receptor binding domains, although detailed analysis of these regions has not been carried out to date.
  • the first 546 amino acids of TcdB contain the enzymatic region, which is followed by a putative translocation and receptor-binding domain. Enzymatic activity appears to require the amino-terminal 546 residues since amino or carboxy terminal deletions of this fragment decrease activity.
  • tryptophan 102 has been shown to be essential for UDP-glucose binding.
  • a conserved DXD motif within LCTs is essential for LCT glucosyltransferase activity.
  • Other studies, involving analysis of chimeras of the TcdB and TcsL enzymatic domain suggest residues 364 to 516 confer substrate specificity.
  • Steps in cell entry by TcdB have been broadly defined, yet events subsequent to entry are not well understood. For example, while we have a profile of the time-course for TcdB cell entry, very little is known about post-entry events that lead to glucosylation. Steps between membrane translocation and substrate interaction are not understood in TcdB intoxication. In fact almost no information exists in this regard for any intracellular toxin. In the cytosol, TcdB is capable of glucosylating multiple substrates, but whether inactivation of Rho, Rac and Cdc42 in combination is necessary for complete intoxication, or if other substrates are targeted, is not known.
  • Rho has been found that overexpression of Rho isoforms protects cells from TcdB, suggesting inactivation of all substrates may not be necessary for cellular intoxication.
  • Rho has also been shown to regulate the suppression of apoptosis, so it is not entirely clear whether overexpression of Rho is protective at the substrate inactivation level or prevents events downstream of glucosylation.
  • TcdB-intoxicating events such as depolymerization of actin, can be attributed to inactivation of Rho, other processes like apoptosis may be linked to activities other than substrate inactivation. Given TcdB's large size ( ⁇ 270 kD), and broad impact on cell physiology, it is possible the toxin may possess yet undefined activities in addition to glucosylation.
  • the invention herein contemplates, in one embodiment, a mutant of native C. difficile TcdB toxin polypeptide wherein the mutant is substituted at position 395, such that the cysteine at position 395 in the native TcdB toxin has been replaced with another amino acid, for example, a tryptophan residue and wherein the mutant is not cytotoxic (non-toxic).
  • the invention further comprises fragments of the TcdB toxin, which are effective in inhibiting TcdB toxin or are effective as a vaccine, and are non-toxic.
  • the invention further contemplates a vaccine generally applicable to the prevention or treatment of C. difficile disease. Additionally, the present invention contemplates a method of inhibiting, modulating, or treating a C. difficile or a C.
  • the present invention contemplates a monoclonal antibody raised against the C. difficile TcdB toxin mutant.
  • the present invention contemplates a method of making an antibody against C. difficile TcdB toxin comprising immunizing an animal with an immunogenic amount of the C. difficile TcdB toxin mutant.
  • FIG. 1 shows chromatography gels of LFnTcdB deletion and site-directed mutants.
  • Panel A Overview of deletion and site directed mutants. Deletion mutants were generated by PCR, cloned in-frame with lfn in pET15b, expressed in E. coli BL-21, and subsequently purified by Ni 2+ affinity chromatography. Site-directed mutants were generated by the Quick-change method, using complementary mutation encoding oligonucleotides, and pLMS200 as template.
  • Panel B DS-PAGE analysis of his-tagged fusions.
  • Lane 1 Molecular Weight Marker
  • Lane 2 TcdB W102A ; Lane 3, TcdB C395W ; Lane 4, TcdB C395S ; Lane 5, TcdB 35-556 ; Lane 6, TcdB 1-70 ; Lane 7, TcdB 1-420 ; Lane 8, TcdB 1-500 ; Lane 9, TcdB 1-556 ; Lane 10, Molecular Weight Marker.
  • FIG. 2 is a gel depicting glucosylation activity of deletion and site-directed mutants on RhoA, Rac1 and Cdc42. Each mutant and TcdB was tested for glucosylation activity on recombinant substrates GST-RhoA, GST-Rac1 and GST-Cdc42, using [ 14 C]UDP-Glucose as cosubstrate. Following a 2 h incubation, the reaction mix was resolved by SDS-PAGE and exposed to film for 48 h.
  • Lane 1 TcdB; Lane 2, TcdB W102A ; Lane 3, TcdB C395W ; Lane 4, TcdB C395S ; Lane 5, TcdB 1-556 ; Lane 6, TcdB 1-500 ; Lane 7, TcdB 1-420 ; Lane 8, TcdB 1-170 ; Lane 9, TcdB 35-556 .
  • FIG. 3 shows inhibition of TcdB cytopathic effects by TcdB mutants.
  • HeLa cells were cotreated with TcdB and each TcdB fusion plus PA. The cells were followed for 7 h and cytopathic effects were determined by visualization.
  • FIG. 4 is a graphical representation depicting sustained inhibition by supplemental treatments with inhibitor.
  • HeLa cells were cotreated with TcdB and TcdB 1-500 plus PA.
  • TcdB 1-500 and PA were added to the cells at 1 h intervals for 12 h. The cells were then followed for 30 h and visualized for cytopathic effects.
  • FIG. 5 is a graphical representation depicting the protection of CHO cells expressing TcdB 1-556 .
  • FIG. 6 is a chart demonstrating the inhibitory effects following inhibitor treatments prior to or following treatment with TcdB.
  • TcdB 1-500 plus PA at time points prior to or following treatment with TcdB.
  • Cells were amended with inhibitor every 30′ and observed for cytopathic effects at 8 h following toxin treatment.
  • FIG. 7 is a graphical representation depicting TcdB 1-500 inhibition of TcsL cytopathic effects.
  • HeLa cells were treated with TcdB 1-500 plus PA for 30 min prior to treatment with TcsL.
  • FIG. 8 Differential glucosylation of extracts from cells treated with TcdB plus inhibitor.
  • HeLa cells were plated in T-25 flasks and grown until semiconfluent, then treated with PA, TcdB 1-500 and TcdB was added to the cells.
  • the invention contemplated herein comprises, in a preferred embodiment, non-cytotoxic C. difficile TcdB toxin derivatives and deletions (mutants) which are deficient in at least one specific function required for toxicity and which are effective intracellular inhibitors of native TcdB toxin or are effective in producing immunity against TcdB toxin.
  • the present invention demonstrates that enzymatically inactive fragments of the TcdB enzymatic domain are effective intracellular inhibitors of native TcdB.
  • the present invention comprises purified derivatives (mutants) of C. difficile TcdB toxin which are deficient in glucosyltransferase and glucosylhydrolase activity. The mutants are considered to be useful as a vaccine for both humans and animals.
  • animals which may be treated are cattle, chickens, turkeys, ostriches, emu, ducks, horses, donkeys, mules, pigs, sheep, goats antelope, buffalo, llamas, cats, lions, tigers, dogs, bears, guinea pigs, hamsters, chinchillas, mink, ferrets, rodents, parrots, parakeets, peacocks, seals, sea lions, orcas, monkeys, chimpanzees, baboons, orangutans, gorillas, reptiles, and other zoo and livestock animals.
  • mutant refers to a fragment, point deletion, point substitution, or a deletion of multiple residues of the TcdB protein sequence, and may be encoded by a nucleotide sequence intentionally made variant from a native sequence.
  • present invention also contemplates nucleotide sequences which encode the mutants.
  • mutants of the present invention preferably have at least one substituted amino acid in the enzymatic domain of the TcdB toxin which includes amino acid position 395 of the sequence of the native TcdB toxin as shown in SEQ ID NO: 1.
  • novel mutants contemplated herein comprise at least one amino acid substitution or deletion of the native C. difficile TcdB toxin.
  • the amino acid at position 395 also referred to herein as the “critical position” of the amino acid sequence of the native C. difficile TcdB toxin (SEQ ID NO: 1) may be substituted with a different amino acid in the same position.
  • the invention comprises mutants wherein the native cysteine at position 395 has been substituted with a tryptophan residue at position 395.
  • any amino acid residue which would provide a mutant effective in inhibiting TcdB toxin, and which is not cytotoxic may be substituted for the cysteine residue at position 395.
  • other amino acids which may be used to substitute the cysteine residue include alanine, valine, leucine, isoleucine, proline, methionine, phenylalanine, glycine, threonine, tyrosine, asparagine, aspartic acid, glutamine, glutamic acid, lysine, arginine, and histidine.
  • Mutants comprising deletions of portions of the enzymatic domain include, for example, a modified C. difficile TcdB toxin having a deletion of amino acid positions 501-556 (SEQ ID NO: 3), 421-556 (SEQ ID NO: 5), 171-556 (SEQ ID NO: 7), or 1-34 (SEQ ID NO: 9) are also contemplated.
  • An especially preferred embodiment comprises a mutant having at least one substitution in the enzymatic domain.
  • mutants of the present invention preferably have deficient glucosyltransferase and glucosylhydrolase activity compared to the native C. difficile TcdB toxin, and are non-toxic, and in an especially preferred embodiment are antigenic, whereby vaccines produced from them induce anti-TcdB toxin antibodies in vivo as explained in more detail below.
  • novel vaccines comprising the TcdB toxin mutants described herein, or antigenic fragments thereof, which when administered to animals or humans, are capable of inducing production of protective antibodies directed against C. difficile TcdB toxin, thereby providing prophylaxis against infection by C. difficile disease states resulting from such infection, and/or from the TcdB toxin itself. It is a particular aim of the present invention to provide such a vaccine that is relatively safe and simple to produce. Antibodies and antisera raised against the mutants are also capable of use in therapy for at least some, if not all, disease states, in which TcdB toxin is involved.
  • recombinant DNA which encode any proteins, fragments, or amino acid sequences thereof described or claimed herein.
  • recombinant DNA is conveniently provided by PCR amplification of the DNA encoding for the desired sequence, using primers targeted at respective ends of the double stranded sequence of which it forms one half, using methods well known to those of ordinary skill in the art.
  • antisera raised to the mutants, or antigenic fragments thereof, of the invention and antibodies derived therefrom.
  • the present invention provides monoclonal antibodies against the mutants, or antigenic fragments thereof, of the invention and hybridoma cells for production thereof as described in more detail below.
  • the present invention further contemplates TcdB toxin mutants which have additional substitutions which are merely conservative substitutions of amino acids.
  • conservative substitution is meant the substitution of an amino acid by another one of the same class; the classes according to Table I.
  • altering any given non-critical amino acid of a protein by conservative substitution may not significantly alter the activity of that protein because the side-chain of the amino acid which is inserted into the sequence may be able to form similar bonds and contacts as the side chain of the amino acid which has been substituted for.
  • Non-conservative substitutions are possible provided that these do not excessively affect the immunogenicity of the polypeptide and/or reduce its effectiveness in inhibiting TcdB toxin.
  • polypeptides of the invention may be prepared synthetically, or more suitable, they are obtained using recombinant DNA technology.
  • the invention further provides a nucleic acid which encodes any of the mutants of TcdB toxin which have at least one substitution and/or deletion as described herein.
  • nucleic acids may be incorporated into an expression vector, such as a plasmid, under the control of a promoter as understood in the art.
  • the vector may include other structures as conventional in the art, such as signal sequences, leader sequences and enhancers, and can be used to transform a host cell, for example a prokaryotic cell such as E coli or a eukaryotic cell. Transformed cells can then be cultured and polypeptide of the invention recovered therefrom, either from the cells or from the culture medium, depending upon whether the desired product is secreted from the cell or not.
  • the terms “complementary” or “complementarity” are used in reference to polynucleotides (i.e., a sequence of nucleotides) related by the base-pairing rules. For example, for the sequence “A-G-T,” is complementary to the sequence “T-C-A.” Complementary may be “partial,” in which only some of the nucleic acids' bases are matched according to the base pairing rules. Or, there may be “complete” or “total” complementary between the nucleic acids. The degree of complementary between nucleic acid strands has significant effects on the efficiency and strength of hybridization between nucleic acid strands. This is of particular importance in amplification reactions, as well as detection methods which depend upon binding between nucleic acids.
  • Nucleic acids of the present invention also include DNA sequences which hybridize to the DNA sequences which encode the mutant polypeptides described herein, or their complementary sequences, under conditions of high or low stringency and which encode proteins having activity against TcdB toxin and/or which preferably can stimulate antibodies against native TcdB toxin.
  • Hybridization and washing conditions are well known and exemplified in Sambrook, J., Fritsch, E. F. and Maniatis, T. Molecular Cloning: A Laboratory Manual, Second Edition, Cold Spring Harbor Laboratory Press, Cold Spring Harbor (1989), particularly Chapter 11 and Table 11.1 therein (expressly entirely incorporated herein by reference).
  • the conditions of temperature and ionic strength determine the “stringency” of the hybridization.
  • high stringency conditions are prehybridization and hybridization at 68° C., washing twice with 0.1 ⁇ SSC, 0.1% SDS for 20 minutes at 22° C. and twice with 0.1 ⁇ SSC, 0.1% SDS for 20 minutes at 50° C. Hybridization is preferably overnight.
  • low stringency conditions comprise conditions equivalent to binding or hybridization at 42° C. in a solution consisting of 5 ⁇ SSPE (43.8 g/l NaCl, 6.9 g/l NaH 2 PO 4 .H 2 O and 1.85 g/l EDTA, pH adjusted to 7.4 with NaOH), 0.1% SDS, 5 ⁇ Denhardt's reagent [50 ⁇ Denhardt's contains per 500 ml: 5 g Ficoll (Type 400, Pharmacia), 5 g BSA (Fraction V; sigma) and 100 ⁇ g/ml denatured salmon sperm DNA] followed by washing in a solution comprising 5 x SSPE, 0 . 1 % SDS at 42° C. when a probe of about 500 nucleotides in length is employed.
  • 5 ⁇ SSPE 43.8 g/l NaCl, 6.9 g/l NaH 2 PO 4 .H 2 O and 1.85 g/l EDTA, pH adjusted to 7.4 with NaOH
  • low stringency conditions are prehybridization and hybridization at 68° C., washing twice with 2 ⁇ SSC, 0.1% SDS for 5 minutes at 22° C., and twice with 0.2 ⁇ SSC, 0.1% SDS for 5 minutes at 22° C. Hybridization is preferably overnight.
  • very low to very high stringency conditions are defined as prehybridization and hybridization at 42° C. in 5 ⁇ SSPE, 0.3% SDS, 200 ug/ml sheared and denatured salmon sperm DNA, and either 25% formamide for very low and low stringencies, 35% formamide for medium and medium-high stringencies, or 50% formamide for high and very high stringencies, following standard Souther blotting procedures.
  • the carrier material is then washed three times each for 15 minutes using 2 ⁇ SSC, 0.2% SDS preferably at least 45° C. (very low stringency), more preferably at least at 50° C. (low stringency), more preferably at least at 55° C. (medium stringency), more preferably at least at 60° C. (medium-high stringency), even more preferably at least at 65° C. (high stringency), and most preferably at least at 70° C. (very high stringency).
  • low stringency conditions factors such as the length and nature (e.g., DNA, RNA, base composition) of the probe and nature of the target (e.g., DNA, RNA, base composition, present in solution or immobilized, etc.) And the concentration of the salts and other components (e.g., the presence or absence of formamide, dextran sulfate, polyethylene glycol) are considered and the hybridization solution may be varied to generate conditions of low stringency hybridization different form, but equivalent to, the above listed conditions.
  • conditions which promote hybridization under conditions of high stringency e.g., increasing the temperature of the hybridization and/or wash steps, the use of formamide in the hybridization solution, etc. are also know in the art.
  • substantially homologous refers to any probe which can hybridize to either or both strands of the double-stranded nucleic acid sequence under conditions of low stringency as described above.
  • substantially homologous refers to any probe which can hybridize (i.e., it is the complement of) the single-stranded nucleic acid sequence under conditions of low stringency as described above.
  • hybridization is used in reference to the pairing of complementary nucleic acids. Hybridization and the strength of hybridization (e.e., the strength of the association between the nucleic acids) is impacted by such factors as the degree of complementary between the nucleic acids, stringency of the conditions involved, the Tm (melting temperature) of the formed hybrid, and the G:C ration within the nucleic acids.
  • stringency is used in reference to the conditions of temperature, ionic strength, and the presence of other compounds such as organic solvents, under which nucleic acid hybridizations are conducted.
  • the terms “cell,” “cell line,” and “cell culture” are used interchangeably and all such designations include progeny.
  • the words “transformants” or “transformed cells” include the primary transformed cell and cultures derived from that cell without regard to the number of transfers. All progeny may not be precisely identical in DNA content, due to deliberate or inadvertent mutations. Mutant progeny that have the same functionality as screened for in the originally transformed cell are included in the definition of transformants.
  • vector is used in reference to nucleic acid molecules that transfer DNA segment(s) from one cell to another.
  • vehicle is sometimes used interchangeably with “vector”.
  • DNA vector refers to DNA sequences containing a desired coding sequence and appropriate DNA sequences necessary for the expression of the operably linked coding sequence in a particular host organism.
  • DNA sequences necessary for expression in prokaryotes include a promoter, optionally and operator sequence, a ribosome binding site and possibly other sequences.
  • Eukaryotic cells are known to utilize promoters, polyadenylation signals and enhancers. It is not intended that the term be limited to any particular type of vector.
  • vectors that remain autonomous within host cells e.g., plasmids
  • vectors that result in the integration of foreign e.g., recombinant nucleic acid sequences
  • expression vector and “recombinant expression vector” as used herein refers to a recombinant DNA molecule containing a desired coding sequence and appropriate nucleic acid sequences necessary for the expression of the operably linked coding sequence in a particular host organism.
  • Nucleic acid sequences necessary for expression in prokaryotes usually include a promoter, an operator (optional), and a ribosome binding site, often along with other sequences.
  • Eukaryotic cells are known to utilize promoters, enhancers, and termination and polyadenylation signals. It is contemplated that the present invention encompasses expression vectors that are integrated into host cell genomes, as well as vectors that remain unintegrated into the host genome.
  • operable combination refers to the linkage of nucleic acid sequences in such a manner that a nucleic acid molecule capable of directing the transcription of a given gene and/or the synthesis of a desired protein molecule is produced.
  • the term also refers to the linkage of amino acid sequences in such a manner so that a functional protein is produced.
  • the mutants described herein may be expressed in either prokaryotic or eukaryotic host cells.
  • Nucleic acids encoding the mutants may be introduced into bacterial host cells by a number of means including transformation or transfection of bacterial cells made competent for transformation by treatment with calcium chloride or by electroporation.
  • nucleic acid encoding the protein or toxin of interest may be introduced into eukaryotic host cells by a number of means including calcium phosphate co-precipitation, spheroplast fusion, electroporation, microinjection, lipofection, protoplast fusion, and retroviral infection, for example.
  • transformation may be affected by treatment of the host cells with lithium acetate or by electroporation, for example.
  • the mutant is characterized as having 50% or less of the glucosyltransferase and glucoslyhydrolase activity of wild type TcdB toxin, as measured by assays described herein. In a more preferred version of the invention, the mutant is characterized as having 30% or less of the glucosyltransferase and glucosylhydrolase activity of wild type TcdB toxin, as measured by assays described herein. In a more preferred version of the invention, the mutant has less than 20% of the glucosyltransferase and glucosylhydrolase activity of wild type TcdB toxin as measured by assays described herein.
  • the mutant has less than 10% of the glucoslytransferase and glucosylhydrolase activity of wild-type TcdB toxin, as measured by assays described herein. More preferably, the mutant has less than 5% of the glucosyltransferase and glucosylhydrolase activity of the wild-type TcdB toxin, as measured by assays described herein. Even more preferably, the mutant has less than 0% of the glucosyltransferase and glucosylhydrolase activity of wild-type TcdB toxin, as measured by assays described herein. More particularly, the invention as contemplated herein is a mutant (mutein) of C.
  • TcdB 1-556 (SEQ ID NO: 1) could be inactivated by n-ethylmaleimide (data not shown), indicating a role for the sole cysteine (position 395) in enzymatic activity, thus site-directed mutants TcdB C395S , TcdB C395W were produced. Amino-terminal and carboxy-terminal deletions were also generated in an attempt to further identify inactive mutants. Since these mutants lacked receptor binding and translocation domains, the fragments were fused to the cell entry proteins of anthrax lethal toxin.
  • This anthrax toxin derivative consists of anthrax protective antigen (PA), and a truncated form of anthrax lethal toxin (LFn), to which heterologous fusions are made.
  • PA-LFn has been used by several groups for the cytosolic delivery of a variety of proteins, and we previously used this system to deliver TcdB 1-556 to cultured mammalian cells [Spyres, 2001]. Using this delivery system, the fusions were tested for cytopathic activity and only TcdB 1-556 and TcdB C395S were cytotoxic (data not shown).
  • mutants were tested for glucosylation of RhoA, Rac1 or Cdc42. As shown in FIG. 2 , only TcdB 1-556 , and TcdB C395S glucosylated substrate. In line with earlier reports carboxy-terminal deletions and TcdB W102A were unable to glucosylate substrate. The remainder of the site-directed and deletion mutants were also deficient in glucosylation. Furthermore, this loss of activity was maintained across all of the shared substrates since these same mutants were attenuated in glucosylation of RhoA, Rac1 and Cdc42.
  • Each mutant was also analyzed for glucosylhydrolase activity using radiolabeled UDP-glucose in the absence of substrate. Fusions were incubated with [ 14 C]UDP-glucose and the liberated sugar was separated by anion-exchange chromatography. As shown in Table 1, even with extended (16 h) incubation glucosylhydrolase activity was significantly reduced for all enzymatically inactive mutants. Without wishing to be constrained by theory, the absence of substrate modification by these mutants could be accounted for, at least in part, by defective hydrolase activity.
  • TcdB inhibitors lack native translocation and receptor binding domains suggested that inhibition occurred within the cytosol. However, inhibition at the cell surface could not be formally excluded since cell surfacing-interacting regions of TcdB have not been fully elucidated.
  • a CHO cell line capable of inducible expression of TcdB 1-556 was generated.
  • a tightly regulated expression system, pSwitch was selected which allows expression only in the presence of the hormone mifepristone.
  • GeneSwitch-CHOpGene/TcdB 1-556 cells showed early toxic effects, such as rounding, at around 4 h following addition of mifepristone and were no longer viable by 24 h.
  • TcdB-Inhibitors as Tools to Dissect the Time-Course of Posttranslocation Events
  • TcsL which is closely related to TcdB, yet modifies a different set of Ras proteins including Ras, and Ra1.
  • TcsL does share Rac as a common substrate with TcdB.
  • TcdB inhibitor's ability to block TcsL intoxicaion In recent work we reported that acid pH enhances TcsL entry [Qa'Dan, 2001], so the initial treatment with TcsL was carried out by providing an extracellular acid pulse to TcsL.
  • TcdB 1-500 was also able to block the activity of TcsL. Similar to results with TcdB, the inhibitor was capable of reducing TcsL's cytopathic effects by almost 50%. These results suggested the TcdB inhibitor was blocking LCT intoxication events that might not be related to substrate targeting or that blocking a single target was sufficient to prevent toxic effects.
  • Attenuated mutants of TcdB inhibit wild-type toxin at an intracellular site. To our knowledge this is the first example of an approach that blocks the activity of an intracellular bacterial toxin within the cytosol of intoxicated cells. This inhibitory effect also suggests some yet undefined aspects of TcdB. Clearly, while unable to modify substrate, the mutants carry out functions within the cytosol, which allow inhibition. The exact mode of inhibition is not clear; however, the preliminary evidence indicates the inhibitor prevents glucosylation of a substrate other than Rho, Rac or Cdc42. This is a feasible possibility since some of the inhibitors do not encompass the region of TcdB reported to interact with Rho, Rac and Cdc42. Work by Hofmann et al.
  • TcdB The region encoding for the enzymatic domain of TcdB i.e., TcdB nucleotides 1-1668 (SEQ ID NO:2)) was genetically fused to lfn, cloned expressed and purified in E. coli as previously described [Spyres, 2001]. Using a similar approach, four other fusions of LFnTcdB were also constructed.
  • fragments encoding regions TcdB 1-500 (SEQ ID NO: 3 encoded by nucleotides 1-1500 (SEQ ID NO: 4)
  • TcdB 1-420 (SEQ ID NO: 5 encoded by nucleotides 1-1260 (SEQ ID NO: 6)
  • TcdB 1-170 (SEQ ID NO: 7 encoded by nucleotides 1-510 (SEQ ID NO: 8)
  • TcDB 35-556 SEQ IDNO: 9 encoded by nucleotides 103-1668 (SEQ ID NO: 10)
  • Plasmids were transformed into E. coli XL1-blue (Stratagene) and candidate clones were sequenced, then transformed into E. coli BL-21 Star (In
  • TcdB1-556 C395S Oligonucleotides for generation of TcdB1-556 C395S (SEQ ID NO: 11, where Xaa at position 395 is serine) were GTTTACTATTAAATTGCTAGAATATGAGTCTTTCACAG (sense) (SEQ ID NO: 13), CTGTGAAGACTCATATTCTAGCAATTTAATAGTAAAAC (antisense) (SEQ ID NO: 14); TcdB1-556 C395W (SEQ ID NO: 11, where Xaa at position 395 is tryptophan) GTTTTACTATTAAATTGCTACCTATGAGTCTTTCACAG (sense) (SEQ ID NO: 15), CTGTGAAAGACTCATATTGGAGCAATTTAATAGTAAAAC (antisense) (SEQ ID NO: 16); TcdB1-556 W102A (SEQ ID NO: 12) AAAAATTTACATTTTGTTGCT
  • LFnTcdB fusions were induced with 0.2 mM iso-propyl- ⁇ -D-thiogalactopyranoside in log phase (OD 600 0.8) cultures at 16° C.
  • Cells were harvested by centrifugation at 8700 ⁇ g, resuspended in binding buffer (5 mM imidazole, 500 mM NaCl, 20 mM Tris-HCl, pH7.9) supplemented with a protease inhibitor cocktail containing 4-(2-aminoethyl)benzenesulfonyl fluoride, phosphoramidon, pepstatin A, bestatin, and E-64 and lysed by sonication.
  • binding buffer 5 mM imidazole, 500 mM NaCl, 20 mM Tris-HCl, pH7.9
  • protease inhibitor cocktail containing 4-(2-aminoethyl)benzenesulfonyl fluoride, phosphoramidon, pe
  • LFnTcdB fusion proteins were isolated using nickel 900 cartridges following the manufacturer's instructions (Novagen). As a second purification step, proteins were fractionated on a high-resolution anion exchange (mono-Q) column (Amersham Pharmacia). Recombinant PA was isolated from E. coli BL-21, harboring the plasmid, pSRB/ET-15b-PA (a generous gift from Steven Blanke), as previously described [Whilhite, 1998]. TcdB and TcsL were purified as previously described [Qa'Dan, 2000]. Recombinant clones of RhoA, Rac1, and Cdc42 (a generous gift of Alan Hall) were expressed and purified as previously described [Spyres, 2001].
  • Glucosylation reactions were carried out as previously described [Spyres, 2001]. Glucosylhydrolase assays were carried out in a reaction mix containing 50 mM n-2hydroxyethylpiperazine-N′-2-ethane sulfonic acid, 100 mM KCl, 1 mM MnCl 2 , 1 mM MgCl 2 , 100 ⁇ g/ml BSA, 0.2 mM GDP, 40 ⁇ M [ 14 C]UDP-glucose (303Ci/mol; ICN Pharmaceuticals), 100 ⁇ M UDP-glucose and 3 pmol of TcdB or 10 pmol of each fusion protein. The assay was allowed to incubated overnight at 37° C. and similar to a previously described protocol [Ciesla, 1998], the cleaved glucose was separated using AG1-X2 anion exchange resin and counted in a liquid scintillation counter.
  • HeLa cells were plated in 96 well microtiter plates (3 ⁇ 10 4 cells/well) and allowed to incubate overnight. The following day the cells were treated with 30 pmol of each fusion plus 8.5 pmol of PA and observed for 48 h for signs of cytopathic effects. For inhibition assays, HeLa cells were plated as before and treated with 4 pmol of the appropriate LFnTcdB fusion plus 8.5 pmol of PA in a final volume of 100 ⁇ l. At the same point the cells were cotreated with 80 fmol of TcdB and observed for cytopathic effects.
  • TcsL competition cells were pretreated with TcdB 1-500 and PA for 30 min at which point cells were treated with TcsL via an acid pulse as previously described [Qa'Dan, 2001]. The cells were then amended with 30 pmol of TcdB 1-500 , 8.5 pmol of PA every 30 min up to 12 h and followed for 16 h.
  • HeLa cells semi-confluent (1 ⁇ 10 6 ) were first treated with 325 pmol of PA and 300 pmol of TcdB 1-500 followed by treatment with 50 fmol of TcdB in a final volume of 10 ml.
  • a DNA sequence coding for the enzymatic domain of TcdB (amino acids 1 to 556) placed downstream and in-frame with a Kozak sequence (GNNATGG) was cloned between the HindIII and EcoRI sites of plasmid pGene/V5-His version B (Invitrogen) multiple cloning site.
  • the recombinant plasmid was linearized with SapI and introduced into GeneSwitch-CHO cells (Invitrogen) by lipofection according to the protocol supplied with the LipofectAMINE PLUS Reagent Kit (Gibco Life Technologies).
  • Stably transfected cells were selected for on selective growth medium consisting of complete F-12 (HAM) medium plus zeocin (300 ⁇ g/ml) and hygromycin (100 ⁇ g/ml) by feeding the cells with selective medium every 3 to 4 days until foci could be seen.
  • Antibiotic resistant cells were treated with trypsin (0.25%) solution for 3 min, diluted with 5 volumes of complete F-12 (HAM) medium and harvested by centrifugation at 250 ⁇ g for 5 min. Cells were then resuspended in complete F-12 (HAM) medium, and diluted to a final cell density of five cells per ml. One hundred microliters of cell suspension was used to seed the wells of two 96-well plates.
  • TcdB was induced in the different cell lineages of transfected CHO cells by the addition of mifepristone (10 ⁇ 8 M), to the selective medium.
  • GeneSwitch-CHOpGene/TcdB1-556 a lineage of transfected cells showing nearly 100% rounding in 24 h in the presence of mifepristone was identified and chosen for the experiments reported herein.
  • Results were analyzed using the statistical software component of Excel 2001. Sample variations are reported as standard deviation from the mean, and significance was confirmed by student's T-test (p ⁇ 0.05).
  • mutants contemplated herein are inactive, and therefore are not lethal, but are effective in binding to native TcdB toxin, they will make excellent therapeutics or vaccines against C. difficile toxins or infections in their pure and partially pure forms.
  • the mutant toxin may be therapeutically administered to inhibit active TcdB in subjects having existing C. difficile infections or circulating TcdB toxin, for example, for treating or inhibiting diarrhea or pseudomembranous colitis.
  • the administration of a human vaccine comprising one or more of the mutants described herein is applicable to the prevention or treatment of a C. difficile infection in a human or animal.
  • the vaccine may be administered by epicutaneous injection, subcutaneous injection, intramuscular injection, interdermal injection (injection by infusion), sustained-release repository, aerosolization, parenteral delivery, inoculation into an egg, and the like, by known techniques in the art. Although this approach is generally satisfactory, other routes of administration, such as i.v. (into the blood stream) may also be used in a manner known to those of ordinary skill in the art.
  • the vaccine can be given together with adjuvants and/or immuno-modulators to boost the activity of the vaccine and the subject's response, the subject being a human or animal as described elsewhere herein.
  • each therapeutic or vaccine dose can be selected as an amount which induces an antitoxin or immunoprotective response without significant, adverse side effects.
  • amount in a vaccine will vary depending upon which specific immunogen is employed, how it is presented, and the size of the subject treated.
  • each therapeutic or immunogenic dose of the protein will comprise 0.1-1000 ⁇ g/kg of weight of the subject, preferably 0.2-100 ⁇ g/kg, and most preferably 1-10 ⁇ g/kg.
  • An optimal amount for a particular vaccine can be ascertained by standard studies involving observation of appropriate immune responses in subjects. Following an initial vaccination, subjects may receive one or several booster immunization adequately spaced.
  • Therapeutic doses for inhibiting TcdB toxin may also be from 10 ⁇ g-1 mg/kg, for example.
  • the invention provides a method of treatment comprising administering an effective amount of a vaccine of the present invention to a subject.
  • the vaccine formulations of the present invention may be used for both prophylactic and therapeutic purposes.
  • the vaccine compositions of the present invention can be formulated according to known methods of preparing pharmaceutically useful compositions, whereby these materials are combined in a mixture with a pharmaceutically acceptable carrier vehicle. Suitable vehicles and their formulation are described, for example, in Remingtons' Pharmaceutical Sciences, (Mack Publishing Co., 1980).
  • the TcdB toxin mutants can be administered in combination with a pharmaceutical carrier compatible with the protein and the subject.
  • suitable pharmacological carriers include, for example, physiological saline (0.85%), phosphate-buffered saline (PBS), Tris hydroxymethyl aminomethane (TRIS), Tris-buffered saline, and the like.
  • the protein may also be incorporated into a carrier which is biocompatible and can incorporate the protein and provide for its controlled release or delivery, for example, a sustained release polymer such as a hydrogel, acrylate, polylactide, polycaprolactone, polyglycolide, or copolymer thereof.
  • a sustained release polymer such as a hydrogel, acrylate, polylactide, polycaprolactone, polyglycolide, or copolymer thereof.
  • An example of a solid matrix for implantation into the subject and sustained release of the protein antigen into the body is a metabolizable matrix, as known in the art.
  • Adjuvants may be included in the vaccine to enhance the immune response in the subject.
  • adjuvants include, for example, aluminum hydroxide, aluminum phosphate, Freund's Incomplete Adjuvant (FCA), liposomes, ISCOM, and the like.
  • FCA Freund's Incomplete Adjuvant
  • the vaccine may also include additives such as buffers and preservatives to maintain isotonicity, physiological pH and stability.
  • Parenteral and intravenous formulations of the vaccine may include an emulsifying and/or suspending agent, together with pharmaceutically-acceptable diluents to control the delivery and the dose amount of the vaccine.
  • Factors bearing on the therapeutic or vaccine dosage include, for example, the age and weight of the subject.
  • the range of a given dose is about 25-5000 ⁇ g of the purified mutant receptor protein per ml, preferably about 100-1000 ⁇ g/ml preferably given in about 0.1-5 ml doses.
  • the vaccine or therapeutic should be administered to the subject in an amount effective to ensure that the subject will develop an immunity to protect against a C. difficle infection or a therapeutic response against a current C. difficile infection.
  • a vaccine for immunizing an about 5-lb. piglet against C. difficile would contain about 100-5000 ⁇ g protein per ml, preferably given in 1-5 ml doses.
  • the immunizing dose would then be followed by a booster given at about 21-28 days after the first injection.
  • the vaccine is formulated with an amount of the TcdB toxin mutant effective for immunizing a susceptible subject against an infection by more than one strain C. difficile.
  • the present invention further contemplates a monoclonal antibody raised against the C. difficile TcdB toxin mutant.
  • the monoclonal antibody may be prepared by a method comprising immunizing a suitable animal or animal cell with an immunogenic C. difficile TcdB toxin mutant to obtain cells for producing an antibody to said mutant, fusing cells producing the antibody with cells of a suitable cell line, and selecting and cloning the resulting cells producing said antibody, or immortalizing an unfused cell line producing said antibody, e.g. by viral transformation, followed by growing the cells in a suitable medium to produce said antibody and harvesting the antibody from the growth medium in a manner well known to those of ordinary skill in the art.
  • the recovery of the polyclonal or monoclonal antibodies may be preformed by conventional procedures well known in the art, for example as described in Kohler and Milstein, Nature 256, 1975, p. 495.
  • the invention relates to a diagnostic agent which comprises a monoclonal antibody as defined above.
  • a diagnostic agent which comprises a monoclonal antibody as defined above.
  • the diagnostic agent is to be employed in an agglutination assay in which solid particles to which the antibody is coupled agglutinate in the presence of a C. difficile toxin in the sample subjected to testing, no labeling of the monoclonal antibody is necessary, it is preferred for most purposes to provide the antibody with a label in order to detect bound antibody.
  • a double antibody (“sandwich”) assay at least one of the antibodies may be provided with a label.
  • Substances useful as labels in the present context may be selected from enzymes, fluorescers, radioactive isotopes and complexing agents such as biotin.
  • the diagnostic agent comprises at least one antibody covalently or non-covalently bonded coupled to a solid support.
  • a solid support This may be used in a double antibody assay in which case the antibody coupled to the solid support is not labeled.
  • the solid support may be selected from a plastic, e.g. latex, polystyrene, polyvinylchloride, nylon, polyvinylidene difluoride, cellulose, e.g. nitrocellulose and magnetic carrier particles such as iron particle coated with polystyrene.
  • the monoclonal antibody of the invention may be used in a method of determining the presence of C. difficile TcdB toxin in a sample, such as blood, plasma, or serum, the method comprising incubating the sample with a monoclonal antibody as described above and detecting the presence of bound toxin resulting from said incubation.
  • the antibody may be provided with a label as explained above and/or may be bound to a solid support as exemplified above.
  • a sample desired to be tested for the presence of C. difficile is incubated with a first monoclonal antibody coupled to a solid support and subsequently with a second monoclonal or polyclonal antibody provided with a label.
  • the sample may be incubated with a monoclonal antibody coupled to a solid support and simultaneously or subsequently with a labeled C. difficile TcdB toxin competing for binding sites on the antibody with any toxin present in the sample.
  • the sample subjected to the present method may be any sample suspected of containing a C. difficile TcdB toxin.
  • the sample may be selected from bacterial suspensions, bacterial extracts, culture supernatants, animal body fluids (e.g. serum, colostrum or nasal mucous) and intermediate or final vaccine products.
  • the monoclonal antibody of the invention it is contemplated to utilize a well-known ability of certain monoclonal antibodies to inhibit or block the activity of biologically active antigens by incorporating the monoclonal antibody in a composition for the passive immunization of a subject against diseases caused by C difficile producing a TcdB toxin, which comprises a monoclonal antibody as described above and a suitable carrier or vehicle.
  • the composition may be prepared by combining a therapeutically effective amount of the antibody or fragment thereof with a suitable carrier or vehicle. Examples of suitable carriers and vehicles may be the ones discussed above in connection with the vaccine of the invention.
  • a C. difficile -specific antibody may be used for prophylactic or therapeutic treatment of a subject having a C. difficile infection or a subject which may potentially incur a C. dfficile infection.
  • a further use of the monoclonal antibody of the invention is in a method of isolating a C. difficile TcdB toxin, the method comprising adsorbing a biological material containing said toxin to a matrix comprising an immobilized monoclonal antibody as described above, eluting said toxin from said matrix and recovering said toxin from the eluate.
  • the matrix may be composed of any suitable material usually employed for affinity chromatographic purposes such as agarose, dextran, controlled pore glass, DEAE cellulose, optionally activated by means of CNBr, divinylsulphone, etc. in a manner known per se.
  • the present invention relates to a method of determining the presence of antibodies against C. difficile TcdB toxin in a sample, the method comprising incubating the sample with C. difficile TcdB toxin and detecting the presence of bound antibody resulting from incubation.
  • a diagnostic agent comprising the TcdB toxin used in this method may otherwise exhibit any of the features described above for diagnostic agents comprising the monoclonal antibody and be used in similar detection methods although these will detect bound antibody rather than bound TcdB toxin as such.
  • the diagnostic agent may be useful, for instance as a reference standard or to detect anti-toxin antibodies in body fluids, e.g. serum, colostrum or nasal mucous, from subjects exposed to the toxin or C. difficile.
  • the monoclonal antibody of the invention may be used in a method of determining the presence of a C. difficile toxin, in a sample, the method comprising incubating the sample with a monoclonal antibody and detecting the presence of bound toxin resulting from said incubation.
  • the present invention further contemplates a nucleic acid sequence encoding a C. difficile TcdB toxin mutant wherein the nucleic acid sequence is a cDNA similar to a cDNA which encodes native C. difficile TcdB toxin, but differs therefrom only in having instead a substituted codon which encodes the substituted amino acid or amino acids in the mutant TcdB toxin, as defined herein, and wherein the substituted codon is any codon known to encode the substitute amino acid residue.
  • the mutant TcdB toxin described herein may be produced by well-known recombinant methods using cDNA encoding the mutant TcdB toxin, the cDNA having been transfected into a host cell in a plasmid or other vector.
  • the present invention contemplates any antigenic Clostridium difficile TcdB toxin mutant wherein the TcdB toxin mutant lacks the toxicity of a native C. difficile TcdB toxin.
  • the invention contemplates a vaccine for use in immunizing a human or an animal against an infection by Clostridium difficile, the vaccine comprising a purified non-toxic C. difficile TcdB toxin mutant.
  • the present invention contemplates a method for immunizing a subject against an infection by Clostridium difficile by administering an effective quantity of a vaccine comprising at least one purified non-toxic C. difficile TcdB toxin mutant as defined elsewhere herein.
  • the vaccine may be administered by epicutaneous injection, subcutaneous injection, intramuscular injection, interdermal injection, intravenous injection, sustained-release repository, aerosolization, parenteral delivery, or inoculation into an egg.
  • the vaccine induces an effective antibody titer to prevent or eliminate the infection without administration of a booster of the vaccine.
  • the present invention further contemplates a serum for treating a subject with an existing a Clostridium difficile infection comprising antibodies against a C. difficile TcdB toxin wherein the antibodies are raised against a C. difficile TcdB toxin mutant as defined elsewhere herein.
  • the present invention further contemplates an antibody against a Clostridium difficile TcdB toxin wherein said antibody is raised against a C. difficile TcdB toxin mutant as defined elsewhere herein.
  • the present invention further contemplates a method of treating a human or animal having, or disposed to having, a Clostridium difficile infection, comprising administering to the subject a therapeutically effective amount of an antibody against to an TcdB toxin of C. difficile, the antibody raised against a C. dfficile TcdB toxin mutant as defined elsewhere herein.
  • the method for a Clostridium difficile infection may comprise administering a serum comprising the antibodies effective against C. difficile TcdB toxin.
  • the present invention further contemplates a method of making a hybridoma which secretes an antibody against C. difficile TcdB toxin, the method comprising fusing a lymphocyte from an animal immunized with a C. difficile TcdB toxin mutant with cells capable of replicating indefinitely in cell culture to produce the hybridoma and further isolating the hybridoma.
  • the hybridoma may further secrete an antibody against C. difficile TcdB toxin.
  • the present invention further contemplates an immunoassay for C. difficile TcdB toxin in which a sample is contacting a sample which may contain a C. difficile TcdB toxin or a portion thereof with an antibody raised against a C. difficile TcdB toxin mutant to form an antibody-TcdB toxin complex and further detecting the antibody-TcdB toxin complex to determine the presence of the C. difficile TcdB toxin.
  • the present invention further contemplates a polynucleotide which encodes a mutant of C. difficile TcdB toxin polypeptide as defined herein.
  • the present invention further contemplates a vector containing a polynucleotide which encodes a mutant of C. difficile TcdB toxin polypeptide as defined herein.
  • the present invention further contemplates a host cell containing a vector containing a polynucleotide which encodes a mutant of C. difficile TcdB toxin polypeptide as defined herein.
  • the present invention further contemplates a process for producing a mutant of C.
  • the present invention further contemplates a non-toxic mutant of C. difficile TcdB toxin comprising a substitution in the cysteine residue of the native form of the toxin.

Abstract

An active, or passive vaccine utilizing purified non-toxic mutant TcdB toxins from Clostridium difficile for humans and animals against infections caused by C. difficile and/or C. sordellii. Persons most potentially affected by C. difficile infections include hospitalized patients, infants, and elderly persons. The TcdB toxin mutant of the vaccine preferably lacks the toxicity of a native C. difficile TcdB toxin. A serum comprising antibodies raised to the TcdB toxin mutant is also available for treating humans or animals against C. difficile infections. The serum may be used in a method for conferring passive immunity against C. difficile. Antibodies to the TcdB toxin mutant may be used in diagnostic tests or in treatments to clear TcdB toxin from bodily fluids. The mutant TcdB toxin may be produced by recombinant methods using cDNA encoding the toxin, the cDNA contained for example in a plasmid or host cell.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • The present application is a continuation of U.S. Ser. No. 10/463,957, filed Jun. 17, 2003, now U.S. Pat. No. 7,226,597, which claims benefit of U.S. Provisional No. 60/389,685, filed Jun. 17, 2002, each of which is explicitly incorporated herein by reference in its entirety.
  • STATEMENT REGARDING FEDERALLY SPONSORED RESEARCH OR DEVELOPMENT
  • Not Applicable.
  • BACKGROUND
  • Intracellular bacterial toxins enter cells, modify targets, and in many cases ultimately destroy the targeted cells thereby contributing to the disease process. Currently, there are no techniques for blocking intracellular virulence factors once they have entered the cytosol of cells. Further, no techniques exist which utilize inactive mutants derived from a toxin in order to inhibit the wild-type toxin at the intracellular cite.
  • Clostridium difficile is the leading cause of hospital acquired diarrhea and pseudomembranous colitis, a multifactorial disease involving steps in colonization, adherence, inflammation and cellular intoxication. TcdA and TcdB are two large clostridial toxins (LCTs) produced by C. difficile and are involved in development of pseudomembranous colitis. TcdB, (SEQ ID NO: 1), the focus of this study, glucosylates isoforms of small GTPases Rho, Rac and Cdc42 within the effector binding region at residues Threonine-37 (Rho) or Threonine-35 (Rac and Cdc42). The physiological impact of TcdB's activity includes disruption of tight junctions, increased epithelial permeability, as well as actin condensation and cell death.
  • TcdB can be divided into enzymatic, translocation and receptor binding domains, although detailed analysis of these regions has not been carried out to date. The first 546 amino acids of TcdB contain the enzymatic region, which is followed by a putative translocation and receptor-binding domain. Enzymatic activity appears to require the amino-terminal 546 residues since amino or carboxy terminal deletions of this fragment decrease activity. Within the enzymatic region, tryptophan 102 has been shown to be essential for UDP-glucose binding. A conserved DXD motif within LCTs is essential for LCT glucosyltransferase activity. Other studies, involving analysis of chimeras of the TcdB and TcsL enzymatic domain suggest residues 364 to 516 confer substrate specificity.
  • Steps in cell entry by TcdB have been broadly defined, yet events subsequent to entry are not well understood. For example, while we have a profile of the time-course for TcdB cell entry, very little is known about post-entry events that lead to glucosylation. Steps between membrane translocation and substrate interaction are not understood in TcdB intoxication. In fact almost no information exists in this regard for any intracellular toxin. In the cytosol, TcdB is capable of glucosylating multiple substrates, but whether inactivation of Rho, Rac and Cdc42 in combination is necessary for complete intoxication, or if other substrates are targeted, is not known. It has been found that overexpression of Rho isoforms protects cells from TcdB, suggesting inactivation of all substrates may not be necessary for cellular intoxication. Interestingly, Rho has also been shown to regulate the suppression of apoptosis, so it is not entirely clear whether overexpression of Rho is protective at the substrate inactivation level or prevents events downstream of glucosylation. Additionally, while some TcdB-intoxicating events, such as depolymerization of actin, can be attributed to inactivation of Rho, other processes like apoptosis may be linked to activities other than substrate inactivation. Given TcdB's large size (˜270 kD), and broad impact on cell physiology, it is possible the toxin may possess yet undefined activities in addition to glucosylation.
  • It would be desirable to have a vaccine or therapeutic composition for inhibiting or preventing action of the C difficile TcdB toxin.
  • SUMMARY OF THE INVENTION
  • The invention herein contemplates, in one embodiment, a mutant of native C. difficile TcdB toxin polypeptide wherein the mutant is substituted at position 395, such that the cysteine at position 395 in the native TcdB toxin has been replaced with another amino acid, for example, a tryptophan residue and wherein the mutant is not cytotoxic (non-toxic). The invention further comprises fragments of the TcdB toxin, which are effective in inhibiting TcdB toxin or are effective as a vaccine, and are non-toxic. The invention further contemplates a vaccine generally applicable to the prevention or treatment of C. difficile disease. Additionally, the present invention contemplates a method of inhibiting, modulating, or treating a C. difficile or a C. sordelii infection in a subject. Further, the present invention contemplates a monoclonal antibody raised against the C. difficile TcdB toxin mutant. In addition, the present invention contemplates a method of making an antibody against C. difficile TcdB toxin comprising immunizing an animal with an immunogenic amount of the C. difficile TcdB toxin mutant. These and other embodiments of the invention will be described further below.
  • DESCRIPTION OF THE DRAWINGS
  • FIG. 1. shows chromatography gels of LFnTcdB deletion and site-directed mutants. Panel A: Overview of deletion and site directed mutants. Deletion mutants were generated by PCR, cloned in-frame with lfn in pET15b, expressed in E. coli BL-21, and subsequently purified by Ni2+ affinity chromatography. Site-directed mutants were generated by the Quick-change method, using complementary mutation encoding oligonucleotides, and pLMS200 as template. Panel B: DS-PAGE analysis of his-tagged fusions. Lane 1, Molecular Weight Marker; Lane 2, TcdBW102A; Lane 3, TcdBC395W; Lane 4, TcdBC395S; Lane 5, TcdB35-556; Lane 6, TcdB1-70; Lane 7, TcdB1-420; Lane 8, TcdB1-500; Lane 9, TcdB1-556; Lane 10, Molecular Weight Marker.
  • FIG. 2. is a gel depicting glucosylation activity of deletion and site-directed mutants on RhoA, Rac1 and Cdc42. Each mutant and TcdB was tested for glucosylation activity on recombinant substrates GST-RhoA, GST-Rac1 and GST-Cdc42, using [14C]UDP-Glucose as cosubstrate. Following a 2 h incubation, the reaction mix was resolved by SDS-PAGE and exposed to film for 48 h. Lane 1, TcdB; Lane 2, TcdBW102A; Lane 3, TcdBC395W; Lane 4, TcdBC395S; Lane 5, TcdB1-556; Lane 6, TcdB1-500; Lane 7, TcdB1-420; Lane 8, TcdB1-170; Lane 9, TcdB35-556.
  • FIG. 3. shows inhibition of TcdB cytopathic effects by TcdB mutants. HeLa cells were cotreated with TcdB and each TcdB fusion plus PA. The cells were followed for 7 h and cytopathic effects were determined by visualization. Panel I is a micrograph depicting CHO cells treated with competitive inhibitors; A, PBS alone; B, TcdB alone; C, PA,LFn plus TcdB; D, PA,TcdB1-170 plus TcdB; E, PA,TcdB1-420 plus TcdB; F, PA,TcdB1-500 plus TcdB; G, PA,TcdB33-556 plus TcdB; H, PA, TcdBC39W, plus TcdB; I, PA, TcdBW102A plus TcdB; Panel II is a summary of inhibitors capable of blocking TcdB cytopathic effects; ▪=TcdB1-420; □=TcdBW102A;
    Figure US20080107673A1-20080508-P00001
  • =TcdBC395W; ▪=TcdB33-556;=TcdB1-500.
  • FIG. 4. is a graphical representation depicting sustained inhibition by supplemental treatments with inhibitor. HeLa cells were cotreated with TcdB and TcdB1-500 plus PA. During the course of the assay TcdB1-500 and PA were added to the cells at 1 h intervals for 12 h. The cells were then followed for 30 h and visualized for cytopathic effects. Open circles=TcdB; open diamonds =PA,TcdB1-500; closed circles=TcdB1-500, plus TcdB.
  • FIG. 5. is a graphical representation depicting the protection of CHO cells expressing TcdB1-556. GeneSwitch-CHOpGene/TcdB1-556 cells were induced with mifepristone in the presence or absence of TcdB1-500 plus PA. Cells were then observed for rounding and cytopathic effects at the indicated time-points. Open Circles=Uninduced Control; Closed Circles=Mifepristone-induced, PA,TcdB1-500; Open Squares=Mifepristone-induced control.
  • FIG. 6. is a chart demonstrating the inhibitory effects following inhibitor treatments prior to or following treatment with TcdB. In a 96-well plate, HeLa cells were treated with TcdB1-500 plus PA at time points prior to or following treatment with TcdB. Cells were amended with inhibitor every 30′ and observed for cytopathic effects at 8 h following toxin treatment.
  • FIG. 7. is a graphical representation depicting TcdB1-500 inhibition of TcsL cytopathic effects. HeLa cells were treated with TcdB1-500 plus PA for 30 min prior to treatment with TcsL. To enhance TcsL cytopathic activity, cells were treated with the toxin using an acid pulse where cells were subjected to TcsL in acid medium (pH 4.0) for 10 min. followed by replacement with neutral medium (pH 7.4) and TcdB1-500 plus PA. The cells were amended with inhibitor every 30′ for 12 h, then followed for 18 h to determine cytopathic effects. Open circles=TcsL; closed circles=PA,TcdB1-500 plus TcsL.
  • FIG. 8. Differential glucosylation of extracts from cells treated with TcdB plus inhibitor. HeLa cells were plated in T-25 flasks and grown until semiconfluent, then treated with PA, TcdB1-500 and TcdB was added to the cells. Three hours after TcdB treatment, cell extracts were collected and subjected to a TcdB glucosylation using [14C]UDP-Glucose as cosubstrate. The reactions were subsequently resolved by SDS-PAGE and exposed to film for 48 h. Lane 1=untreated HeLa cells; Lane 2=TcdB-treated cells; Lane 3=TcdB plus inhibitor treated cells.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The invention contemplated herein comprises, in a preferred embodiment, non-cytotoxic C. difficile TcdB toxin derivatives and deletions (mutants) which are deficient in at least one specific function required for toxicity and which are effective intracellular inhibitors of native TcdB toxin or are effective in producing immunity against TcdB toxin. The present invention demonstrates that enzymatically inactive fragments of the TcdB enzymatic domain are effective intracellular inhibitors of native TcdB. The present invention comprises purified derivatives (mutants) of C. difficile TcdB toxin which are deficient in glucosyltransferase and glucosylhydrolase activity. The mutants are considered to be useful as a vaccine for both humans and animals.
  • Examples of animals which may be treated are cattle, chickens, turkeys, ostriches, emu, ducks, horses, donkeys, mules, pigs, sheep, goats antelope, buffalo, llamas, cats, lions, tigers, dogs, bears, guinea pigs, hamsters, chinchillas, mink, ferrets, rodents, parrots, parakeets, peacocks, seals, sea lions, orcas, monkeys, chimpanzees, baboons, orangutans, gorillas, reptiles, and other zoo and livestock animals.
  • The term “mutant”, where used herein, refers to a fragment, point deletion, point substitution, or a deletion of multiple residues of the TcdB protein sequence, and may be encoded by a nucleotide sequence intentionally made variant from a native sequence. The present invention also contemplates nucleotide sequences which encode the mutants.
  • The mutants of the present invention preferably have at least one substituted amino acid in the enzymatic domain of the TcdB toxin which includes amino acid position 395 of the sequence of the native TcdB toxin as shown in SEQ ID NO: 1.
  • As noted above, the novel mutants contemplated herein comprise at least one amino acid substitution or deletion of the native C. difficile TcdB toxin. For example, the amino acid at position 395 (also referred to herein as the “critical position”) of the amino acid sequence of the native C. difficile TcdB toxin (SEQ ID NO: 1) may be substituted with a different amino acid in the same position.
  • In particular, the invention comprises mutants wherein the native cysteine at position 395 has been substituted with a tryptophan residue at position 395. However, any amino acid residue which would provide a mutant effective in inhibiting TcdB toxin, and which is not cytotoxic, may be substituted for the cysteine residue at position 395. Examples of other amino acids which may be used to substitute the cysteine residue include alanine, valine, leucine, isoleucine, proline, methionine, phenylalanine, glycine, threonine, tyrosine, asparagine, aspartic acid, glutamine, glutamic acid, lysine, arginine, and histidine. Mutants which are cytotoxic, e.g., a serine-substitute, also comprise the invention, particularly when they are used in a diagnostic assay as described below. Mutants comprising deletions of portions of the enzymatic domain include, for example, a modified C. difficile TcdB toxin having a deletion of amino acid positions 501-556 (SEQ ID NO: 3), 421-556 (SEQ ID NO: 5), 171-556 (SEQ ID NO: 7), or 1-34 (SEQ ID NO: 9) are also contemplated. An especially preferred embodiment comprises a mutant having at least one substitution in the enzymatic domain. The mutants of the present invention preferably have deficient glucosyltransferase and glucosylhydrolase activity compared to the native C. difficile TcdB toxin, and are non-toxic, and in an especially preferred embodiment are antigenic, whereby vaccines produced from them induce anti-TcdB toxin antibodies in vivo as explained in more detail below.
  • As noted above, it is an object of the present invention to provide novel vaccines comprising the TcdB toxin mutants described herein, or antigenic fragments thereof, which when administered to animals or humans, are capable of inducing production of protective antibodies directed against C. difficile TcdB toxin, thereby providing prophylaxis against infection by C. difficile disease states resulting from such infection, and/or from the TcdB toxin itself. It is a particular aim of the present invention to provide such a vaccine that is relatively safe and simple to produce. Antibodies and antisera raised against the mutants are also capable of use in therapy for at least some, if not all, disease states, in which TcdB toxin is involved.
  • In further aspects of the present invention there is provided recombinant DNA which encode any proteins, fragments, or amino acid sequences thereof described or claimed herein. Such recombinant DNA is conveniently provided by PCR amplification of the DNA encoding for the desired sequence, using primers targeted at respective ends of the double stranded sequence of which it forms one half, using methods well known to those of ordinary skill in the art.
  • In a further aspect of the present invention there are provided antisera raised to the mutants, or antigenic fragments thereof, of the invention and antibodies derived therefrom. Furthermore, the present invention provides monoclonal antibodies against the mutants, or antigenic fragments thereof, of the invention and hybridoma cells for production thereof as described in more detail below.
  • The present invention further contemplates TcdB toxin mutants which have additional substitutions which are merely conservative substitutions of amino acids. By “conservative substitution” is meant the substitution of an amino acid by another one of the same class; the classes according to Table I.
  • TABLE I
    Table I. Classes of amino acids suitable for conservative substitution.
    CLASS AMINO ACID
    Nonpolar: Ala, Val, Leu, Ile, Pro, Met, Phe, Trp
    Uncharged polar: Gly, Ser, Thr, Cys, Tyr, Asn, Gln
    Acidic: Asp, Glu
    Basic: Lys, Arg, His
  • As is well known to those skilled in the art, altering any given non-critical amino acid of a protein by conservative substitution may not significantly alter the activity of that protein because the side-chain of the amino acid which is inserted into the sequence may be able to form similar bonds and contacts as the side chain of the amino acid which has been substituted for.
  • Non-conservative substitutions (outside the classes of Table I) are possible provided that these do not excessively affect the immunogenicity of the polypeptide and/or reduce its effectiveness in inhibiting TcdB toxin.
  • The polypeptides of the invention may be prepared synthetically, or more suitable, they are obtained using recombinant DNA technology. Thus, the invention further provides a nucleic acid which encodes any of the mutants of TcdB toxin which have at least one substitution and/or deletion as described herein.
  • Such nucleic acids may be incorporated into an expression vector, such as a plasmid, under the control of a promoter as understood in the art. The vector may include other structures as conventional in the art, such as signal sequences, leader sequences and enhancers, and can be used to transform a host cell, for example a prokaryotic cell such as E coli or a eukaryotic cell. Transformed cells can then be cultured and polypeptide of the invention recovered therefrom, either from the cells or from the culture medium, depending upon whether the desired product is secreted from the cell or not.
  • As used herein, the terms “complementary” or “complementarity” are used in reference to polynucleotides (i.e., a sequence of nucleotides) related by the base-pairing rules. For example, for the sequence “A-G-T,” is complementary to the sequence “T-C-A.” Complementary may be “partial,” in which only some of the nucleic acids' bases are matched according to the base pairing rules. Or, there may be “complete” or “total” complementary between the nucleic acids. The degree of complementary between nucleic acid strands has significant effects on the efficiency and strength of hybridization between nucleic acid strands. This is of particular importance in amplification reactions, as well as detection methods which depend upon binding between nucleic acids.
  • Nucleic acids of the present invention also include DNA sequences which hybridize to the DNA sequences which encode the mutant polypeptides described herein, or their complementary sequences, under conditions of high or low stringency and which encode proteins having activity against TcdB toxin and/or which preferably can stimulate antibodies against native TcdB toxin.
  • Hybridization and washing conditions are well known and exemplified in Sambrook, J., Fritsch, E. F. and Maniatis, T. Molecular Cloning: A Laboratory Manual, Second Edition, Cold Spring Harbor Laboratory Press, Cold Spring Harbor (1989), particularly Chapter 11 and Table 11.1 therein (expressly entirely incorporated herein by reference). The conditions of temperature and ionic strength determine the “stringency” of the hybridization.
  • In one embodiment, high stringency conditions are prehybridization and hybridization at 68° C., washing twice with 0.1×SSC, 0.1% SDS for 20 minutes at 22° C. and twice with 0.1×SSC, 0.1% SDS for 20 minutes at 50° C. Hybridization is preferably overnight.
  • In one example, low stringency conditions comprise conditions equivalent to binding or hybridization at 42° C. in a solution consisting of 5×SSPE (43.8 g/l NaCl, 6.9 g/l NaH2PO4.H2O and 1.85 g/l EDTA, pH adjusted to 7.4 with NaOH), 0.1% SDS, 5×Denhardt's reagent [50×Denhardt's contains per 500 ml: 5 g Ficoll (Type 400, Pharmacia), 5 g BSA (Fraction V; sigma) and 100 μg/ml denatured salmon sperm DNA] followed by washing in a solution comprising 5x SSPE, 0.1% SDS at 42° C. when a probe of about 500 nucleotides in length is employed.
  • In another embodiment, low stringency conditions are prehybridization and hybridization at 68° C., washing twice with 2×SSC, 0.1% SDS for 5 minutes at 22° C., and twice with 0.2×SSC, 0.1% SDS for 5 minutes at 22° C. Hybridization is preferably overnight.
  • In an alternative embodiment, very low to very high stringency conditions are defined as prehybridization and hybridization at 42° C. in 5×SSPE, 0.3% SDS, 200 ug/ml sheared and denatured salmon sperm DNA, and either 25% formamide for very low and low stringencies, 35% formamide for medium and medium-high stringencies, or 50% formamide for high and very high stringencies, following standard Souther blotting procedures.
  • The carrier material is then washed three times each for 15 minutes using 2×SSC, 0.2% SDS preferably at least 45° C. (very low stringency), more preferably at least at 50° C. (low stringency), more preferably at least at 55° C. (medium stringency), more preferably at least at 60° C. (medium-high stringency), even more preferably at least at 65° C. (high stringency), and most preferably at least at 70° C. (very high stringency).
  • It is well known in the art that numerous equivalent conditions may be employed to comprise low stringency conditions; factors such as the length and nature (e.g., DNA, RNA, base composition) of the probe and nature of the target (e.g., DNA, RNA, base composition, present in solution or immobilized, etc.) And the concentration of the salts and other components (e.g., the presence or absence of formamide, dextran sulfate, polyethylene glycol) are considered and the hybridization solution may be varied to generate conditions of low stringency hybridization different form, but equivalent to, the above listed conditions. In addition, conditions which promote hybridization under conditions of high stringency (e.g., increasing the temperature of the hybridization and/or wash steps, the use of formamide in the hybridization solution, etc.) are also know in the art.
  • When used in reference to a double-stranded nucleic acid sequence such as a cDNA or genomic clone, the term “substantially homologous” refers to any probe which can hybridize to either or both strands of the double-stranded nucleic acid sequence under conditions of low stringency as described above.
  • When used in reference to a single-stranded nucleic acid sequence, the term “substantially homologous” refers to any probe which can hybridize (i.e., it is the complement of) the single-stranded nucleic acid sequence under conditions of low stringency as described above.
  • As used herein, the term “hybridization” is used in reference to the pairing of complementary nucleic acids. Hybridization and the strength of hybridization (e.e., the strength of the association between the nucleic acids) is impacted by such factors as the degree of complementary between the nucleic acids, stringency of the conditions involved, the Tm (melting temperature) of the formed hybrid, and the G:C ration within the nucleic acids. As used herein the term “stringency” is used in reference to the conditions of temperature, ionic strength, and the presence of other compounds such as organic solvents, under which nucleic acid hybridizations are conducted.
  • As used herein, the terms “cell,” “cell line,” and “cell culture” are used interchangeably and all such designations include progeny. The words “transformants” or “transformed cells” include the primary transformed cell and cultures derived from that cell without regard to the number of transfers. All progeny may not be precisely identical in DNA content, due to deliberate or inadvertent mutations. Mutant progeny that have the same functionality as screened for in the originally transformed cell are included in the definition of transformants.
  • As used herein, the term “vector” is used in reference to nucleic acid molecules that transfer DNA segment(s) from one cell to another. The term “vehicle” is sometimes used interchangeably with “vector”.
  • The terms “recombinant DNA vector” as used herein refers to DNA sequences containing a desired coding sequence and appropriate DNA sequences necessary for the expression of the operably linked coding sequence in a particular host organism. DNA sequences necessary for expression in prokaryotes include a promoter, optionally and operator sequence, a ribosome binding site and possibly other sequences. Eukaryotic cells are known to utilize promoters, polyadenylation signals and enhancers. It is not intended that the term be limited to any particular type of vector. Rather, it is intended that the term encompass vectors that remain autonomous within host cells (e.g., plasmids), as well as vectors that result in the integration of foreign (e.g., recombinant nucleic acid sequences) into the genome of the host cell.
  • The term “expression vector” and “recombinant expression vector” as used herein refers to a recombinant DNA molecule containing a desired coding sequence and appropriate nucleic acid sequences necessary for the expression of the operably linked coding sequence in a particular host organism. Nucleic acid sequences necessary for expression in prokaryotes usually include a promoter, an operator (optional), and a ribosome binding site, often along with other sequences. Eukaryotic cells are known to utilize promoters, enhancers, and termination and polyadenylation signals. It is contemplated that the present invention encompasses expression vectors that are integrated into host cell genomes, as well as vectors that remain unintegrated into the host genome.
  • The terms “in operable combination,” “in operable order,” and “operably linked,” as used herein refer to the linkage of nucleic acid sequences in such a manner that a nucleic acid molecule capable of directing the transcription of a given gene and/or the synthesis of a desired protein molecule is produced. The term also refers to the linkage of amino acid sequences in such a manner so that a functional protein is produced.
  • The mutants described herein may be expressed in either prokaryotic or eukaryotic host cells. Nucleic acids encoding the mutants may be introduced into bacterial host cells by a number of means including transformation or transfection of bacterial cells made competent for transformation by treatment with calcium chloride or by electroporation. If the mutants are to be expressed in eukaryotic host cells, nucleic acid encoding the protein or toxin of interest may be introduced into eukaryotic host cells by a number of means including calcium phosphate co-precipitation, spheroplast fusion, electroporation, microinjection, lipofection, protoplast fusion, and retroviral infection, for example. When the eukaryotic host cell is a yeast cell, transformation may be affected by treatment of the host cells with lithium acetate or by electroporation, for example.
  • In a preferred version of the invention, the mutant is characterized as having 50% or less of the glucosyltransferase and glucoslyhydrolase activity of wild type TcdB toxin, as measured by assays described herein. In a more preferred version of the invention, the mutant is characterized as having 30% or less of the glucosyltransferase and glucosylhydrolase activity of wild type TcdB toxin, as measured by assays described herein. In a more preferred version of the invention, the mutant has less than 20% of the glucosyltransferase and glucosylhydrolase activity of wild type TcdB toxin as measured by assays described herein. In a more preferred version of the invention, the mutant has less than 10% of the glucoslytransferase and glucosylhydrolase activity of wild-type TcdB toxin, as measured by assays described herein. More preferably, the mutant has less than 5% of the glucosyltransferase and glucosylhydrolase activity of the wild-type TcdB toxin, as measured by assays described herein. Even more preferably, the mutant has less than 0% of the glucosyltransferase and glucosylhydrolase activity of wild-type TcdB toxin, as measured by assays described herein. More particularly, the invention as contemplated herein is a mutant (mutein) of C. difficile TcdB toxin polypeptide which comprises: (a) a polypeptide having a substitution at position 395 of the amino acid sequence of native C. difficile TcdB toxin, wherein the cysteine at position 395 has been replaced with tryptophan (SEQ ID NO: 11) or with another amino acid; or (b) a modified C. difficile TcdB toxin having a deletion of amino acid positions 501-556 (SEQ ID NO: 3), 421-556 (SEQ ID NO: 5), 171-556 (SEQ ID NO: 7), or 1-34 (SEQ ID NO: 9), and wherein the mutant of (a) or (b) is effective in inhibiting or modulating the cytotoxic effect of C. difficile TcdB toxin or is effective as a vaccine against C. difficile and wherein the mutant is not cytotoxic.
  • While the invention will now be described in connection with certain preferred embodiments in the following examples so that aspects thereof may be more fully understood and appreciated, it is not intended to limit the invention to these particular embodiments. On the contrary, it is intended to cover all alternatives, modifications and equivalents as may be included within the scope of the invention as defined by the appended claims. Thus, the following examples, which include preferred embodiments will serve to illustrate the practice of this invention, it being understood that the particulars shown are by way of example and for purposes of illustrative discussion of preferred embodiments of the present invention only and are presented in the cause of providing what is believed to be the most useful and readily understood description of formulation procedures as well as of the principles and conceptual aspects of the invention.
  • EXAMPLES
  • During analysis of the TcdB enzymatic domain a set of mutants were identifiable which were unable to modify substrate, yet were capable of blocking TcdB cytopathic effects. Herein are described generation and analyses of these mutants and the demonstration that these proteins are potent intracellular inhibitors of TcdB and block glucosylation of a previously undescribed target. These mutants show, for the first time, that a toxin derivative can be used to effectively block the activity of the native toxin within the cell. This inhibitory activity also suggests a new paradigm for a therapeutic approach to treat toxin-based diseases.
  • Enzymatic and Cytopathic Activity of Mutants
  • As summarized in FIG. 1, 4 deletion and 3 site-directed mutants in the TcdB enzymatic domain were constructed, cloned and isolated from E. coli. The nomenclature for each of these mutants is summarized in panel A of FIG. 1. One site-directed mutant, TcdBW102A wherein the tryptophan at position 102 is substituted with alanine has been previously characterized and served as a control in cytotoxicity and enzymatic assays [Busch, 2000]. Experiments conducted in the present work suggested TcdB1-556 (SEQ ID NO: 1) could be inactivated by n-ethylmaleimide (data not shown), indicating a role for the sole cysteine (position 395) in enzymatic activity, thus site-directed mutants TcdBC395S, TcdBC395W were produced. Amino-terminal and carboxy-terminal deletions were also generated in an attempt to further identify inactive mutants. Since these mutants lacked receptor binding and translocation domains, the fragments were fused to the cell entry proteins of anthrax lethal toxin. This anthrax toxin derivative consists of anthrax protective antigen (PA), and a truncated form of anthrax lethal toxin (LFn), to which heterologous fusions are made. PA-LFn has been used by several groups for the cytosolic delivery of a variety of proteins, and we previously used this system to deliver TcdB1-556 to cultured mammalian cells [Spyres, 2001]. Using this delivery system, the fusions were tested for cytopathic activity and only TcdB1-556 and TcdBC395S were cytotoxic (data not shown).
  • To determine if lack of cytotoxicity was due to attenuation of enzymatic activity, mutants were tested for glucosylation of RhoA, Rac1 or Cdc42. As shown in FIG. 2, only TcdB1-556, and TcdBC395S glucosylated substrate. In line with earlier reports carboxy-terminal deletions and TcdBW102A were unable to glucosylate substrate. The remainder of the site-directed and deletion mutants were also deficient in glucosylation. Furthermore, this loss of activity was maintained across all of the shared substrates since these same mutants were attenuated in glucosylation of RhoA, Rac1 and Cdc42.
  • Each mutant was also analyzed for glucosylhydrolase activity using radiolabeled UDP-glucose in the absence of substrate. Fusions were incubated with [14C]UDP-glucose and the liberated sugar was separated by anion-exchange chromatography. As shown in Table 1, even with extended (16 h) incubation glucosylhydrolase activity was significantly reduced for all enzymatically inactive mutants. Without wishing to be constrained by theory, the absence of substrate modification by these mutants could be accounted for, at least in part, by defective hydrolase activity.
  • TcdB Mutants as Inhibitors of Native Toxin
  • Since the inactive mutants could be effectively delivered to the cytosol of cells via the PA, LFn system, we were presented with the unique opportunity to examine the effects these mutants might have when administered in combination with wild-type TcdB. Thus, HeLa cells were treated with TcdB in the presence or absence of PA plus each attenuated mutant. As shown in FIG. 3(I), PA-delivered TcdB1-500, TcdB1-420, TcdBW102A, TcdBC395W, or TcdB35-556, attenuated TcdB cytopathic effects suggesting the mutants had an antagonistic impact on TcdB intoxication. The inhibitor effects were dependent on the presence of inactive enzymatic domain mutants since PA-LFn alone did not inhibit TcdB.
  • It was clear from the results in FIG. 3(II), that approximately 7 h after delivery of inhibitory fragments to the TcdB treated cells that the protective effect began to decrease. This observation suggested that the inhibitory effect of the enzymatically inactive mutants has a limited lifetime. To address this possibility, the initial competition was set-up as before and the inhibitor (TcdB1-500) was added to the cells at 1 h time intervals during the course of the assay. As shown in FIG. 4, using this approach greater than 50% of the cells demonstrated no cytopathic effects and appeared to be protected from the wild-type toxin during the course of the assay (>30 h). Hence, continued administration maintained the protective effect against TcdB. Continued addition of the inhibitor after 12 h, did not improve or change the inhibition of TcdB, suggesting TcdB had lost activity or that the accumulated inhibitor was in sufficient excess so that its protective effect was extended.
  • Inactive Mutants Protect CHO Cells Expressing TcdB1-556
  • The fact that the TcdB inhibitors lack native translocation and receptor binding domains suggested that inhibition occurred within the cytosol. However, inhibition at the cell surface could not be formally excluded since cell surfacing-interacting regions of TcdB have not been fully elucidated. To determine if inhibition of TcdB was occurring within the cytosol, a CHO cell line capable of inducible expression of TcdB1-556 was generated. A tightly regulated expression system, pSwitch, was selected which allows expression only in the presence of the hormone mifepristone. GeneSwitch-CHOpGene/TcdB1-556 cells showed early toxic effects, such as rounding, at around 4 h following addition of mifepristone and were no longer viable by 24 h. To test the inhibitor on these cells, mifepristone was added to the cells and inhibitor was added 2 h later and subsequently added every 30 min for an additional 3 h. As shown in FIG. 5, mifepristone treated GeneSwitch-CHOpGene/TcdB1-556 cells were protected from the effects of TcdB1-556 when the inhibitor was added at 2 h following induction. The inhibitor clearly slows the cytopathic activity of these cells following induction. Cells eventually show cytopathic effects similar to that control since the cell continues to express TcdB1-556. These results demonstrate that the inhibitor is capable of blocking TcdB intoxication at a site within the cell.
  • TcdB-Inhibitors as Tools to Dissect the Time-Course of Posttranslocation Events
  • In earlier studies we reported on the time-course of entry by TcdB, based on results from lysosomotropic inhibitor assays [Qa'Dan, 2000]. The inhibitors now provided a reagent to determine the time-course of events occurring after translocation into the cytosol. At a given time-point, if intoxication events have been initiated, then addition of the inhibitor should no longer have an effect. In this experiment cells were pretreated with the inhibitor or treated with the inhibitor at time-points following TcdB treatment. As shown in FIG. 6, protection occurs in cells when the inhibitor is added as early as 40 min before treatment with TcdB. Protection also occurs when the inhibitor is added up 40 min after treatment with TcdB. Only when the inhibitor is added over 40 min prior to treatment with TcdB or over 40 min after treatment with TcdB is there a noticeable cytopathic effect. Given that cell entry takes approximately 20 min, these results suggested intoxication events require at least a 40 min time period after translocation to initiate cytotoxic effects.
  • Inhibition of Intoxication by C. sordellii Lethal Toxin (TcsL)
  • A variety of events, including substrate related and substrate unrelated, could occur during the 40 min posttranslocation time-period. If the inhibitor blocked processes unrelated to substrate interaction, we suspected the mutant might block another highly related LCT, which does not share similar substrate targets with TcdB. An ideal candidate for this experiment was TcsL, which is closely related to TcdB, yet modifies a different set of Ras proteins including Ras, and Ra1. TcsL does share Rac as a common substrate with TcdB. We tested the TcdB inhibitor's ability to block TcsL intoxicaion. In recent work we reported that acid pH enhances TcsL entry [Qa'Dan, 2001], so the initial treatment with TcsL was carried out by providing an extracellular acid pulse to TcsL. In this assay cells were pretreated with the inhibitor, then acid-pulse treated with TcsL, and subsequently treated with additional inhibitor during the time-course of the assay. As can be seen in FIG. 7, TcdB1-500 was also able to block the activity of TcsL. Similar to results with TcdB, the inhibitor was capable of reducing TcsL's cytopathic effects by almost 50%. These results suggested the TcdB inhibitor was blocking LCT intoxication events that might not be related to substrate targeting or that blocking a single target was sufficient to prevent toxic effects.
  • Effects of Inhibitor on Substrate Glucosylation in Cultured Cells
  • The results from the TcsL inhibition assay suggested the inhibitor prevented toxin-specific activities that might not be related to targeting Rho, Rac and Cdc42. For this reason it was important to determine if the inhibitor prevented glucosylation of these substrates during TcdB intoxication. Thus, a set of differential glucosylation reactions were carried out that involved examining extracts from cells treated with TcdB, or treated with TcdB plus the inhibitor, for a decrease in substrate that could be glucosylated. As shown in FIG. 8 using a minimal intoxicating dose of TcdB, cells showed a relatively equal amount of Rho substrate that could be glucosylated from both TcdB-treated and TcdB-plus-inhibitor treated cells. While there did not appear to be a difference in targeting of Rho, Rac or Cdc42 there was a change in the ability to glucosylate a second protein that migrated at a size larger than the Rho proteins. The larger protein was below the level of detection in extracts from TcdB treated cells yet this protein was glucosylated in extracts from cells treated with TcdB plus the inhibitor. These results further suggest the inhibitor prevents an LCT activity other than glucosylation of Rho, Rac and Cdc42.
  • Attenuated mutants of TcdB inhibit wild-type toxin at an intracellular site. To our knowledge this is the first example of an approach that blocks the activity of an intracellular bacterial toxin within the cytosol of intoxicated cells. This inhibitory effect also suggests some yet undefined aspects of TcdB. Clearly, while unable to modify substrate, the mutants carry out functions within the cytosol, which allow inhibition. The exact mode of inhibition is not clear; however, the preliminary evidence indicates the inhibitor prevents glucosylation of a substrate other than Rho, Rac or Cdc42. This is a feasible possibility since some of the inhibitors do not encompass the region of TcdB reported to interact with Rho, Rac and Cdc42. Work by Hofmann et al. [Hofmann, 1998] using chimeric derivatives between the enzymatic domains of TcsL and TcdB, suggested residues 365-516 conferred substrate specificity. Our deletion analysis shows residues 1-420 are able to inhibit TcdB intoxication, while the 1-170 deletion has no inhibitory effect. Finally, the mutants also inhibit TcsL, which shares only one substrate, Rac, with TcdB. If inhibition were due to Rho, Rac and Cdc42 interaction then the inhibitor should be less effective on TcsL, but it is not. The amino terminal domains of these two proteins are homologous (78% homology) and could share activities, and yet undefined common substrates.
  • Experimental Procedures Tissue Culture, Bacterial Strains and Chemical Reagents
  • Human cervical adenocarcinoma cells American Type Culture Collection (ATCC) Manassas, Va CCL-2 (HeLa) were grown in supplemented RPMI 1640 (RP-10) [Starnbach, 1994] with 10% fetal bovine serum at 37° C. in a humid atmosphere with 7% CO2. Clostridium difficile strain VPI 10463, and Clostridium sordellii strain 9714 were obtained from ATCC and used as a source of culture supernatant, genomic DNA, TcdB and TcsL. All reagents were of molecular biology grade and were purchased from Sigma Chemical Co., St. Louis, Mo. unless otherwise noted.
  • Construction of Recombinant LFn-TcdB Fusions
  • The region encoding for the enzymatic domain of TcdB i.e., TcdB nucleotides 1-1668 (SEQ ID NO:2)) was genetically fused to lfn, cloned expressed and purified in E. coli as previously described [Spyres, 2001]. Using a similar approach, four other fusions of LFnTcdB were also constructed. Briefly, fragments encoding regions TcdB1-500(SEQ ID NO: 3 encoded by nucleotides 1-1500 (SEQ ID NO: 4)), TcdB1-420 (SEQ ID NO: 5 encoded by nucleotides 1-1260 (SEQ ID NO: 6)), TcdB1-170 (SEQ ID NO: 7 encoded by nucleotides 1-510 (SEQ ID NO: 8)), and TcDB35-556 (SEQ IDNO: 9 encoded by nucleotides 103-1668 (SEQ ID NO: 10)), were PCR amplified and cloned into the BamHI site of pABII [Spyres, 2001] to make the plasmids pLMS201, pLMS202, pLMS204, and pLMS301 respectively. Plasmids were transformed into E. coli XL1-blue (Stratagene) and candidate clones were sequenced, then transformed into E. coli BL-21 Star (Invitrogen) for expression.
  • Site-directed mutants were generated using Pfu Turbo DNA polymerase and the QuickChange mutagenesis approach (Stratagene). Oligonucleotides for generation of TcdB1-556C395S (SEQ ID NO: 11, where Xaa at position 395 is serine) were GTTTACTATTAAATTGCTAGAATATGAGTCTTTCACAG (sense) (SEQ ID NO: 13), CTGTGAAGACTCATATTCTAGCAATTTAATAGTAAAAC (antisense) (SEQ ID NO: 14); TcdB1-556C395W (SEQ ID NO: 11, where Xaa at position 395 is tryptophan) GTTTTACTATTAAATTGCTACCTATGAGTCTTTCACAG (sense) (SEQ ID NO: 15), CTGTGAAAGACTCATATTGGAGCAATTTAATAGTAAAAC (antisense) (SEQ ID NO: 16); TcdB1-556W102A (SEQ ID NO: 12) AAAAATTTACATTTTGTTGCTATTGGAGGTCAA (sense) (SEQ ID NO: 17), TTGACCTCCAATAGCAACAAAATGTAAATTTTT (antisense) (SEQ ID NO: 18). Mutants were selected in E. coli XL1-blue and confirmed by sequencing, followed by transformation into E. coli BL-21 Star for expression.
  • Purification of Recombinant Proteins and TcdB
  • Expression of LFnTcdB fusions was induced with 0.2 mM iso-propyl-β-D-thiogalactopyranoside in log phase (OD600 0.8) cultures at 16° C. Cells were harvested by centrifugation at 8700×g, resuspended in binding buffer (5 mM imidazole, 500 mM NaCl, 20 mM Tris-HCl, pH7.9) supplemented with a protease inhibitor cocktail containing 4-(2-aminoethyl)benzenesulfonyl fluoride, phosphoramidon, pepstatin A, bestatin, and E-64 and lysed by sonication. LFnTcdB fusion proteins were isolated using nickel 900 cartridges following the manufacturer's instructions (Novagen). As a second purification step, proteins were fractionated on a high-resolution anion exchange (mono-Q) column (Amersham Pharmacia). Recombinant PA was isolated from E. coli BL-21, harboring the plasmid, pSRB/ET-15b-PA (a generous gift from Steven Blanke), as previously described [Whilhite, 1998]. TcdB and TcsL were purified as previously described [Qa'Dan, 2000]. Recombinant clones of RhoA, Rac1, and Cdc42 (a generous gift of Alan Hall) were expressed and purified as previously described [Spyres, 2001].
  • Glucosylhydrolase/Glucosylation Assays
  • Glucosylation reactions were carried out as previously described [Spyres, 2001]. Glucosylhydrolase assays were carried out in a reaction mix containing 50 mM n-2hydroxyethylpiperazine-N′-2-ethane sulfonic acid, 100 mM KCl, 1 mM MnCl2, 1 mM MgCl2, 100 μg/ml BSA, 0.2 mM GDP, 40 μM [14C]UDP-glucose (303Ci/mol; ICN Pharmaceuticals), 100 μM UDP-glucose and 3 pmol of TcdB or 10 pmol of each fusion protein. The assay was allowed to incubated overnight at 37° C. and similar to a previously described protocol [Ciesla, 1998], the cleaved glucose was separated using AG1-X2 anion exchange resin and counted in a liquid scintillation counter.
  • Assay for Cytopathic Effects and Inhibitor Assays
  • To determine the cytopathic activity of each fusion and site-directed mutant, HeLa cells were plated in 96 well microtiter plates (3×104 cells/well) and allowed to incubate overnight. The following day the cells were treated with 30 pmol of each fusion plus 8.5 pmol of PA and observed for 48 h for signs of cytopathic effects. For inhibition assays, HeLa cells were plated as before and treated with 4 pmol of the appropriate LFnTcdB fusion plus 8.5 pmol of PA in a final volume of 100 μl. At the same point the cells were cotreated with 80 fmol of TcdB and observed for cytopathic effects. In a second competition assay, 30 pmol of TcdB1-500 plus 8.5 pmol of PA were added to cells in a final volume of 100 μl and allowed to incubate 30 min, at which point 20 fmol of TcdB was added to the cells. Following the initial treatment, 30 pmol of TcdB1-500 and 8.5 pmol of PA were added every 30 min for the first 90 min and every hour thereafter up to 12 h. The cells were observed for cytopathic effects for an additional 18 h. Similar competition assays were carried out using 2 pmol of TcsL. For inhibition assays with TcsL, cells were subjected to a brief acid-pulse, which enhances cytotoxic activity for this toxin. For TcsL competition, cells were pretreated with TcdB1-500 and PA for 30 min at which point cells were treated with TcsL via an acid pulse as previously described [Qa'Dan, 2001]. The cells were then amended with 30 pmol of TcdB1-500, 8.5 pmol of PA every 30 min up to 12 h and followed for 16 h. For differential glucosylation assays, HeLa cells semi-confluent (1×106) were first treated with 325 pmol of PA and 300 pmol of TcdB1-500 followed by treatment with 50 fmol of TcdB in a final volume of 10 ml. Following 3 h of treatment cells were washed 3 times in ice-cold PBS, scraped and extracts were prepared as previously described [Spyres, 2001]. Using each extract as target substrate, glucosylation reactions were identical to those previously described with changes only to reaction volume (30 μl) and amount of substrate (80 μg).
  • Generation of TcdB-Expressing CHO Cells
  • A DNA sequence coding for the enzymatic domain of TcdB (amino acids 1 to 556) placed downstream and in-frame with a Kozak sequence (GNNATGG) was cloned between the HindIII and EcoRI sites of plasmid pGene/V5-His version B (Invitrogen) multiple cloning site. The recombinant plasmid was linearized with SapI and introduced into GeneSwitch-CHO cells (Invitrogen) by lipofection according to the protocol supplied with the LipofectAMINE PLUS Reagent Kit (Gibco Life Technologies). Stably transfected cells were selected for on selective growth medium consisting of complete F-12 (HAM) medium plus zeocin (300 μg/ml) and hygromycin (100 μg/ml) by feeding the cells with selective medium every 3 to 4 days until foci could be seen. Antibiotic resistant cells were treated with trypsin (0.25%) solution for 3 min, diluted with 5 volumes of complete F-12 (HAM) medium and harvested by centrifugation at 250×g for 5 min. Cells were then resuspended in complete F-12 (HAM) medium, and diluted to a final cell density of five cells per ml. One hundred microliters of cell suspension was used to seed the wells of two 96-well plates. Only wells containing single foci were subcultured on selective medium in 12 and 24-well plates. Expression of TcdB was induced in the different cell lineages of transfected CHO cells by the addition of mifepristone (10−8 M), to the selective medium. GeneSwitch-CHOpGene/TcdB1-556 a lineage of transfected cells showing nearly 100% rounding in 24 h in the presence of mifepristone was identified and chosen for the experiments reported herein.
  • Statistical Analysis
  • Results were analyzed using the statistical software component of Excel 2001. Sample variations are reported as standard deviation from the mean, and significance was confirmed by student's T-test (p<0.05).
  • Utility
  • Since the preferred embodiments of the mutants contemplated herein are inactive, and therefore are not lethal, but are effective in binding to native TcdB toxin, they will make excellent therapeutics or vaccines against C. difficile toxins or infections in their pure and partially pure forms. The mutant toxin may be therapeutically administered to inhibit active TcdB in subjects having existing C. difficile infections or circulating TcdB toxin, for example, for treating or inhibiting diarrhea or pseudomembranous colitis.
  • The administration of a human vaccine comprising one or more of the mutants described herein is applicable to the prevention or treatment of a C. difficile infection in a human or animal. The vaccine may be administered by epicutaneous injection, subcutaneous injection, intramuscular injection, interdermal injection (injection by infusion), sustained-release repository, aerosolization, parenteral delivery, inoculation into an egg, and the like, by known techniques in the art. Although this approach is generally satisfactory, other routes of administration, such as i.v. (into the blood stream) may also be used in a manner known to those of ordinary skill in the art. In addition, the vaccine can be given together with adjuvants and/or immuno-modulators to boost the activity of the vaccine and the subject's response, the subject being a human or animal as described elsewhere herein.
  • The amount of protein in each therapeutic or vaccine dose can be selected as an amount which induces an antitoxin or immunoprotective response without significant, adverse side effects. Such amount in a vaccine will vary depending upon which specific immunogen is employed, how it is presented, and the size of the subject treated. Generally, it is expected that each therapeutic or immunogenic dose of the protein will comprise 0.1-1000 μg/kg of weight of the subject, preferably 0.2-100 μg/kg, and most preferably 1-10 μg/kg. An optimal amount for a particular vaccine can be ascertained by standard studies involving observation of appropriate immune responses in subjects. Following an initial vaccination, subjects may receive one or several booster immunization adequately spaced. Therapeutic doses for inhibiting TcdB toxin may also be from 10 μg-1 mg/kg, for example.
  • Accordingly in one aspect, the invention provides a method of treatment comprising administering an effective amount of a vaccine of the present invention to a subject. The vaccine formulations of the present invention may be used for both prophylactic and therapeutic purposes. The vaccine compositions of the present invention can be formulated according to known methods of preparing pharmaceutically useful compositions, whereby these materials are combined in a mixture with a pharmaceutically acceptable carrier vehicle. Suitable vehicles and their formulation are described, for example, in Remingtons' Pharmaceutical Sciences, (Mack Publishing Co., 1980).
  • The TcdB toxin mutants can be administered in combination with a pharmaceutical carrier compatible with the protein and the subject. Suitable pharmacological carriers include, for example, physiological saline (0.85%), phosphate-buffered saline (PBS), Tris hydroxymethyl aminomethane (TRIS), Tris-buffered saline, and the like. The protein may also be incorporated into a carrier which is biocompatible and can incorporate the protein and provide for its controlled release or delivery, for example, a sustained release polymer such as a hydrogel, acrylate, polylactide, polycaprolactone, polyglycolide, or copolymer thereof. An example of a solid matrix for implantation into the subject and sustained release of the protein antigen into the body is a metabolizable matrix, as known in the art.
  • Adjuvants may be included in the vaccine to enhance the immune response in the subject. Such adjuvants include, for example, aluminum hydroxide, aluminum phosphate, Freund's Incomplete Adjuvant (FCA), liposomes, ISCOM, and the like. The vaccine may also include additives such as buffers and preservatives to maintain isotonicity, physiological pH and stability. Parenteral and intravenous formulations of the vaccine may include an emulsifying and/or suspending agent, together with pharmaceutically-acceptable diluents to control the delivery and the dose amount of the vaccine.
  • Factors bearing on the therapeutic or vaccine dosage include, for example, the age and weight of the subject. The range of a given dose is about 25-5000 μg of the purified mutant receptor protein per ml, preferably about 100-1000 μg/ml preferably given in about 0.1-5 ml doses. The vaccine or therapeutic should be administered to the subject in an amount effective to ensure that the subject will develop an immunity to protect against a C. difficle infection or a therapeutic response against a current C. difficile infection. For example, a vaccine for immunizing an about 5-lb. piglet against C. difficile would contain about 100-5000 μg protein per ml, preferably given in 1-5 ml doses. The immunizing dose would then be followed by a booster given at about 21-28 days after the first injection. Preferably, the vaccine is formulated with an amount of the TcdB toxin mutant effective for immunizing a susceptible subject against an infection by more than one strain C. difficile.
  • The present invention further contemplates a monoclonal antibody raised against the C. difficile TcdB toxin mutant. The monoclonal antibody may be prepared by a method comprising immunizing a suitable animal or animal cell with an immunogenic C. difficile TcdB toxin mutant to obtain cells for producing an antibody to said mutant, fusing cells producing the antibody with cells of a suitable cell line, and selecting and cloning the resulting cells producing said antibody, or immortalizing an unfused cell line producing said antibody, e.g. by viral transformation, followed by growing the cells in a suitable medium to produce said antibody and harvesting the antibody from the growth medium in a manner well known to those of ordinary skill in the art. The recovery of the polyclonal or monoclonal antibodies may be preformed by conventional procedures well known in the art, for example as described in Kohler and Milstein, Nature 256, 1975, p. 495.
  • In a further aspect, the invention relates to a diagnostic agent which comprises a monoclonal antibody as defined above. Although in some cases when the diagnostic agent is to be employed in an agglutination assay in which solid particles to which the antibody is coupled agglutinate in the presence of a C. difficile toxin in the sample subjected to testing, no labeling of the monoclonal antibody is necessary, it is preferred for most purposes to provide the antibody with a label in order to detect bound antibody. In a double antibody (“sandwich”) assay, at least one of the antibodies may be provided with a label. Substances useful as labels in the present context may be selected from enzymes, fluorescers, radioactive isotopes and complexing agents such as biotin. In a preferred embodiment, the diagnostic agent comprises at least one antibody covalently or non-covalently bonded coupled to a solid support. This may be used in a double antibody assay in which case the antibody coupled to the solid support is not labeled. The solid support may be selected from a plastic, e.g. latex, polystyrene, polyvinylchloride, nylon, polyvinylidene difluoride, cellulose, e.g. nitrocellulose and magnetic carrier particles such as iron particle coated with polystyrene.
  • The monoclonal antibody of the invention may be used in a method of determining the presence of C. difficile TcdB toxin in a sample, such as blood, plasma, or serum, the method comprising incubating the sample with a monoclonal antibody as described above and detecting the presence of bound toxin resulting from said incubation. The antibody may be provided with a label as explained above and/or may be bound to a solid support as exemplified above.
  • In a preferred embodiment of the method, a sample desired to be tested for the presence of C. difficile is incubated with a first monoclonal antibody coupled to a solid support and subsequently with a second monoclonal or polyclonal antibody provided with a label. In an alternative embodiment (a so-called competitive binding assay), the sample may be incubated with a monoclonal antibody coupled to a solid support and simultaneously or subsequently with a labeled C. difficile TcdB toxin competing for binding sites on the antibody with any toxin present in the sample. The sample subjected to the present method may be any sample suspected of containing a C. difficile TcdB toxin. Thus, the sample may be selected from bacterial suspensions, bacterial extracts, culture supernatants, animal body fluids (e.g. serum, colostrum or nasal mucous) and intermediate or final vaccine products.
  • Apart from the diagnostic use of the monoclonal antibody of the invention, it is contemplated to utilize a well-known ability of certain monoclonal antibodies to inhibit or block the activity of biologically active antigens by incorporating the monoclonal antibody in a composition for the passive immunization of a subject against diseases caused by C difficile producing a TcdB toxin, which comprises a monoclonal antibody as described above and a suitable carrier or vehicle. The composition may be prepared by combining a therapeutically effective amount of the antibody or fragment thereof with a suitable carrier or vehicle. Examples of suitable carriers and vehicles may be the ones discussed above in connection with the vaccine of the invention. It is contemplated that a C. difficile-specific antibody may be used for prophylactic or therapeutic treatment of a subject having a C. difficile infection or a subject which may potentially incur a C. dfficile infection.
  • A further use of the monoclonal antibody of the invention is in a method of isolating a C. difficile TcdB toxin, the method comprising adsorbing a biological material containing said toxin to a matrix comprising an immobilized monoclonal antibody as described above, eluting said toxin from said matrix and recovering said toxin from the eluate. The matrix may be composed of any suitable material usually employed for affinity chromatographic purposes such as agarose, dextran, controlled pore glass, DEAE cellulose, optionally activated by means of CNBr, divinylsulphone, etc. in a manner known per se.
  • In a still further aspect, the present invention relates to a method of determining the presence of antibodies against C. difficile TcdB toxin in a sample, the method comprising incubating the sample with C. difficile TcdB toxin and detecting the presence of bound antibody resulting from incubation. A diagnostic agent comprising the TcdB toxin used in this method may otherwise exhibit any of the features described above for diagnostic agents comprising the monoclonal antibody and be used in similar detection methods although these will detect bound antibody rather than bound TcdB toxin as such. The diagnostic agent may be useful, for instance as a reference standard or to detect anti-toxin antibodies in body fluids, e.g. serum, colostrum or nasal mucous, from subjects exposed to the toxin or C. difficile.
  • The monoclonal antibody of the invention may be used in a method of determining the presence of a C. difficile toxin, in a sample, the method comprising incubating the sample with a monoclonal antibody and detecting the presence of bound toxin resulting from said incubation.
  • The present invention further contemplates a nucleic acid sequence encoding a C. difficile TcdB toxin mutant wherein the nucleic acid sequence is a cDNA similar to a cDNA which encodes native C. difficile TcdB toxin, but differs therefrom only in having instead a substituted codon which encodes the substituted amino acid or amino acids in the mutant TcdB toxin, as defined herein, and wherein the substituted codon is any codon known to encode the substitute amino acid residue. The mutant TcdB toxin described herein may be produced by well-known recombinant methods using cDNA encoding the mutant TcdB toxin, the cDNA having been transfected into a host cell in a plasmid or other vector.
  • In particular, the present invention contemplates any antigenic Clostridium difficile TcdB toxin mutant wherein the TcdB toxin mutant lacks the toxicity of a native C. difficile TcdB toxin.
  • As noted above, the invention contemplates a vaccine for use in immunizing a human or an animal against an infection by Clostridium difficile, the vaccine comprising a purified non-toxic C. difficile TcdB toxin mutant.
  • Alternatively, the present invention contemplates a method for immunizing a subject against an infection by Clostridium difficile by administering an effective quantity of a vaccine comprising at least one purified non-toxic C. difficile TcdB toxin mutant as defined elsewhere herein. In this method, the vaccine may be administered by epicutaneous injection, subcutaneous injection, intramuscular injection, interdermal injection, intravenous injection, sustained-release repository, aerosolization, parenteral delivery, or inoculation into an egg. In one embodiment of the method, the vaccine induces an effective antibody titer to prevent or eliminate the infection without administration of a booster of the vaccine.
  • The present invention further contemplates a serum for treating a subject with an existing a Clostridium difficile infection comprising antibodies against a C. difficile TcdB toxin wherein the antibodies are raised against a C. difficile TcdB toxin mutant as defined elsewhere herein.
  • The present invention further contemplates an antibody against a Clostridium difficile TcdB toxin wherein said antibody is raised against a C. difficile TcdB toxin mutant as defined elsewhere herein.
  • The present invention further contemplates a method of treating a human or animal having, or disposed to having, a Clostridium difficile infection, comprising administering to the subject a therapeutically effective amount of an antibody against to an TcdB toxin of C. difficile, the antibody raised against a C. dfficile TcdB toxin mutant as defined elsewhere herein. The method for a Clostridium difficile infection may comprise administering a serum comprising the antibodies effective against C. difficile TcdB toxin.
  • The present invention further contemplates a method of making a hybridoma which secretes an antibody against C. difficile TcdB toxin, the method comprising fusing a lymphocyte from an animal immunized with a C. difficile TcdB toxin mutant with cells capable of replicating indefinitely in cell culture to produce the hybridoma and further isolating the hybridoma. The hybridoma may further secrete an antibody against C. difficile TcdB toxin.
  • Additionally, the present invention further contemplates an immunoassay for C. difficile TcdB toxin in which a sample is contacting a sample which may contain a C. difficile TcdB toxin or a portion thereof with an antibody raised against a C. difficile TcdB toxin mutant to form an antibody-TcdB toxin complex and further detecting the antibody-TcdB toxin complex to determine the presence of the C. difficile TcdB toxin.
  • The present invention further contemplates a polynucleotide which encodes a mutant of C. difficile TcdB toxin polypeptide as defined herein. In addition, the present invention further contemplates a vector containing a polynucleotide which encodes a mutant of C. difficile TcdB toxin polypeptide as defined herein. The present invention further contemplates a host cell containing a vector containing a polynucleotide which encodes a mutant of C. difficile TcdB toxin polypeptide as defined herein. The present invention further contemplates a process for producing a mutant of C. difficile TcdB toxin polypeptide by culturing the host cell described herein thereby expressing the mutant and purifying the mutant from the cultured host cell. The present invention further contemplates a non-toxic mutant of C. difficile TcdB toxin comprising a substitution in the cysteine residue of the native form of the toxin.
  • The present invention is not to be limited in scope by the specific embodiments described herein, since such embodiments are intended as but single illustrations of one aspect of the invention and any functionally equivalent embodiments are within the scope of this invention. Indeed, various modifications of the invention in addition to those shown and described herein will become apparent to those skilled in the art from the foregoing description and accompanying drawings.

Claims (29)

1. An isolated mutant of Clostridium difficile TcdB toxin polypeptide comprising:
a modified Clostridium difficile TcdB toxin having SEQ ID NO: 3, SEQ ID NO: 5, (SEQ ID NO: 7, or SEQ ID NO: 9, wherein the mutant is effective in inhibiting or modulating the cytotoxic effect of C difficile TcdB toxin and C sordellii TcsL toxin.
2. A composition comprising the mutant of claim 1 disposed within a pharmaceutically-acceptable carrier.
3. A method of inhibiting, modulating or treating a Clostridium difficile and/or a Clostridium sordellii infection or the symptoms or toxin thereof in a subject, comprising administering an effective amount of the mutant composition of claim 1 to the subject, wherein the mutant is non-cytotoxic.
4. The method of claim 3 wherein the subject is a human.
5. The method of claim 3 wherein the subject is an animal.
6. The method of claim 3 wherein the method comprises administering at least two mutants.
7. A vaccine composition comprising at least one mutant of claim 1 and a pharmaceutically effective carrier, and wherein the mutant is non-toxic.
8. The vaccine composition of claim 7 further comprising an adjuvant.
9. The vaccine composition of claim 7 further comprising mote than one mutant of claim 1.
10. A vaccine composition comprising the mutant of claim 1 or an immunogenic fragment thereof which is effective in generating an antibody which is effective against Clostridium difficile TcdB toxin.
11. A method of immunizing a human or animal against a Clostridium difficile infection comprising treating the human or animal with an immunogenic amount of the vaccine of claim 10.
12. An antibody raised against the mutant of claim 1 wherein the antibody binds to Clostridium difficile TcdB toxin.
13. The antibody of claim 12 wherein the antibody also binds to Clostridium sordellii TcsL toxin.
14. A method of making an antibody against Clostridium difficile TcdB toxin comprising:
immunizing an animal with an immunogenic amount of the mutant of claim 1, wherein the mutant is non-cytotoxic; and
obtaining the antibody from the animal.
15. The method of claim 14 wherein the antibody is also effective against Clostridium sordellii TcsL toxin.
16. A serum comprising the antibody made by the method of claim 14.
17. A method of making a hybridoma which secretes an antibody against Clostridium dfficile TcdB toxin, comprising:
fusing a lymphocyte from an animal immunized with a mutant of claim 1 with cells capable of replicating indefinitely in cell culture to produce the hybridoma, wherein the mutant is non-cytotoxic; and
isolating the hybridoma.
18. A hybridoma produced by the method of claim 17, which hybridoma secretes an antibody against Clostridium difficile TcdB toxin.
19. A method of making a monoclonal antibody that recognizes Clostridium difficile TcdB toxin, comprising isolating the antibody produced by the hybridoma of claim 18.
20. The method of claim 19 wherein the monoclonal antibody also recognizes Clostridium sordellii TcsL toxin.
21. The antibody produced by the method of claim 20 wherein the antibody is humanized.
22. An immunoassay for Clostridium difficile TcdB toxin, comprising:
contacting a sample to be tested for a Clostridium difficile TcdB toxin or a portion thereof with an antibody of claim 12 to form an antibody-TcdB toxin complex; and
detecting the antibody-TcdB toxin complex to determine the presence or absence of the Clostridium difficile TcdB toxin in the sample.
23. The immunoassay of claim 22 wherein the immunoassay is also effective in detecting Clostridium sordellii TcsL toxin.
24. A polynucleotide which encodes the mutant of Clostridium difficile TcdB toxin polypeptide as defined in claim 1.
25. A vector containing the polynucleotide of claim 24.
26. The vector of claim 25 wherein the polynucleotide is operatively associated with an expression control sequence.
27. A host cell containing the vector of claim 25.
28. A process for producing a mutant of Clostridium difficile TcdB toxin polypeptide, comprising:
culturing the host cell of claim 27 thereby expressing the mutant, and wherein the mutant is non-cytotoxic; and
purifying the mutant from the cultured host cell.
29. The mutant of Clostridium difficile TcdB toxin produced by the process of claim 28.
US11/809,970 2002-06-17 2007-06-04 Mutants of clostridium difficile toxin B and methods of use Abandoned US20080107673A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US11/809,970 US20080107673A1 (en) 2002-06-17 2007-06-04 Mutants of clostridium difficile toxin B and methods of use

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US38968502P 2002-06-17 2002-06-17
US10/463,957 US7226597B2 (en) 2002-06-17 2003-06-17 Mutants of Clostridium difficile toxin B and methods of use
US11/809,970 US20080107673A1 (en) 2002-06-17 2007-06-04 Mutants of clostridium difficile toxin B and methods of use

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US10/463,957 Continuation US7226597B2 (en) 2002-06-17 2003-06-17 Mutants of Clostridium difficile toxin B and methods of use

Publications (1)

Publication Number Publication Date
US20080107673A1 true US20080107673A1 (en) 2008-05-08

Family

ID=32312426

Family Applications (2)

Application Number Title Priority Date Filing Date
US10/463,957 Expired - Fee Related US7226597B2 (en) 2002-06-17 2003-06-17 Mutants of Clostridium difficile toxin B and methods of use
US11/809,970 Abandoned US20080107673A1 (en) 2002-06-17 2007-06-04 Mutants of clostridium difficile toxin B and methods of use

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US10/463,957 Expired - Fee Related US7226597B2 (en) 2002-06-17 2003-06-17 Mutants of Clostridium difficile toxin B and methods of use

Country Status (3)

Country Link
US (2) US7226597B2 (en)
AU (1) AU2003299527A1 (en)
WO (1) WO2004041857A2 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2011068953A3 (en) * 2009-12-02 2011-11-03 Tufts University Atoxic recombinant holotoxins of clostridium difficile as immunogens
US8603824B2 (en) 2004-07-26 2013-12-10 Pfenex, Inc. Process for improved protein expression by strain engineering
US20140348844A1 (en) * 2011-09-16 2014-11-27 Ucb Pharma S.A. Neutralising antibodies to the major exotoxins tcda and tcdb of clostridium difficile
WO2014197651A1 (en) * 2013-06-05 2014-12-11 Tufts University Atoxic recombinant holotoxins of clostridium difficile as immunogens
US20150290319A1 (en) * 2012-11-28 2015-10-15 Cnj Holdings, Inc, Antibodies against clostridium difficile
US9394571B2 (en) 2007-04-27 2016-07-19 Pfenex Inc. Method for rapidly screening microbial hosts to identify certain strains with improved yield and/or quality in the expression of heterologous proteins
US9453251B2 (en) 2002-10-08 2016-09-27 Pfenex Inc. Expression of mammalian proteins in Pseudomonas fluorescens
US9580719B2 (en) 2007-04-27 2017-02-28 Pfenex, Inc. Method for rapidly screening microbial hosts to identify certain strains with improved yield and/or quality in the expression of heterologous proteins
US10364298B2 (en) 2011-11-18 2019-07-30 National Research Council Of Canada Clostridium difficile lipoteichoic acid and uses thereof

Families Citing this family (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7653377B1 (en) * 2000-07-07 2010-01-26 Bellsouth Intellectual Property Corporation Pre-paid wireless interactive voice response system with variable announcements
WO2004041857A2 (en) 2002-06-17 2004-05-21 Ballard Jimmy D Mutant of clostridium difficile toxin b and methods of use
MX2011005758A (en) * 2008-12-03 2011-06-28 Boehringer Ingelheim Vetmed Process for production of vaccines.
CN102365097A (en) 2009-02-20 2012-02-29 卫生防护机构 Antibodies to clostridium difficile toxins
GB0921288D0 (en) 2009-12-04 2010-01-20 Health Prot Agency Therapies for preventing or suppressing clostridium difficile infection
KR20130072201A (en) 2010-03-30 2013-07-01 피페넥스 인크. High level expression of recombinant toxin proteins
RU2754446C2 (en) 2011-04-22 2021-09-02 ВАЙЕТ ЭлЭлСи Compositions related to mutant toxin clostridium difficile and their application methods
WO2013084071A2 (en) 2011-12-08 2013-06-13 Novartis Ag Clostridium difficile toxin-based vaccine
AR089797A1 (en) 2012-01-27 2014-09-17 Merck Sharp & Dohme VACCINES AGAINST CLOSTRIDUM DIFFICILE THAT INCLUDE RECOMBINANT TOXINS
US9169304B2 (en) 2012-05-01 2015-10-27 Pfenex Inc. Process for purifying recombinant Plasmodium falciparum circumsporozoite protein
CN104582722A (en) * 2012-09-19 2015-04-29 诺华股份有限公司 Clostridium difficile polypeptides as vaccine
BR122016023101B1 (en) * 2012-10-21 2022-03-22 Pfizer Inc Polypeptide, immunogenic composition comprising it, as well as recombinant cell derived from Clostridium difficile
AU2014257172B2 (en) * 2013-04-22 2019-01-31 Board Of Regents Of The University Of Oklahoma Clostridium difficile vaccine and methods of use
EP3344276B1 (en) 2015-09-03 2020-04-22 The Board of Regents of the University of Oklahoma Peptide inhibitors of clostridium difficile tcdb toxin
US10933126B2 (en) 2018-05-03 2021-03-02 The Board Of Regents Of The University Of Oklahoma Clostridium difficile immunogenic compositions and methods of use

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2004041857A2 (en) 2002-06-17 2004-05-21 Ballard Jimmy D Mutant of clostridium difficile toxin b and methods of use

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10041102B2 (en) 2002-10-08 2018-08-07 Pfenex Inc. Expression of mammalian proteins in Pseudomonas fluorescens
US9458487B2 (en) 2002-10-08 2016-10-04 Pfenex, Inc. Expression of mammalian proteins in pseudomonas fluorescens
US9453251B2 (en) 2002-10-08 2016-09-27 Pfenex Inc. Expression of mammalian proteins in Pseudomonas fluorescens
US8603824B2 (en) 2004-07-26 2013-12-10 Pfenex, Inc. Process for improved protein expression by strain engineering
US9109229B2 (en) 2004-07-26 2015-08-18 Pfenex Inc. Process for improved protein expression by strain engineering
US9394571B2 (en) 2007-04-27 2016-07-19 Pfenex Inc. Method for rapidly screening microbial hosts to identify certain strains with improved yield and/or quality in the expression of heterologous proteins
US10689640B2 (en) 2007-04-27 2020-06-23 Pfenex Inc. Method for rapidly screening microbial hosts to identify certain strains with improved yield and/or quality in the expression of heterologous proteins
US9580719B2 (en) 2007-04-27 2017-02-28 Pfenex, Inc. Method for rapidly screening microbial hosts to identify certain strains with improved yield and/or quality in the expression of heterologous proteins
EP2519257A4 (en) * 2009-12-02 2014-10-15 Univ Tufts Atoxic recombinant holotoxins of clostridium difficile as immunogens
WO2011068953A3 (en) * 2009-12-02 2011-11-03 Tufts University Atoxic recombinant holotoxins of clostridium difficile as immunogens
JP2013512916A (en) * 2009-12-02 2013-04-18 タフツ ユニバーシティー Clostridium difficile non-toxic recombinant holotoxin as immunogen
EP2519257A2 (en) * 2009-12-02 2012-11-07 Tufts University Atoxic recombinant holotoxins of clostridium difficile as immunogens
US20140348844A1 (en) * 2011-09-16 2014-11-27 Ucb Pharma S.A. Neutralising antibodies to the major exotoxins tcda and tcdb of clostridium difficile
US10752676B2 (en) * 2011-09-16 2020-08-25 Ucb Biopharma Sprl Neutralising antibodies to the major exotoxins TCDA and TCDB of Clostridium difficile
US10364298B2 (en) 2011-11-18 2019-07-30 National Research Council Of Canada Clostridium difficile lipoteichoic acid and uses thereof
US20150290319A1 (en) * 2012-11-28 2015-10-15 Cnj Holdings, Inc, Antibodies against clostridium difficile
US10117933B2 (en) * 2012-11-28 2018-11-06 Emergent Biosolutions Canada Inc. Antibodies against Clostridium difficile
WO2014197651A1 (en) * 2013-06-05 2014-12-11 Tufts University Atoxic recombinant holotoxins of clostridium difficile as immunogens

Also Published As

Publication number Publication date
WO2004041857A2 (en) 2004-05-21
US20040028705A1 (en) 2004-02-12
US7226597B2 (en) 2007-06-05
AU2003299527A1 (en) 2004-06-07
WO2004041857A3 (en) 2004-10-28

Similar Documents

Publication Publication Date Title
US20080107673A1 (en) Mutants of clostridium difficile toxin B and methods of use
JP2008029344A (en) Novel compound
JPH10210987A (en) New histidinol dehydrogenase
JPH11253179A (en) New glucosaminidase
MXPA97009338A (en) Transmembrane-seven type receiver
JP2002504304A (en) ribH
JPH11103871A (en) New compound
JPH1189585A (en) New defi
JPH10309194A (en) Topoisomerase i
JP2002262891A (en) Two-component signal transduction system protein derived from staphylococcus aureus
JPH10113189A (en) New rnasep
US20030086946A1 (en) Antigenic, non-toxic mutants of Clostridium septicum alpha toxin and vaccines, antibodies, sera, and methods of treatment therewith
JPH10201484A (en) New ftsl
JPH11243969A (en) New murd
JPH11192091A (en) Seca
CA2247072A1 (en) Bacterial elastin binding protein, nucleic acid sequence encoding same and diagnostic and therapeutic methods of use thereof
JPH11253173A (en) New histidine kinase
JPH11103870A (en) New compound
US6350857B1 (en) Fifty-four homologue (ffh) from streptococcus pneumoniae
JPH11123088A (en) New compound
JPH10313879A (en) New compound
JPH11155582A (en) New glmu
JP2000500326A (en) Fibronectin binding protein B compound
JPH1198991A (en) Novel compound
JPH11137248A (en) Mura

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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