Defined therapeutic microbiota and methods of use thereof

Information

  • Patent Grant
  • 11666611
  • Patent Number
    11,666,611
  • Date Filed
    Tuesday, December 11, 2018
    5 years ago
  • Date Issued
    Tuesday, June 6, 2023
    11 months ago
Abstract
Described herein are methods and compositions for the use of treating and/or preventing Clostridium difficile infections, including recurrent C. difficile infections, in a subject. Aspects of the technology relate to administering to a subject in need thereof a composition comprising a defined therapeutic microbiota comprising, e.g. Clostridial species. Also described herein are biomarker profiles, including a biomarker profile comprising two groups of Clostridial species, that is predictive of the likelihood of recurrent C. difficile infection and/or susceptibility to initial C. difficile infection.
Description
TECHNICAL FIELD

The technology described herein relates to the treatment or prevention of host pathology involving bacterial toxin production.


SEQUENCE LISTING

The instant application contains a Sequence Listing which has been submitted electronically in ASCII format and is hereby incorporated by reference in its entirety. Said ASCII copy, created on Dec. 28, 2018, is named 043214-090870WOPT_SL.txt and is 116,476 bytes in size.


BACKGROUND

The healthy human gastrointestinal tract is generally host to 300 to 1,000 or more different species of microorganisms. These commensal organisms serve a wide range of functions increasingly recognized as mutualistic and directly connected to the health of the host, including assisting digestion, metabolism, immune function, and colonization resistance against pathogens, among others (Guarner, F. “Enteric flora in health and disease.” Digestion 73 Suppl 1:5-12 (2006)).



Clostridium difficile (C. difficile) infection is the most common cause of nosocomial diarrhea, most often arising in individuals treated with antibiotics, accounting for 10-20% of antibiotic-associated diarrhea and most cases of colitis associated with antibiotic use. C. difficile can be present in the healthy gut microbiota, but the pathogen's biomass and toxin production is kept in check by the indigenous gut flora. Further, when stressed, C. difficile can generate spores that can tolerate extreme conditions many active bacteria cannot, and C. difficile spores are also resistant to antibiotics and many cleaners. Thus, as the normal intestinal flora is disturbed by antibiotic use, the C. difficile spores can remain, leading to their germination and proliferation a large population of C. difficile; perturbation of the healthy microbiota caused by antibiotic use is believed to provide an advantage to C. difficile, allowing it to proliferate and elaborate toxins. The mortality for C. difficile infection is estimated at 1-2.5%, contributing to 15,000-30,000 deaths annually in the U.S. (Ananthakrishnan, A. N. “Clostridium difficile infection: epidemiology, risk factors and management,” Nat Rev Gastroenterol Hapatol, 8:17-26 (2011).


While the importance of other microbial species in suppressing C. difficile populations is generally acknowledged, and some species, including Lactobacilli, Enterococci, and some Bifidobacteria and Bacteroides species, have shown varying degrees of inhibitory activity against C. difficile (Borriello, S. P., and Barclay, F. E. 1986. Ibid.; Naaber, P. et al. “Inhibition of Clostridium difficile strains by intestinal Lactobacillus species.” J Med Microbiol 53:551-4 (2004); Rolfe, R. D. et al. “Bacterial interference between Clostridium difficile and normal fecal flora.” J. Infect. Dis. 143:470-475 (1981); Lee, Y. J. et al. “Identification and screening for antimicrobial activity against Clostridium difficile of Bifidobacterium and Lactobacillus species isolated from healthy infant faeces.” Int J Antimicrob Agents 21:340-6 (2003)), it is not clear which species are essential for suppressing C. difficile in the healthy gut.


SUMMARY

The technology described herein is related to the discovery of commensal bacteria that can suppress toxin production by Gram-positive toxigenic bacteria such as C. difficile and thereby treat or prevent the development of toxin-mediated pathology. Indeed, it was found that as few as a single species of bacterium can provide complete protection from otherwise fatal C. difficile infection in murine models described herein. Suppression of toxin production provides an alternative route to treatment of C. difficile-mediated pathology, in that it can be sufficient for treatment to just suppress production of the pathology-generating toxin without necessarily killing the microbe.


Described herein are defined therapeutic microbiota that treat and/or prevent C. difficile infection, as the term is used herein, including recurrent C. difficile infection. Highly effective therapeutic microbiota include species that are highly proteolytic and that promote Stickland fermentation by C. difficile. In one embodiment, the single, highly proteolytic, Stickland fermenting species Clostridium bifermentans can provide complete protection against fatal C. difficile infection. In another embodiment, a defined consortium of just two species, C. bifermentans and C. scindens, another proteolytic, Stickland fermenting Clostridial species, provides highly effective protection against pathology caused by C. difficile and its toxin production. It is anticipated that other proteolytic, Stickland fermenting species can also provide benefits, including, as non-limiting example, other proteolytic, Stickland fermenting Clostridia.


The examination of the mechanisms through which the identified bacterial species affect C. difficile toxin production provide insight into the ability to suppress the expression of toxins by other toxigenic species of Gram positive, spore-forming anaerobes, as well as insights into the properties of other species likely to perform the same toxin-suppression.


Also described herein is the identification of a biomarker profile, comprising two groups of Clostridial species, that is predictive of the likelihood of recurrent C. difficile infection and/or susceptibility to initial C. difficile infection. When the relative abundance of these groups in a microbiota sample are low relative to healthy gut microbiota, it is considerably more likely that C. difficile infection will arise and/or be recurrent.


Also described herein are diagnostic methods that exploit markers such as proteolytic activity or protease expression, markers of Stickland fermentation (at the protein, nucleic acid, and/or metabolite levels), including: i) methods for the determination of the efficacy of bacteriotherapy for infection with a toxigenic, Gram positive, spore-forming bacterium such as C. difficile; ii) methods for predicting the likelihood of recurrent infection with a toxigenic, Gram positive, spore-forming bacterium such as C. difficile; and iii) methods of determining the likelihood of infection with a toxigenic, Gram positive, spore-forming bacterium such as C. difficile.


In one aspect, described herein is a pharmaceutical composition comprising an oral formulation comprising C. scindens and C. bifermentans bacteria.


In another aspect, described herein is a composition comprising C. scindens and C. bifermentans bacteria, wherein one or both of the C. scindens and C. bifermentans bacteria is in dried, viable form.


In another aspect, described herein is a pharmaceutical composition comprising a formulation comprising C. scindens and C. bifermentans bacteria, wherein the composition does not comprise Bacteroides species or Escherichia species.


In one embodiment of these aspects and all such aspects described herein, one or both of the C. scindens and C. bifermentans bacteria are in spore form.


In another embodiment of these aspects and all such aspects described herein, one or both of the C. scindens and C. bifermentans bacteria are not in spore form.


In another embodiment of these aspects and all such aspects described herein, the C. scindens and C. bifermentans bacteria are present as a mixture of metabolically active and spore forms.


In another embodiment of these aspects and all such aspects described herein, the composition comprises a capsule or microcapsule, or a composition formulated for enteric delivery.


In another embodiment of these aspects and all such aspects described herein, wherein one or both of the C. scindens and C. bifermentans bacteria are in dried viable form.


In another embodiment of these aspects and all such aspects described herein, the composition does not comprise C. sardiniensis bacteria.


In another embodiment of these aspects and all such aspects described herein, the composition does not comprise any other Clostridium species.


In another embodiment of these aspects and all such aspects described herein, the composition does not contain Bacteroides species or Escherichia coli.


In another embodiment of these aspects and all such aspects described herein, the formulation comprises no other bacteria.


In another aspect, described herein is a pharmaceutical composition comprising an oral formulation comprising a first bacterial population that has 16S rDNA with a sequence at least 97% identical to a 16S rDNA sequence present in a reference C. scindens bacterium, and a second bacterial population that has 16S rDNA with a sequence at least 97% identical to a 16S rDNA sequence present in a reference C. bifermentans bacterium.


In another aspect described, herein is a composition comprising a first bacterial population that has 16S rDNA with a sequence at least 97% identical to a 16S rDNA sequence present in a reference C. scindens bacterium, and a second bacterial population that has 16S rDNA with a sequence at least 97% identical to a 16S rDNA sequence present in a reference C. bifermentans bacterium, wherein one or both of the C. scindens and C. bifermentans bacteria is in dried, viable form.


In another aspect, described herein is a pharmaceutical composition comprising a formulation comprising a first bacterial population that has 16S rDNA with a sequence at least 97% identical to a 16S rDNA sequence present in a reference C. scindens bacterium, and a second bacterial population that has 16S rDNA with a sequence at least 97% identical to a 16S rDNA sequence present in a reference C. bifermentans bacterium wherein the composition does not comprise Bacteroides species or Escherichia species.


In one embodiment of these aspects and all such aspects described herein, one or both of the C. scindens and C. bifermentans bacteria are in spore form.


In another embodiment of these aspects and all such aspects described herein, one or both of the C. scindens and C. bifermentans bacteria are not in spore form.


In another embodiment of these aspects and all such aspects described herein, the C. scindens and C. bifermentans bacteria are present as a mixture of metabolically active and spore forms.


In another embodiment of these aspects and all such aspects described herein, the composition comprises a capsule or microcapsule, or a composition formulated for enteric delivery.


In another embodiment of these aspects and all such aspects described herein, one or both of the C. scindens and C. bifermentans bacteria are in dried viable form.


In another embodiment of these aspects and all such aspects described herein, the composition does not comprise C. sardiniensis bacteria.


In another embodiment of these aspects and all such aspects described herein, the composition does not comprise Bacteroides species or Escherichia coli.


In another embodiment of these aspects and all such aspects described herein, the composition does not comprise any other Clostridium species.


In another embodiment of these aspects and all such aspects described herein, the composition in which the formulation comprises no other bacteria.


In another embodiment of these aspects and all such aspects described herein, further comprises a prebiotic.


In another embodiment of these aspects and all such aspects described herein, the composition further comprises a microbe that supports C. scindens and/or C. bifermentans.


In another embodiment of these aspects and all such aspects described herein, the microbe that supports C. scindens is Ruminococcus obeum.


In another embodiment of these aspects and all such aspects described herein, the composition is for use in the treatment of C. difficile infection.


In one embodiment of this aspect and all such aspects described herein, the use comprises suppressing the expression of C. difficile toxin.


In another embodiment of this aspect and all such aspects described herein, the use comprises promoting a shift towards use of the proline reductase pathway of Stickland fermentation in C. difficile.


In another embodiment of this aspect and all such aspects described herein, the use comprises inducing CodY activity or expression in C. difficile.


In another embodiment of this aspect and all such aspects described herein, the use comprises promoting ethanolamine utilization by C. difficile.


In another aspect described herein is a method comprising administering a composition of any embodiment of the aspects above to a subject in need thereof.


In one embodiment of this aspect and all such aspects described herein is a method, the subject has or has been diagnosed with C. difficile infection.


In another embodiment of this aspect and all such aspects described herein, the C. difficile infection is recurrent.


In another embodiment of this aspect and all such aspects described herein, the subject is at risk of C. difficile infection or recurrent C. difficile infection.


In another embodiment of this aspect and all such aspects described herein, the administration is oral.


In another embodiment of this aspect and all aspects described herein the method comprises administering a composition directly to the colon of a subject in need thereof.


In another embodiment of this aspect and all such aspects described herein, administration is via colonoscopy or enema.


In another embodiment of this aspect and all such aspects described herein, the method the subject is receiving or has recently received antibiotic treatment.


In another aspect, described herein is a method of treating an infection, the method comprising administering an antibiotic and a composition of the aspects described herein.


In another aspect, as described herein is a method of treating an infection compromising administering an antibiotic and a composition as described herein, wherein the composition is administered before or concurrently with the antibiotic.


In another aspect, described herein is a method of treating an infection compromising administering an antibiotic and a composition as described herein, wherein the composition is administered after a course of an antibiotic.


In another aspect, described herein is a method of predicting recurrence of C. difficile infection in a subject, the method comprising:

    • (a) determining the relative abundance of all operational taxonomic units (OTUs) in a sample of the subject's stool that are >90% identical to a reference sequence of C. scindens;
    • (b) determining the relative abundance of all operational taxonomic units (OTUs) in a sample of the subject's stool that are >90% identical to a reference sequence of C. hylemonae; and
    • (c) summing the relative abundances determined in steps (a) and (b), wherein a sum of relative abundances less than or equal to 1% indicates an increased risk of C. difficile recurrence relative to a subject in which the sum of relative abundances is greater than 1%.


In another embodiment of this aspect, the reference sequences for C. scindens and C. hylemonae are 16S rDNA sequences.


In another embodiment of this aspect, the determining steps are performed on samples taken before, during or after the subject has been treated with antibiotics for C. difficile infection.


In another embodiment of this aspect, the determining steps are performed on samples taken after the subject has been treated with antibiotics for C. difficile infection.


In another embodiment of this aspect, and all such aspects described herein, the method further comprises the step, when the sum of relative abundances is at or below 1%, of administering a composition as described herein above.


In another aspect, described herein is a method of suppressing expression of a bacterial toxin in a subject, the method comprising administering a defined bacterial microbiota comprising at least one bacterial organism that encodes and secretes a protease and/or performs Stickland fermentation.


In another aspect, described herein is a method of treating or preventing a pathology caused by expression of a bacterial toxin, the method comprising administering a defined bacterial microbiota comprising at least one bacterial organism that encodes and secretes a protease and/or performs Stickland fermentation.


In another aspect, described herein is a method of promoting CodY expression or activity in a C. difficile bacterium in a subject, the method comprising administering a defined bacterial microbiota comprising a bacterial organism that encodes and secretes a protease and/or performs Stickland fermentation.


In another aspect, described herein is a method of promoting ethanolamine utilization by a C. difficile bacterium in a subject, the method comprising administering a defined bacterial microbiota comprising a bacterial organism that encodes and secretes a protease and/or performs Stickland fermentation.


In one embodiment of these aspects and all such aspects described herein, at least one bacterial organism encodes and secretes at least one protease selected from the group consisting of: a protease of PATRIC ID fig|186802.30.peg.279; a protease of PATRIC ID fig|186802.30.peg.290; a protease of PATRIC ID fig|186802.30.peg.313; a protease of PATRIC ID fig|186802.30.peg.414; a protease of PATRIC ID fig|186802.30.peg.543; a protease of PATRIC ID fig|186802.30.peg.2205; a protease of PATRIC ID fig|186802.30.peg.2313; a protease of PATRIC ID fig|186802.30.peg.2680; a protease of PATRIC ID fig|186802.30.peg.2745; a protease of PATRIC ID fig|186802.30.peg.2746; a protease of PATRIC ID fig|186802.30.peg.830; a protease of PATRIC ID fig|186802.30.peg.921; a protease of PATRIC ID fig|186802.30.peg.936; a protease of PATRIC ID fig|186802.30.peg.3000; a protease of PATRIC ID fig|186802.30.peg.3018; a protease of PATRIC ID fig|186802.30.peg.3019; and a protease of PATRIC ID fig|186802.30.peg.3065.


In another embodiment of these aspects and all such aspects described herein, at least one protease performs the proteolysis reaction of enzymes of Enzyme Commission number (E.C. number) EC 3.4.21.-; EC 3.4.21.53; or EC 3.4.21.92.


In another embodiment of these aspects and all such aspects described herein, at least one bacterial organism encodes and expresses one or more of D-proline reductase, Glycine reductase, Thioredoxin, or Choloylglycine hydrolase.


In another embodiment of these aspects and all such aspects described herein, the at least one bacterial organism falls within Clostridial cluster I, XI, or XIVa, and does not express a pathology-causing bacterial toxin.


In another embodiment of these aspects and all such aspects described herein, the bacterial organism in Clostridial cluster I is selected from C. sporogenes, and C. histolyticum.


In another embodiment of these aspects and all such aspects described herein, the bacterial organism in Clostridial cluster XI is selected from C. bifermentans, C. hiranonis, and P. anaerobius.


In another embodiment of these aspects and all such aspects described herein, the bacterial organism in Clostridial cluster XIVa is selected from C. scindens, C. clostriiforme, and C. nexile.


In another embodiment of these aspects and all such aspects described herein, the at least one bacterial organism inhibits sorbitol/mannitol fermentation by C. difficile.


In another embodiment of these aspects and all such aspects described herein, the at least one bacterial organism promotes Stickland fermentation through the acceptor amino acid proline, or activation of proline reductase.


In another embodiment of these aspects and all such aspects described herein, the at least one bacterial organism promotes 5-aminovalerate production.


In another embodiment of these aspects and all such aspects described herein, the bacterial toxin is a C. difficile toxin.


In another embodiment of these aspects and all such aspects described herein, the bacterial organism is C. bifermentans and/or C. scindens.


In another embodiment of these aspects and all such aspects described herein, suppressing expression of a bacterial toxin comprises inhibition of butyrate, codY, ccpA, tcdR, and/or tcdA production.


In another aspect, described herein is a method of suppressing expression of a bacterial toxin in the gut of a subject, the method comprising administering at least one amino acid that is metabolized by Stickland fermentation.


In another aspect, described herein is a method of treating or preventing a pathology caused by expression of a bacterial toxin, the method comprising administering at least one amino acid that is metabolized by Stickland fermentation.


In one embodiment of this aspect and all such aspects described herein, the at least one amino acid is a Stickland donor or Stickland acceptor.


In another embodiment of this aspect and all such aspects described herein, the Stickland donor is selected from the group consisting of: alanine, leucine, valine, isoleucine, tryptophan, tyrosine and phenylalanine.


In another embodiment of this aspect and all such aspects described herein, the Stickland acceptor is selected from the group consisting of: glycine and proline.


In another embodiment of this aspect and all such aspects described herein, the amino acid is a branched-chain amino acid, a branched-keto amino acid, or an aromatic amino acid.


In another embodiment of this aspect and all such aspects described herein, the at least one amino acid promotes 5-aminovalerate production.


In another embodiment of this aspect and all such aspects described herein, the bacterial toxin is a C. difficile toxin.


In another embodiment of this aspect and all such aspects described herein, suppression of the expression of a bacterial toxin comprises inhibition of butyrate, codY, ccpA, tcdR, and/or tcdA activity or production.


In another aspect, described herein is a method of determining the therapeutic efficacy of a bacterial organism for treatment of a pathology involving expression of a toxin, produced by a Gram-positive spore-forming bacterium, the method comprising measuring in a biological sample obtained from an individual administered the bacterial organism one or more of:

    • a) the amount and/or activity of a secreted proteolytic enzyme;
    • b) the amount and/or activity of bacterial proline reductase;
    • c) the amount or concentration of one or more branched short-chain fatty acids;
    • d) the amount or concentration of one or more branched keto acids; and
    • e) the amount or concentration of Stickland donor and/or Stickland acceptor amino acids and/or 5-aminovalerate;


In one embodiment of this aspect and all such aspects described herein, the bacterial toxin is produced by a Gram-positive, spore-forming bacterium.


In another embodiment of this aspects and all such aspects described herein, the bacterial toxin is a C. difficile toxin.


In another embodiment of this aspect and all such aspects described herein, the pathology comprises expression of a toxin by C. difficile.


In another embodiment of this aspect and all such aspects described herein, Stickland donor amino acids are selected from leucine, isoleucine, valine, alanine, phenylalanine, tryptophan, tyrosine and Stickland acceptor amino acids are selected from glycine, proline, and hydroxyproline.


In another embodiment of this aspect and all such aspects described herein, the sample is a stool sample or a sample from within the colon of the individual.


In another aspect, described herein is a method to predict the risk of developing a disease involving a toxin produced by a Gram positive, spore-forming bacterium, the method comprising measuring in a biological sample obtained from an individual one or more of the following:

    • a) the amount and/or activity of a secreted proteolytic enzyme;
    • b) the amount and/or activity of bacterial proline reductase;
    • b) the amount or concentration of one or more branched short-chain fatty acids;
    • c) the amount or concentration of one or more branched keto acids; and
    • e) the amount or concentration of Stickland donor and/or Stickland acceptor amino acids; and


comparing the amount, concentration or activity measured in one or more of (a)-(e) to a reference, wherein an amount, concentration or activity in one or more of (a)-(e) below the reference indicates increased risk of developing a disease involving a toxin produced by a Gram positive, spore-forming bacterium.


In one embodiment of this aspect and all such aspects described herein, the disease involves expression of a toxin by C. difficile.


In another embodiment of this aspect and all such aspects described herein, the reference comprises a biological sample from a healthy individual.


In another embodiment of this aspect and all such aspects described herein, the biological sample is a stool sample or a sample from within the colon of the individual.


In another embodiment of this aspect and all such aspects described herein, two or more of (a)-(e) are measured.


In another embodiment of this aspect and all such aspects described, three or more of (a)-(e) are measured.


In one embodiment of this aspect and all such aspects described herein, four or more, of (a)-(e) are measured.


In another aspect, described herein is a method of identifying a candidate bacterial organism that is likely to suppress the expression of a toxin by a Gram-positive, spore-forming bacterial pathogen, the method comprising:

    • a) identifying from a database of bacterial genetic information a candidate bacterial organism having in its genome:
      • i) one or more genes encoding a secreted protease enzyme; and/or
      • ii) a gene encoding a proline reductase enzyme; and
    • b) assaying a sample comprising the candidate bacterial organism for the expression of a secreted protease enzyme and/or the proline reductase enzyme;


      wherein the detection of expression of the secreted protease enzyme and/or the expression of the proline reductase enzyme indicates that the candidate bacterial organism is likely to suppress expression of a toxin by a Gram-positive, spore-forming bacterial pathogen.


In one embodiment of this aspect and all such aspects described herein, the candidate bacterial organism is not an opportunistic gut pathogen in humans.


In one embodiment of this aspect and all such aspects described herein, the proline reductase enzyme is an enzyme of E.C. 1.21.4.1.


In another aspect, described herein is a method to predict the risk of developing a spore-forming, toxin-producing Gram-positive bacterial pathogen in the gut or other location, or its recurrence, comprising measuring in a biological sample

    • (a) amounts or unit activity of proteolytic activity;
    • (b) concentrations of branched short chain fatty acids;
    • (c) concentrations of branched keto acids; and/or
    • (d) concentrations of Stickland donor and Stickland acceptor amino acids,


wherein an increase in the amount or activity of at least one of (a)-(d) relative to a biological sample obtained prior to administration identifies a risk of developing a spore-forming, toxin-producing Gram-positive bacterial pathogen in the gut or other location.


In one embodiment of this aspect and all such aspects described herein, the method further comprising, prior to measuring, administering the bacterial organism or amino acid to the subject.


In another embodiment of this aspect and all such aspects described herein, the biological sample is obtained from a subject.


In another embodiment of this aspect and all such aspects described herein, the biological sample is a stool sample.


In another embodiment of this aspect and all such aspects described herein, the biological sample is obtained from the gut.


In another embodiment of this aspect and all such aspects described herein, the gram-positive bacterial pathogen is C. difficile infection.


In another embodiment of a composition comprising a defined bacterial microbiota as described herein, the composition further comprises an amount of one or more free Stickland donor and/or Stickland acceptor amino acids effective to promote Stickland fermentation by a species in the composition or by C. difficile after the composition is administered to a subject.


In another embodiment of a composition comprising a defined bacterial microbiota as described herein, the composition further comprises an amount of a polypeptide substrate effective for proteolysis by proteolytic activity of a bacterial species in the composition to generate amino acids fermentable by Stickland fermentation.


In one embodiment, the polypeptide substrate comprises casein, collagen and/or gelatin. In another embodiment, the polypeptide substrate comprises a synthetic polymer or copolymer polypeptide hydrolysable by a proteolytic activity of a species in the composition to generate Stickland fermentable amino acids.


In one embodiment, the synthetic polymer comprises a poly[N] polymer, where N is a Stickland donor amino acid selected from leucine, isoleucine, valine, alanine, phenylalanine, tryptophan, and tyrosine or a Stickland acceptor amino acids selected from glycine and proline.


In another embodiment, the synthetic copolymer comprises a poly[N,X] copolymer, where N and X are Stickland donor amino acids selected from leucine, isoleucine, valine, alanine, phenylalanine, tryptophan, and tyrosine or Stickland acceptor amino acids selected from glycine and proline.


Definitions

For convenience, the meaning of some terms and phrases used in the specification, examples, and appended claims, are provided below. Unless stated otherwise, or implicit from context, the following terms and phrases include the meanings provided below. The definitions are provided to aid in describing particular embodiments, and are not intended to limit the claimed technology, because the scope of the technology is limited only by the claims. Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this technology belongs. If there is an apparent discrepancy between the usage of a term in the art and its definition provided herein, the definition provided within the specification shall prevail.


Definitions of common terms in microbiology, molecular biology and medicine can be found, for example, in The Merck Manual of Diagnosis and Therapy, 19th Edition, published by Merck Sharp & Dohme Corp., 2011 (ISBN 978-O-911910-19-3); Robert S. Porter et al. (eds.), Black, Jacquelyn G. Microbiology: Principles and Explorations, 9th Edition: Wiley; 9th Edition, 2014, Moore, Veranus A. Principles of Microbiology: A Treatise on Bacteria, Fungi and Protozoa: Forgotten Books, 2012, The Encyclopedia of Molecular Cell Biology and Molecular Medicine, published by Blackwell Science Ltd., 1999-2012 (ISBN 9783527600908); and Robert A. Meyers (ed.), Molecular Biology and Biotechnology: a Comprehensive Desk Reference, published by VCH Publishers, Inc., 1995 (ISBN 1-56081-569-8); Lewin's Genes XI, published by Jones & Bartlett Publishers, 2014 (ISBN-1449659055); Michael Richard Green and Joseph Sambrook, Molecular Cloning: A Laboratory Manual, 4th ed., Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y., USA (2012) (ISBN 1936113414); Davis et al., Basic Methods in Molecular Biology, Elsevier Science Publishing, Inc., New York, USA (2012) (ISBN 044460149X); Laboratory Methods in Enzymology: DNA, Jon Lorsch (ed.) Elsevier, 2013 (ISBN 0124199542); Current Protocols in Molecular Biology (CPMB), Frederick M. Ausubel (ed.), John Wiley and Sons, 2014 (ISBN 047150338X, 9780471503385), Current Protocols in Protein Science (CPPS), John E. Coligan (ed.), John Wiley and Sons, Inc., 2005; and Current Protocols in Immunology (CPI) (John E. Coligan, ADA M Kruisbeek, David H Margulies, Ethan M Shevach, Warren Strober (eds.) John Wiley and Sons, Inc., 2003 (ISBN 0471142735, 9780471142737), the contents of which are all incorporated by reference herein in their entireties.


It should be understood, as discussed elsewhere herein and as known in the art, that healthy individuals can have detectable levels of C. difficile bacteria in their gut, but that the pathogen biomass and toxin production are kept in balance by components of the healthy gut microbiota and do not cause disease until that balance is disturbed, often, but not necessarily exclusively, by antibiotic treatment for a different infection. Thus, as used herein, the term “C. difficile infection” or “CDI” refers not to the mere presence of C. difficile bacteria, but to expression of C. difficile toxin by such bacteria at a level that causes symptoms, and/or to an overgrowth of C. difficile bacteria relative to levels in healthy individuals, with accompanying gastrointestinal pathology, including diarrhea, among other signs and symptoms. C. difficile overgrowth includes or results in the elaboration of C. difficile toxins (which include C. difficile toxins A and B, and cytolethal distending toxins A and B), which are detectable in stool samples of those with C. difficile infection using, for example, commercially available immunoassays (e.g., lateral flow “dipstick” assays). C. difficile overgrowth also increases (as that term is used herein) the relative biomass of C. difficile in the stool as compared to healthy stool.


“Recurrent” C. difficile infection refers to C. difficile infection that occurs after an initial treatment for C. difficile infection, generally antibiotic therapy. Recurrence can occur, for example, days, weeks or months after an initial infection has been treated, including up to 8 months or more after the initial infection.


As used herein, “bacteriotherapy” refers to the administration of live bacteria for the treatment or prevention of a disease or disorder, including, but not limited to C. difficile infection. As the term is used herein, the bacteria administered are viable and metabolically active, or become metabolically active after administration. Thus, bacteriotherapy can include administration of metabolically active bacteria and/or the administration of dried (e.g., lyophilized) viable bacteria or bacterial spores or a mixture or combination of these. It is preferred that bacteriotherapy as described herein result in the proliferation of the administered bacteria, and preferably establishment of a persistent presence of a population of such bacteria in the gut. A “persistent” presence is one that is detectable at least one week or more after administration, including, for example, at least two weeks, at least three weeks, at least four weeks, at least five weeks, at least six weeks, at least seven weeks, at least eight weeks or more, including essentially indefinite establishment of the administered species or strain in the gut of the subject. To the extent that administration does not result in a persistent presence, re-administration, including re-administration at regular intervals is specifically contemplated.


As used herein, the term “oral formulation” refers to a pharmaceutical formulation formulated to be suitable for oral administration. Oral formulations include, for example, elixirs and suspensions including an active agent or agents that render the agent or agents suitable, or more suitable than the agent alone, for oral administration, and can include, for example, cryoprotectants, reducing agents to protect against oxygen exposure, sweeteners or other agents to enhance palatability. Oral formulations also include, for example, formulations in capsules or microcapsules, which can be standard or designed to dissolve substantially only after passage through the acidic environment of the stomach—i.e., enteric encapsulation. Oral formulations can also include, for example, an antacid, which can modify the pH of the stomach and thereby promote survival of a bacteriotherapy composition. Oral formulations also include, for example, an active agent or agents in admixture with or as a component of a foodstuff.


As used herein, the term “viable” encompasses metabolically active, proliferative as well as dried forms of bacteria that can be reconstituted to provide metabolically active, proliferative bacteria, and spores that can germinate to provide metabolically active, proliferative bacteria. It is contemplated that non-viable, killed cells or an extract or preparation derived therefrom may have benefit for inhibiting C. difficile infection. It is also contemplated that while not proliferative, bacterial cells that have been rendered non-proliferative, e.g., by irradiation, yet retain metabolic activity, can also provide a benefit for inhibiting C. difficile infection. In such instances, regular administration of preparations of killed or proliferatively inactivated bacteria as described herein would be specifically contemplated.


As used herein, the term “dried, viable form” refers to a preparation of bacteria that have been dried, e.g., lyophilized, yet retain the capacity, upon re-hydration, to become metabolically and proliferatively active. Methods for the preparation of dried, viable bacteria are known to those of skill in the art. Indeed, C. scindens, C. bifermentans and C. hylemonae species available from American Type Culture Collection (ATCC) are shipped in dried, viable form.


As used herein, the term “does not comprise species X,” “does not contain species X,” means, at a minimum, that a composition does not comprise viable species X. In one embodiment, the composition does not contain species X at all—i.e., even dead or inactivated bacteria of species X are excluded. Similarly, the term “comprises no other bacteria,” means, at a minimum, that the subject composition does not comprise any viable bacteria other than those specified. In one embodiment, “comprises no other bacteria” means the formulation does not comprise any other bacteria at all, whether dead or alive.


As used herein, a “prebiotic” refers to an ingredient that allows or promotes specific changes, both in the composition and/or activity in the gastrointestinal microbiota that may (or may not) confer benefits upon the host. A prebiotic is not directly digestible by humans, but that is readily digestible by and promotes the growth or establishment of one or more probiotic and/or commensal microbes. Non-limiting examples of prebiotics include but are not limited to inulin, fructooligosaccharides, galactooligosaccharides, hemicelluloses (e.g., arabinoxylan, xylan, xyloglucan, and glucomannan), chitin, lactulose, mannan oligosaccharides, oligofructose-enriched inulin, gums (e.g., guar gum, gum arabic and carrageenan), oligofructose, oligodextrose, tagatose, resistant maltodextrins (e.g., resistant starch), trans-galactooligosaccharide, pectins (e.g., xylogalactouronan, citrus pectin, apple pectin, and rhamnogalacturonan-I), dietary fibers (e.g., soy fiber, sugarbeet fiber, pea fiber, corn bran, and oat fiber) and xylooligosaccharides. In one embodiment, a prebiotic comprises a sugar or carbohydrate e.g., a starch or other carbohydrate polymer, or protein formulation that can be digested by C. bifermentans and C. scindens, but not readily by other commensals, and therefore promotes selective expansion of these species.


As used herein, the term “defined bacterial microbiota” refers to one or a combination of known bacteria in a composition to be administered for bacteriotherapy. In a defined bacterial microbiota, the component bacteria are all known and prepared from pure culture; a defined bacterial microbiota excludes stool or fecal material. In one embodiment, the relative proportions of members of a defined microbial microbiota are defined and deliberately set.


As used herein, the term “probiotic” refers to microorganisms that form at least a part of the transient or endogenous flora or microbial consortium and thereby exhibit a beneficial prophylactic and/or therapeutic effect on the host organism. Probiotics are generally known to be clinically safe (i.e., non-pathogenic) by those skilled in the art.


As used herein, a microbe that “supports” another produces one or more metabolites that promotes the growth or maintenance of another microbe. Alternatively, or in addition, a microbe that “supports” another microbe provides or contributes to an environment, e.g., a pH, redox status, nutritional status, or other environmental component that promotes growth of the other, or, for example, inhibits the growth of a third species or class of microbes that inhibits the desired species.


As used herein, the term “at risk” as applied to risk of C. difficile infection refers to an individual in a high risk category for C. difficile infection. These include, for example, those who are receiving or have recently completed a course of antibiotic therapy for a different infection (or for a first infection with C. difficile), those of advanced age, those who are hospitalized or under nursing home care, those who have had prior C. difficile infection, those taking medication for gastric acid suppression (e.g., with proton pump inhibitors or histamine-2 receptor antagonists), those undergoing gastrointestinal procedures, those undergoing chemotherapy, those with inflammatory bowel disease, and those who are immunosuppressed. Additional risk factors include low relative abundance of the biomarker or indicator species C. scindens and/or C. hylemonae, as described herein below.


As used herein, the term “recently received,” in reference to antibiotic treatment refers to having received the last dose of a course of antibiotics to treat C. difficile or another infection. In this context, one has “recently received” antibiotic treatment if the last dose of a course was given two days, three days, four days, five days, six days, one week, eight days, nine days, ten days, eleven days, twelve days, thirteen days, two weeks, fifteen days, sixteen days, seventeen days, eighteen days, nineteen days, twenty days, three weeks, twenty-two days, twenty-three days, twenty-four days, twenty-five days, twenty-six days, twenty-seven days or four weeks previously.


As used herein, the term “operational taxonomic unit (OTU, plural OTUs)” refers to a terminal leaf in a phylogenetic tree and is defined by a specific genetic sequence and all sequences that share a specified degree of sequence identity to this sequence. The specific genetic sequence may be the 16S rRNA sequence or a portion of the 16S rRNA sequence, or it may be a functionally conserved housekeeping gene found broadly across the eubacterial kingdom. OTUs generally share at least 97%, 98%, or 99% sequence identity in the reference sequence, although lower numbers can be applied—it follows that the lower the % identity to the reference sequence, the more species will be encompassed, and the more variation between members of the OTU grouping.


As used herein, the term “clade” refers to the set of OTUs or members of a phylogenetic tree downstream of a statistically valid node in a phylogenetic tree. The clade comprises a set of terminal leaves in the phylogenetic tree that is a distinct monophyletic evolutionary unit.


In microbiology, “16S sequencing” or “16S rRNA” or “16S-rRNA” or “16S” refers to sequence derived by characterizing the nucleotides that comprise the 16S ribosomal RNA gene(s). The bacterial 16S rDNA is approximately 1500 nucleotides in length and is used in reconstructing the evolutionary relationships and sequence similarity of one bacterial isolate to a second isolate using phylogenetic approaches. 16S sequences are used for phylogenetic reconstruction as they are in general highly conserved, but contain specific hypervariable regions that harbor sufficient nucleotide diversity to differentiate genera and species of most bacteria, as well as fungi.


The “V1-V9 regions” of the 16S rRNA refers to the first through ninth hypervariable regions of the 16S rRNA gene that are used for genetic typing of bacterial samples. These regions in bacteria are defined by nucleotides 69-99, 137-242, 433-497, 576-682, 822-879, 986-1043, 1117-1173, 1243-1294 and 1435-1465 respectively using numbering based on the E. coli system of nomenclature. Brosius et al., Complete nucleotide sequence of a 16S ribosomal RNA gene from Escherichia coli, PNAS 75(10):4801-4805 (1978). In some embodiments, at least one of the V1, V2, V3, V4, V5, V6, V7, V8, and V9 regions are used to characterize an OTU. In one embodiment, the V1, V2, and V3 regions are used to characterize an OTU. In another embodiment, the V3, V4, and V5 regions are used to characterize an OTU. In another embodiment, the V4 region is used to characterize an OTU. In another embodiment, the full length of a 16S rRNA is used to characterize an OTU. A person of ordinary skill in the art can identify the specific hypervariable regions of a candidate 16S rRNA by comparing the candidate sequence in question to the reference sequence and identifying the hypervariable regions based on similarity to the reference hypervariable regions.


As used herein, a “protease” is an enzyme which degrades proteins or peptides into smaller components or amino acids. A “proteolytic” species of bacteria or a “proteolytic” anaerobe is an anaerobic bacterial species that expresses and secretes a protease enzyme that generates amino acids fermentable by Stickland fermentation. A “highly” proteolytic species expresses and secretes proteolytic activity at least as great or greater than that of C. scindens as measured by methods known in the art or described herein. In one embodiment, a “highly” proteolytic species expresses and secretes a proteolytic activity at least as great or greater than that of C. bifermentans.


The terms “decrease”, “reduced”, “reduction”, or “inhibit” are all used herein to mean a decrease by a statistically significant amount. In some embodiments, “reduce,” “reduction” or “decrease” or “inhibit” typically means a decrease by at least 10% as compared to a reference level (e.g. the absence of a given treatment or agent) and can include, for example, a decrease by at least about 10%, at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, at least about 99%, or more. As used herein, “reduction” or “inhibition” does not encompass a complete inhibition or reduction as compared to a reference level. “Complete inhibition” is a 100% inhibition as compared to a reference level. Where applicable, a decrease can be preferably down to a level accepted as within the range of normal for an individual without a given disorder.


The terms “increased”, “increase”, “enhance”, or “activate” are all used herein to mean an increase by a statically significant amount. In some embodiments, the terms “increased”, “increase”, “enhance”, or “activate” can mean an increase of at least 10% as compared to a reference level, for example an increase of at least about 20%, or at least about 30%, or at least about 40%, or at least about 50%, or at least about 60%, or at least about 70%, or at least about 80%, or at least about 90% or up to and including a 100% increase or any increase between 10-100% as compared to a reference level, or at least about a 2-fold, or at least about a 3-fold, or at least about a 4-fold, or at least about a 5-fold or at least about a 10-fold increase, or any increase between 2-fold and 10-fold or greater as compared to a reference level. In the context of a marker or symptom, an “increase” is a statistically significant increase in such level.


As used herein, a “subject” means a human or animal. Usually the animal is a vertebrate mammal such as a non-human primate, rodent, domestic animal or game animal. Primates include, for example, chimpanzees, cynomologous monkeys, spider monkeys, and macaques, e.g., Rhesus. Rodents include, for example, mice, rats, woodchucks, ferrets, rabbits and hamsters. Domestic and game animals include, for example, cows, horses, pigs, deer, bison, buffalo, feline species, e.g., domestic cat, or canine species, e.g., dog, fox or wolf. Mammals other than humans can be advantageously used as subjects that represent models of human disease e.g., C. difficile infection. A subject can be male or female. The terms “individual,” “patient” and “subject” are used interchangeably herein.


A subject can be one who has been previously diagnosed with or identified as suffering from or having a condition in need of treatment (e.g. C. difficile infection) or one or more complications related to such a condition, and optionally, have already undergone treatment for the condition or the one or more complications related to the condition. Alternatively, a subject can also be one who has not been previously diagnosed as having such condition or related complications. For example, a subject can be one who exhibits one or more risk factors for the condition or one or more complications related to the condition or a subject who does not exhibit risk factors. Risk factors for C. difficile infection include, but are not limited to, receiving antibiotic therapy for a different infection (or for a first infection with C. difficile), advanced age, hospitalization or nursing home care, prior C. difficile infection, gastric acid suppression (e.g., with proton pump inhibitors or histamine-2 receptor antagonists), gastrointestinal procedures, chemotherapy, inflammatory bowel disease, and immunosuppression. In addition, in those infected with C. difficile, low levels of vitamin D have been shown to be an independent predictor of poor outcome and are associated with higher recurrence. Additional risk factors include low relative abundance of the biomarker or indicator species C. scindens and/or C. hylemonae as described herein below.


A “subject in need” of treatment for a particular condition can be a subject having that condition, diagnosed as having that condition, or at risk of developing that condition.


As used herein, the term “suppressing expression of a bacterial toxin” refers to conditions or treatment(s) that inhibit or reduce, as the terms are defined herein, the expression of a toxin by a bacterium in a subject. In one embodiment, the inhibition or reduction is sufficient to reduce one or more symptoms or one or more markers of bacterial infection in a subject. In one embodiment, the inhibition or reduction is to a level that does not cause symptoms or detectable pathology in the subject. In another embodiment, the suppression acts upon toxin production without necessarily reducing biomass or viability of the bacterium.


As used herein, the terms “treat,” “treatment,” “treating,” or “amelioration” refer to therapeutic treatments, wherein the object is to reverse, alleviate, ameliorate, inhibit, slow down or stop the progression or severity of a condition associated with a disease or disorder, e.g. C. difficile infection, including recurrent C. difficile infection. The term “treating” includes reducing or alleviating at least one adverse effect or symptom of a condition, disease or disorder. Treatment is generally “effective” if one or more symptoms or clinical markers are reduced. Alternatively, treatment is “effective” if the progression of a disease is reduced or halted. That is, “treatment” includes not just the improvement of symptoms or markers, but also a cessation of, or at least slowing of, progress or worsening of symptoms compared to what would be expected in the absence of treatment. Beneficial or desired clinical results include, but are not limited to, alleviation of one or more symptom(s), diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, delay or slowing of disease progression, amelioration or palliation of the disease state, remission (whether partial or total), and/or decreased mortality. Treatment can include suppression of bacterial toxin production, e.g., to a degree such that pathology or symptoms caused by the toxin are reduced.


As used herein, the term “pharmaceutical composition” refers to an active agent in combination with a pharmaceutically acceptable carrier e.g. a carrier commonly used in the pharmaceutical industry. The phrase “pharmaceutically acceptable” is employed herein to refer to those compounds, materials, compositions, and/or dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, allergic response, or other problem or complication, commensurate with a reasonable benefit/risk ratio. In some embodiments of any of the aspects, a pharmaceutically acceptable carrier can be a carrier other than water. In some embodiments of any of the aspects, a pharmaceutically acceptable carrier can be an artificial or engineered carrier, e.g., a carrier in which the active ingredient would not be found to occur in nature. Where the active agent includes one or more bacterial species, whether in active metabolic, dried viable or spore form, the carrier will be compatible with the bacteria and will not interfere with viability or activity of the bacteria.


As used herein, the term “administering,” refers to the placement of a therapeutic or pharmaceutical composition as disclosed herein into a subject by a method or route which results in at least partial delivery of the agent at a desired site. Pharmaceutical compositions comprising agents as disclosed herein can be administered by any appropriate route which results in an effective treatment in the subject. Common routes of administration for bacteriotherapies include oral administration, often, but not necessarily in the form of one or more capsules, and direct administration to the lower GI tract, e.g., via enema or colonoscope.


The term “statistically significant” or “significantly” refers to statistical significance and generally means a two standard deviation (2SD) or greater difference.


Other than in the operating examples, or where otherwise indicated, all numbers expressing quantities of ingredients or reaction conditions used herein should be understood as modified in all instances by the term “about.” The term “about” when used in connection with percentages can mean±1%.


As used herein, the term “comprising” means that other elements can also be present in addition to the defined elements presented. The use of “comprising” indicates inclusion rather than limitation.


The term “consisting of” refers to compositions, methods, and respective components thereof as described herein, which are exclusive of any element not recited in that description of the embodiment.


As used herein the term “consisting essentially of” refers to those elements required for a given embodiment. The term permits the presence of additional elements that do not materially affect the basic and novel or functional characteristic(s) of that embodiment of the technology.


The singular terms “a,” “an,” and “the” include plural referents unless context clearly indicates otherwise. Similarly, the word “or” is intended to include “and” unless the context clearly indicates otherwise. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of this disclosure, suitable methods and materials are described below. The abbreviation, “e.g.” is derived from the Latin exempli gratia, and is used herein to indicate a non-limiting example. Thus, the abbreviation “e.g.” is synonymous with the term “for example.”


Other terms are defined within the description of the various aspects and embodiments of the technology of the following.





BRIEF DESCRIPTION OF THE DRAWINGS


FIGS. 1A-1J presents experimental data of C. difficile gnotobiotic mouse survival studies. FIG. 1A shows a schematic of the Gnotobiotic mouse colonization model of C. difficile infection. Adult Swiss-Webster mice were pre-colonized with a commensal species (C. bifermentans, C. scindens or C. sardiniense for 7 days, prior to oral challenge with 1000 C. difficile spores (ATCC 43255 strain). The survival curve shows the survival post-challenge with C. difficile. At least 8 mice across 2 experiments were assessed per timepoint. Biological samples for metabolomics and microbial gene expression analysis were taken at the following time points: GF controls (first arrow), after 7 days of commensal colonization (second arrow), at 20 hours of C. difficile infection (third arrow), alone or with the given pre-colonized commensal. At least 8 mice across 2 experiments were assessed per timepoint. FIG. 1B: Swiss-Webster germfree mice were associated with a commensal: C. bifermentans, C. sardiniense or C. scindens for 7 days, prior to challenge with 1000 spores of C. difficile strain ATCC43255. Control germfree mice received C. difficile alone. The survival curve shows the survival post-challenge with C. difficile. FIG. 1C. Body condition scores (BCS) of the mice were monitored daily to assess activity, feeding, grooming and tissue turgor for additional clinical symptoms of infection. Swiss-Webster germfree mice associated with a commensal: C. bifermetans, C. sardiniense, C. scindens, prior to challenge with 1000 spores of C. difficile strain ATCC43255. Control germfree mice received C. difficile alone. FIGS. 1D-1J show representative images of H&E stained sections of the colon of the mice. FIG. 1D shows a representative image of a healthy colon from a conventional mouse, 200× magnification showing intact epithelial crypts, mucosal and muscular layers. FIG. 1E shows a representative image of a colon of a Gnotobiotic mouse 24 hours after C. difficile infection showing denudation of the surface epithelium and massive neutrophil influx (200× magnification). FIG. 1F shows a representative image of colon of a Gnotobiotic mouse pre-colonized with C. sardiniense, 24 hr after C. difficile challenge showing massive tissue edema (arrow), epithelial denudation and neutrophil influx, consistent with a toxic megacolon picture (100× magnification). FIG. 1G shows a representative image of colon of a Gnotobiotic mouse pre-colonized with C. bifermentans, 24 hr after C. difficile challenge with some epithelial ballooning but no overt epithelial disruptions or tissue edema (200×). FIG. 1H shows a representative image of colon of a Gnotobiotic mouse with C. scindens 28 days after C. difficile challenge showing intact epithelium and residual inflammatory mucosal infiltrate consisting of neutrophils and lymphocytes (200×). FIG. 1I shows a representative image of colon of a Gnotobiotic mouse precolonized with C. scindens and 28 days after C. difficile challenge, showing a focal area of epithelial damage with surrounding intact epithelium and submucosal (200×). FIG. 1J shows a representative image of colon of a Gnotobiotic mouse precolonized with C. bifermentans at 28 days post-challenge with intact epithelium and submucosa. Resolving inflammatory infiltrates from infection, which are largely lymphocytic, can be seen. However, ongoing areas of focus epithelial damage were not noted.



FIGS. 2A-2C present experimental data that shows that the commensal bacterium C. bifermentans suppresses toxin production by C. difficile. FIG. 2A shows the results of an ELISA of cecal contents from germfree Swiss-Webster mice that were collected at 24 and 48 hours after oral challenge with 1000 spores of the C. difficile ATCC-43255 strain. ToxinB was detected by ELISA and concentration in cecal contents calculated against a standard curve of purified toxinB. 4-8 mice were assayed per condition. Toxin levels in C. sardiniense pre-colonized mice were assessed from those that had survived to 24 or 48 hours. FIGS. 2B-2C present experimental data that show C. bifermentans suppresses toxin production without altering C. difficile biomass. FIG. 2B shows a bar graph showing C. difficile biomass in shed stool samples in the indicated days post-challenge. FIG. 2C shows the levels of C. difficile toxin production with indicated conditions. C. bifermentans precolonization prevents a spike in C. difficile toxin production.



FIGS. 3A-3B show a schematic outlining of the infectious mouse model protocols using conventional mice and the therapeutic intervention. FIG. 3A shows a schematic of the therapeutic intervention: adult conventional mice were treated with intraperitoneal clindamycin 24 hours before receiving 1×10{circumflex over ( )}4 spores of C. difficile strain ATCC 43255. Approximately 20 hours after dosing, as mice first developed signs of symptomatic infection, animals received 5×10{circumflex over ( )}7 CFU of C. bifermentans or C. sardiniense, or control vehicle alone, by gavage and were monitored for 2 additional weeks. FIG. 3B shows the survival curves of clindamycin-treated conventional mice infected with C. difficile and gavaged 20 hours later, upon onset of symptomatic infection, with vehicle alone, 5×10{circumflex over ( )}7 CFU of C. bifermentans or 5×107 CFU of C. sardiniense. At least 8 mice across 2 experimental replicates were studied for each condition.



FIGS. 4A-4B present experimental data showing that Clostridium bifermentans is a highly proteolytic Stickland fermenting species. FIG. 4A shows representative images of chopped meat anaerobic culturing broth inoculated and incubated with either C. bifermentans, C. hiranonis, C. sardiniense, C. scindens, C. ramosum, or C. difficile and assessed for their proteolytic activity using a biochemical protease assay protease in a biological sample. FIG. 4B lists features of the strains with C. difficile, C. bifermentans, C. sardiniense and C. scindens.



FIGS. 5A-5I present experimental data showing that Clostridium bifermentans promotes Stickland fermentation by the Gram-positive toxigenic bacterium C. difficile. The cecal contents of germfree Swiss-Webster mice were collected 20 hours after infection with C. difficile and untargeted metabolomic analysis of cecal samples was performed. FIGS. 5A-5D show the MassSpectometry profiles of Stickland acceptor amino acids in cecal contents of germfree Swiss-Webster mice at 20 hours of infection with C. difficile. Y axis is Log10 MassSpec units of detected compounds. X axis indicates experimental condition: GF-germfree controls (no bacteria); Cdiff-challenge with 1000 C. difficile spores of strain ATCC43255; CSAR-7 days mono-association with C. sardiniense; CBI-7 days mono-association with C. bifermentans; Cdiff+CSAR-mice mono-associated with C. sardiniense for 7 days followed by C. difficile challenge; Cdiff+CBI-mice mono-associated with C. bifermentans for 7 days followed by C. difficile challenge. Each group has 8 mice across two experimental replicates. FIG. 5E shows the Mass Spectrometry profiles of cecal branched-chain amino acids and degradation products in gnotobiotic colonized mice. MassSpectrometry profiles of Stickland donor, branched chain amino acids in cecal contents of germfree Swiss-Webster mice at 20 hours of infection with C. difficile. Y axis is Log10 MassSpec units of detected compounds. X axis indicates experimental condition: GF-germfree controls (no bacteria); Cdiff-challenge with 1000 C. difficile spores of strain ATCC43255; CSAR-7 days mono-association with C. sardiniense; CBI-7 days mono-association with C. bifermentans; Cdiff+CSAR-mice mono-associated with C. sardiniense for 7 days followed by C. difficile challenge; Cdiff+CBI-mice mono-associated with C. bifermentans for 7 days followed by C. difficile challenge. Each group has 8 mice across two experimental replicates. Leucine, isoleucine and valine; levels elevated in Cdiff+CSAR mice, consistent with toxic megacolon picture. Cdiff and CSAR+C. difficile infected mice, which have a more severe clinical and histologic picture have 10× elevated keto-acid derivatives of the BCAA over Cdiff+CBI colonized mice, providing potential biomarkers for a more severe infection. Hydroxy-acid intermediates showing elevation with CSAR alone and CSAR+Cdiff. Isolavaerate, branched-short chain fatty acid product of Stickland fermentations, present in cecal contents of mice colonized with C. difficile and/or C. bifermentans, indicating in vivo Stickland fermenting of leucine as a donor amino acids. Other branched SCFA, sobutyrate, isocaproate and Valerate were not assayable by this method and are not shown. FIG. 5F shows that C. difficile and C. bifermentans use aromatic amino acids in vivo in Stickland reactions. Cecal aromatic amino acids and metabolites in specifically-associated GF mice. Y-axis shows log 10 MS Units. X-axis indicates the colonized state of the mice. 8 adult Swiss-Webster mice at baseline (GF), +7 days commensal colonization with C. sardiniense (CSAR) or C. bifermentans (CBI), 20 hours of infection with C. difficile or 20 hours of infection after 7 days of commensal colonization (Cdiff CSAR, Cdiff+CBI). phenylalanine in cecal contents. Elevated amounts with Cdiff+CSAR relating to toxic megacolon picture. Stickland phenylalanine metabolites that are only present or elevated in mice associated with a Stickland fermenter. C. difficile is the dominant producer of phenylacetate, phenyllactate and phenylpyruvate. C. bifermentans is the dominant producer of phenylpropionate. Levels of C. diff dominant metabolites are reduced in the presence of C. bifermentans. Tryptophan levels and indoleacetate Stickland metabolite; other Stickland metabolites not present or detectable in the Metabolon panel. Both C. diff and CBI use tryptophan in Stickland reactions per elevated production of indoleacetate. tyrosine and Stickland tyrosine metabolites. Both species produce 4-hydroxphenylacetate. Cdiff specifically produces para-cresol (host sulfated derivative shown, native molecule not detected in Metabolon panel). CBI specifically produces 3(4-hydroyphenyl-propionate). FIG. 5G shows the commensal and C. difficile carbohydrate utilization in vivo. Cecal sugars and metabolites in specifically-associated GF mice. Y-axis shows log 10 MS Units. X-axis indicates the colonized state of the mice. 8 adult Swiss-Webster mice at baseline (GF), +7 days commensal colonization with C. sardiniense (CSAR) or C. bifermentans (CBI), 20 hours of infection with C. difficile (C. difficile) or 20 hours of infection after 7 days of commensal colonization (Cdiff CSAR, Cdiff+CBI). Baseline levels of sugars and metabolites shown in germfree mice. When metabolized microbially, amounts decrease from baseline. Levels of glucose in germfree mice versus those colonized with single species show some glucose use by commensals or C. difficile but levels increase when two species are co-colonized. Commensal and C. difficile metabolize fructose in cecal contents with inhibition of metabolism when co-colonized. C. difficile alone metabolizes mannitol/sorbitol with some inhibition of metabolism when co-colonized with C. saridniense. Elevated pyruvate produced in infections with C. difficile or C. difficile+CSAR. Lactate levels increase with C. difficile infection by itself or with commensals. Succinate levels increase with C. difficile by itself or with CSAR but do not change significantly with CBI as compared to CBI-alone. FIG. 5H-5I shows a representative Mass Spectrometry plot for C. sardiniense (CSAR) (FIG. 5H) and for C. bifermentans (CBI) (FIG. 5I).



FIGS. 6A-6B presents the experimental results of C. difficile Short Chain Fatty Acids (SCFA) profile produced in vitro. FIG. 6A shows the concentrations of C. difficile Short Chain Fatty Acids (SCFA) produced in vitro (mM). Cultures were grown in 10 mL of pre-reduced peptone-yeast (PY) broth at starting pH=7 with 1% glucose, 1% mannitol or 1% sorbitol for 72 hours at 37° C. under anaerobic conditions. 100 uL of broth supernatant was extracted and run on an Agilent GC/LC flame ionization detector (FID) instrument with internal standard to quantitate production of short chain fatty acids acetate, propionate and butyrate and the branched short chain fatty acids isobutyrate, isovalerate, isocaproate, valerate or capropate. ND=Not detected below the threshold of detection. FIG. 6B shows the experimental results of the commensal Short Chain Fatty Acids (SCFA) profiles produced in vitro (mM). Millimolar concentrations of SCFA in liquid culture supernatants. Cultures were grown in 10 mL of pre-reduced peptone-yeast (PY) broth or PY+1% glucose (PYG), for 24 hours at 37° C. under anaerobic conditions. 100 uL of broth supernatant was extracted and run on an Agilent GCLC flame ionization detector (FID) instrument with internal standard to quantitate production of short chain fatty acids acetate, propionate and butyrate and the branched short chain fatty acids isobutyrate, isovalerate and isocaproate. ND=Not detected below the threshold of detection; caproate and valerate were not detected in the commensals (data not shown).



FIG. 7 presents the experimental results showing the gene expression of the C. difficile toxin PaLoc and eut (ethanolamine) operon. Comparison of PaLoc and eut gene expression by bacterial RNAseq of cecal contents from C. difficile-infected gnotobiotic Swiss-Webster mice at 20 hours post-inoculation, compared with C. bifermentans colonized mice for 7 days prior to challenge with C. difficile for 20 hours. At least 4 mice/group evaluated. C. bifermentans colonized mice show a >48× decrease in C. difficile tcdR gene expression with concomitant >10× decreases in toxinA (tcdA) and toxinB (tcdB) gene expression. In contrast, C. difficile structural proteins for the ethanolamine carboxysome (eutH, eutK, eutL, eutN) are up-regulated >10× when C. difficile is inoculated into a C. bifermentans-colonized mouse.



FIGS. 8A-8B show a schematic of Stickland Donor Branched Chain Amino Acids (FIG. 8A) and Stickland Donor Aromatic Amino Acids (FIG. 8B).



FIG. 9 shows a schematic of the metabolic pathways utilized by C. difficile.





DETAILED DESCRIPTION

By its very name, C. difficile is notorious for being difficult to treat. Not only is the C. difficile organism resistant to multiple antibiotics commonly used to treat bacterial infections in clinical settings (e.g., aminoglycosides, lincomycin, tetracyclines, erythromycin, clindamycin, penicillins, cephalosporins, and fluoroquinolones), but the use of such antibiotics to treat other infections is one of the greatest risk factors for developing C. difficile pathology. Add to that the ability of C. difficile to sporulate and essentially wait until even an antibiotic that kills its metabolically active form is gone, and it truly deserves its name and reputation.


Toxigenic strains of C. difficile cause pseudomembranous colitis, a severe infectious disease of the colon. Infection often arises with disruptions to the gut microbiota, commonly after use of antibiotics that ablate protective commensals. C. difficile elaborates toxins when starved for carbon, nitrogen or energy (=high levels of NAD+). The toxins rapidly destroy the gut epithelial barrier enabling release of host-derived proteins and carbohydrate sources into the gut lumen which the pathogen then metabolizes. When co-existing with “beneficial” commensal species, C. difficile remains energetically stable and does not elaborate significant toxin. For patients failing multiple rounds of antibiotic treatment, who develop recurrent C. difficile infections, fecal microbiota transplant (FMT) is highly efficacious through its restoration of healthy gut microbial communities. However, the mechanisms of action of FMT, and specific microbes needed to halt toxin production and reduce pathogen biomass remain ill-defined. While microbial conversion of primary host bile acids to secondary acids have been hypothesized to play a key role, mechanisms of action have highlighted effects upon germination of C. difficile spores (primary bile acids stimulate; secondary bile acids inhibit) and less so on metabolic factors that modulate toxin production or overall pathogen biomass.


The technology described herein is related to the discovery of commensal bacteria that can suppress toxin production by Gram-positive toxigenic bacteria such as C. difficile and thereby treat or prevent the development of toxin-mediated pathology. Indeed, it was found that as few as a single species of bacteria can provide complete protection from otherwise fatal C. difficile infection in murine models described herein. Suppression of toxin production provides an alternative route to treatment of C. difficile-mediated pathology, in that it can be sufficient for treatment to just suppress production of the pathology-generating toxin without necessarily killing the microbe.


It is discovered that commensal bacteria that suppress toxin production by Gram-positive toxigenic bacteria such as C. difficile strongly express and secrete protease enzyme activity that generates free amino acids that can be fermented by C. difficile via Stickland fermentation, which metabolizes free amino acids to generate energy. Without wishing to be bound by theory, the picture that emerges is that in order to avoid toxin production by Gram-positive toxigenic bacteria such as C. difficile, the gut environment needs to provide energy and nutrients sufficient to keep such bacteria from becoming stressed and responding by toxin production. Thus, commensal bacteria that secrete proteolytic enzymes can provide therapeutic benefits in suppressing toxin production by Stickland-fermenting Gram-positive toxigenic bacteria such as C. difficile. It is noted that among the protective commensal species found to have protective effects were bacteria that are themselves capable of Stickland fermentation, including, for example, C. bifermentans and C. scindens.


Examination of the effects of protective commensals as described herein provides insights into the mechanisms of protection and further targets for manipulation of toxin production and biomarkers useful for evaluation of, for example, the likelihood of C. difficile recurrence, or the efficacy of treatment.


The following provides additional details regarding C. difficile and the discoveries of highly protective/therapeutic commensal species, and considerations to facilitate the performance of the methods and compositions for the prevention, treatment, prognosis and diagnosis that exploit these discoveries.



Clostridium difficile



Clostridium is a genus of Gram-positive bacteria including around 100 species, which includes several significant human pathogens, including the causative agent of botulism and an important cause of diarrhea, Clostridium difficile. Pathogenic Clostridium strains include but are not limited to C. botulinum that can produce botulinum toxin in food or wounds and can cause botulism. Clostridium difficile can flourish when other members of the gut microbiota are killed during antibiotic therapy, leading to superinfection and potentially fatal pseudomembranous colitis (a severe necrotizing disease of the large intestine). Clostridium perfringens causes a wide range of symptoms, from food poisoning to cellulitis, fasciitis, and gas gangrene. Clostridium tetanii causes tetanus. Clostridium sordellii can cause a fatal infection in rare cases after medical abortions.



C. difficile is a Gram-positive, anaerobic, spore-forming and toxin-producing bacillus, belonging to cluster XI of the Clostridium genus and commonly occurs in the hospital environment, and in the intestines of humans and domesticated animals. C. difficile can cause a spectrum of clinical conditions in humans, collectively known as C. difficile infections (CDI), which range from mild and possibly recurrent diarrhea to life-threatening complications such as pseudomembranous colitis (PMC), toxic megacolon and colonic perforation. As discussed elsewhere herein, C. difficile can occur in the gut of healthy individuals, but be maintained at a level that does not cause illness and/or have the expression of C. difficile toxin suppressed to a degree that the organism does not cause illness.


The clinical spectrum of C. difficile ranges from asymptomatic colonization, mild and self-limiting disease to a severe, life-threatening pseudomembranous colitis, toxic megacolon, sepsis and death. CDI is defined when there is the presence of symptomatic diarrhea defined by three or more unformed stools per 24 h and at least one of the following criteria: a positive laboratory assay for C. difficile toxin A and/or B or toxin-producing C. difficile organism in a stool sample or pseudomembranous colitis or colonic histopathology characteristic of CDI revealed by endoscopy. Toxin is detected, e.g., by immunoassay for the A and or B toxin proteins, and/or by RT-PCR for the toxin-encoding nucleic acids. CDI can be associated with an increased abundance of toxin-producing C. difficile strains, leading to high toxin concentrations within the colon resulting in inflammation and damage of the colonocytes.


The various strains of C. difficile may be classified by a number of methods. One of the most commonly used is polymerase chain reaction (PCR) ribotyping in which PCR is used to amplify the 16S-23S rRNA gene intergenic spacer region of C. difficile. Reaction products from this provide characteristic band patterns identifying the bacterial ribotype of isolates. Toxinotyping is another typing method in which the restriction patterns derived from DNA coding for the C. difficile toxins are used to identify strain toxinotype. The differences in restriction patterns observed between toxin genes of different strains are also indicative of sequence variation within the C. difficile toxin family. Toxin B shows sequence variation in some regions. For example, there's an approximate 13% sequence difference with the C-terminal 60 kDa region of toxinotype 0 Toxin B compared to the same region in toxinotype III.



C. difficile uses a variety of carbon and nitrogen sources for energy and metabolism. Within the gut environment, these sources can originate from the diet, from other commensals, or from the host, particularly when elaborating toxin to disrupt mucosal barriers. Known carbon sources include sugars such as glucose, fructose, mannose, mannitol or sorbitol, the latter two of which are poorly absorbed in the gut and reach the colon. Sources of carbon and nitrogen include amino groups and ethanolamine, a breakdown product of host phosphatidyl ethanolamine from eukaryotic cell membranes, other commensals, or the diet. As noted elsewhere herein, when preferred energy sources or nutrients for C. difficile run low or become limiting, C. difficile toxin production is induced.



C. difficile Toxins


As noted above, C. difficile encodes two toxin proteins, referred to as TcdA and TcdB, or simply herein as C. difficile toxin A and toxin B. TcdA and TcdB are broadly classified as AB toxins, wherein a B subunit is involved in the delivery of an enzymatic A subunit into the cytosol of a target cell. The enzymatic A subunit of TcdA is an N-terminal glucosyltransferase domain (GTD) that inactivates members of the Rho family of small GTPases by glucosylation. The B subunit is composed of three regions: a combined repetitive oligopeptides (CROPS) domain, a delivery/pore-forming domain and an autoprocessing domain (APD).


TcdA is encoded by the tcdA gene. Sequences for TcdA are known for a number of species, e.g., for C. difficile 630 (the TcdA NCBI Gene ID is 4914076) and polypeptide sequence (e.g., YP_001087137.1 (SEQ ID NO: 1). The sequence for the TcdA polypeptide is as follows (SEQ ID NO: 1):










(SEQ ID No: 1)










   1
msliskeeli klaysirpre neyktiltnl deynklttnn nenkylqlkk lnesidvfmn






  61
kyktssrnra lsnlkkdilk eviliknsnt spveknlhfv wiggevsdia leyikqwadi





 121
naeyniklwy dseaflvntl kkaivesstt ealqlleeei qnpqfdnmkf ykkrmefiyd





 181
rqkrfinyyk sqinkptvpt iddiikshlv seynrdetvl esyrtnslrk insnhgidir





 241
anslfteqel lniysqelln rgnlaaasdi vrllalknfg gvyldvdmlp gihsdlfkti





 301
srpssigldr wemikleaim kykkyinnyt senfdkldqq lkdnfkliie sksekseifs





 361
klenlnvsdl eikiafalgs vinqaliskq gsyltnlvie qvknryqfln qhlnpaiesd





 421
nnftdttkif hdslfnsata ensmfltkia pylqvgfmpe arstislsgp gayasayydf





 481
inlqentiek tlkasdlief kfpennlsql teqeinslws fdqasakyqf ekyvrdytgg





 541
slsedngvdf nkntaldkny llnnkipsnn veeagsknyv hyiiqlqgdd isyeatcnlf





 601
sknpknsiii qrnmnesaks yflsddgesi lelnkyripe rlknkekvkv tfighgkdef





 661
ntsefarlsv dslsneissf ldtikldisp knvevnllgc nmfsydfnve etypgkllls





 721
imdkitstlp dvnknsitig anqyevrins egrkellahs gkwinkeeai msdlsskeyi





 781
ffdsidnklk aksknipgla sisediktll ldasvspdtk filnnlklni essigdyiyy





 841
eklepvknii hnsiddlide fnllenvsde lyelkklnnl dekylisfed isknnstysv





 901
rfinksnges vyvetekeif skysehitke istiknsiit dvngnlldni qldhtsqvnt





 961
lnaaffiqsl idyssnkdvl ndlstsvkvq lyaqlfstgl ntiydsiqlv nlisnavndt





1021
invlptiteg ipivstildg inlgaaikel ldehdpllkk eleakvgvla inmslsiaat





1081
vasivgigae vtifllpiag isagipslvn nelilhdkat svvnyfnhls eskkygplkt





1141
eddkilvpid dlviseidfn nnsiklgtcn ilameggsgh tvtgnidhff sspsisship





1201
slsiysaigi etenldfskk immlpnapsr vfwwetgavp glrslendgt rlldsirdly





1261
pgkfywrfya ffdyaittlk pvyedtniki kldkdtrnfi mptittneir nklsysfdga





1321
ggtyslllss ypistninls kddlwifnid nevreisien gtikkgklik dvlskidink





1381
nkliignqti dfsgdidnkd ryifltceld dkisliiein lvaksyslll sgdknylisn





1441
lsniiekint lgldskniay nytdesnnky fgaisktsqk siihykkdsk nilefyndst





1501
lefnskdfia edinvfmkdd intitgkyyv dnntdksidf sislvsknqv kvnglylnes





1561
vyssyldfvk nsdghhntsn fmnlfldnis fwklfgfeni nfvidkyftl vgktnlgyve





1621
ficdnnknid iyfgewktss skstifsgng rnvvvepiyn pdtgedists ldfsyeplyg





1681
idryinkvli apdlytslin intnyysney ypeiivlnpn tfhkkvninl dsssfeykws





1741
tegsdfilvr yleesnkkil qkirikgils ntqsfnkmsi dfkdikklsl gyimsnfksf





1801
nseneldrdh lgfkiidnkt yyydedsklv kglininnsl fyfdpiefnl vtgwqtingk





1861
kyyfdintga alisykiing khfyfnndgv mqlgvfkgpd gfeyfapant qnnniegqai





1921
vyqskfltln gkkyyfdnds kavtgwriin nekyyfnpnn aiaavglqvi dnnkyyfnpd





1981
taiiskgwqt vngsryyfdt dtaiafngyk tidgkhfyfd sdcvvkigvf stsngfeyfa





2041
pantynnnie gqaivyqskf ltlngkkyyf dnnskavtgw qtidskkyyf ntntaeaatg





2101
wqtidgkkyy fntntaeaat gwqtidgkky yfntntaias tgytiingkh fyfntdgimq





2161
igvfkgpngf eyfapantda nniegqaily gnefltlngk kyyfgsdska vtgwriinnk





2221
kyyfnpnnai aaihlctinn dkyyfsydgi lqngyitier nnfyfdanne skmvtgvfkg





2281
pngfeyfapa nthnnniegq aivyqnkflt lngkkyyfdn dskavtgwqt idgkkyyfnl





2341
ntaeaatgwq tidgkkyyfn lntaeaatgw qtidgkkyyf ntntfiastg ytsingkhfy





2401
fntdgimqig vfkgpngfey fapanthnnn iegqailyqn kfltlngkky yfgsdskavt





2461
glrtidgkky yfntntavav tgwqtingkk yyfntntsia stgytiisgk hfyfntdgim





2521
qigvfkgpdg feyfapantd anniegqair yqnrflylhd niyyfgnnsk aatgwvtidg





2581
nryyfepnta mgangyktid nknfyfrngl pqigvfkgsn gfeyfapant danniegqai





2641
ryqnrflhll gkiyyfgnns kavtgwqtin gkvyyfmpdt amaaagglfe idgviyffgv





2701
dgvkapgiyg






The tcdA gene sequence for C. difficile 630 is as follows (SEQ ID NO:2):










(SEQ ID NO: 2)










   1
atgtctttaa tatctaaaga agagttaata aaactcgcat atagcattag accaagagaa






  61
aatgagtata aaactatact aactaattta gacgaatata ataagttaac tacaaacaat





 121
aatgaaaata aatatttaca attaaaaaaa ctaaatgaat caattgatgt ttttatgaat





 181
aaatataaaa cttcaagcag aaatagagca ctctctaatc taaaaaaaga tatattaaaa





 241
gaagtaattc ttattaaaaa ttccaataca agccctgtag aaaaaaattt acattttgta





 301
tggataggtg gagaagtcag tgatattgct cttgaataca taaaacaatg ggctgatatt





 361
aatgcagaat ataatattaa actgtggtat gatagtgaag cattcttagt aaatacacta





 421
aaaaaggcta tagttgaatc ttctaccact gaagcattac agctactaga ggaagagatt





 481
caaaatcctc aatttgataa tatgaaattt tacaaaaaaa ggatggaatt tatatatgat





 541
agacaaaaaa ggtttataaa ttattataaa tctcaaatca ataaacctac agtacctaca





 601
atagatgata ttataaagtc tcatctagta tctgaatata atagagatga aactgtatta





 661
gaatcatata gaacaaattc tttgagaaaa ataaatagta atcatgggat agatatcagg





 721
gctaatagtt tgtttacaga acaagagtta ttaaatattt atagtcagga gttgttaaat





 781
cgtggaaatt tagctgcagc atctgacata gtaagattat tagccctaaa aaattttggc





 841
ggagtatatt tagatgttga tatgcttcca ggtattcact ctgatttatt taaaacaata





 901
tctagaccta gctctattgg actagaccgt tgggaaatga taaaattaga ggctattatg





 961
aagtataaaa aatatataaa taattataca tcagaaaact ttgataaact tgatcaacaa





1021
ttaaaagata attttaaact cattatagaa agtaaaagtg aaaaatctga gatattttct





1081
aaattagaaa atttaaatgt atctgatctt gaaattaaaa tagctttcgc tttaggcagt





1141
gttataaatc aagccttgat atcaaaacaa ggttcatatc ttactaacct agtaatagaa





1201
caagtaaaaa atagatatca atttttaaac caacacctta acccagccat agagtctgat





1261
aataacttca cagatactac taaaattttt catgattcat tatttaattc agctaccgca





1321
gaaaactcta tgtttttaac aaaaatagca ccatacttac aagtaggttt tatgccagaa





1381
gctcgctcca caataagttt aagtggtcca ggagcttatg cgtcagctta ctatgatttc





1441
ataaatttac aagaaaatac tatagaaaaa actttaaaag catcagattt aatagaattt





1501
aaattcccag aaaataatct atctcaattg acagaacaag aaataaatag tctatggagc





1561
tttgatcaag caagtgcaaa atatcaattt gagaaatatg taagagatta tactggtgga





1621
tctctttctg aagacaatgg ggtagacttt aataaaaata ctgccctcga caaaaactat





1681
ttattaaata ataaaattcc atcaaacaat gtagaagaag ctggaagtaa aaattatgtt





1741
cattatatca tacagttaca aggagatgat ataagttatg aagcaacatg caatttattt





1801
tctaaaaatc ctaaaaatag tattattata caacgaaata tgaatgaaag tgcaaaaagc





1861
tactttttaa gtgatgatgg agaatctatt ttagaattaa ataaatatag gatacctgaa





1921
agattaaaaa ataaggaaaa agtaaaagta acctttattg gacatggtaa agatgaattc





1981
aacacaagcg aatttgctag attaagtgta gattcacttt ccaatgagat aagttcattt





2041
ttagatacca taaaattaga tatatcacct aaaaatgtag aagtaaactt acttggatgt





2101
aatatgttta gttatgattt taatgttgaa gaaacttatc ctgggaagtt gctattaagt





2161
attatggaca aaattacttc cactttacct gatgtaaata aaaattctat tactatagga





2221
gcaaatcaat atgaagtaag aattaatagt gagggaagaa aagaacttct ggctcactca





2281
ggtaaatgga taaataaaga agaagctatt atgagcgatt tatctagtaa agaatacatt





2341
ttttttgatt ctatagataa taagctaaaa gcaaagtcca agaatattcc aggattagca





2401
tcaatatcag aagatataaa aacattatta cttgatgcaa gtgttagtcc tgatacaaaa





2461
tttattttaa ataatcttaa gcttaatatt gaatcttcta ttggtgatta catttattat





2521
gaaaaattag agcctgttaa aaatataatt cacaattcta tagatgattt aatagatgag





2581
ttcaatctac ttgaaaatgt atctgatgaa ttatatgaat taaaaaaatt aaataatcta





2641
gatgagaagt atttaatatc ttttgaagat atctcaaaaa ataattcaac ttactctgta





2701
agatttatta acaaaagtaa tggtgagtca gtttatgtag aaacagaaaa agaaattttt





2761
tcaaaatata gcgaacatat tacaaaagaa ataagtacta taaagaatag tataattaca





2821
gatgttaatg gtaatttatt ggataatata cagttagatc atacttctca agttaataca





2881
ttaaacgcag cattctttat tcaatcatta atagattata gtagcaataa agatgtactg





2941
aatgatttaa gtacctcagt taaggttcaa ctttatgctc aactatttag tacaggttta





3001
aatactatat atgactctat ccaattagta aatttaatat caaatgcagt aaatgatact





3061
ataaatgtac tacctacaat aacagagggg atacctattg tatctactat attagacgga





3121
ataaacttag gtgcagcaat taaggaatta ctagacgaac atgacccatt actaaaaaaa





3181
gaattagaag ctaaggtggg tgttttagca ataaatatgt cattatctat agctgcaact





3241
gtagcttcaa ttgttggaat aggtgctgaa gttactattt tcttattacc tatagctggt





3301
atatctgcag gaataccttc attagttaat aatgaattaa tattgcatga taaggcaact





3361
tcagtggtaa actattttaa tcatttgtct gaatctaaaa aatatggccc tcttaaaaca





3421
gaagatgata aaattttagt tcctattgat gatttagtaa tatcagaaat agattttaat





3481
aataattcga taaaactagg aacatgtaat atattagcaa tggagggggg atcaggacac





3541
acagtgactg gtaatataga tcactttttc tcatctccat ctataagttc tcatattcct





3601
tcattatcaa tttattctgc aataggtata gaaacagaaa atctagattt ttcaaaaaaa





3661
ataatgatgt tacctaatgc tccttcaaga gtgttttggt gggaaactgg agcagttcca





3721
ggtttaagat cattggaaaa tgacggaact agattacttg attcaataag agatttatac





3781
ccaggtaaat tttactggag attctatgct tttttcgatt atgcaataac tacattaaaa





3841
ccagtttatg aagacactaa tattaaaatt aaactagata aagatactag aaacttcata





3901
atgccaacta taactactaa cgaaattaga aacaaattat cttattcatt tgatggagca





3961
ggaggaactt actctttatt attatcttca tatccaatat caacgaatat aaatttatct





4021
aaagatgatt tatggatatt taatattgat aatgaagtaa gagaaatatc tatagaaaat





4081
ggtactatta aaaaaggaaa gttaataaaa gatgttttaa gtaaaattga tataaataaa





4141
aataaactta ttataggcaa tcaaacaata gatttttcag gcgatataga taataaagat





4201
agatatatat tcttgacttg tgagttagat gataaaatta gtttaataat agaaataaat





4261
cttgttgcaa aatcttatag tttgttattg tctggggata aaaattattt gatatccaat





4321
ttatctaata ttattgagaa aatcaatact ttaggcctag atagtaaaaa tatagcgtac





4381
aattacactg atgaatctaa taataaatat tttggagcta tatctaaaac aagtcaaaaa





4441
agcataatac attataaaaa agacagtaaa aatatattag aattttataa tgacagtaca





4501
ttagaattta acagtaaaga ttttattgct gaagatataa atgtatttat gaaagatgat





4561
attaatacta taacaggaaa atactatgtt gataataata ctgataaaag tatagatttc





4621
tctatttctt tagttagtaa aaatcaagta aaagtaaatg gattatattt aaatgaatcc





4681
gtatactcat cttaccttga ttttgtgaaa aattcagatg gacaccataa tacttctaat





4741
tttatgaatt tatttttgga caatataagt ttctggaaat tgtttgggtt tgaaaatata





4801
aattttgtaa tcgataaata ctttaccctt gttggtaaaa ctaatcttgg atatgtagaa





4861
tttatttgtg acaataataa aaatatagat atatattttg gtgaatggaa aacatcgtca





4921
tctaaaagca ctatatttag cggaaatggt agaaatgttg tagtagagcc tatatataat





4981
cctgatacgg gtgaagatat atctacttca ctagattttt cctatgaacc tctctatgga





5041
atagatagat atatcaataa agtattgata gcacctgatt tatatacaag tttaataaat





5101
attaatacca attattattc aaatgagtac taccctgaga ttatagttct taacccaaat





5161
acattccaca aaaaagtaaa tataaattta gatagttctt cttttgagta taaatggtct





5221
acagaaggaa gtgactttat tttagttaga tacttagaag aaagtaataa aaaaatatta





5281
caaaaaataa gaatcaaagg tatcttatct aatactcaat catttaataa aatgagtata





5341
gattttaaag atattaaaaa actatcatta ggatatataa tgagtaattt taaatcattt





5401
aattctgaaa atgaattaga tagagatcat ttaggattta aaataataga taataaaact





5461
tattactatg atgaagatag taaattagtt aaaggattaa tcaatataaa taattcatta





5521
ttctattttg atcctataga atttaactta gtaactggat ggcaaactat caatggtaaa





5581
aaatattatt ttgatataaa tactggagca gctttaatta gttataaaat tattaatggt





5641
aaacactttt attttaataa tgatggtgtg atgcagttgg gagtatttaa aggacctgat





5701
ggatttgaat attttgcacc tgccaatact caaaataata acatagaagg tcaggctata





5761
gtttatcaaa gtaaattctt aactttgaat ggcaaaaaat attattttga taatgactca





5821
aaagcagtca ctggatggag aattattaac aatgagaaat attactttaa tcctaataat





5881
gctattgctg cagtcggatt gcaagtaatt gacaataata agtattattt caatcctgac





5941
actgctatca tctcaaaagg ttggcagact gttaatggta gtagatacta ctttgatact





6001
gataccgcta ttgcctttaa tggttataaa actattgatg gtaaacactt ttattttgat





6061
agtgattgtg tagtgaaaat aggtgtgttt agtacctcta atggatttga atattttgca





6121
cctgctaata cttataataa taacatagaa ggtcaggcta tagtttatca aagtaaattc





6181
ttaactttga atggtaaaaa atattacttt gataataact caaaagcagt taccggatgg





6241
caaactattg atagtaaaaa atattacttt aatactaaca ctgctgaagc agctactgga





6301
tggcaaacta ttgatggtaa aaaatattac tttaatacta acactgctga agcagctact





6361
ggatggcaaa ctattgatgg taaaaaatat tactttaata ctaacactgc tatagcttca





6421
actggttata caattattaa tggtaaacat ttttatttta atactgatgg tattatgcag





6481
ataggagtgt ttaaaggacc taatggattt gaatattttg cacctgctaa tacggatgct





6541
aacaacatag aaggtcaagc tatactttac caaaatgaat tcttaacttt gaatggtaaa





6601
aaatattact ttggtagtga ctcaaaagca gttactggat ggagaattat taacaataag





6661
aaatattact ttaatcctaa taatgctatt gctgcaattc atctatgcac tataaataat





6721
gacaagtatt actttagtta tgatggaatt cttcaaaatg gatatattac tattgaaaga





6781
aataatttct attttgatgc taataatgaa tctaaaatgg taacaggagt atttaaagga





6841
cctaatggat ttgagtattt tgcacctgct aatactcaca ataataacat agaaggtcag





6901
gctatagttt accagaacaa attcttaact ttgaatggca aaaaatatta ttttgataat





6961
gactcaaaag cagttactgg atggcaaacc attgatggta aaaaatatta ctttaatctt





7021
aacactgctg aagcagctac tggatggcaa actattgatg gtaaaaaata ttactttaat





7081
cttaacactg ctgaagcagc tactggatgg caaactattg atggtaaaaa atattacttt





7141
aatactaaca ctttcatagc ctcaactggt tatacaagta ttaatggtaa acatttttat





7201
tttaatactg atggtattat gcagatagga gtgtttaaag gacctaatgg atttgaatac





7261
tttgcacctg ctaatactca taataataac atagaaggtc aagctatact ttaccaaaat





7321
aaattcttaa ctttgaatgg taaaaaatat tactttggta gtgactcaaa agcagttacc





7381
ggattgcgaa ctattgatgg taaaaaatat tactttaata ctaacactgc tgttgcagtt





7441
actggatggc aaactattaa tggtaaaaaa tactacttta atactaacac ttctatagct





7501
tcaactggtt atacaattat tagtggtaaa catttttatt ttaatactga tggtattatg





7561
cagataggag tgtttaaagg acctgatgga tttgaatact ttgcacctgc taatacagat





7621
gctaacaata tagaaggtca agctatacgt tatcaaaata gattcctata tttacatgac





7681
aatatatatt attttggtaa taattcaaaa gcagctactg gttgggtaac tattgatggt





7741
aatagatatt acttcgagcc taatacagct atgggtgcga atggttataa aactattgat





7801
aataaaaatt tttactttag aaatggttta cctcagatag gagtgtttaa agggtctaat





7861
ggatttgaat actttgcacc tgctaatacg gatgctaaca atatagaagg tcaagctata





7921
cgttatcaaa atagattcct acatttactt ggaaaaatat attactttgg taataattca





7981
aaagcagtta ctggatggca aactattaat ggtaaagtat attactttat gcctgatact





8041
gctatggctg cagctggtgg acttttcgag attgatggtg ttatatattt ctttggtgtt





8101
gatggagtaa aagcccctgg gatatatggc taa






TcdB is encoded by the tcdB gene. Sequences for TcdB are known for a number of species, e.g., for C. difficile 630 (the TcdB NCBI Gene ID is 4914074) and polypeptide sequence (e.g., YP_001087135.1 (SEQ ID NO: 3). The sequence for the TcdB gene product is as follows (SEQ ID NO: 3):










(SEQ ID No: 3)










   1
mslvnrkqle kmanvrfrtq edeyvailda leeyhnmsen tvvekylklk dinsltdiyi






  61
dtykksgrnk alkkfkeylv tevlelknnn ltpveknlhf vwiggqindt ainyinqwkd





 121
vnsdynvnvf ydsnaflint lkktvvesai ndtlesfren lndprfdynk ffrkrmeiiy





 181
dkqknfinyy kaqreenpel iiddivktyl sneyskeide lntyieesln kitqnsgndv





 241
rnfeefknge sfnlyeqelv erwnlaaasd ilrisalkei ggmyldvdml pgiqpdlfes





 301
iekpssvtvd fwemtkleai mkykeyipey tsehfdmlde evqssfesvl asksdkseif





 361
sslgdmeasp levkiafnsk giinqglisv kdsycsnliv kqienrykil nnslnpaise





 421
dndfntttnt fidsimaean adngrfmmel gkylrvgffp dvkttinlsg peayaaayqd





 481
llmfkegsmn ihlieadlrn feisktnisq steqemaslw sfddarakaq feeykrnyfe





 541
gslgeddnld fsqnivvdke yllekissla rssergyihy ivqlqgdkis yeaacnlfak





 601
tpydsvlfqk niedseiayy ynpgdgeiqe idkykipsii sdrpkikltf ighgkdefnt





 661
difagfdvds lsteieaaid lakedispks ieinllgcnm fsysinveet ypgklllkvk





 721
dkiselmpsi sqdsiivsan qyevrinseg rrelldhsge winkeesiik disskeyisf





 781
npkenkitvk sknlpelstl lqeirnnsns sdieleekvm lteceinvis nidtqiveer





 841
ieeaknltsd sinyikdefk liesisdalc dlkqqneled shfisfedis etdegfsirf





 901
inketgesif vetektifse yanhiteeis kikgtifdtv ngklvkkvnl dtthevntln





 961
aaffiqslie ynsskeslsn lsvamkvqvy aqlfstglnt itdaakvvel vstaldetid





1021
llptlseglp iiatiidgvs lgaaikelse tsdpllrqei eakigimavn lttattaiit





1081
sslgiasgfs illvplagis agipslvnne lvlrdkatkv vdyfkhvslv etegvftlld





1141
dkimmpqddl viseidfnnn sivlgkceiw rmeggsghtv tddidhffsa psityrephl





1201
siydvlevqk eeldlskdlm vlpnapnrvf awetgwtpgl rslendgtkl ldrirdnyeg





1261
efywryfafi adalittlkp ryedtnirin ldsntrsfiv piitteyire klsysfygsg





1321
gtyalslsqy nmginielse sdvwiidvdn vvrdvtiesd kikkgdlieg ilstlsieen





1381
kiilnshein fsgevngsng fvsltfsile ginaiievdl lsksykllis gelkilmlns





1441
nhiqqkidyi gfnselqkni pysfvdsegk engfingstk eglfvselpd vvliskvymd





1501
dskpsfgyys nnlkdvkvit kdnvniltgy ylkddikisl sltlqdekti klnsvhldes





1561
gvaeilkfmn rkgntntsds lmsflesmni ksifvnflqs nikfildanf iisgttsigq





1621
feficdendn iqpyfikfnt letnytlyvg nrqnmivepn ydlddsgdis stvinfsqky





1681
lygidscvnk vvispniytd einitpvyet nntypevivl danyinekin vnindlsiry





1741
vwsndgndfi lmstseenkv sqvkirfvnv fkdktlankl sfnfsdkqdv pvseiilsft





1801
psyyedglig ydlglvslyn ekfyinnfgm mvsgliyind slyyfkppvn nlitgfvtvg





1861
ddkyyfnpin ggaasigeti iddknyyfnq sgvlqtgvfs tedgfkyfap antldenleg





1921
eaidftgkli ideniyyfdd nyrgavewke ldgemhyfsp etgkafkgln qigdykyyfn





1981
sdgvmqkgfv sindnkhyfd dsgvmkvgyt eidgkhfyfa engemqigvf ntedgfkyfa





2041
hhnedlgnee geeisysgil nfnnkiyyfd dsftavvgwk dledgskyyf dedtaeayig





2101
lslindgqyy fnddgimqvg fvtindkvfy fsdsgiiesg vqniddnyfy iddngivqig





2161
vfdtsdgyky fapantvndn iygqaveysg lvrvgedvyy fgetytietg wiydmenesd





2221
kyyfnpetkk ackginlidd ikyyfdekgi mrtglisfen nnyyfnenge mqfgyinied





2281
kmfyfgedgv mqigvfntpd gfkyfahqnt ldenfegesi nytgwldlde kryyftdeyi





2341
aatgsviidg eeyyfdpdta qlvise






The tcdB gene sequence for C. difficile 630 is as follows (SEQ ID NO: 4):










(SEQ ID NO: 4)










   1
atgagtttag ttaatagaaa acagttagaa aaaatggcaa atgtaagatt tcgtactcaa






  61
gaagatgaat atgttgcaat attggatgct ttagaagaat atcataatat gtcagagaat





 121
actgtagtcg aaaaatattt aaaattaaaa gatataaata gtttaacaga tatttatata





 181
gatacatata aaaaatctgg tagaaataaa gccttaaaaa aatttaagga atatctagtt





 241
acagaagtat tagagctaaa gaataataat ttaactccag ttgagaaaaa tttacatttt





 301
gtttggattg gaggtcaaat aaatgacact gctattaatt atataaatca atggaaagat





 361
gtaaatagtg attataatgt taatgttttt tatgatagta atgcattttt gataaacaca





 421
ttgaaaaaaa ctgtagtaga atcagcaata aatgatacac ttgaatcatt tagagaaaac





 481
ttaaatgacc ctagatttga ctataataaa ttcttcagaa aacgtatgga aataatttat





 541
gataaacaga aaaatttcat aaactactat aaagctcaaa gagaagaaaa tcctgaactt





 601
ataattgatg atattgtaaa gacatatctt tcaaatgagt attcaaagga gatagatgaa





 661
cttaatacct atattgaaga atccttaaat aaaattacac agaatagtgg aaatgatgtt





 721
agaaactttg aagaatttaa aaatggagag tcattcaact tatatgaaca agagttggta





 781
gaaaggtgga atttagctgc tgcttctgac atattaagaa tatctgcatt aaaagaaatt





 841
ggtggtatgt atttagatgt tgatatgtta ccaggaatac aaccagactt atttgagtct





 901
atagagaaac ctagttcagt aacagtggat ttttgggaaa tgacaaagtt agaagctata





 961
atgaaataca aagaatatat accagaatat acctcagaac attttgacat gttagacgaa





1021
gaagttcaaa gtagttttga atctgttcta gcttctaagt cagataaatc agaaatattc





1081
tcatcacttg gtgatatgga ggcatcacca ctagaagtta aaattgcatt taatagtaag





1141
ggtattataa atcaagggct aatttctgtg aaagactcat attgtagcaa tttaatagta





1201
aaacaaatcg agaatagata taaaatattg aataatagtt taaatccagc tattagcgag





1261
gataatgatt ttaatactac aacgaatacc tttattgata gtataatggc tgaagctaat





1321
gcagataatg gtagatttat gatggaacta ggaaagtatt taagagttgg tttcttccca





1381
gatgttaaaa ctactattaa cttaagtggc cctgaagcat atgcggcagc ttatcaagat





1441
ttattaatgt ttaaagaagg cagtatgaat atccatttga tagaagctga tttaagaaac





1501
tttgaaatct ctaaaactaa tatttctcaa tcaactgaac aagaaatggc tagcttatgg





1561
tcatttgacg atgcaagagc taaagctcaa tttgaagaat ataaaaggaa ttattttgaa





1621
ggttctcttg gtgaagatga taatcttgat ttttctcaaa atatagtagt tgacaaggag





1681
tatcttttag aaaaaatatc ttcattagca agaagttcag agagaggata tatacactat





1741
attgttcagt tacaaggaga taaaattagt tatgaagcag catgtaactt atttgcaaag





1801
actccttatg atagtgtact gtttcagaaa aatatagaag attcagaaat tgcatattat





1861
tataatcctg gagatggtga aatacaagaa atagacaagt ataaaattcc aagtataatt





1921
tctgatagac ctaagattaa attaacattt attggtcatg gtaaagatga atttaatact





1981
gatatatttg caggttttga tgtagattca ttatccacag aaatagaagc agcaatagat





2041
ttagctaaag aggatatttc tcctaagtca atagaaataa atttattagg atgtaatatg





2101
tttagctact ctatcaacgt agaggagact tatcctggaa aattattact taaagttaaa





2161
gataaaatat cagaattaat gccatctata agtcaagact ctattatagt aagtgcaaat





2221
caatatgaag ttagaataaa tagtgaagga agaagagaat tattggatca ttctggtgaa





2281
tggataaata aagaagaaag tattataaag gatatttcat caaaagaata tatatcattt





2341
aatcctaaag aaaataaaat tacagtaaaa tctaaaaatt tacctgagct atctacatta





2401
ttacaagaaa ttagaaataa ttctaattca agtgatattg aactagaaga aaaagtaatg





2461
ttaacagaat gtgagataaa tgttatttca aatatagata cgcaaattgt tgaggaaagg





2521
attgaagaag ctaagaattt aacttctgac tctattaatt atataaaaga tgaatttaaa





2581
ctaatagaat ctatttctga tgcactatgt gacttaaaac aacagaatga attagaagat





2641
tctcatttta tatcttttga ggacatatca gagactgatg agggatttag tataagattt





2701
attaataaag aaactggaga atctatattt gtagaaactg aaaaaacaat attctctgaa





2761
tatgctaatc atataactga agagatttct aagataaaag gtactatatt tgatactgta





2821
aatggtaagt tagtaaaaaa agtaaattta gatactacac acgaagtaaa tactttaaat





2881
gctgcatttt ttatacaatc attaatagaa tataatagtt ctaaagaatc tcttagtaat





2941
ttaagtgtag caatgaaagt ccaagtttac gctcaattat ttagtactgg tttaaatact





3001
attacagatg cagccaaagt tgttgaatta gtatcaactg cattagatga aactatagac





3061
ttacttccta cattatctga aggattacct ataattgcaa ctattataga tggtgtaagt





3121
ttaggtgcag caatcaaaga gctaagtgaa acgagtgacc cattattaag acaagaaata





3181
gaagctaaga taggtataat ggcagtaaat ttaacaacag ctacaactgc aatcattact





3241
tcatctttgg ggatagctag tggatttagt atacttttag ttcctttagc aggaatttca





3301
gcaggtatac caagcttagt aaacaatgaa cttgtacttc gagataaggc aacaaaggtt





3361
gtagattatt ttaaacatgt ttcattagtt gaaactgaag gagtatttac tttattagat





3421
gataaaataa tgatgccaca agatgattta gtgatatcag aaatagattt taataataat





3481
tcaatagttt taggtaaatg tgaaatctgg agaatggaag gtggttcagg tcatactgta





3541
actgatgata tagatcactt cttttcagca ccatcaataa catatagaga gccacactta





3601
tctatatatg acgtattgga agtacaaaaa gaagaacttg atttgtcaaa agatttaatg





3661
gtattaccta atgctccaaa tagagtattt gcttgggaaa caggatggac accaggttta





3721
agaagcttag aaaatgatgg cacaaaactg ttagaccgta taagagataa ctatgaaggt





3781
gagttttatt ggagatattt tgcttttata gctgatgctt taataacaac attaaaacca





3841
agatatgaag atactaatat aagaataaat ttagatagta atactagaag ttttatagtt





3901
ccaataataa ctacagaata tataagagaa aaattatcat attctttcta tggttcagga





3961
ggaacttatg cattgtctct ttctcaatat aatatgggta taaatataga attaagtgaa





4021
agtgatgttt ggattataga tgttgataat gttgtgagag atgtaactat agaatctgat





4081
aaaattaaaa aaggtgattt aatagaaggt attttatcta cactaagtat tgaagagaat





4141
aaaattatct taaatagcca tgagattaat ttttctggtg aggtaaatgg aagtaatgga





4201
tttgtttctt taacattttc aattttagaa ggaataaatg caattataga agttgattta





4261
ttatctaaat catataaatt acttatttct ggcgaattaa aaatattgat gttaaattca





4321
aatcatattc aacagaaaat agattatata ggattcaata gcgaattaca gaaaaatata





4381
ccatatagct ttgtagatag tgaaggaaaa gagaatggtt ttattaatgg ttcaacaaaa





4441
gaaggtttat ttgtatctga attacctgat gtagttctta taagtaaggt ttatatggat





4501
gatagtaagc cttcatttgg atattatagt aataatttga aagatgtcaa agttataact





4561
aaagataatg ttaatatatt aacaggttat tatcttaagg atgatataaa aatctctctt





4621
tctttgactc tacaagatga aaaaactata aagttaaata gtgtgcattt agatgaaagt





4681
ggagtagctg agattttgaa gttcatgaat agaaaaggta atacaaatac ttcagattct





4741
ttaatgagct ttttagaaag tatgaatata aaaagtattt tcgttaattt cttacaatct





4801
aatattaagt ttatattaga tgctaatttt ataataagtg gtactacttc tattggccaa





4861
tttgagttta tttgtgatga aaatgataat atacaaccat atttcattaa gtttaataca





4921
ctagaaacta attatacttt atatgtagga aatagacaaa atatgatagt ggaaccaaat





4981
tatgatttag atgattctgg agatatatct tcaactgtta tcaatttctc tcaaaagtat





5041
ctttatggaa tagacagttg tgttaataaa gttgtaattt caccaaatat ttatacagat





5101
gaaataaata taacgcctgt atatgaaaca aataatactt atccagaagt tattgtatta





5161
gatgcaaatt atataaatga aaaaataaat gttaatatca atgatctatc tatacgatat





5221
gtatggagta atgatggtaa tgattttatt cttatgtcaa ctagtgaaga aaataaggtg





5281
tcacaagtta aaataagatt cgttaatgtt tttaaagata agactttggc aaataagcta





5341
tcttttaact ttagtgataa acaagatgta cctgtaagtg aaataatctt atcatttaca





5401
ccttcatatt atgaggatgg attgattggc tatgatttgg gtctagtttc tttatataat





5461
gagaaatttt atattaataa ctttggaatg atggtatctg gattaatata tattaatgat





5521
tcattatatt attttaaacc accagtaaat aatttgataa ctggatttgt gactgtaggc





5581
gatgataaat actactttaa tccaattaat ggtggagctg cttcaattgg agagacaata





5641
attgatgaca aaaattatta tttcaaccaa agtggagtgt tacaaacagg tgtatttagt





5701
acagaagatg gatttaaata ttttgcccca gctaatacac ttgatgaaaa cctagaagga





5761
gaagcaattg attttactgg aaaattaatt attgacgaaa atatttatta ttttgatgat





5821
aattatagag gagctgtaga atggaaagaa ttagatggtg aaatgcacta ttttagccca





5881
gaaacaggta aagcttttaa aggtctaaat caaataggtg attataaata ctatttcaat





5941
tctgatggag ttatgcaaaa aggatttgtt agtataaatg ataataaaca ctattttgat





6001
gattctggtg ttatgaaagt aggttacact gaaatagatg gcaagcattt ctactttgct





6061
gaaaacggag aaatgcaaat aggagtattt aatacagaag atggatttaa atattttgct





6121
catcataatg aagatttagg aaatgaagaa ggtgaagaaa tctcatattc tggtatatta





6181
aatttcaata ataaaattta ctattttgat gattcattta cagctgtagt tggatggaaa





6241
gatttagagg atggttcaaa gtattatttt gatgaagata cagcagaagc atatataggt





6301
ttgtcattaa taaatgatgg tcaatattat tttaatgatg atggaattat gcaagttgga





6361
tttgtcacta taaatgataa agtcttctac ttctctgact ctggaattat agaatctgga





6421
gtacaaaaca tagatgacaa ttatttctat atagatgata atggtatagt tcaaattggt





6481
gtatttgata cttcagatgg atataaatat tttgcacctg ctaatactgt aaatgataat





6541
atttacggac aagcagttga atatagtggt ttagttagag ttggtgaaga tgtatattat





6601
tttggagaaa catatacaat tgagactgga tggatatatg atatggaaaa tgaaagtgat





6661
aaatattatt tcaatccaga aactaaaaaa gcatgcaaag gtattaattt aattgatgat





6721
ataaaatatt attttgatga gaagggcata atgagaacgg gtcttatatc atttgaaaat





6781
aataattatt actttaatga gaatggtgaa atgcaatttg gttatataaa tatagaagat





6841
aagatgttct attttggtga agatggtgtc atgcagattg gagtatttaa tacaccagat





6901
ggatttaaat actttgcaca tcaaaatact ttggatgaga attttgaggg agaatcaata





6961
aactatactg gttggttaga tttagatgaa aagagatatt attttacaga tgaatatatt





7021
gcagcaactg gttcagttat tattgatggt gaggagtatt attttgatcc tgatacagct





7081
caattagtga ttagtgaata g






Outbreaks of CDI have been reported with Toxin A-negative/Toxin B-positive strains, which indicates that Toxin B is also capable of playing a key role in the disease pathology. TcdA and TcdB are 308 and 270 kDa proteins, respectively. The toxins belong to the family of large clostridial toxins (LCTs), which are a group of homologous, high molecular weight proteins that further include the lethal and hemorrhagic toxins from C. sordellii (TcsL and TcsH, respectively), α-toxin from C. novyi (Tcnα) and a cytotoxin from C. perfringens (TpeL). The homologous proteins intoxicate host cells through a multistep mechanism that involves (i) receptor binding and endocytosis, (ii) pore formation and translocation of the GTD across the endosomal membrane, (iii) autoprocessing and release of GTD into the cytosol, and (iv) glucosylation of host Rho GTPase. Both Toxins A and B also contain a second enzyme activity in the form of a cysteine protease which appears to play a role in the release of the effector domain into the cytosol after translocation. The C. difficile binary toxin modifies cell actin by a mechanism which involves the transfer of an ADP-ribose moiety from NAD onto its target protein. Given the similarities in toxin structure and the genetic similarities of Clostridial species, it is likely that the expression of toxins of other spore-forming toxigenic Clostridium species are regulated in a similar manner to that of C. difficile, i.e., in a manner sensitive to environmental conditions that can be influenced by commensal bacteria as described herein.


Additional bacterial toxins, including additional Clostridial toxins, are described in Table 1.









TABLE 1







Bacterial toxins and their mechanism of action.










Toxin
Organism/Result of Gram stain
Mechanism
Clinical Features





Toxin A/toxin B

Clostridium difficile

Inhibits cytoskeletal
Diarrhea, vomiting



(gram-positive)
action in epithelial cells


Anthrax toxins

Bacillus anthracis

Adenylyl cyclase (EF),
Edema and skin necrosis; shock


(edema toxin [EF],
(gram-positive)
metalloprotease (LF)


lethal toxin [LF])


Adenylate cyclase toxin

Bordetella pertussis

Adenylyl cyclase
Tracheobronchitis



(gram-negative)


Botulinum toxins

Clostridium botulinum

Blocks release of acetylcholine,
Muscle paralysis, botulism


(C2/C3 toxin)
(gram-positive)
ADP-ribosyltransferase


Lecithinase

Clostridium perfringens

Phospholipase
Gangrene; destraction of phagocytes


(α-toxin; perfringolysin O)
(gram-positive)


Tetanus toxin

Clostridium tetani

Blocks release of
Spasms and rigidity of the



(gram-positive)
inhibitory
voluntary muscles;




neurotransmitters
characteristic symptom of





“lockjaw”


Diphtheria toxin

Corynebacterium diphtheria

ADP-ribosylates
Respiratory infection;



(gram-positive)
EF-2, inhibiting
complicating myocarditis with




protein synthesis
accompanying neurologic toxicity


CNF-1, CNF-2

Escherichia coli

Affects ρ-GTP-binding regulators
Diarrhea



(gram-negative)


Heat-stable toxin

Escherichia coli

Secondary message regulation
Diarrhea



(gram-negative)


Hemolysin

Escherichia coli

Heptameric pore-forming complex
Urinary tract infections



(gram-negative)
(hemolysin)


Shiga-like toxin

Escherichia coli

Stops host protein synthesis
Hemolytic-uremic syndrome, dysentery



(gram-negative)


Exotoxin A

Pseudomonas aeruginosa

ADP-ribosylates elongation
Respiratory distress; possible



(gram-negative)
factor-2 (EF-2),
role as virulence factor in lung




inhibiting protein synthesis
infections of cystic fibrosis





patients


Shiga toxin

Shigella dysenteriae

Stops host protein synthesis
Dysentery



(gram-negative)


α-Toxin

Staphylococcus aureus

Heptameric pore-forming complex
Abscess formation



(gram-positive)
(hemolysin)


Toxic shock syndrome toxin 1

Staphylococcus aureus

Superantigen activates T-cell
Cytokine cascade elicits


(TSST-1)
(gram-positive)
populations, cross-linking
shock; capillary leak




VβTCR and class II MHC
syndrome and hypotension


Pneumolysin

Streptococcus pneumonia

Pore-forming complex
Pneumonia



(gram-positive)
(hemolysin)


Pyrogenic exotoxin

Streptococcus pyogenes

Superantigen activates T-cell
Cytokine cascade elicits



(gram-positive)
populations, cross-linking
shock; capillary leak




VβTCR and class II MHC
syndrome and hypotension


Streptolysin O

Streptococcus pyogenes

Pore-forming complex
“Strep” throat, scarlet fever



(gram-positive)
(hemolysin)


Cholera toxin

Vibrio cholera

Disrupts adenylyl cyclase
Watery diarrhea, loss of



(gram-negative)

electrolytes and fluids





Abbreviations used: ADP, adenosine diphosphate;


EF, elongation factor;


LF, lethal factor;


GTP, guanosine triphosphate;


TCR, T-cell receptor;


MHC, major histocompatability complex







Regulation of C. difficile Toxin Gene Expression



C. difficile's pathogenicity locus, PaLoc, contains the toxin operon with genes tcdA, tcdB and tcdE that respectively encode the A and B portions of the toxin and a putative holin involved in toxin export. In addition to the toxins TcdA and TcdB, the PaLoc in pathogenic strains encodes TcdR, a member of the extracytoplasmic function family of alternative sigma factors that plays a critical role in activating the expression of tcdA and tcdB; transcription of the tcdA and tcdB genes requires TcdR to enable RNA polymerase to have specificity to bind the toxin gene promoters. Within C. difficile, multiple nutritional regulators influence PaLoc gene expression at the level of tcdR and the tcdAEB operon genes. In particular, C. difficile primarily elaborates toxin under starvation conditions to extract nutrients from the host and to also enable shedding of spores. Exogenously added glucose, amino acids—proline and leucine in particular, as well as cysteine, inhibit toxin production through codY, ccpA, rex and/or prdR activation. Exogenously added butyrate induces toxin production, while butanol does not. The symptoms of CDI correlate with the expression of TcdR. The genes encoding TcdA (tcdA) and TcdB (tcdB) are located within a 19.6-kb chromosomal region that makes up the PaLoc. The activity of TcdR is modulated by TcdC, an anti-sigma factor that destabilizes the TcdR-core RNA polymerase complex. TcdC seems to be most active in rapidly growing cells.


Stickland Fermentation


Stickland reactions couple metabolism of pairs of amino acids in which one amino acid, acting as an electron donor, is oxidatively deaminated or decarboxylated and a second amino acid, acting as an electron acceptor, is reduced or reductively deaminated (Stickland, L. H., Biochem. J. 28: 1746-1759 (1934). The most efficient electron donors are leucine, isoleucine, and alanine, and the most efficient acceptors are glycine, proline. Hydroxyproline is also an efficient Stickland acceptor.



C. difficile, like other cluster XI species of clostridia, uses Stickland fermentations to extract energy from amino acids. Donor amino acids include the electron-rich branched chain amino acids leucine, isoleucine and valine (BCAA), aromatic amino acids phenylalanine, tyrosine and tryptophan, and acceptor amino acids glycine and proline. Cellular proline and glycine reductases transfer electrons from Stickland donor amino acids to recipients proline and glycine. The reaction consumes one NADH to one NAD+ in the reductive pathway, with release of ammonia and the branched short-chain fatty acids isocaproate, isobutyrate and isovalerate, and regenerates 2 NAD+ to NADH in the oxidative pathway. to regenerate NAD+ to NADH.


The reduction of the Stickland acceptors glycine and proline is performed by two selenium-dependent reductases, glycine reductase (GR) and D-Proline reductase (PR), respectively. GR catalyzes the reductive deamination of glycine to acetyl phosphate and ammonium, and PR reductively cleaves D-proline to 5-aminovalerate. The glycine reductase and proline reductase operons, respectively, mediate these reactions. Each reductase is comprised of multiple polypeptides and is dependent upon selenocysteine residues for activity. FIG. 5A-5I show metabolites from the input donor or acceptor amino acids. In one embodiment described is that the Stickland donor amino acids proline and glycine have a powerful inhibitory effect on C. difficile toxin production.


Genes encoding the C. difficile GR and PR subunits are clustered in two distinct genetic loci (grd and prd, respectively) on the chromosome of C. difficile strain 630. The grd locus contains eight genes. Two of the genes, grdA and grdB, likely encode the selenocysteine-containing subunits of GR. The seven genes of the prd operon include prdF, that encodes a d-proline racemase, and prdB, that encodes the selenium-containing subunit of PR.


In one embodiment, prdR activates the proline reductase operon and represses the glycine reductase operon. prdR suppresses the expression of C. difficile toxins by inhibiting butyrate, coDY, ccpA, tcdR, and/or tcdA production. The Stickland fermentation pathway using proline as the Stickland acceptor generates NADH and the metabolite 5-aminovalerate from proline. The acetate-generating pathway generates NADH and the metabolite acetate from glycine. C. difficile consumes large amounts of NADH in the carbohydrate metabolism pathway from acetate. Therefore, acetate-generating pathways are more prone to inducing a stress response pathway, leading to the generation of butyrate, codY, ccpA, tcdR, and/or tcdA toxins.


In one embodiment, the method of treating or preventing a pathology caused by expression of a bacterial toxin compromises administering at least one bacterial organism that encodes and expresses one or more of D-Proline reductase (PR), Glycine reductase (GR), thioredoxin, or choloylglycine.


As noted above, the C. difficile pathogen can also utilize sugars, sugar alcohols, and ethanolamine for energy production (carbohydrate metabolism and other energy production approaches used by C. difficile are discussed further below). Toxin production in C. difficile responds to environmental conditions, including the availability of specific nutrients, temperature changes, and alteration of the redox potential. The presence of a rapidly metabolizable carbon source or certain amino acids inhibits toxin gene expression. When cells are grown in rich medium, the toxin genes are transcribed only when the cells reach stationary phase. While not wishing to be bound by theory, this is fully consistent with the discovery that protective commensal species are highly proteolytic and induce genes in C. difficile that, for example, promote the use of ethanolamine in the gut environment as an energy source.


Nutritional regulators within C. difficile, including codY, ccpA, and rex sense aspects of amino acid, sugar, Stickland reactions and NAD+/NADH pools respectively. Each of these regulons also exerts effects on PaLoc gene expression through direct and indirect mechanisms.


Upon binding of BCAA and GTP, codY strongly represses tcdR transcription, with subsequent reduction of tcdAEB gene expression. ccpA, active under carbon starvation conditions, binds the tcdAEB operon promoter, enhancing toxin expression if the tcdR sigma factor has been expressed. Thus, starvation where carbohydrates and Stickland amino acids are limiting induces expression of toxin. Under conditions of nutrient sufficiency, codY, ccpA and rex act coordinately through direct and indirect mechanisms to repress the expression of PaLoc genes. Notably, exogenous in vitro addition of Stickland amino acids or carbohydrate energy sources represses C. difficile toxin expression through the above mechanisms, while exogenous addition of butyrate, a key end product of anaerobic carbohydrate fermentation, can alone induce toxin expression. Together, these gene regulatory systems promote C. difficile toxin expression under conditions of starvation and energy limitation by sensing preferred sources for intracellular energy production.


NAD+/NADH


The balance of NAD+ to NADH influences the expression of C. difficile toxin. The Stickland reaction involves the coupled oxidation and reduction of pairs of amino acids to generate ATP and NAD+. The oxidative pathway generates ATP and NADH, while the reductive pathway regenerates NAD+ from NADH. Proline reductase (PR) and glycine reductase (GR) expression are specifically induced in the presence of proline and glycine, respectively, and carry out the respective reduction of these amino acids. Moreover, the addition of proline to the growth medium decreases the expression of GR-encoding genes, suggesting a preferential utilization of proline for NAD+ regeneration. PrdR, which is a regulator that responds to proline, mediates both the proline-dependent activation of PR and the proline-dependent repression of toxin genes and the GR operon. When proline is limiting in the medium or if PrdR or PR is inactive, the alternative reductive pathways are induced. In fact, both PrdR and a functional PR are indirectly required for the proline-dependent regulation of the alternative reductive pathways in response to the intracellular concentration of NADH and NAD+. This process involves the global redox-sensing regulator Rex. In a number of Gram-positive bacteria, Rex acts as a repressor of genes that are important for growth using fermentation. Rex directly senses changes in redox status and is only active as a DNA-binding protein when the intracellular NADH/NAD+ ratio is low. The protein Rex is stimulated by NAD+ but inhibited by NADH. Although Rex, like PrdR, controls the proline-responsive expression of these alternative reductive pathways, Rex also mediates the proline-dependent repression of toxin gene expression, probably through the regulation of butyrate production. As a result, when proline is not limiting the NADH/NAD+ ratio is low and Rex is active as a repressor of the alternative NAD+ regeneration pathways. In contrast, if proline becomes limiting, the NADH/NAD+ ratio increases and NADH prevents Rex-dependent repression of the alternative pathways. The regeneration of NAD+ using these alternative reductive pathways leads to an accumulation of butyrate, a compound that stimulates toxin synthesis as shown in FIG. 9.


In one embodiment, the method of treating or preventing a pathology caused by expression of a bacterial toxin compromises administering at least one bacterial organism that promotes Stickland fermentation by C. difficile. In another embodiment, the bacterial organism itself performs Stickland fermentation—bacteria that have evolved to perform Stickland fermentation tend to express, among other things, extracellular proteolytic activities that can feed amino acids into the Stickland fermentation pathway for C. difficile.


The following discusses the C. difficile toxin regulatory factors involved in sensing environmental conditions.


TcdR


TcdR or “alternative RNA polymerase sigma factor TcdR” is encoded by the tcdR gene. Sequences for TcdR are known for a number of species, e.g., for C. difficile 630 (the TcdR NCBI Gene ID is 4914073) and polypeptide sequence (e.g., YP_001087134.1 (SEQ ID NO: 5). The sequence for the TcdR gene product is as follows (SEQ ID NO: 5):










(SEQ ID No: 5)










  1
mqksfyeliv larnnsvddl qeilfmfkpl vkklsrvlhy eegetdliif fieliknikl






 61
ssfseksdai ivkyihksll nktfelsrry skmkfnfvef denilnmknn yqsksvfeed





121
icffeyilke lsgiqrkvif ykylkgysdr eisvklkisr qavnkaknra fkkikkdyen





181
yfnl






The tcdR gene sequence for C. difficile 630 is as follows (SEQ ID NO: 6):










(SEQ ID NO: 6)










  1
atgcaaaagt ctttttatga attaattgtt ttagcaagaa ataactcagt agatgatttg






 61
caagaaattt tatttatgtt taagccatta gtaaaaaaac ttagtagagt tttacattat





121
gaagagggag aaacagattt aataatattt tttattgaat taataaaaaa tattaaatta





181
agtagctttt cagaaaaaag cgatgctatt atagtcaaat atattcataa atcattactg





241
aataagactt ttgagttgtc tagaagatat tctaaaatga agtttaattt tgtagaattt





301
gatgaaaata tcttaaatat gaaaaataat tatcaaagta agtctgtttt tgaggaagat





361
atttgttttt tcgaatatat tttgaaagaa ttatctggta ttcaaagaaa agttattttt





421
tataaatatt taaaaggata ttctgataga gaaatatcag tgaaattaaa aatatctaga





481
caagctgtta ataaggctaa aaatagagca tttaaaaaaa taaaaaaaga ctatgaaaat





541
tattttaact tgtaa






TcdR polypeptide expression can be detected or measured via immunoassay, e.g., via an ELISA using antibodies raised against TcdR. TcdR expression can also be detected or measured by RT-PCR using primers specific for the tcdR mRNA.


CodY


CodY or “transcriptional repressor CodY” refers to a protein encoded by the codY gene. CodY is a sensor of carbon and nitrogen, and binds GTP and leucine to become active. When active, it represses transcription through a number of genes. C. difficile actively undergoing Stickland fermentations (abundant leucine and energy) represses toxin production in part through CodY's effects on tcdR and tcd operon gene expression. Transcription of the tcdR gene is repressed during the rapid exponential growth phase by CodY. CodY is active in cells with an excess of branched-chain amino acids (isoleucine, leucine, and valine) and GTP. When the cells reach stationary phase, the intracellular concentrations of these ligands decrease and CodY is less able to bind as a repressor, leading to derepression of tcdR transcription.


Sequences for CodY are known for a number of species, e.g., for C. difficile 630 (the codY NCBI Gene ID 4915868) and polypeptide sequence (e.g., YP_001087769.1 (SEQ ID NO: 7). The sequence for the codY gene product is as follows (SEQ ID NO: 7):










(SEQ ID No: 7)










  1
masevlqktr kinktlqtsg gssvsfdlla galgdvlssn vyvvsakgkv lglhlndvqd






 61
ssviedeytk qkkfsdeytq nvlkidetle nlngekilei fpeehgrlqk yttvvpilgs





121
gqrlgtlvls rysnsfnddd lviaeysatv vgleilraig eeleeemrkk avvqmaigtl





181
syseleaveh ifaeldgkeg llvaskiadr vgitrsvivn alrkfesagv iesrslgmkg





241
thirilndkl tdelkklknn q






The coDY gene sequence for C. difficile 630 is as follows (SEQ ID NO: 8):










(SEQ ID NO: 8)










  1
atggcaagtg aagtgttaca aaaaacaagg aaaataaata aaacattaca aacaagtggt






 61
ggaagcagtg tctcttttga tttactggcc ggagcattgg gcgacgtttt aagttctaat





121
gtttatgtag taagtgcaaa aggtaaagta ctaggtcttc atttaaatga tgttcaagac





181
agttcagtta tagaagatga gtatactaag caaaagaaat tttcagatga atatactcaa





241
aatgtgttaa aaattgatga aacattagaa aatttaaatg gtgagaagat attagaaatc





301
tttcctgaag aacatggaag attacaaaaa tatactacag tagttccaat attaggaagc





361
ggtcaaagat taggaacatt ggtactttca agatattcaa attcattcaa tgatgatgat





421
ttagtaatag ctgaatacag tgcaactgtt gttggtcttg aaatattaag agcaataggt





481
gaagaattag aagaagaaat gagaaagaaa gctgtagttc aaatggcaat aggcactctg





541
tcctactccg agcttgaagc agttgaacat atttttgctg aattggatgg aaaagaaggt





601
ctacttgtag caagtaagat agctgataga gttggtataa ctaggtctgt aatagtaaat





661
gcacttagaa aatttgagag tgcaggtgtg atagaatcaa gatcattagg tatgaaaggt





721
actcatataa gaatacttaa tgacaaactt acagatgaat taaaaaaatt aaaaaacaat





781
caataa






CodY polypeptide expression can be detected or measured via immunoassay, e.g., via an ELISA using antibodies raised against CodY. CodY expression can also be detected or measured by RT-PCR using primers specific for the codY mRNA.


CcpA


Catabolite control protein A, CcpA, senses carbon state within the cell and is a global regulator of carbon metabolism in Gram-positive bacteria. In B. subtilis, fructose derivatives of glucose bind and activate CcpA. Regulation of C. difficile toxin production by carbon source is mediated at least in part by CcpA, which is a direct repressor of the tcdA and tcdB genes.


CcpA protein is encoded by the ccpA gene. Sequences for CcpA are known for a number of species, e.g., for C. difficile 630 (the CcpA NCBI Gene ID 4915199) and polypeptide sequence (e.g., YP_001087548.1 (SEQ ID NO: 9). The sequence for the CcpA gene product is as follows (SEQ ID NO: 9):










(SEQ ID No: 9)










  1
mkgnitikdv akqagvsist vsrvindskp vtdevkqkvl eviketgyip nplarslvtk






 61
ksqligvivp evsdsfvnev lngieevakm ydydillant ysdkeqelks inllrakqve





121
givmiswive qehinyiqnc gipatyiskt arnydiytvs tsneeatfdm tehlikkghe





181
kiafimtskd dtvlemerla gyekalsnnn ieldksliky ggtdyesgyn smkellddgi





241
iphaafvtgd eaaigainai cdagykvped isvagfndvk iarmyrpklt tvyqplydmg





301
avairmvikl inkelienkk ielpyrivdr esvterkk






The CcpA gene sequence for C. difficile 630 is as follows (SEQ ID NO:10):










(SEQ ID NO: 10)










  1
atgaaaggca atataacgat aaaagatgtt gctaaacaag caggagtgtc aatatctact






 61
gtatctagag ttataaatga ttcaaaacct gtaactgatg aagtcaaaca aaaagtttta





121
gaggttataa aagagactgg atatatacca aatccacttg ctagaagctt agtaacaaag





181
aagagtcaat taataggggt aatagttcca gaagtttcag attcttttgt taatgaggtg





241
ttaaatggga tagaagaggt tgctaaaatg tatgactatg atattctttt agcgaataca





301
tactctgata aggaacaaga acttaagagt ataaatctat tgagagcaaa acaagtggaa





361
ggtatagtta tgatttcatg gatagttgaa caagaacata tcaactatat acaaaattgt





421
ggaataccag cgacatatat aagtaaaact gctagaaatt atgatatata tacagtaagt





481
actagcaacg aagaagctac ttttgatatg acagagcatc ttataaagaa aggtcatgaa





541
aagatagctt ttataatgac gagtaaagat gatactgttt tagaaatgga aagacttgct





601
ggttatgaga aagcactttc aaataacaat atagaattag acaagagttt gattaagtat





661
ggtggaactg attatgagag tggatacaat agtatgaaag aactattaga tgatggaata





721
atacctcatg cggcttttgt aacaggtgat gaggctgcca taggtgctat aaatgctata





781
tgtgatgctg gatataaggt tccagaagac atatctgttg caggatttaa tgatgttaag





841
atagctagaa tgtatagacc taaacttact acagtatatc aacctctata cgatatggga





901
gcagtagcaa taagaatggt tataaaatta ataaataagg aattaattga aaataagaaa





961
atagaattac cttatagaat tgttgataga gaaagtgtta cagaaagaaa aaaataa






CcpA polypeptide expression can be detected or measured via immunoassay, e.g., via an ELISA using antibodies raised against CcpA. CcpA expression can also be detected or measured by RT-PCR using primers specific for the ccpA mRNA.


Rex


Rex or “redox-sensing transcriptional repressor” senses energy state of the cell, particularly the concentration of NADH/NAD+. When activated by low energy (high concentrations of NAD+), Rex is known to indirectly lead to toxin expression, though the mechanisms of action are not well described. High NAD+ and butyrate levels, the latter possibly reflective of NADH consumption, are believed to promote C. difficile toxin production through this pathway.


Sequences for Rex and the gene sequence encoding it are known for a number of species, e.g., for C. difficile 630 (the rex NCBI Gene ID 4914836) and polypeptide sequence (e.g., YP_001086640.1 (SEQ ID NO:11). The sequence for the Rex gene product is as follows (SEQ ID NO: 11):










(SEQ ID NO: 11)










  1
mlgnknisma virrlpkyhr ylgdlldrdi qrisskelsd iigftasqir qdlnnfggfg






 61
qqgygynvea lhteigkilg ldrpynavlv gagnlgqaia nyagfrkagf eikalfdanp





121
rmiglkiref evldsdtled fiknnnidia vlcipkngaq evinrvvkag ikgvwnfapl





181
dlevpkgviv envniteslf tlsylmkegk






The rex gene sequence for C. difficile 630 is as follows (SEQ ID NO:12):










(SEQ ID NO: 12)










  1
atgttgggaa ataaaaatat atcaatggca gttataagaa ggctcccaaa atatcataga






 61
tatcttggag acttattaga tagggatata caaagaatat cttctaaaga attgagtgat





121
ataatagggt ttaccgcttc tcaaataaga caagatttaa acaactttgg tggatttgga





181
caacaaggat atggttataa tgtagaagct cttcatactg agataggtaa aattcttggg





241
ttggatcgac catacaacgc agttcttgta ggagcaggta acttaggaca agctatagcc





301
aattatgcag gatttagaaa agctggattc gagataaaag ctttatttga tgcaaatcct





361
agaatgatag gtttaaagat aagagagttt gaagtattag attcagatac tttagaagac





421
tttataaaaa acaataatat agatattgct gtattatgta tacctaaaaa tggagcacaa





481
gaagttatta atagagttgt aaaagctgga atcaaaggtg tatggaattt tgcaccttta





541
gatttagaag ttccgaaagg tgttatagtt gaaaatgtaa acttaacaga aagtttattt





601
accttatcgt atttaatgaa agaaggaaag tag






Rex polypeptide expression can be detected or measured via immunoassay, e.g., via an ELISA using antibodies raised against Rex. Rex expression can also be detected or measured by RT-PCR using primers specific for the rex mRNA.


Energy status of C. difficile is important for determining whether the pathogen expresses toxin. The following considers various pathways in addition to Stickland fermentation that C. difficile uses to extract energy needed for metabolism from its environment.



C. difficile Carbohydrate Utilization



C. difficile metabolizes sugars such as glucose and fructose, and sugal alcohols such as sorbitol and mannitol for energy production. End metabolites and points of energy production are shown in FIG. 5G and in FIG. 9. Glycolytic pathways generate ATP and NADH and lead to production of pyruvate, lactate, propionate, acetate or ethanol as metabolites. The succinate pathway generates ATP and GTP and consumes NADH in the conversion of two pyruvates to succinate. Butyrate production can occur from condensation of two acetates into acetylCoA to butyrate, or from conversion of succinate to crotonylCoA to butyrylCoA and butyrate; while some ATP is produced in the process of butyrate production, substantive NADH is consumed in these pathways.



C. difficile Ethanolamine Utilization


Ethanolamine occurs abundantly in gut secretions, primarily from the breakdown of phosphatidyl ethanolamine from sloughed epithelial cells, as well as from dietary and other commensal sources. The C. difficile eut operons include a two-component ethanolamine sensing system and transporter, and downstream genes that encode the carboxysome, a polyhedral protein complex in which ethanolamine is metabolized to ammonia and acetaldehyde, resulting in the generation of NADH from NAD+. A schematic of the pathway is shown in FIG. 9.



C. difficile Reductive Leucine Pathway



C. difficile also uses the reductive leucine pathway for energy. Mediated by the had gene operon, the pathway generates 2 ATP from 3 leucine molecules. No net NADH/NAD+ is consumed, and the end product of this pathway is isocaproate.


Cysteine and Threonine Use



C. difficile can convert these amino acids to pyruvate with branch points for conversion of pyruvate to acetate, propionate or butyrate, that follow the glycolytic pathways.


SCFA, bSCFA and 5-Aminovalerate


Exogenous acetate and succinate are hypothesized to be taken up and converted to butyrate by C. difficile if it needs energy and does not have sugars or amino acids readily available. The capacity of C. difficile to use exogenous branched short-chain fatty acids and 5-aminovalerate is not known. Other organisms can use these compounds for further energy derivation. Without wishing to be bound by theory, another potential mechanism of protection provided by Stickland fermenting species could be cross-feeding these compounds to C. difficile for energy, or through other mechanisms of C. difficile sensing its extracellular environment.


PrdR


PrdR regulates that proline and glycine reductase enzymes, and also has indirect effects on toxin expression. With abundant proline, PrdR activates the proline reductase operon and inhibits the glycine reductase operon. PrdR activates the proline reductase operon and represses the glycine reductase operon. prdR suppresses the expression of C. difficile toxins by inhibiting butyrate, coDY, ccpA, tcdR, and/or tcdA production. The Stickland fermentation pathway using proline as the Stickland acceptor generates NADH and the metabolite 5-aminovalerate from proline. The acetate-generating pathway generates NADH and the metabolite acetate from glycine. C. difficile consumes large amounts of NADH in the carbohydrate metabolism pathway from acetate


Therapeutic Microbiota


Described herein are therapeutic microbiota that suppress toxin production by Gram positive, spore-forming bacteria such as C. difficile. As described herein above, commensal species that provide C. difficile with amino acids and other energy sources that promote Stickland fermentation can provide therapeutic benefits by suppressing C. difficile toxin production. Such species include, for example, species that express and secrete one or more protease enzymes that generate a supply of amino acids for C. difficile to derive energy from via Stickland fermentation, as well as species that themselves perform Stickland fermentation.


Proteases Expressed by Therapeutic Species


In one embodiment, a method of suppressing bacterial toxin production or treating or preventing a pathology caused by expression of a bacterial toxin compromises administering at least one bacterial organism that encodes and secretes a protease enzyme. Protease enzymes catalyze protein catabolism by hydrolysis of peptide bonds. Proteases are produced by all living organisms, in which they display many physiological functions ranging from generalized protein degradation to more specific and regulated processes such as blood coagulation, hormone activation, or transport of secretory proteins across membranes. Proteases can be either exopeptidases, whose actions are directed by the amino- or carboxyl terminus of proteins, or endopeptidases, which cleave internal peptide bonds. Extracellular proteases are made as precursors, containing an amino-terminal signal peptide, which is removed during the export to generate a mature, extracellular protease.


Proteolytic Clostridia elaborate a number of extracellular and membrane-bound metalloprotein, serine and other classes of proteases. The proteases generate free amino acids from available extracellular protein. An estimated 10-15 grams/day of undigested dietary protein enters the large intestine and remains available for microbial digestion and growth. The proteolytic species commonly use Stickland fermentations as core metabolic processes for energy generation under anaerobic conditions. Protease activity can be measured biochemically with lysis assays of casein, gelatin and other proteins, and with, e.g., FITC-conjugated substrates that release the fluorophore for detection. Microbiologically, protease activity can be measured on solid agar (casein hydrolysis agar), or with casein gelatin or meat granule hydrolysis in liquid media.


Among Stickland-fermenting Cluster XI Clostridia, Clostridium bifermentans is among the most highly proteolytic species, preferring Stickland fermentations over glycolytic pathways for energy extraction. In contrast, C. sardiniense, a non-proteolytic and strongly glycolytic Cluster I Clostridial species, produces abundant butyrate through anaerobic fermentation of available carbon sources.


In one embodiment, a bacterial organism administered as described herein, e.g., to suppress C. difficile toxin production, encodes and secretes at least one protease selected from the group shown in Table 2 or a homologue thereof. In another embodiment, the species administered encodes and secretes two or more, three or more, four or more, five or more, six or more, seven or more, eight or more, nine or more, ten or more, eleven or more, twelve or more, thirteen or more, fourteen or more, or all of the protease enzymes listed in Table 2. In another embodiment, the species administered expresses one or more (up to and including all) enzymes with the same Enzyme Commission (E.C.) number as an enzyme in Table 2. That is, the administered species can express and secrete one or more enzymes that catalyze the same reaction as an enzyme listed in Table 2. Additional bacteria that express and secrete such enzymes can be identified, for example, by analysis of bacterial genomic sequences for sequence encoding such secreted enzymes. For example, the sequence encoding a representative listed C. bifermentans protease can be used to query the genomic sequences of other non-pathogenic bacteria, including but not limited to other commensal bacteria, including but not limited to other Clostridial bacteria, for sequence encoding a homologous enzyme.









TABLE 2







Representative secreted proteases encoded by



Clostridium bifermentans MHMC14 and the corresponding



PATRIC (Pathosystems Resource Integration Center)


ID Number (https://www.patricbrc.org/).










Extracellular Proteases
PATRIC ID







Protease PrsW
fig|186802.30.peg.279



ATP-dependent protease La
fig|186802.30.peg.290



(EC 3.4.21.53) Type II



Protease
fig|186802.30.peg.313



ATP-dependent Zn protease
fig|186802.30.peg.414



CAAX amino terminal protease
fig|186802.30.peg.543



family protein



CAAX amino terminal protease
fig|186802.30.peg.2205



family protein



CAAX amino terminal protease
fig|186802.30.peg.2313



family protein



Putative membrane protease YugP
fig|186802.30.peg.2680



Uncharacterized zinc protease YmfH
fig|186802.30.peg.2745



FIG001621: Zinc protease
fig|186802.30.peg.2746



Major intracellular serine
fig|186802.30.peg.830



protease precursor (EC 3.4.21.—)



CAAX amino terminal protease
fig|186802.30.peg.921



family protein



Uncharacterized zinc protease YmxG
fig|186802.30.peg.936



Serine protease, DegP/HtrA,
fig|186802.30.peg.3000



do-like (EC 3.4.21.)



ATP-dependent Clp protease
fig|186802.30.peg.3018



ATP-binding subunit ClpX



ATP-dependent Clp protease
fig|186802.30.peg.3019



proteolytic subunit (EC 3.4.21.92)



Tail-specific protease (EC 3.4.21.—)
fig|186802.30.peg.3065










In one embodiment, a bacterial organism administered as described herein, e.g., to suppress C. difficile toxin production, encodes an enzyme that participates in Stickland fermentation. Such enzymes include, for example, D-proline reductase and glycine reductase. Species that encode one or both of such enzymes can be identified, for example, by analysis of genomic sequences for sequence that encodes the enzymes.


D-Proline Reductase (PR)


“prdA” or “D-Proline reductase (PR)” is encoded by the prdA gene. Sequences for prdA are known for a number of species, e.g., for C. difficile 630 (the prdA NCBI Gene ID is 4916399 and polypeptide sequence e.g., YP_001089760.1), as well as for C. scindens ATCC 35704 (the prdA NCBI polypeptide sequence e.g., EDS06621.1) and for C. bifermentans ATCC 638 (the prdA NCBI polypeptide sequence e.g., EQK41327.1). Proline reductase levels can be measured, for example, via immunoassay or by measurement of RNA encoding the enzyme, e.g., via RT-PCR. The sequence for the prdA gene product for C. difficile 630 is as follows (SEQ ID NO: 13):










(SEQ ID NO: 13)










  1
msitletaqa handpavccc rfeagtiiap enledpaifa dledsgllti pengltigqv






 61
lgaklketld alspmttdnv egykageake evveetveea apvseaavvp vstgvagetv





121
kihigegkni sleiplsvag qagvaapvan vaapvasaaa evapkveekk llrsltkkhf





181
kidkvefade tkiegttlyi rnaeeickea netqelvvdm kleiitpdky etyseavldi





241
qpiatkeege lgsgitrvid gavmvltgtd edgvqigefg ssegelntti mwgrpgaadk





301
geifikgqvt ikagtnmerp gplaahrafd yvtqeireal kkvdnslvvd eevieqyrre





361
gkkkvvvike imgqgamhdn lilpvepvgt lgaqpnvdlg nmpvvlsple vldggihalt





421
cigpaskems rhywreplvi ramedeeidl vgvvfvgspq vnaekfyvsk rlgmlveame





481
vdgavvtteg fgnnhidfas hieqigmrgi pvvgvsfsav qgalvvgnky mthmvdnnks





541
kqgieneils nntlapedav rimamlknai egvevkaper kwnpnvklnn ieaiekvtge





601
kivleeneqs lpmskkrrei yekden






The prdA gene sequence for C. difficile 630 is as follows (SEQ ID NO:14):










(SEQ ID NO: 14)










   1
atgtcaataa ctttagaaac agctcaagcc catgcaaatg acccagcagt ttgttgttgt






  61
agatttgaag cgggaacaat tatagcgcca gaaaacttag aagatccagc aatatttgca





 121
gacttagagg attctggatt attaacaata ccagaaaatg gattaactat aggtcaagta





 181
ctaggagcta agttaaaaga aactttagat gcactttctc caatgactac agataacgta





 241
gaaggataca aagcaggaga ggctaaagaa gaagtagtag aagaaacagt agaagaagca





 301
gctccagtat cagaagcagc agtagttcca gtaagcacag gagttgcagg tgaaacagtt





 361
aaaatacaca taggtgaagg taagaacata agcttagaga tacctttatc agtagctggt





 421
caagcaggag ttgctgctcc agtagcaaac gttgctgctc cagtggcaag tgcagcagca





 481
gaagtagctc caaaagttga agaaaagaaa cttttaagaa gcttaactaa aaaacacttt





 541
aaaatagata aagttgaatt tgctgatgaa actaaaatag aaggaactac tttatacatc





 601
agaaacgcag aagaaatatg taaagaagct aatgaaactc aagagttagt tgtagatatg





 661
aagttagaaa taataactcc tgataaatat gaaacttaca gtgaagctgt attagatata





 721
caaccaatcg ctactaaaga agaaggcgaa ttaggttcag gtataactag agttatagat





 781
ggagctgtaa tggtattaac tggtacagat gaagatggag ttcaaatagg tgaatttggt





 841
tcttcagaag gtgagttaaa tactactata atgtggggta gaccaggtgc tgctgacaaa





 901
ggtgaaatat tcatcaaagg tcaagtaaca ataaaagcag gaactaacat ggaaagacca





 961
ggacctttag ctgctcaccg tgcatttgac tatgtaactc aagaaataag agaagcatta





1021
aagaaagttg acaactcttt agtagttgat gaagaagtaa tagagcaata cagaagagaa





1081
ggtaaaaaga aagttgttgt tataaaagaa ataatgggac aaggtgcaat gcatgataac





1141
ctaatattac cagttgagcc agttggtaca ttaggagctc aaccaaacgt tgacttagga





1201
aacatgccag ttgtattatc tccacttgaa gtattagatg gtggtatcca tgcattaact





1261
tgtataggac ctgcatcaaa agaaatgtca agacattact ggagagagcc attagtaata





1321
agagctatgg aagacgaaga aatagattta gtaggtgttg tatttgttgg ttctccacaa





1381
gtaaatgctg agaaattcta tgtatctaag agattaggta tgttagttga agctatggaa





1441
gttgatggag ctgtagtaac tactgaaggt ttcggaaaca accatataga tttcgcatct





1501
cacatagagc aaataggtat gagaggtata ccagtagttg gtgtaagttt ctcagctgtt





1561
caaggtgctc tagttgttgg taataaatac atgactcaca tggtagacaa caataagtct





1621
aagcaaggta tagagaatga aatattatct aacaacactt tagctccaga agatgctgtt





1681
agaataatgg ctatgcttaa aaatgctata gaaggtgtag aagttaaagc tcctgaaaga





1741
aaatggaatc caaatgttaa attaaataac atagaagcta tagaaaaagt tacaggagaa





1801
aaaatagtat tagaagagaa tgagcaatct ctaccaatga gtaagaagag aagagaaata





1861
tacgaaaaag acgaaaacta a






Each reductase is comprised of multiple polypeptides of which the sequences are listed below. In one embodiment, the prdA gene (D-proline reductase (EC 1.21.4.1)) sequence for C. bifermentans 638 is as follows (SEQ ID NO:15):









(SEQ ID NO: 15)


atgggtataggaccatcaactaaagaaacatcattacatcactttagaga





tccgcttcttgatatagttagtaatgacaaagacatagatcttctgggga





tagtagtagtaggaacacctcaggacaacaaagaaaaagaatttgttgga





caaagaacagctgcatggctagaagctatgagagcagatggtgttataat





ttcatgtgatgggtggggaaactcacacgtagattatgctaatactattg





aagaaataggaaaaagagagatcccggtagttggacttacatttaatgga





acacaagctaagtttgtagttacaaataaatatatggacacaatagtaga





ttttaataaatcagacaaggggatagaaacagaagttgtcggagagaaca





ctgtaagcgagttagacgcaaaaaaatcattagccttattaaaattaaaa





atgcaaagaaataataaaaaataa






In one embodiment, the prdA gene (D-proline reductase (EC 1.21.4.1)) sequence for C. bifermentans 638 is as follows (SEQ ID NO:16):









(SEQ ID NO: 16)


atgtcaataactgtagaaacagctaaagctcatgctaaagatccagcggt





atgctgctgtagatttgaagctgggactgtactagaaccatcaaatttag





aagatccagcaatattcgctgacttagaggattcaggattattaacaata





gcagatgattgtttaacaatagagcaagttttaggagctaaactattaaa





aactttagatgctttaactccaataactgctgactgtgtagaaggtgtag





tagcagtagctgaagaggctaaagaagaagttaaggaagaagttaaagaa





gtagcaccagttgcttcagtagctccagtatctcaaatagctccagtaaa





tggacaaactataaagatacatataggtgaaggtagagatataaacttag





aaatacctttaaatgtagctcaaggaatgggtgtagcaccagttgctcct





gtagctgtagcagaaaatgcagaagctgtagaagttaaagctgagccagt





tcaagaagctaaagcaatgagaagcttaactaaaaaacattttaaaatag





aaaaagtagttttcgctgaagaaactaaaatagatggaactactttatac





ttaagaactccagaagaattaactaaagaagctgtaaattcagaagaatt





agttgttgatatgaagttagaaataataactccagctgaatacaacaaat





acagtgaaactataatggatgttcaacctatagctgctaaagaagaagga





gaaataggagaaggtgtaacaagagttatagacggagttataatgatggt





aactggtactgatgaaaacggagttcaaataggtgaattcggttcttcag





aaggtgtattagaaactaacataatgtggggaagaccaggtgctcctgat





aaaggtgatatattcatcaaaactcaagtaacagttaaagctggtactaa





catggaaagaccaggaccattagctgctcactgtgcatctgattatataa





ctcaagaaataagagaagcattaaagaacgctgaagagtctttagtagtt





gatactgaagaattaactcaatatagaagacctggtaagaaaaaggttgt





tgtagttaaagagataatgggacaaggggcaatgcatgataacttaatat





tacctgttgagccagttggaacattaggagctaaaccaaacgttgactta





ggaaacgttccagtagtattatctccacttgaagtattagatggtggtat





acatgcattaacttgtataggacctgcatctaaagaaaactctagacatt





actggagagagccattagtaatagaagctatgcatgatgaagaaatagat





ttagtaggtgttatatttgtaggatctccacaagtaaatgctgagaaatt





ctatgtatctaagagattaggtatgatgatagaagctatgggtgttgatg





gtgctatagtaacaactgaaggattcggaaacaaccatatagatttcgct





tctcatatagagcaaataggtaagagagatgtagctgtagtaggtgtaag





tttctctgctgttcaaggtgctctagttgttggtaatgaatacatgaaat





acatgatagacaacaacaagtctaaacaaggtatagaaaatgaagtatta





tcaaacaatacattatgcccagaagatgctgtaagatctttagcaatgtt





aaagacagtaatgggtggagaagaagttaaagctgctgagagaaaatgga





atgctaacgttaaattaaataacgttgaattaatagaaaaagaaactggt





aagaagttagaacttgttgaaaacgagcaaactttaccaatgagtgaaaa





aagaaagaatatatacgaaaaagacgctaaatag






In one embodiment, the prdA gene (D-proline reductase (EC 1.21.4.1)) sequence for C. bifermentans 638 is as follows (SEQ ID NO:17):









(SEQ ID NO: 17)


atggaagagaaaatacttagacgtttggtaattaaaccatttcatataa





ataatgttgaattcaatgaaaagttctcaataaaaaaaggtacactatc





cataaacaatgactacataaatgaaattaaaaattcacatgaattaata





acggacataaaattagatataatcaaaccaggagattataacaaggaaa





ttaatactatcatggatataatccctatatctactaaagttttaggtag





attaggtgaaggaataacacacactttaacaggtgtttatgttatgctt





actggtgttgatgaagatggaagacaaatgcatgaatttggatcttcag





aaggtatactttctgagcaaatggtgtttggaagatatggtactccatc





tactaatgattacataattcattttgatgttacagttaaaggtgggttg





ccatatgagagaaaacttccgatgatgacatttaaggcatgtgatactt





ttatacaaggtataagaaatgttttaaaacagcaagacggaagagatgc





tacagaaattcgtgaatattttgacaaaattagacctgacgctaaaaaa





gttgtaatagtaaaacaaatagcaggtcagggtgcaatgtatgacaatc





aattattttctcatgaaccaagtggtttagagggaggtacatccattat





tgatatgggaaatgtaccgatgataatatcacctaatgaatacagagat





ggcgccttgagagctatgacttaa






In one embodiment, the prdA gene (D-proline reductase (EC 1.21.4.1)) sequence for C. bifermentans 638 is as follows (SEQ ID NO:18):









(SEQ ID NO: 18)


atgagccttacaacaataaaaggacttcaatctgaaatatttgtaccaat





aacacctcctcctgtttggactcctgtaactaaagaactaaaggatatga





ctatagctttagctacagcgtcaggtgtacatttaaaagctgataagaga





ttcaacctagcaggtgactttacatttagagaaataccagacacagcaac





tactgatgagatgatggtatctcacggaggatatgataacgctgatgtta





ataaagatataaactgtatgttccctatagacagactacatgaattagct





aaagaaggatttataaaatctgtagctccagttcatataggattcatggg





tggtggtggagaccaaactaaattcactgaagaaactggtcctgaaatcg





ctaaaagattaaaagatgagggagtagacggtgtagttctaacagctggc





tgaggtacttgccatagaactgccgtgatcgtgcagagagcaatagaaga





agctggtataccaactataataatagcagctcttcctccagtagttagac





aaaacggaactccaagagcagttgctccactagttccaatgggtgctaat





gctggtgaaccaaacaataaagaaatgcaaatgcatatattaagagatac





tttagagcaattaatagctataccatctgctggtaagataattcaattac





catacgagtatgtagctcaagtataa






In one embodiment, the sequence for the prdA gene product for C. scindens 35704 is as follows (SEQ ID NO: 19):










(SEQ ID NO: 19)










  1
msitaetake handpavlcc raeagitiea anledpaifd dlvdsgllnl dgaltieevl






 61
gakltktcds lcpltadvve gakaptapaa eeaeeeapaa papaaapvag paaggtlkih





121
igegkdidle ipvgalggga avaplpagae avvagaaape aageekvvrs ltrkhftite





181
vkrgpetkie gttlyiregi esevidnqel vkdfkleiit pdlyhtyset vmdvqpiatk





241
egddelgtgv trvldgvvmm ltgvdeggvq igefgssegy ldenimwnrp scpdkgeifi





301
kgniviqekt nmerrgpmaa htafdvitqe irevmkkldd slvadteelk qvrrpgkkkv





361
vivkeimgqg amhdnfilpv epvgvlgara nvdlgnvpvc vsplevldgc ihaltcigpa





421
skemsrhywr eplvlealhd pevdlcgvvf vgspqinaek fyvsrrvght vemmdadgaf





481
vttegfgnnh idfashieqi gmrgipvvgm sycavqgalv vgnkymtymv dnnkseagie





541
neilgnntlc pedavralam lktamagedv kaaekkwnpn vkstnvelie stygtkvdlv





601
eneqalpmse krrlkys






In one embodiment, the prdA gene (D-proline reductase (EC 1.21.4.1)) sequence for C. scindens 35704 is as follows (SEQ ID NO:20):









(SEQ ID NO: 20)


ttggctgaagaggtaaaagacctgagacgtcttgtaattaaagcgttcca





catgaatgatgtagagtggggtgaacataatgatattactgttgacggta





atatgacagtcagtaaagaaatgattgatcagctggtggctcaggaggaa





cacattgaaaaaattgatattcagattattaagccgggggatcatgaccg





ttggacgaatacgattatggatatcataccgatctctacaaaggtacttg





gaaaattaggggagggcattacccataccattaccggcgtatatgtaatg





cttaccggcgttgacgtaaatggaaagcaatgccatgaattcggttcttc





tgaggggaatctgaaagaccagctgtacttgaaccgtgcaggcacgccgg





gggatgatgattacataatttcctttgatgtaacgcttgcagccggaatg





gggcaggagaggcctggaccgactgccgcacatagggcgtgcgataagtt





tatccagacataccgtgataagatgaagaagttcaaaggcgagaagtgta





cggaacgccatgagtaccatgatgtggtaaggccgggaaagaaacgcgtc





ctgatcgtaaagcaggtggcaggacagggagcaatgtatgatacgcatct





gttttccaaagagccgtctggcgtagagggcggacgttcaattatcgata





tgggcaatatgccgatccttgtaactccaaatgagtacagagacggtatt





atccgctccatgcagtag






In one embodiment, the prdA gene (D-proline reductase (EC 1.21.4.1)) sequence for C. scindens 35704 is as follows (SEQ ID NO:21):









(SEQ ID No: 21)


atgtcaatcacagctgaaacagcgaaagaacatgctcatgatcctgcggt





attatgttgtagagccgaagcaggcattacaatcgaagctgctaatcttg





aagatccggcgatctttgatgacttggtagattcaggattattgaacctg





gatggtgcattgaccatcgaagaagttttgggagcaaaacttacaaaaac





atgtgattctctttgcccgttaactgcagatgtagttgaaggtgcaaaag





cgccgactgctccagcagcagaagaggcagaagaggaagcgccggcagca





ccggcaccggctgcagcacctgtagcaggacctgcggcaggcggaacact





taagatccacattggagaaggcaaggacattgatcttgagatcccagttg





gagcgcttggcggcggagcagcagttgcaccattgccggcaggagcagag





gcagttgttgcaggagcagcagcaccagaagcagctggagaagaaaaggt





tgtaagaagtttaacaagaaaacacttcacgatcacagaggttaagagag





gaccagagaccaagatcgaaggaacaactctttacatccgtgaaggcatt





gagtcagaagttattgacaaccaggagcttgtaaaagatttcaaactgga





aatcatcactcctgatttatatcacacatattccgagactgttatggacg





ttcagccaatcgctacaaaagaaggcgatgatgaactcggaacaggtgtt





acaagagtacttgacggcgttgttatgatgctgacaggtgttgacgaagg





cggagttcagattggcgagttcggttcttcagaaggataccttgatgaga





acattatgtggaatcgtccgagctgcccagataaaggcgagatctttatc





aagggtaacatcgtaatccaggaaaagacaaacatggaacgtcgtggacc





tatggctgctcatacagcatttgatgtaatcacacaggaaatccgcgaag





ttatgaagaaacttgatgacagccttgttgctgatacggaagaactgaag





caggttcgccgtccgggcaagaagaaagtcgttatcgttaaggaaatcat





gggacagggagctatgcatgacaactttatccttcctgtagagcctgttg





gcgttctaggcgcaagagctaacgtagacttaggaaacgtaccggtttgc





gtatctccattggaagttcttgatggatgtatccatgcattaacatgtat





cggacctgcatctaaggaaatgtccagacattactggagagagccattgg





ttctggaagcattgcatgacccggaagttgacctttgcggcgttgtattt





gtaggatctcctcagatcaatgctgagaaattctatgtatcccgtcgtgt





aggccataccgtagaaatgatggatgctgatggagctttcgttacaacgg





aaggttttggaaacaaccacatcgatttcgcaagccatatcgagcagatc





ggtatgagaggaattccggttgttggcatgtcttactgtgcagttcaggg





cgctctggttgttggtaacaagtatatgacatacatggttgacaataaca





agtctgaagctggtatcgagaacgagattcttggtaacaatacgctttgc





ccggaagatgctgttcgtgcacttgctatgcttaagactgcaatggcagg





cgaagacgttaaggctgctgagaagaagtggaatccaaacgttaagtcta





caaacgtagagttaattgagagcacatacggtacaaaggttgatcttgtt





gaaaatgagcaggctcttccgatgagtgaaaaacgtagattaaaatacag





ctaa






In one embodiment, the prdA gene (D-proline reductase (EC 1.21.4.1)) sequence for C. scindens 35704 is as follows (SEQ ID NO:22):









(SEQ ID NO: 22)


atgaatgtaggatcaaggctgacggttaaggcgtaccctgtcacagaagt





gtgctatggggaggagaaccgagtgacggtggatggccggatgacggtct





gtaagaacatagcagaaaagattctggcgcaggagccattgataaaggag





attgatatccgtattatcatgccggatgagcaccgacagcataccaacac





ggtgatggatgtgattcctctggcaaccaaagtgctgggacgggtggggg





agggcattacccataccctgacaggcgtatacgtgatccttaccggtgtg





gatgagagcgggcgtcagatatgtaattttggcgccagcgacggaatact





cgaggagaagattgcctgggggcgggcgggaacgccgcttaggagcgacg





tgctgatctcctttgacgtggttcttaaggaaggatcctgggcggatcgt





ccgggtccggaagcagcccatcgcgcctgcgatacatactgccagatatt





ccgggagcagataaagaagtttaatggatacaagtgcgcggaaaagcatg





tctttcaggagacgtatgagccggggaaaaaagatgtctatattgtgaaa





gaagtatccgggcaaggtgccgtatacgatacccggatgttcggacatga





gccttgcggattcgaaggcgggaagtctgttattgatatgggctgcatgc





ctgcgctggtgacgcccaatgaatttagggatggcattatgcgcgcgatg





gattag






In one embodiment, the prdA gene (D-proline reductase (EC 1.21.4.1)) sequence for C. scindens 35704 is as follows (SEQ ID NO:23):









(SEQ ID NO: 23)


atgtctattacagcagaaactgcaaaagaacatgcaaatgacccggctgt





attatgctgccgggcagaagagggcattacaatacaggcttccaacttgg





aagatcctgctatttttgacgagttagtggattcagggctgctatctttg





gatggctgtctgacaatcggacaagtcttaggggcaaccctgacaaagac





aagcgattctttatgtccattgactgcagataacgtagggggcttcaaag





aggtagttgaggaagaagagcctgcatcagagccagtcgaagaagcggta





gccgcagatattaatattgggggcgcggtcaccacgatcaaaaatggaaa





agttgttatttcaatcaaagaaggaaaagatatctatttagaacttcctg





tttaa






In one embodiment, the prdA gene (D-proline reductase (EC 1.21.4.1)) sequence for C. scindens 35704 is as follows (SEQ ID NO:24):









(SEQ ID NO: 24)


atgggaaatgtacagattttattacgtcagcatgttggtgcaccctgtga





ggcaatcgtaaaggctggggataaggtggaaaaaggtaccttgattgcaa





ctcctacaggacttggcgctaacatcttttccagcgtctatggcgtggtg





gaagaagtcttggaagaccgaatcgttatcaagccggatgaagagcagaa





agatgagtttgtacctattaaggaaggcagcaagcttgagatggttaagg





aagccggaatcgtaggtatgggcggcgcaggattcccaactggcgtgaag





attggaacggaccttcacggcggatatatcctggtaaatgctgcagaatg





cgagcctggacttcgccacaatatccagcagattgaagaaaagacagata





tcacaatccgcggattgaaatactgcatggagatatccaatgcggcaaaa





ggaattattgctattaagaagaagaacgaaaaagcgatcgaatttctcag





agaggcaatcaaggatgaagacaatatcacgatccatcttcttccggata





tttacccaatgggagaggaaagagcggtagtaagagaatgcctcggaaaa





ctgcttgatcctacacaacttccgtcagcagcagatgcagtcgtaatcaa





ctgcgagaccctgcttcgtatcgcagaggcgatcgaacttaagaaacctt





gctttagcaagaatatgacggttattggaaagattaacggtggaaacgag





ccgcatgtattcatggatgttccggttggaacctgtgttgcagacatgat





cgagaaggcaggcggaattgatggtacatatggcgagattatcatgggtg





gagcatttactggaaagtccaccacattagacgcgcctactacgaagacg





acaggcggaatcatcgttacggtagagttcccggatcttcacggagcgcc





ggtaggattgcttgtctgtgcgtgcggcggaagcgaagaccgtatgcgcg





aactttgcgaaaagatgaatggaaaggtcgtttctgtggcaagatgtaaa





caggcggttgagccgaagccgggcgcagcgcttaagtgcgagaatcctgg





aaactgtcctggacaggcacagaaatgtctgcagtttaagaaggacggcg





cagagtacatcatcatcggtaactgctcagactgttccaacacagttatg





ggatctgcaccaaagttaaaactgaagacattccatcagacagaccatgt





gatgagaacaatcggtcatccattatacagaagactgaccgtgtccaaag





aagttgaccagctgcccaacggcaaataa






In one embodiment, the prdA gene (D-proline reductase (EC 1.21.4.1)) sequence for C. scindens 35704 is as follows (SEQ ID NO:25):









(SEQ ID NO: 25)


atgggtataggaccatcaacaaaagaaacatcattgcatcacttcaggga





tccgctgctggatgtagtctcttcggatacagatctggatctgatgggaa





ttatcatcgtaggaacaccggacgataatgaggataagatgcttgtagga





accaggacggctgtttgggccgaggcaatgcgtgcggacggcgtaatcat





ctcttcggacggatggggaaacagcgacgtggattacacgaatacatgcg





agcaggtggggacgagaggcatcgcggtgacgggccttaatttcagcggt





acggtagctcaatttgtagttgtaaataattacctggatggaattgtgga





tatcaataagagcgcggacgggacagagaccaatgtggttggggaaaaca





atatggtcgagctggattgcaaaaaggcgactgcgcttctgaaacttaag





atgcgaaagaatgagaaaaagtag






In one embodiment, the prdA gene (D-proline reductase (EC 1.21.4.1)) sequence for C. scindens 35704 is as follows (SEQ ID NO:26):









(SEQ ID NO: 26)


atgagtttaacggttgttaaaggtttacaatctgaaatattcgttcctat





tactccaccatcagtatggactcctgtaacaaaagagttgaaagacatgt





ctatcgctcttgcaacagctgccggtgttcataagaaggatcaggaaaga





ttcaatcttgctggtgactttacatggagaaaaatagagaacacaacacc





atctagcgaactgatggtatcccatggtggatatgataacagtgatgtta





acaaagatatcaactgtatgttcccgattgacagaattcatgaattggct





gctgaaggatttatcagggcttgtgctccggtacatgcaggattcatggg





tggtggcggaaaccaggagaagttcaaaggcgaaactggtccggctatcg





cgcagatgttcaaagaagaggacgttgacgcagtaattctcaccgctggc





tgaggaacctgccaccgctctgcagtattggtgcagagagcgattgaaga





agctggaattcctactattattattgcagctcttccaccagttgttcgcc





agactggtactcctcgtgcagttgctccattggtacctatgggtgctaat





gcaggtggaccgcacaatgttgaacagcagacacagatcgtaaaggcaac





tctggagcagttagttgaaatccagacacctggaaagattgttccactgc





cattcgagtatgtagctaagatttaa






The sequence for the prdA gene product for C. bifermentans 638 is as follows (SEQ ID NO: 27):










(SEQ ID NO: 27)










  1
meekilrrlv ikpfhinnve fnekfsikkg tlsinndyin eiknshelit dikldiikpg






 61
dynkeintim diipistkvl grlgegitht ltgvyvmltg vdedgrqmhe fgssegilse





121
qmvfgrygtp stndyiihfd vtvkgglpye rklpmmtfka cdtfiqgirn vlkqqdgrda





181
teireyfdki rpdakkvviv kqiagqgamy dnqlfsheps gleggtsiid mgnvpmiisp





241
neyrdgalra mt






In some embodiments, it is useful to measure proline reductase levels or activity, e.g., as an indicator of the activity of the pathway in a sample, or, e.g., to identify a bacterial species as one that likely performs Stickland fermentation and/or can aid in suppressing C. difficile toxin expression. Measurement of proline reductase levels can be performed, for example, by immunoassay or by RT-PCR for the mRNA encoding the enzyme. Measurement of proline reductase activity can be performed, for example, with the fluorometric assay described by Jackson et al., J. Bacteriol. 188: 8487-8495 (2006), which is incorporated herein by reference. The assay follows the DTT- and d-proline-dependent production of δ-aminovaleric acid, which reacts with o-phthalaldehyde to generate a fluorescent product.


The nucleic acid and polypeptide sequences for the proline reductase expressed by C. difficile 630 are NCBI Gene ID 4916399, YP_001089760.1, respectively. The nucleic acid and polypeptide sequences for the proline reductase expressed by C. scindens 35704 NCBI Gene ID 167662491 and EDS06621.1, respectively. The nucleic acid and polypeptide sequences for the proline reductase expressed by C. bifermentans 638 are NCBI Gene ID 531765064 and EQK41327.1, respectively.


Glycine Reductase (GR)


The activity or expression of glycine reductase is important for Stickland fermentation via the glycine reductase pathway. In some embodiments, it is useful to measure glycine reductase levels or activity, e.g., as an indicator of the activity of the pathway in a sample, or, for example, to identify a bacterial species as one that likely performs Stickland fermentation. Measurement of glycine reductase levels in a sample can be performed, for example, by immunoassay and/or via RT-PCR for the mRNA encoding the enzyme. A biochemical assay for glycine reductase activity is described, for example, by Stadtman & Davis, J Biol Chem. 266(33):22147-53 (1991).


The nucleic acid and polypeptide sequences for the glycine reductase expressed by C. difficile 630 are NCBI Gene ID is 4915147 and YP_001088866.2, respectively.


The “grdA” or “glycine/sarcosine/betaine reductase complex protein A” glycine reductase is encoded by the grdA gene. Sequences for grdA are known for a number of species, e.g., for C. difficile 630 (the grdA NCBI Gene ID is 4915147) and polypeptide sequence (e.g., YP_001088866.2 (SEQ ID NO: 28). The sequence for the grdA gene product is as follows (SEQ ID NO: 28):










(SEQ ID NO: 28)










  1
msllsnkkvl iigdrdgipg paieecvktv egaevvfsst ecfvutaaga mdlenqnrvk






 61
daadkfgaen vvillgaaea eaaglaaetv tagdptfagp lagvalglsv yhvveepiks





121
lfdesvyedq ismmemvlev eeieeemsgi reefckf






The grdA gene sequence for C. difficile 630 is as follows (SEQ ID NO:29):










(SEQ ID NO: 29)










  1
atgagtttac ttagtaataa aaaggttctt ataataggtg accgtgatgg tataccagga






 61
cctgcgatag aagaatgtgt aaaaacagta gaaggagcag aggttgtttt ctcatctaca





121
gaatgctttg tctgaacagc tgctggggct atggacttag aaaatcaaaa cagagttaaa





181
gatgctgctg ataaattcgg agctgaaaat gttgtgattt tactaggtgc tgctgaagcc





241
gaagctgcag gtcttgcagc cgaaacagta actgctggag atccaacttt cgctggacca





301
cttgctggag ttgccttagg attaagtgtt taccacgttg ttgaggaacc aataaaatca





361
ttatttgatg aaagtgtata tgaagaccaa ataagtatga tggaaatggt tttagaagtt





421
gaagaaatag aagaagaaat gtctggtata agagaagaat tttgtaaatt ttaa







Defined Therapeutic Microbiota


Described herein are defined therapeutic microbiota that can be administered to suppress toxin expression by Gram positive, spore forming toxigenic bacteria such as C. difficile. In one embodiment, a defined therapeutic microbiota is or consists essentially of the single species, C. bifermentans. In another embodiment, the defined therapeutic microbiota comprises, consists essentially of or consists of C. scindens. In another embodiment, the defined therapeutic microbiota consists of, consists essentially of or comprises C. bifermentans and C. scindens. Strains of these species and others that express and secrete proteolytic enzymes into their surroundings and/or themselves perform Stickland fermentation are expected to promote suppression of C. difficile toxin expression. The following describes these species in further detail.



Clostridium Taxonomy: Clustering. As a starting point, species of the Genus Clostridium encompass a large number of anaerobic, spore-forming bacteria. In 1994, Collins et al. (Int. J. Systematic Biol. 44: 812-826 (1994), incorporated herein by reference) described a classification system that placed the species then known, and for which there was 16S rRNA sequence data available, into 19 “clusters,” termed Clostridium Clusters I-XIX, based upon similarities and differences in 16S rRNA sequences. This taxonomy and nomenclature has been retained to date, with some refinement, e.g., classifications of some of the larger clusters into smaller sub-clusters given alphabetic identifiers, e.g., Cluster XIVa. C. difficile is in Cluster XI.



Clostridium bifermentans



C. bifermentans are anaerobic, motile, Gram positive bacteria that colonize the healthy human gut, and are not commonly found to be pathogenic. C. bifermentans is a member of Clostridium Cluster XI, the same cluster as C. difficile. C. bifermentans induces the C. difficile ethanolamine pathway genes and maintains proline reductase activity, glucose and sorbitol metabolism in vivo. Through the greater than 10× repression of tcdR expression and greater than 30× suppression of toxin production, strong codY activation and repression of the PaLoc genes is indicated. The C. difficile metabolic pathways influenced by C. bifermentans are also indicated to activate ccpA, rex and prdR to also inhibit toxin production.


Both C. bifermentans and C. scindens possess the enzymatic machinery for Stickland fermentations. Of the two, C. bifermentans preferentially uses Stickland fermentations over glycolytic pathways, including in the presence of glucose or other sugars (see FIG. 6A and FIG. 6B), while in vitro C. scindens is preferentially glycolytic and produces abundant acetate (FIG. 6B).


A genome of C. bifermentans was sequenced as part of the Human Microbiome Project, and can be found at, e.g., accession #PRJNA212658.


In one embodiment, the C. bifermentans strain is 76 (ATCC #638). The 16S rRNA gene sequence for C. bifermentans strain 76 (SEQ ID NO: 30) is as follows:










(SEQ ID NO: 30)










   1
catrgctcag gatgaacgct ggcggcgtgc ctaacacatg caagtcgagc gatctcttcg






  61
gagagagcgg cggacgggtg agtaacgcgt gggtaacctg ccctgtacac acggataaca





 121
taccgaaagg tatactaata cgggataaca tatgaaagtc gcatggcttt tgtatcaaag





 181
ctccggcggt acaggatgga cccgcgtctg attagctagt tggtaaggta atggcttacc





 241
aaggcaacga tcagtagccg acctgagagg gtgatcggcc acactggaac tgagacacgg





 301
tccagactcc tacgggaggc agcagtgggg aatattgcac aatgggcgaa agcctgatgc





 361
agcaacgccg cgtgagcgat gaaggccttc gggtcgtaaa gctctgtcct caaggaagat





 421
aatgacggta cttgaggagg aagccccggc taactacgtg ccagcagccg cggtaatatg





 481
tagggggcta gcgttatccg gaattactgg gcgtaaaggg tgcgtaggtg gttttttaag





 541
tcagaagtga aaggctacgg ctcaaccgta gtaagctttt gaaactagag aacttgagtg





 601
caggagagga gagtagaatt cctagtgtag cggtgaaatg cgtagatatt aggaggaata





 661
ccagtagcga aggcggctct ctggactgta actgacactg aggcacgaaa gcgtggggag





 721
caaacaggat tagataccct ggtagtccac gccgtaaacg atgagtacta ggtgtcgggg





 781
gttacccccc tcggtgccgc actaacgcat taagtactcc gcctgggaag tacgctcgca





 841
agagtgaaac tcaaaggaat ttdcggggac ccgcacaagt agcggagcat gtggtttaat





 901
tcgaagcaac gcgaagaacc ttacctaagc ttgacatccc actgacctct ccctaatcgg





 961
agatttccct tcggggacag tggtgacagg tggtgcatgg ttgtcgtcag ctcgtgtcgt





1021
gagatgttgg gttaagtccc gcaacgagcg caacccttgc ctttagttgc cagcattaag





1081
ttgggcactc tagagggact gccgaggata actcggagga aggtggggat gacgtcaaat





1141
catcatgccc cttatgctta gggctacaca cgtgctacaa tgggtggtac agagggttgc





1201
caagccgcga ggtggagcta atcccttaaa gccattctca gttcggattg taggctgaaa





1261
ctcgcctaca tgaagctgga gttactagta atcgcagatc agaatgctgc ggtgaatgcg





1321
ttcccgggtc ttgtacacac cgcccgtcac accatggaag ttgggggcgc ccgaagccgg





1381
ttagctaacc ttttaggaag cggccgtcga aggtgaacaa atgactgggg tgaagtcgta





1441
acaaggtanc cgtatcggaa ggtgcggcbg gatcaa






In another embodiment, the C. bifermentans strain is a bacterial strain comprising a 16S rRNA sequence that is at least 97%, at least 98%, at least 99%, or more identical to the sequence of SEQ ID NO: 30. A C. bifermentans strain useful in the methods and compositions described herein should express at least 75% of the extracellular proteolytic activity of C. bifermentans strain 76 (ATCC #638) when assayed with a meat granule digestion microbiological assay as described herein. In one embodiment, the C. bifermentans strain expresses at least 80%, at least 85%, at least 90%, at least 95%, at least 100% or more of the proteolytic activity of C. bifermentans strain 76 (ATCC #638).



C. bifermentans is an anaerobic bacterium that can be cultured under anaerobic and microaerophilic conditions, and thus should be cultured accordingly, e.g., in a manner that limits or inhibits the bacteria's exposure to oxygen. (C. bifermentans is described as being able to tolerate up to 8% O2— see, e.g., Leja, K., Advances in Microbiology, 2014, however, growth under substantially anaerobic conditions is readily performed). Media and conditions for C. bifermentans growth in culture are known to those of ordinary skill in the art. As examples, C. bifermentans can be cultured under anaerobic conditions in, e.g., ATCC medium 2107 modified reinforced Clostridial agar/broth or ATCC medium 260 Trypticase soy agar/broth with defibrinated sheep blood (ATCC; Manassas, Va.), or grown on, e.g., Brucella Blood agar plates.



Clostridium scindens



C. scindens is an anaerobic, motile bacterium often found in the human gut, as well as in the soil. C. scindens is a member of Clostridium Cluster XIVa. C. scindens produces a 7-alpha bile acid dehydratase which converts primary bile acids to secondary bile acids that are strong inhibitors of C. difficile germination. It is noted that C. bifermentans has a choloylglycine deconjugating enzyme (which removes glycine from glycine-conjugated bile acids), but genomically does not possess comparable bile salt dehydroxylating enzymes.


While not wishing to be bound by theory, it is likely that at least part of the mechanism by which C. scindens promotes suppression of C. difficile infection is through bile acid metabolism to produce secondary bile acids (e.g., deoxycholic acid and lithocholic acid) that maintain an environment that is hostile to or not compatible with C. difficile and/or C. difficile infection. In this regard, C. scindens bacteria possess a complete secondary bile acid synthesis pathway, producing at least two enzymes active on the side-chain of the bile acid steroid nucleus and at least two enzymes active on the hydroxyl groups of the 7-position of bile acids. C. scindens expresses bile salt hydrolase/Choloylglycine hydrolase activity (E.C. #3.5.1.24), which catalyzes hydrolysis of the amide bond in conjugated bile salts, resulting in the release of free amino acids. This activity adds another source of free amino acid generation in the gut that can at least potentially contribute further substrate amino acids that promote Stickland fermentation.


A genome of C. scindens was sequenced as part of the Human Microbiome Project, and can be found at, e.g., accession #PRJNA18175.



C. scindens is an anaerobic bacterium, and thus should be cultured accordingly, e.g., in a manner the limits or inhibits the bacteria's exposure to oxygen. Media and conditions for C. scindens growth in culture are known to those of ordinary skill in the art. As examples, C. scindens can be cultured under anaerobic conditions in, e.g., ATCC medium 2107 modified reinforced Clostridial agar/broth or ATCC medium 260 Trypticase soy agar/broth with defibrinated sheep blood (ATCC; Manassas, Va.), or grown on, e.g., Brucella Blood agar plates.


In one embodiment, the C. scindens strain is VPI 13733 (ATCC #35704). The 16S rRNA gene sequence for C. scindens strain VPI 13733 (SEQ ID NO: 31) is as follows:









(SEQ ID NO: 31)


gagagtttgatcctggctcaggatgaacgctggcggcgtgcctaacaca





tgcaagtcgaacgaagcgcctggccccgacttcttcggaacgaggagcc





ttgcgactgagtggcggacgggtgagtaacgcgtgggcaacctgccttg





cactgggggataacagccagaaatggctgctaataccgcataagaccga





agcgccgcatggcgcggcggccaaagccccggcggtgcaagatgggccc





gcgtctgattaggtagttggcggggtaacggcccaccaagccgacgatc





agtagccgacctgagagggtgaccggccacattgggactgagacacggc





ccagactcctacgggaggcagcagtggggaatattgcacaatgggggaa





accctgatgcagcgacgccgcgtgaaggatgaagtatttcggtatgtaa





acttctatcagcagggaagaagatgacggtacctgactaagaagccccg





gctaactacgtgccagcagccgcggtaatacgtagggggcaagcgttat





ccggatttactgggtgtaaagggagcgtagacggcgatgcaagccagat





gtgaaagcccggggctcaaccccgggactgcatttggaactgcgtggct





ggagtgtcggagaggcaggcggaattcctagtgtagcggtgaaatgcgt





agatattaggaggaacaccagtggcgaaggcggcctgctggacgatgac





tgacgttgaggctcgaaagcgtggggagcaaacaggattagataccctg





gtagtccacgccgtaaacgatgactactaggtgtcgggtggcaaggcca





ttcggtgccgcagcaaacgcaataagtagtccacctggggagtacgttc





gcaagaatgaaactcaaaggaattgacggggacccgcacaagcggtgga





gcatgtggtttaattcgaagcaacgcgaagaaccttacctgatcttgac





atcccgatgccaaagcgcgtaacgcgctctttcttcggaacatcggtga





caggtggtgcatggttgtcgtcagctcgtgtcgtgagatgttgggttaa





gtcccgcaacgagcgcaacccctatcttcagtagccagcattttggatg





ggcactctggagagactgccagggagaacctggaggaaggtggggatga





cgtcaaatcatcatgccccttatgaccagggctacacacgtgctacaat





ggcgtaaacaaagggaggcgaacccgcgagggtgggcaaatcccaaaaa





taacgtctcagttcggattgtagtctgcaactcgactacatgaagttgg





aatcgctagtaatcgcgaatcagaatgtcgcggtgaatacgttcccggg





tcttgtacacaccgcccgtcacaccatgggagtcagtaacgcccgaagc





cggtgacccaacccgtaagggagggagccgtcgaaggtgggaccgataa





ctggggtgaagtcgtaacaaggtagccgtatcggaaggtgcggctggat





cacctccttc






In another embodiment, the C. scindens strain is a bacterial strain comprising a 16S rRNA sequence that is at least 97%, at least 98%, at least 99%, or more identical to the sequence of SEQ ID NO: 1. In one embodiment, the C. scindens strain expresses at least the level of bile salt hydrolase/Choloylglycine hydrolase expressed by C. scindens strain VPI 13733 (ATCC #35704). C. scindens is also proteolytic, albeit to a lesser degree than C. bifermentans. In one embodiment, the C. scindens strain used in a defined microbiota consortium as described herein has at least the proteolytic activity of C. scindens strain VPI 13733 (ATCC #35704).



Clostridium hylemonae



C. hylemonae is a naturally-occurring anaerobic commensal bacterium of the human gut. As described herein below, the relative abundance of C. hylemonae has been found, in combination with that of C. scindens, to be reliably predictive of the recurrence of C. difficile infection in human patients. C. hylemonae is a member of Clostridium Cluster XIVa. The 16S rRNA gene sequence for the C. hylemonae strain (SEQ ID NO: 32) is as follows:









(SEQ ID NO: 32)


aggatgaacgctgccgccgtgcttaacacatgcaagtcgaacgaagcaa





tactgtgtgaagagattagcttgctaagatcagaactttgtattgactg





agtggcggacgggtgagtaacgcgtgggcaacctgccttacacaggggg





ataacagctagaaatggctgctaataccgcataagacctcagtaccgca





tggtagaggggtaaaaactccggtggtgtaagatgggcccgcgtctgat





taggtagttggtagggtaacggcctaccaagccgacgatcagtagccga





cctgagagggtgaccggccacattggactgagacacggcccaaactcct





acgggaggcagcagtggggaatattgcacaatgggggaaaccctgatgc





agcgacgccgcgtgaaggatgaagtatttcggtatgtaaacttctatca





gcagggaagaagatgacggtacctgactaagaagccccggctaactacg





tgccagcagccgcggtaatacgtagggggcaagcgttatccggatttac





tgggtgtaaagggagcgtagacggcatggcaagtctgaagtgaaagccc





ggggctcaaccccgggactgctttggaaactgtcaggctagagtgtcgg





agaggcaagtggaattcctagtgtagcggtgaaatgcgtagatattagg





aggaacaccagtggcgaagcggcttgctggacgatgactgacgttgagg





ctcgaaagcgtggggagcaaacaggattagataccctggtagtccacgc





cgtaaacgatgattactaggtgtcgggaagcaaagcttttcggtgccgc





agccaacgcaataagtaatccacctggggagtacgttcgcaagaatgaa





actcaaaggaattgacggggacccgcacaagcggtggagcatgtggttt





aattcgaagcaacgcgaagaaccttacctgatcttgacatcccggtgac





aaagtatgtaacgtactctttcttcggaacaccggtgacaggtggtgca





tggttgtcgtcagctcgtgtcgtgagatgttgggttaagtcccgcaacg





gcgcaacccttatctttagtagccagcatttgaggtgggcactctagag





agactgccagggataacctggaggaaggtggggatgacgtcaaatcatc





atgccccttatgaccagggctacacacgtgctacaatggcgtaaacaaa





gggaagcgaccctgtgaaggcaagcaaatcccaaaaataacgtctcagt





tcggattgtagtctgcaactcgactacatgaagctggaatcgctagtaa





tcgcgaatcagaatgtcgcggtgaatacgttcccgggtcttgtacacac





cgcccgtcacaccatggggtcagtaacgcccgaagccggtgacctaacc





gcaaaggaggagccgtcgaaggtg.






In one embodiment, the C. hylemonae bacterium useful in the reliable prediction of C. difficile infection recurrence or in the reliable prediction of initial C. difficile infection has a 16S rRNA gene sequence of SEQ ID NO: 32. In another embodiment, a C. hylemonae bacterium useful in the reliable prediction of C. difficile infection recurrence or in the reliable prediction of initial C. difficile infection has a 16S rRNA gene sequence at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 32. It is also contemplated that C. hylemonae can provide therapeutic benefit, alone, or together with one or both of C. bifermentans and C. scindens.


Other potentially therapeutic Clostridial species that perform Stickland fermentation include, for example, C. cadaveris (Cluster I), and C. hiranonis, C. sticklandii, Peptostreptococcus anaerobius and C. sporogenes (Cluster XI).


The outcome of the studies described herein indicates that any or all of the following can have therapeutic benefit in combatting toxin production by Gram positive, spore-forming bacteria such as C. difficile:


A) Maintain metabolic and energy state in C. difficile through commensal proteolytic activity to release amino acids for Stickland fermentations (preferentially via the proline reductase pathway). Metabolites of Stickland fermentations can also be used or sensed by C. difficile through mechanisms that also reduce toxin production.


B) Induce or maintain expression of the C. difficile ethanolamine utilization pathway in C. difficile to help maintain energy state.


C) Prevent or limit production of butyrate by C. difficile and other commensals, to limit a stimulus of C. difficile toxin production.


D) Prevent C. difficile spore germination through production of secondary bile acids by C. scindens or other species that promote or produce such products.


E) Reduce biomass of C. difficile through, e.g., direct competition with C. difficile and by permitting recovery of other gut commensal species that directly compete and/or prevent spore germination through innate bile salt hydrolase activities.


F) Glycine Reductase activity or expression.


Defined Therapeutic Microbiota, and Compositions Comprising Them


It is demonstrated herein that C. bifermentans can completely protect GF and conventional mice from otherwise fatal infection with C. difficile.


In one aspect, then, described herein are compositions, including but not limited to pharmaceutical compositions, comprising an oral formulation comprising C. bifermentans bacteria. In one embodiment the C. bifermentans species is the only bacterial species in the formulation. In another embodiment, the C. bifermentans is the only Clostridial XI species in the formulation. In another embodiment, the C. bifermentans is the only Clostridial species in the formulation. In another embodiment, the C. bifermentans is the only bacterial species in the formulation. Such compositions can be used to treat or prevent C. difficile infection, including recurrent C. difficile infection, and/or toxin production.


Methods for the growth or preparation of C. bifermentans bacteria are known in the art. As noted herein above, this species is an obligate anaerobe, so culture under anaerobic conditions is required. Media for culture of Clostridium species are also known in the art, and are commercially available, as also noted elsewhere herein.


In another aspect, described herein is a composition, including a pharmaceutical composition, comprising C. bifermentans bacteria, wherein the C. bifermentans bacteria are in dried, viable form. Methods of drying Clostridium species in a manner that maintains viability upon re-hydration are known in the art.


In another aspect, described herein is a pharmaceutical composition comprising a formulation comprising C. bifermentans bacteria, wherein the composition does not comprise Bacteroides species or Escherichia species.


It is contemplated that killed or proliferatively inactive, but metabolically active C. bifermentans bacteria could have beneficial effect, e.g., if administered repeatedly. However, in each of the aspects noted above, it is preferred that the bacteria are viable as the term is used herein. In this context, the bacterial species for any of the compositions described herein can be present in vegetative, metabolically and proliferatively active forms, dried viable form, spore form, or a combination of these forms. Thus, in one embodiment, the C. bifermentans bacteria are in spore form. In another embodiment, the C. bifermentans bacteria are in metabolically active form, including, but not limited to vegetative and/or actively proliferative forms. In another embodiment, the C. bifermentans bacteria are not in spore form. In another embodiment, the C. bifermentans bacteria are present as a mixture of vegetative, metabolically active and spore forms. Methods for inducing sporulation of Clostridium species are known to those of ordinary skill in the art.


Another aspect described herein provides a pharmaceutical composition comprising an oral formulation comprising a bacterial population that has 16S rDNA with a sequence at least 97% identical to a 16S rDNA sequence present in a reference C. bifermentans bacterium. In various embodiments, the bacterial population has 16S rDNA with a sequence at least 97%, 98%, 99% or more identical to the 16S rDNA of the reference bacterium.


Another aspect described herein provides a pharmaceutical composition comprising a bacterial population that has 16S rDNA with a sequence at least 97% identical to a 16S rDNA sequence present in a reference C. bifermentans bacterium, wherein the C. bifermentans bacteria is in dried, viable form. In various embodiments, either or both of the bacterial populations has 16S rDNA with a sequence at least 97%, 98%, 99% or more identical to the 16S rDNA of the reference bacterium.


Another aspect described herein provides a pharmaceutical composition comprising a formulation comprising a bacterial population that has 16S rDNA with a sequence at least 97% identical to a 16S rDNA sequence present in a reference C. bifermentans bacterium, wherein the composition does not comprise Bacteroides species or Escherichia species. In various embodiments, the bacterial population has 16S rDNA with a sequence at least 97%, 98%, 99% or more identical to the 16S rDNA of the reference bacterium.


In one embodiment of this or any other aspect described herein, the compositions described herein do not comprise viable C. sardiniense bacteria. As described herein, it was found that the presence of C. sardiniense in otherwise germ-free mice comprising pathogenic C. difficile bacteria increased the severity of the resulting C. difficile infection. Thus, in one embodiment, the technology described herein excludes C. sardiniense from a therapeutic composition as described herein.


In one embodiment of this or any aspect described herein, a therapeutic microbiota composition does not comprise a Bacteroides species or Escherichia species. Bacteroides is a genus of Gam-negative, obligate anaerobic bacteria. Bacteroides species can be identified by sphingolipids in their membranes, as well as by 16S rDNA sequence. They make up a substantial proportion of the gut microbiota, and are commonly mutualistic. Escherichia species are Gram-negative, non-spore forming, facultative anaerobic bacteria from the family Enterobacteriaceae. The majority of Escherichia species are commensal gut flora, although certain Escherichia strains are human pathogens. A skilled person can identify Bacteroides and Escherichia species, e.g., using PCR-based methods to quantify 16S rDNA genetic markers, among others.


In one embodiment of this or any aspect described herein, the compositions described herein do not comprise any additional viable Clostridium species. In another embodiment of any aspect described herein, the compositions described herein do not comprise any additional viable bacteria of any kind. A skilled person can identify the presence of other viable bacteria (e.g., additional Clostridium species, among others) in the composition, e.g., using anaerobic culture, and identify organisms in such culture using, for example, PCR-based methods to quantify 16S rDNA genetic markers.


It is also demonstrated herein that a defined consortium of Clostridium species comprising C. scindens and C. bifermentans, as those species are defined herein, is sufficient to treat or prevent C. difficile infection, including, but not limited to recurrent C. difficile infection, and to suppress toxin production by C. difficile.


In one aspect, then, described herein are compositions, including but not limited to pharmaceutical compositions, comprising an oral formulation comprising C. scindens and C. bifermentans bacteria. In another embodiment of this and all other aspects described herein that involve the C. scindens and C. bifermentans species, the only bacterial species in the formulation are C. scindens and C. bifermentans. Methods for the growth or preparation of C. scindens and C. bifermentans bacteria are known in the art. As noted herein above, these species are obligate anaerobes, so culture under anaerobic conditions is required. Media for culture of Clostridium species are also known in the art, and are commercially available, as also noted herein above.


In another aspect, described herein is a composition, including a pharmaceutical composition, comprising C. scindens and C. bifermentans bacteria, wherein one or both of the C. scindens and C. bifermentans bacteria is in dried, viable form. Methods of drying Clostridium species in a manner that maintains viability upon re-hydration are known in the art.


In another aspect, described herein is a pharmaceutical composition comprising a formulation comprising C. scindens and C. bifermentans bacteria, wherein the composition does not comprise Bacteroides species or Escherichia species.


It is contemplated that killed or proliferatively inactive, but metabolically active bacteria of these species could have beneficial effect if administered repeatedly. However, in each of the aspects noted above, it is preferred that the bacteria are viable as the term is used herein. In this context, the bacterial species for any of the compositions described herein can be present in vegetative, metabolically and proliferatively active forms, dried viable form, spore form, or a combination of these forms. Thus, in one embodiment, both of the C. scindens and C. bifermentans bacteria are in spore form. In another embodiment, both of the C. scindens and C. bifermentans bacteria are in metabolically active form, including, but not limited to vegetative and/or actively proliferative forms. In another embodiment, one or both of the C. scindens and C. bifermentans bacteria are not in spore form. In another embodiment, the C. scindens and C. bifermentans bacteria are present as a mixture of vegetative, metabolically active and spore forms. Methods for inducing sporulation of Clostridium species are known to those of ordinary skill in the art.


Another aspect described herein provides a pharmaceutical composition comprising an oral formulation comprising a first bacterial population that has 16S rDNA with a sequence at least 97% identical to a 16S rDNA sequence present in a reference C. scindens bacterium, and a second bacterial population that has 16S rDNA with a sequence at least 97% identical to a 16S rDNA sequence present in a reference C. bifermentans bacterium. In various embodiments, either or both of the bacterial populations has 16S rDNA with a sequence at least 97%, 98%, 99% or more identical to the 16S rDNA of the reference bacterium.


Another aspect described herein provides a pharmaceutical composition comprising a first bacterial population that has 16S rDNA with a sequence at least 97% identical to a 16S rDNA sequence present in a reference C. scindens bacterium, and a second bacterial population that has 16S rDNA with a sequence at least 97% identical to a 16S rDNA sequence present in a reference C. bifermentans bacterium, wherein one or both of the C. scindens and C. bifermentans bacteria is in dried, viable form. In various embodiments, either or both of the bacterial populations has 16S rDNA with a sequence at least 97%, 98%, 99% or more identical to the 16S rDNA of the reference bacterium.


Another aspect described herein provides a pharmaceutical composition comprising a formulation comprising a first bacterial population that has 16S rDNA with a sequence at least 97% identical to a 16S rDNA sequence present in a reference C. scindens bacterium, and a second bacterial population that has 16S rDNA with a sequence at least 97% identical to a 16S rDNA sequence present in a reference C. bifermentans bacterium wherein the composition does not comprise Bacteroides species or Escherichia species. In various embodiments, either or both of the bacterial populations has 16S rDNA with a sequence at least 97%, 98%, 99% or more identical to the 16S rDNA of the reference bacterium.


The above bacterial compositions can include, for example but are not limited to metabolically active bacteria, wet bacteria, dry viable bacteria (e.g., preparations including viable spray-dried cells, freeze-dried cells, vacuum-dried cells, drum-dried cells, vitrified etc.), and the like. Preparations of Clostridium species described herein can include, for example, suspensions of Clostridium bacteria, cultured cells of Clostridium bacteria (including bacterial cells, and optionally, supernatant and medium ingredients), and, for example, Clostridium culture biomass, removed from suspension culture, e.g., by centrifugation, filtration, or the like. While viable Clostridium bacteria are used in most applications considered herein, it is contemplated that in some embodiments, processed cells of Clostridium bacteria can include, for example, ground cells, crushed cells, liquefied cells (extracts etc.) and concentrates and preparations thereof, and the like.


Dried preservation removes water from the culture by evaporation (in the case of spray drying or ‘cool drying’) or by sublimation (e.g., for freeze drying, spray freeze drying). Removal of water improves long-term bacterial composition storage stability at temperatures elevated above cryogenic. If the bacterial composition comprises spore forming species and results in the production of spores, the final composition can be purified by additional means such as density gradient centrifugation.


Species effective for treatments as described herein are readily available. However, to maintain strain integrity over time, bacterial composition banking can be done by culturing and preserving the strains individually, or by mixing the strains together to create a combined bank. As an example of cryopreservation, a bacterial composition culture can be harvested by centrifugation to pellet the cells from the culture medium, the supernate decanted and replaced with fresh culture broth containing 15% glycerol. The culture can then be aliquoted into 1 mL cryotubes, sealed, and placed at −80° C. for long-term viability retention. This procedure achieves acceptable viability upon recovery from frozen storage.



C. bifermentans and/or C. scindens can be in spore form or not in spore form in the compositions described herein. Bacterial spores are dormant, non-reproductive structures produced by certain bacteria from the Firmicute phylum, for example in an environment lacking nutrients. Spores can be preserved as described above, and can be reactivated, e.g., by heating the endospore. In addition, C. bifermentans and/or C. scindens can be present in a mixture of metabolically active bacteria and spores. Metabolically active bacteria can actively metabolize nutrients, and will have little lag time from administration to active participation in treating or preventing C. difficile infection. They will also likely have little lag time in beginning to proliferate in the gut if conditions are appropriate.


In one embodiment, any of the defined therapeutic microbiota compositions described herein further comprises a prebiotic. Prebiotics promote the growth, survival, and activity of beneficial microorganisms, or probiotics. Prebiotics have been shown to alter the compositions of microorganisms (microflora) in the gut microbiota, alone or in combination with probiotic organisms. In addition, prebiotics have been shown to increase calcium and magnesium absorption in the gut, increase bone density, enhance the immune system, reduce blood triglyceride levels, and control hormone levels. Prebiotics include any of a number of compositions that are generally not directly digestible by humans, but that are readily digestible by and promote the growth or establishment of probiotic microbes. In one embodiment, a preferred prebiotic comprises a sugar or carbohydrate, e.g., a starch or other carbohydrate-comprising polymer, that can be digested by C. bifermentans and/or C. scindens, but not readily so by other commensals, to thereby favor an increase in the relative proportion or abundance of the administered species. Non-limiting examples of prebiotics include but are not limited to inulin, fructooligosaccharides, galactooligosaccharides, hemicelluloses (e.g., arabinoxylan, xylan, xyloglucan, and glucomannan), chitin, lactulose, mannan oligosaccharides, oligofructose-enriched inulin, gums (e.g., guar gum, gum arabic and carregenaan), oligofructose, oligodextrose, tagatose, resistant maltodextrins (e.g., resistant starch), trans-galactooligosaccharide, pectins (e.g., xylogalactouronan, citrus pectin, apple pectin, and rhamnogalacturonan-I), dietary fibers (e.g., soy fiber, sugarbeet fiber, pea fiber, corn bran, and oat fiber) and xylooligosaccharides, and dietary proteins able to be digested by C. bifermentans and/or C. scindens.


In one embodiment, any of the defined therapeutic microbiota compositions described herein further comprise an effective amount of one or more free Stickland fermentable amino acids, e.g. a preparation of free proline alone or any combination of free amino acids selected from the group consisting of: alanine, leucine, valine, isoleucine, tryptophan, tyrosine, phenylalanine, proline or glycine.


In one embodiment, any of the defined therapeutic microbiota compositions described herein further comprise an effective amount of a polypeptide that can be proteolyzed by the administered species to generate free amino acids fermentable by Stickland fermentation. Inclusion of such a polypeptide provides a ready source of protein for an administered Stickland-fermenting bacterium to digest to amino acids useful for Stickland fermentation by C. difficile. Non-limiting examples of such polypeptides include, but are not limited to casein, gelatin, collagen, and an artificial polymer comprising Stickland acceptor amino acids and/or Stickland donor amino acids. A proline-rich or proline+ leucine-rich protein can also be used. Where an artificial polymer is used, the polymer can comprise Stickland donor amino acids selected from the group consisting of: alanine, leucine, valine, isoleucine, tryptophan, tyrosine and phenylalanine, and/or Stickland acceptor amino acids including proline and/or glycine. The polymer can comprise e.g., a poly[N] amino acid polymer, e.g. a poly[alanine], poly[leucine], etc., or e.g. a copolymer of one or more of the Stickland fermentable amino acids. As a non-limiting example, copolymers can include, e.g. poly[alanine, leucine], poly[alanine, isoleucine],[poly[alanine, tryptophan], etc. Polymers rich in proline would be expected to preferentially promote fermentation via the Stickland proline reductase pathway and repression of C. difficile toxin production.


In one embodiment, any of the compositions described herein, except those which expressly exclude any species other than C. scindens and/or C. bifermentans, further comprise a microbe that supports (e.g., the growth, or viability) C. bifermentans and/or C. scindens. Ruminococcus obeum is an exemplary microbe that has been shown to support C. scindens. Ruminococcus obeum is a genus of bacteria in the class Clostridia found in an abundance in the human gut. A skilled person will be able to determine if a microbe supports C. bifermentans and/or C. scindens, e.g., using complementation assays known in the art.


Establishment of administered C. scindens and/or C. bifermentans as described herein can be evaluated by monitoring the proportion of these species in gut microbiota samples taken over time. An administered species can be considered to be established if that species remains at a level increased relative to its level pre-administration for at least two weeks, preferably at least three weeks, one month, five weeks, six weeks, seven weeks, two months, nine weeks, ten weeks, eleven weeks, three months or more following administration. The presence of the administered species at a relative level of abundance of at least 0.3%, at least 0.4%, 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, 1% or more, maintained over time as noted is preferred. Tracking of the administered species can be facilitated by modification of the species to carry a genetic difference or barcode, but this is optional.


Diagnostic Methods


One aspect of the present technology is a method of predicting or determining the likelihood of C. difficile infection or recurrence of C. difficile infection in a subject. Such a prediction can guide prophylactic and subsequent therapeutic treatment decisions. Markers of likely infection or recurrence include, but are not limited to markers in the following classes:


Proteolytic activity of the subject's commensal microbiota, measured, for example, by biochemical assay for proteolysis of a substrate, e.g., one including but not limited to casein or gelatin. Relatively high proteolytic activity is a predictor of reduced likelihood of infection or recurrence.


DNA- or RNA-based studies for commensal proteases and Stickland reductase genes as described herein. The presence and/or expression of commensal protease genes is a predictor of reduced likelihood of infection or recurrence, with higher levels providing benefit.


Microbiologic, biochemical or molecular assays for proteolytic Clostridia, Stickland fermenters, or other highly proteolytic commensal species. The presence of such species is a predictor of reduced likelihood of infection or recurrence, with higher levels providing benefit.


Detection of energy-producing substrates for C. difficile, e.g., substrates selected from proline, Stickland donor amino acids, ethanolamine, glucose, fructose, mannitol, sorbitol, cysteine and threonine. Greater energy-producing substrate levels, and particularly greater levels of substrates preferred by C. difficile are predictors of reduced likelihood of infection or recurrence.


Detection of metabolites of Stickland fermentation including, for example, 5-amino valerate, branched SCFA amino acid metabolites and other Stickland amino acid metabolites. Greater levels of such metabolites are predictive of reduced risk of infection or recurrence. Greater levels of metabolites associated with the proline reductive pathway are particularly predictive.


Detection of metabolites of anaerobic carbohydrate metabolism including, for example the volatile SCFA acetate, propionate or butyrate, and non-volatile SCFA succinate, lactate or pyruvate, where increased levels of volatile SCFA, and of succinate, are predictive of increased likelihood of infection or recurrence.


Detection of microbial energy transporters including NADH/NAD+, NADPH/NADP+, ATP/ADP and GDP/GTP as indicative of the energy state in tested materials from microbial metabolism.


In one embodiment, any or all of the markers noted above can be used with detection of toxigenic C. difficile by microbiologic, toxin ELISA or molecular methods to predict likelihood of infection or recurrence. The presence and/or levels of the various markers can be compared, for example, to a reference to determine likelihood of infection or recurrence. The reference can be, for example, a sample from a healthy individual, or as the case may be a sample from an individual with active C. difficile infection.


In one embodiment, a method is provided for determining the efficacy of therapy for C. difficile. In one embodiment, the therapy is a bacteriotherapy, for example, as described herein. In another embodiment, the therapy is or comprises administration of a pre-biotic, and/or administration of an amino acid or amino acid derivative. In one embodiment, the method comprises measuring in a sample from an individual being treated for C. difficile infection, one or more markers from one or more, two or more, three or more, four or more, or from each of the classes of the markers listed above. In one embodiment, the reference is a sample from an individual with active C. difficile infection. The reference can, but does not necessarily have to be, a sample from the subject being treated, taken before treatment began. A level of activity or expression of one or more markers or classes of markers that is increased relative to the reference indicates effective therapy. In one embodiment, the sample is a stool sample.


In another embodiment, a method is provided for predicting the likelihood of recurrence in a subject being treated or who has been treated for C. difficile infection. In one embodiment, the method comprises measuring in a sample from an individual who has been treated for C. difficile infection, one or more markers from one or more, two or more, three or more, four or more, or from each of the classes of the markers listed above. In one embodiment, the reference is a sample from a healthy individual. A healthy individual is one without active C. difficile infection and who has not received antibiotic treatment within the past three months. A level of one or more biomarkers from one or more of the classes listed above that is below that of the reference indicates an increased risk of recurring C. difficile infection. In one embodiment, the sample is a stool sample. The method can further comprise administering a bacteriotherapy as described herein to an individual for whom the level of such marker(s) is below the reference.


In another embodiment, a method is provided for predicting the risk of an individual for developing a first infection with C. difficile. This is applicable, for example, to those who are elderly, immunocompromised, hospitalized, in a nursing home, receiving antibiotics, or receiving proton pump inhibitors. In one embodiment, the method comprises measuring in a sample from a subject in one or more of these categories one or more markers in one or more of the classes listed above, and the subject is at increased risk of contracting active C. difficile infection if the level of such marker(s) is below a reference. In one embodiment, the reference can be a sample from a healthy individual as above. In another embodiment, the sample from the subject is assayed to determine the presence and/or amount of Stickland fermenting and/or proteolytic bacterial species, including, for example, non-pathogenic Clostridial species that are proteolytic and/or Stickland fermenting. A lack of such species indicates an increased risk for developing active C. difficile infection. A reduced number relative to a healthy reference also indicates increased risk. In one embodiment the method further comprises administering a bacteriotherapy as described herein to an individual for whom such species are lacking or for whom the level of such marker(s) is below the reference.


Further, the markers noted above can be used to define and monitor the efficacious activity of a therapeutic regimen, including, but not limited to a therapeutic regimen comprising administration of bacteriotherapeutic products, e.g., a defined therapeutic microbiota composition or product as described herein.


Also described herein are predictive methods that examine the presence of certain commensal species as a marker of likelihood of C. difficile infection and/or C. difficile toxin production. In one embodiment, such a method comprises: (a) determining the relative abundance of all operational taxonomic units (OTUs) in a sample of the subject's stool that are >90% identical to a reference sequence of C. scindens; (b) determining the relative abundance of all operational taxonomic units (OTUs) in a sample of the subject's stool that are >90% identical to a reference sequence of C. hylemonae; and (c) summing the relative abundances determined in steps (a) and (b), wherein a sum of relative abundances less than or equal to 1% indicates an increased risk of C. difficile recurrence relative to a subject in which the sum of relative abundances is greater than 1%. In one embodiment, relative abundance of the marker species indicative of increased risk is between 1% and 0.01%. As used herein, “relative abundance” refers to comparison with all species of microbes identified in the subject's sample, and is not limited to Clostridium species. The relative abundance of species in a sample can be measured, for example using Roche/454 pyrosequencing or Illumina sequencing for 16S rRNA gene sequencing. This approach, combined with multiplexing produces thousands of 16S rRNA sequences per sample. Microbiome sequencing techniques are further reviewed in, e.g., Grice, E A, and Segre J A. Annu Rev Genomics Hum Genet. 2012; 13:151-170. Relative abundance can additionally be measured using, e.g., using amplicons for microbiologic or microbial products and/or other gene-level targets (e.g., qPCR for genes (e.g., the bai gene, or the gene encoding a Stickland enzyme, a bile acid hydrolase, etc.)).


The predictive method can also be applied to prediction of susceptibility to a first C. difficile infection. Thus, in one embodiment, a sample can be taken from a patient who is at risk of having, has, or has previously had at least one C. difficile infection. A sample can be taken from a subject who has never had a C. difficile infection, but who is in a risk category as noted herein. A stool sample can be collected using standard techniques, e.g., passing stool directly into a clean, dry container.


At least one sample is taken from the subject for the predictive method. However, repeated sampling can also be performed. For example, a sample can be taken from a subject once a day, once a week, twice a month, once a month, or every 3 months following a C. difficile infection to assess the risk of a recurrent C. difficile infection, or a sample can be taken from a subject once a year following a C. difficile infection to assess the risk of a recurrent C. difficile infection. It has even been found that measurement of the relative abundance of the noted species (C. scindens and C. hylemonae) during treatment for an initial C. difficile infection can be predictive of likelihood of a recurrence. Thus, at least one sample can be taken from a subject during a C. difficile infection, e.g., a sample can be taken once a day for the entirety of the infection.


A sample can be taken from a subject who has not previously been treated with antibiotics to treat a C. difficile infection. Alternatively, a sample can be taken from a subject who has been treated with antibiotics to treat a C. difficile infection. The sample can be taken from the subject before, during, or after administration of antibiotics to treat a C. difficile infection. A sample can be taken from a subject before, during, and after administration of an antibiotic (e.g., a sample is taken from a subject during and after administration of an antibiotic).


The method can further comprise the step of administering a further therapeutic or prophylactic treatment, including, for example, an FMT or any of the bacterial compositions described herein to a subject when the sum of relative abundances relative level is at or below 0.01%, 0.1%, 0.2%, 0.3%, 0.4%, 0.5%, 0.6%, 0.7%, 0.8%, 0.9%, or 1.0%.


In this context, the reference sequence of C. scindens can be a sequence comprising at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity to the sequence of SEQ ID NO: 31. Similarly, in this context, the reference sequence of C. hylemonae can be a sequence comprising at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, at least 98%, at least 99%, or 100% sequence identity to the sequence of SEQ ID NO: 32.


In another aspect of any of the embodiments, described herein is a method of treating a pathology involving expression of a bacterial toxin from a Gram positive, spore-forming species in a subject in need thereof, the method comprising: a) determining that the subject has a reduced amount and/or activity of secreted proteolytic enzymes in a gut or stool sample relative to healthy individual as described herein; and b) administering a therapeutic bacterial species as described herein to the subject. In some embodiments of any of the aspects, the step of determining that the subject has a reduced amount and/or activity of a secreted proteolytic enzyme can comprise i) obtaining or having obtained a biological sample from the subject and ii) performing or having performed an assay on the sample obtained from the subject to determine/measure the level of the proteolytic enzyme in the subject. Methods to measure amount of secreted proteolytic enzymes and/or activity are known to a skilled artisan. Such methods to measure gene expression products, e.g., protein level, include ELISA (enzyme linked immunosorbent assay), western blot, immunoprecipitation, and immunofluorescence using detection reagents such as an antibody or protein binding agents. Alternatively, a peptide can be detected in a subject by introducing into a subject a labeled anti-peptide antibody. For example, the antibody can be labeled with a detectable marker whose presence and location in the subject is detected by standard imaging techniques. Methods to measure the activity of secreted proteolytic enzymes are known to a skilled artisan. For example, a protease activity assay that uses casein or gelatin as a substrate can be used to measure the activity of a protease in a biological sample (Cat. No. Ab111750; Abcam, Cambridge Mass.).


In another aspect, described herein is a method of treating a pathology involving expression of a bacterial toxin brom a Gram positive spore-forming bacterium in a subject in need thereof, the method comprising: a) determining that a sample from the subject has a decreased amount and/or activity of proline reductase relative to a sample from a healthy individual; and b) administering a therapeutic bacterial species as described herein to the subject. In one embodiment, the step of determining that a sample from the subject has reduced level or activity of proline reductase can comprise i) obtaining or having obtained a biological sample from the subject and ii) performing or having performed an assay on the sample obtained from the subject to determine/measure the level of the proline reductase in the subject. Methods to measure amount or activity of proline reductase are known to a skilled artisan and/or described herein. Such methods can include measurement of gene expression products, e.g., protein level, and include for example, ELISA (enzyme linked immunosorbent assay), western blot, immunoprecipitation, and immunofluorescence using detection reagents such as an antibody or protein binding agents.


In another aspect of any of the embodiments, described herein is a method of treating a pathology involving expression of a bacterial toxin brom a Gram positive spore-forming bacterium in a subject in need thereof, the method comprising: a) determining that a sample from the subject has a decreased amount and/or activity of glycine reductase relative to a sample from a healthy individual; and b) administering a therapeutic bacterial species as described herein to the subject. In one embodiment, the step of determining that a sample from the subject has reduced level or activity of glycine reductase can comprise i) obtaining or having obtained a biological sample from the subject and ii) performing or having performed an assay on the sample obtained from the subject to determine/measure the level of the glycine reductase in the subject. Methods to measure amount or activity of glycine reductase are known to a skilled artisan and/or described herein. Such methods can include measurement of gene expression products, e.g., protein level, and include for example, ELISA (enzyme linked immunosorbent assay), western blot, immunoprecipitation, and immunofluorescence using detection reagents such as an antibody or protein binding agents.


Dosage, Administration and Formulations


In one aspect, any of the compositions described herein is administered to a subject that has, or has been diagnosed with C. difficile infection. In one embodiment, the C. difficile infection is a recurrent C. difficile infection. Recurrent C. difficile infections can be caused by the same or a different C. difficile strain that caused a previous C. difficile infection. In one embodiment, the subject is at risk for a C. difficile infection or a recurrent C. difficile infection.


A clinician can diagnose a subject as having a C. difficile infection using standard methods to detect toxins that are produced by C. difficile bacteria. For example, a stool sample from subject suspected of having a C. difficile infection can be analyzed via an enzyme immunoassay or even dipstick/lateral flow immunoassay for the C. difficile toxin(s), PCR-based assays, GDH/EIA tests, or cell cytotoxicity assays are also commonly used to definitively determine C. difficile infection. Imaging, e.g., colonoscopy or abdominal x-ray or CT scan can also be used to assist the diagnosis of C. difficile infection.


A clinician can determine is a subject is at risk of having a C. difficile infection by assessing a subject's risk factors, including but not limited to the subject's proximity to an individual who has or has recently had a C. difficile infection, current medications that promote C. difficile growth in the intestine, age, antibiotic use (length of antibiotic regimen, use of broad-spectrum antibiotics, or use of multiple antibiotics, use of gastric acid inhibitors such as proton pump-inhibitors and or histamine-2 receptor antagonists. At present, a subject who has had previous C. difficile infection is at risk of a recurrent C. difficile infection; approximately 20% of subjects who have had a C. difficile infection will have a recurrent C. difficile infection. In a further embodiment, risk of recurrence can be evaluated with the method described herein above that measures the relative abundance of C. scindens and C. hylemonae. Risk of recurrence can also be predicted via one or more of the diagnostic methods described herein.


Dosage


The dosage ranges for the bacterial species described herein in a defined therapeutic microbiota composition depends upon the potency (including viability), and includes amounts large enough to produce the desired effect, e.g., reduction in at least one symptom of a C. difficile infection in a treated subject. The dosage should not be so large as to cause unacceptable adverse side effects. Generally, the dosage can vary with the type of illness, e.g., first infection vs. recurrent infection, and with the age, condition, and sex of the patient. The dosage can be determined by one of skill in the art and can also be adjusted by the individual physician in the event of any complication.


For use in the various aspects described herein, an effective amount of cells in a composition as described herein comprises at least 1×105 bacterial cells, at least 1×106 bacterial cells, at least 1×107 bacterial cells, at least 1×108 bacterial cells, at least 1×109 bacterial cells, at least 1×1010 bacterial cells, at least 1×1011 bacterial cells, at least 1×1012 bacterial cells or more. In one embodiment, the microbial consortium or the individual bacterial components thereof can be obtained from a microbe bank. Members of a therapeutic or preventive/prophylactic consortium are generally administered together, e.g., in a single admixture. However, it is specifically contemplated herein that members of a given consortium can be administered as separate dosage forms or sub-mixtures or sub-combinations of the consortium members (e.g., C. scindens and C. bifermentans can be comprised in two separate compositions that are administered as separate doses). Thus, a consortium of e.g., C. scindens and C. bifermentans, can be administered, for example, as a single preparation including all members (in one or more dosage units, e.g., one or more capsules) or as two separate preparations that, in sum, include all members of the given consortium. While administration as a single admixture is preferred, a potential advantage of the use of e.g., individual units for each member of a consortium, is that the species administered to any given subject can be tailored, if necessary, by selecting the appropriate combination of, for example, single species dosage units that together comprise the desired consortium. With respect to the administration of two separate preparations, the route of administration can be the same for each preparation (e.g., the first and second preparation can be administered orally), or different for each preparation (e.g., the first preparation can be administered orally and the second preparation is administered directly to the colon via colonoscope, to the small intestine via endoscope, or to the colon via suppository or enema).


From the conventional mouse model (administer microbes after onset of symptomatic Cdiff infection) it was found that orally administered C. bifermentans rapidly changes the cecal environment 6-7 hr after administration (transit time through the gut). This does not occur with C. sardiniense. It is noted that C. bifermentans persistence in most conventional mice is only a few days but is sufficient to correct the gut environment and cause Cdiff to rapidly halt toxin production. The administered bacteriotherapy can, for example, provide long-term engraftment, e.g., weeks, months or longer, or, it can provide shorter term persistence or presence of the administered species. Where long-term engraftment does not occur, repeat dosing is warranted, for example to provide continuing protection for as long as needed, including days, weeks, months, years or longer, e.g., indefinitely if needed. The need for such continued dosing can be evaluated, e.g., using methods of evaluating risk of infection or recurrent infection as described herein.


One can also easily adjust the ratio of one species to another if separate dosage forms are administered. It is contemplated that the ratios of C. scindens to C. bifermentans, for example, can be varied, e.g. over a range of 1:10 to 10:1 with benefit to the patient under certain circumstances, especially where, for example, the persistence of one species is found to be less than that of the other. Such a finding would warrant, for example, increasing the ratio in favor of the species with the lower degree of persistence. In one embodiment, the ratio of C. scindens to C. bifermentans in a composition described herein is 1:1. The ratio of C. scindens to C. bifermentans in a composition described herein can be 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, or 1:10. The ratio of C. bifermentans to C. scindens in a composition described herein can be 1:1, 1:2, 1:3, 1:4, 1:5, 1:6, 1:7, 1:8, 1:9, or 1:10. The ratio should be such that the amount of each bacteria in the composition is sufficient to promote engraftment (i.e., in vivo replication) of each species in a subject's GI tract following administration of the composition.


A pharmaceutical composition comprising a microbial consortium can be administered by any method suitable for depositing in the gastrointestinal tract, preferably the colon, of a subject (e.g., human, mammal, animal, etc.). Examples of routes of administration include rectal administration by colonoscopy, suppository, enema, upper endoscopy, or upper push enteroscopy. Additionally, intubation through the nose or the mouth by nasogastric tube, nasoenteric tube, or nasal jejunal tube can be utilized. Oral administration by a solid such as a pill, tablet, a suspension, a gel, a geltab, a semisolid, a tablet, a sachet, a lozenge or a capsule or microcapsule, or as an enteral formulation, or re-formulated for final delivery as a liquid, a suspension, a gel, a geltab, a semisolid, a tablet, a sachet, a lozenge or a capsule, or as an enteral formulation can be utilized as well. Also contemplated herein are food items that are inoculated with a microbial consortium as described herein.


In some embodiments, the compositions described herein can be administered in a form containing one or more pharmaceutically acceptable carriers. Suitable carriers are well known in the art and vary with the desired form and mode of administration of the composition. For example, pharmaceutically acceptable carriers can include diluents or excipients such as fillers, binders, wetting agents, disintegrators, surface-active agents, glidants, lubricants, and the like. The carrier may be a solid (including powder), liquid, or combinations thereof. Each carrier is preferably “acceptable” in the sense of being compatible with the other ingredients in the composition and not injurious to the subject. The carrier may be biologically acceptable and inert (e.g., it permits the composition to maintain viability of the biological material until delivered to the appropriate site).


Oral compositions can include an inert diluent or an edible carrier. For the purpose of oral therapeutic administration, the active compound can be incorporated with excipients and used in the form of tablets, lozenges, pastilles, troches, or capsules, e.g., gelatin capsules. Oral compositions can also be prepared by combining a composition of the present disclosure with a food. In one embodiment a food used for administration is chilled, for instance, iced flavored water. Pharmaceutically compatible binding agents, and/or adjuvant materials can be included as part of the composition. The tablets, pills, capsules, troches and the like can contain any of the following ingredients, or compounds of a similar nature: a binder such as microcrystalline cellulose, gum tragacanth or gelatin; an excipient such as starch or lactose, a disintegrating agent such as alginic acid, primogel, or corn starch; a lubricant such as magnesium stearate or sterotes; a glidant such as colloidal silicon dioxide; a sweetening agent such as sucrose or saccharin; or a flavoring agent such as peppermint, methyl salicylate, orange flavoring, or other suitable flavorings. These are for purposes of example only and are not intended to be limiting.


The compositions comprising a microbial consortium can also be prepared in the form of suppositories (e.g., with conventional suppository bases such as cocoa butter and other glycerides) or retention enemas for rectal delivery. Suppositories can comprise one a number of species in a microbial consortium (e.g., a suppository can comprise a C. scindens and a C. bifermentans, or a suppository can comprise a C. scindens or a C. bifermentans). Suppositories comprising only one species of the microbial consortium can be co-administered with another composition comprising another species of the consortium, such that co-administration will equal the sum of the given consortium. The compositions can be prepared with carriers that will protect the consortium against rapid elimination from the body, such as a controlled release formulation, including implants. Biodegradable, biocompatible polymers can be used, such as ethylene vinyl acetate, polyanhydrides, polyglycolic acid, collagen, polyorthoesters, and polylactic acid. Such formulations can be prepared using standard techniques. The materials can also be obtained commercially from, for instance, Alza Corporation and Nova Pharmaceuticals, Inc. Liposomal suspensions can also be used as pharmaceutically acceptable carriers. These can be prepared according to methods known to those skilled in the art.


In some embodiments, a composition can be encapsulated, e.g., enteric-coated formulations). For instance, when the composition is to be administered orally, the dosage form is formulated so the composition is not exposed to conditions prevalent in the gastrointestinal tract before the small intestine, e.g., high acidity and digestive enzymes present in the stomach. The encapsulation of compositions for therapeutic use is routine in the art. Encapsulation can include hard-shelled capsules, which can be used for dry, powdered ingredients soft-shelled capsules. Capsules can be made from aqueous solutions of gelling agents such as animal protein (e.g., gelatin), plant polysaccharides or derivatives like carrageenans and modified forms of starch and cellulose. Other ingredients can be added to a gelling agent solution such as plasticizers (e.g., glycerin and or sorbitol), coloring agents, preservatives, disintegrants, lubricants and surface treatment.


In one embodiment, a microbial consortium as described herein is formulated with an enteric coating. An enteric coating can control the location of where a microbial consortium is released in the digestive system. Thus, an enteric coating can be used such that a microbial consortium-containing composition does not dissolve and release the microbes in the stomach, which can be a toxic environment for many microbes, but rather travels to the small intestine, where it dissolves and releases the microbes in an environment where they can survive. An enteric coating can be stable at low pH (such as in the stomach) and can dissolve at higher pH (for example, in the small intestine). Material that can be used in enteric coatings includes, for example, alginic acid, cellulose acetate phthalate, plastics, waxes, shellac, and fatty acids (e.g., stearic acid, palmitic acid). Enteric coatings are described, for example, in U.S. Pat. Nos. 5,225,202, 5,733,575, 6,139,875, 6,420,473, 6,455,052, and 6,569,457, all of which are herein incorporated by reference in their entirety. The enteric coating can be an aqueous enteric coating. Examples of polymers that can be used in enteric coatings include, for example, shellac (trade name EmCoat 120 N, Marcoat 125); cellulose acetate phthalate (trade names AQUACOAT™, AQUACOAT ECD™, SEPIFILM™, KLUCEL™, and METOLOSE™); polyvinylacetate phthalate (trade name SURETERIC™); and methacrylic acid (trade name EUDRAGIT™).


In one embodiment, an enteric coated prebiotic composition that additionally comprises members of a microbial consortium as described herein is administered to a subject. In another embodiment, an enteric coated probiotic and prebiotic composition is administered to a subject.


Formulations suitable for rectal administration include gels, aqueous or oily suspensions, dispersible powders or granules, emulsions, dissolvable solid materials, enemas, and the like. The formulations are preferably provided as unit-dose suppositories comprising the active ingredient in one or more solid carriers forming the suppository base, for example, cocoa butter. Suitable carriers for such formulations include petroleum jelly, lanolin, polyethyleneglycols, alcohols, and combinations thereof, provided they are compatible with the bacterial species preparation being administered.


In some embodiments, the microbial consortium can be formulated in a food item. Some non-limiting examples of food items to be used with the methods and compositions described herein include: popsicles, cheeses, creams, chocolates, milk, meat, drinks, yogurt, pickled vegetables, kefir, miso, sauerkraut, etc. In other embodiments, the food items can be juices, refreshing beverages, tea beverages, drink preparations, jelly beverages, and functional beverages; alcoholic beverages such as beers; carbohydrate-containing foods such as rice food products, noodles, breads, and pastas; paste products such as fish, hams, sausages, paste products of seafood; retort pouch products such as curries, food dressed with a thick starchy sauce, and Chinese soups; soups; dairy products such as milk, dairy beverages, ice creams, cheeses, and yogurts; fermented products such as fermented soybean pastes, fermented beverages, and pickles; bean products; various confectionery products including biscuits, cookies, and the like, candies, chewing gums, gummies, cold desserts including jellies, cream caramels, and frozen desserts; instant foods such as instant soups and instant soy-bean soups; and the like. It is preferred that food preparations not require cooking after admixture with the microbial consortium to avoid killing the microbes.


Formulations of a microbial consortium can be prepared by any suitable method, typically by uniformly and intimately admixing the consortium with liquids or finely divided solid carriers or both, in the required proportions and then, if necessary, shaping the resulting in mixture into the desired shape. In addition, the microbial consortium can be treated to prolong shelf-life, preferably the shelf-life of the pre-determined gut flora will be extended via freeze drying.


In some embodiments, the microbial consortium as described herein is combined with one or more additional probiotic organisms prior to treatment of a subject.


A nutrient supplement comprising the microbial consortium as described herein can include any of a variety of nutritional agents, including vitamins, minerals, essential and nonessential amino acids, carbohydrates, lipids, foodstuffs, dietary supplements, short chain fatty acids and the like. Preferred compositions comprise vitamins and/or minerals in any combination. Vitamins for use in a composition as described herein can include vitamins B, C, D, E, folic acid, K, niacin, and like vitamins. The composition can contain any or a variety of vitamins as may be deemed useful for a particularly application, and therefore, the vitamin content is not to be construed as limiting. Typical vitamins are those, for example, recommended for daily consumption and in the recommended daily amount (RDA), although precise amounts can vary. The composition can preferably include a complex of the RDA vitamins, minerals and trace minerals as well as those nutrients that have no established RDA, but have a beneficial role in healthy human or mammal physiology. The amount of material included in the composition can vary widely depending upon the material and the intended purpose for its absorption, such that the composition is not to be considered as limiting.


Also contemplated herein are kits comprising, at a minimum, a biotherapeutic microbial species or a consortium prep or formulations comprising the members of the consortium, e.g., C. scindens and C. bifermentans species, in an admixture or comprising the members of the consortium in sub-combinations or sub-mixtures. In some embodiments, the kit further comprises empty capsules to be filled by the practitioner and/or one or more reagents for enteric coating such capsules. It is also contemplated herein that the microbe preparation is provided in a dried, lyophilized or powdered form. In one embodiment, the kit comprises a strain of C. bifermentans. In another embodiment, the kit comprises a strain of C. scindens and a strain of C. bifermentans. The C. scindens strain comprised in the kit can be a C. scindens strain comprising a 16S rRNA sequence that is at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 1. The C. bifermentans strain comprised in the kit can be a C. bifermentans strain comprising a 16S rRNA sequence that is at least 97%, at least 98%, at least 99%, or 100% identical to the sequence of SEQ ID NO: 2. In another embodiment, the kit comprises at least one reducing agent such as N-acetylcysteine, cysteine, or methylene blue for growing, maintaining and/or encapsulating the microbes under anaerobic conditions. The kits described herein are also contemplated to include cell growth media and supplements necessary for expanding the microbial preparation. The kits described herein are also contemplated to include one or more prebiotics as described herein.


Prior to administration of the bacterial composition, the patient may optionally have a pretreatment protocol to prepare the gastrointestinal tract to receive the bacterial composition. In these instances, the pretreatment protocol can enhance the ability of the bacterial composition to affect the patient's microbiota balance. In an alternative embodiment, the subject is not pre-treated with an antibiotic.


Generally, the defined therapeutic microbiota described herein can be administered after the completion of a course of antibiotics for the treatment of C. difficile infection. However, use of the defined therapeutic microbiota alone to prevent or, for that matter, directly combat the C. difficile infection is specifically contemplated. If a patient has received antibiotics for treatment of an infection other than C. difficile or for C. difficile, in one embodiment the antibiotic should be stopped in sufficient time to allow the antibiotic to be substantially reduced in concentration in the gut before the bacterial composition is administered. In one embodiment, the antibiotic may be discontinued 1, 2, or 3 days before the administration of the bacterial composition. In one embodiment, the antibiotic can be discontinued 3, 4, 5, 6, or 7 antibiotic half-lives before administration of the bacterial composition.


In another embodiment, the bacterial compositions described herein are administered before or concurrently with an antibiotic. In one embodiment, administration of therapeutic microbiota before or concurrently with antibiotic might be contemplated where the administered species are at least somewhat resistant to the effects of the antibiotic administered. In another embodiment, an antibiotic is administered before the administration to the bacterial composition (e.g., less than 1 day, 1, 2, 3, 4, 5, 6, or 7 days before administration of the bacterial composition). Longer times can help to prevent the antibiotic from killing the administered bacteriotherapeutic organism(s). In one embodiment, the antibiotic administered is an antibiotic used to treat C. difficile infection (e.g., metronidazole (Flagyl), vancomycin (Vancocin), or fidaxomicin (Dificid)). In another embodiment, the antibiotic is not specific to treatment of a C. difficile infection, but is an antibiotic known in the art to have therapeutic effects on the intestinal system (e.g., norfloxacin, cephalexin, trimethoprim-sulfamethoxazole, or levofloxacin). Antibiotics listed herein are for purposes of example only and are not intended to be limiting.


In one embodiment, the bacterial compositions described herein are administered with an antacid or proton pump inhibitor (PPI). An antacid works to neutralize the stomach acid, which can interfere to efficient delivery of the bacterial compositions described herein. An antacid can be administered prior to, in combination with, or after the administration of the bacterial compositions described herein. In one embodiment, the bacterial composition can be formulation in a composition that further comprises an antacid. Antacids are known in the art and can comprise the following active ingredients: calcium carbonate, aluminum, magnesium, sodium bicarbonate, and/or alginic acid. Proton pump inhibitors block activity of the H+/K+ ATPase proton pump in stomach epithelium.


Any of the preparations described herein can be administered once on a single occasion or on multiple occasions, such as once a day for several days or more than once a day on the day of administration (including twice daily, three times daily, or up to five times daily). Or the preparation can be administered intermittently according to a set schedule, e.g., once weekly, once monthly, or when the patient relapses from the primary illness. In another embodiment, the preparation can be administered on a long-term basis to assure the maintenance of a protective or therapeutic effect.


Efficacy


Typically, a C. difficile infection can manifest with one of more of the following clinical symptoms or indicators: (i) mild (at least 3 times a day) to severe (4 or more times a day) watery diarrhea, (ii) abdominal pain, (iii) blood and/or pus in the stool, (iv) fever, and (v) loss of appetite. Quantitatively, a C. difficile infection can be assessed by quantitative factors (i) detectable levels of C. difficile toxin, and (ii) detectable levels of C. difficile bacteria. Thus, efficacious treatment and/or prevention of a C. difficile infection using the methods and compositions described herein can reduce or eliminate at least one of the symptoms or indicators associated with a C. difficile infection, as described above. Methods for the measurement of each of these parameters (e.g., measuring the levels of C. difficile toxins) are known to those of ordinary skill in the art and/or described herein.


Effective treatment can be determined by an overall decrease in the Bristol Score from 7 or 6 to a lower value. Alternatively, or in addition, effective treatment can be determined by a decrease, as the term is used herein, in C. difficile biomass or relative abundance in the stool, or by a decrease in C. difficile toxin in the stool.


Efficacy can also be measured by failure of a subject to worsen as assessed by need for medical interventions (e.g., progression of infection is halted or at least slowed). Methods of measuring these indicators are known to those of skill in the art and/or described herein. Example methods include PCR-based or Enzyme-linked immunosorbent assay (ELISA) to detect C. difficile toxin. Treatment includes: (1) inhibiting the infection, e.g., arresting, or slowing symptoms of the infection, for example watery diarrhea; or (2) relieving the infection, e.g., causing regression of symptoms, reducing the symptoms by at least 10%, and/or reducing C. difficile toxin levels by at least 10% compared to a reference level (e.g., a C. difficile toxin level prior to administration; and (4) restoring healthy intestinal flora, thus preventing future C. difficile infection or production. It is expected that the levels of C. difficile toxin and/or C. difficile bacteria present in a subject's intestine should be reduced to levels seen in healthy individuals or below detectable levels at least 1 week, at least 2 weeks, at least 3, weeks, at least 4 weeks following administration of any of the therapeutic compositions described herein.


Therapeutic microbiota, including defined therapeutic microbiota as described herein, are administered in an amount sufficient, or an amount effective, to provide therapeutic benefit. An effective amount of a composition for the treatment of C. difficile infection means that amount which, when administered to a mammal in need thereof, is sufficient to result in effective treatment as that term is defined herein, for that infection. Efficacy of the composition can be determined by a physician by assessing physical indicators of C. difficile infection as described above.


The term “effective amount” as used herein refers to the amount of a therapeutic microbiota composition as described herein needed to alleviate at least one or more symptoms or reduces one or more indicator of a C. difficile infection, and relates to a sufficient amount of pharmacological composition to provide the desired effect. The term “therapeutically effective amount” therefore refers to an amount of a composition that is sufficient to provide a particular effect when administered to a typical subject. An effective amount as used herein, in various contexts, would also include an amount sufficient to delay the development of a symptom of the infection, alter the course of a symptom (for example but not limited to, slowing the progression of a symptom of the infection), or reverse a symptom of the infection. Thus, it is not generally practicable to specify an exact “effective amount.” However, for any given case, an appropriate “effective amount” can be determined by one of ordinary skill in the art using only routine experimentation. The term “effective amount” is used interchangeably with the term “therapeutically effective amount” and refers to the amount of at least one agent, e.g., a bacterial composition that treats a C. difficile infection, at dosages and for periods of time necessary to achieve the desired therapeutic result, for example, to reduce or stop at least one symptom or indicator of such C. difficile infection, in the subject.


Repeated administration of the defined therapeutic microbiota composition may be beneficial to maintain a protective or curative effect.


Effective amounts, toxicity, and therapeutic efficacy of drug agents, e.g., for formulations or treatments using antibiotics in addition to microbes, can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dosage can vary depending upon the dosage form employed and the route of administration utilized. The dose ratio between toxic and therapeutic effects is the therapeutic index and can be expressed as the ratio LD50/ED50. Compositions and methods that exhibit large therapeutic indices are preferred. A therapeutically effective dose can be estimated initially from in vivo assays. It is contemplated that the relevant level for an agent that treat a C. difficile infection may also be the level achieved in the lumen of the gut. The effects of any particular dosage can be monitored by a suitable bioassay or by measurement of administered and stable biomass (engraftment, persistence) and microbial metabolic activities (Stickland metabolite production, free amino acids, protease activities, associate gene content or expression levels)


The dosage can be determined by a physician and adjusted, as necessary, to suit observed effects of the treatment.


All patents and other publications; including literature references, issued patents, published patent applications, and co-pending patent applications; cited throughout this application are expressly incorporated herein by reference for the purpose of describing and disclosing, for example, the methodologies described in such publications that might be used in connection with the technology described herein. These publications are provided solely for their disclosure prior to the filing date of the present application. Nothing in this regard should be construed as an admission that the inventors are not entitled to antedate such disclosure by virtue of prior invention or for any other reason. All statements as to the date or representation as to the contents of these documents is based on the information available to the applicants and does not constitute any admission as to the correctness of the dates or contents of these documents.


The description of embodiments of the disclosure is not intended to be exhaustive or to limit the disclosure to the precise form disclosed. While specific embodiments of, and examples for, the disclosure are described herein for illustrative purposes, various equivalent modifications are possible within the scope of the disclosure, as those skilled in the relevant art will recognize. For example, while method steps or functions are presented in a given order, alternative embodiments may perform functions in a different order, or functions may be performed substantially concurrently. The teachings of the disclosure provided herein can be applied to other procedures or methods as appropriate. The various embodiments described herein can be combined to provide further embodiments. Aspects of the disclosure can be modified, if necessary, to employ the compositions, functions and concepts of the above references and application to provide yet further embodiments of the disclosure. Moreover, due to biological functional equivalency considerations, some changes can be made in protein structure without affecting the biological or chemical action in kind or amount. These and other changes can be made to the disclosure in light of the detailed description. All such modifications are intended to be included within the scope of the appended claims.


Specific elements of any of the foregoing embodiments can be combined or substituted for elements in other embodiments. Furthermore, while advantages associated with certain embodiments of the disclosure have been described in the context of these embodiments, other embodiments may also exhibit such advantages, and not all embodiments need necessarily exhibit such advantages to fall within the scope of the disclosure.


The technology described herein is further illustrated by the following examples which in no way should be construed as being further limiting.


Some embodiments of the technology described herein can be defined according to any of the following numbered paragraphs:


1. A pharmaceutical composition comprising an oral formulation comprising C. scindens and C. bifermentans bacteria.


2. A composition comprising C. scindens and C. bifermentans bacteria, wherein one or both of the C. scindens and C. bifermentans bacteria is in dried, viable form.


3. A pharmaceutical composition comprising a formulation comprising C. scindens and C. bifermentans bacteria, wherein the composition does not comprise Bacteroides species or Escherichia species.


4. The composition of any one of paragraphs 1-3, wherein one or both of the C. scindens and C. bifermentans bacteria are in spore form.


5. The composition of any one of paragraphs 1-3, wherein one or both of the C. scindens and C. bifermentans bacteria are not in spore form.


6. The composition of any one of paragraphs 1-3, wherein the C. scindens and C. bifermentans bacteria are present as a mixture of metabolically active and spore forms.


7. The composition of any one of paragraphs 1-3, wherein the composition comprises a capsule or microcapsule, or a composition formulated for enteric delivery.


8. The composition of paragraph 1 or paragraph 3, wherein one or both of the C. scindens and C. bifermentans bacteria are in dried viable form.


9. The composition of any one of paragraphs 1-3, which does not comprise C. sardiniensis bacteria.


10. The composition of any one of paragraphs 1-3, which does not comprise any other Clostridium species.


11. The composition of either of paragraphs 1 or 2, which does not contain Bacteroides species or Escherichia coli.


12. The composition of any one of paragraphs 1-3, in which the formulation comprises no other bacteria.


13. A pharmaceutical composition comprising an oral formulation comprising a first bacterial population that has 16S rDNA with a sequence at least 97% identical to a 16S rDNA sequence present in a reference C. scindens bacterium, and a second bacterial population that has 16S rDNA with a sequence at least 97% identical to a 16S rDNA sequence present in a reference C. bifermentans bacterium.


14. A composition comprising a first bacterial population that has 16S rDNA with a sequence at least 97% identical to a 16S rDNA sequence present in a reference C. scindens bacterium, and a second bacterial population that has 16S rDNA with a sequence at least 97% identical to a 16S rDNA sequence present in a reference C. bifermentans bacterium, wherein one or both of the C. scindens and C. bifermentans bacteria is in dried, viable form.


15. A pharmaceutical composition comprising a formulation comprising a first bacterial population that has 16S rDNA with a sequence at least 97% identical to a 16S rDNA sequence present in a reference C. scindens bacterium, and a second bacterial population that has 16S rDNA with a sequence at least 97% identical to a 16S rDNA sequence present in a reference C. bifermentans bacterium wherein the composition does not comprise Bacteroides species or Escherichia species.


16. The composition of any one of paragraphs 13-15, wherein one or both of the C. scindens and C. bifermentans bacteria are in spore form.


17. The composition of any one of paragraphs 13-15, wherein one or both of the C. scindens and C. bifermentans bacteria are not in spore form.


18. The composition of any one of paragraphs 13-15, wherein the C. scindens and C. bifermentans bacteria are present as a mixture of metabolically active and spore forms.


19. The composition of any one of paragraphs 13-15, wherein the composition comprises a capsule or microcapsule, or a composition formulated for enteric delivery.


20. The composition of paragraph 13 or paragraph 15, wherein one or both of the C. scindens and C. bifermentans bacteria are in dried viable form.


21. The composition of any one of paragraphs 13-15, which does not comprise C. sardiniense bacteria


22. The composition of either of paragraphs 13 or 14, which does not comprise Bacteroides species or Escherichia coli.

23. The composition of any one of paragraphs 13-15, which does not comprise any other Clostridium species.


24. The composition of any one of paragraphs 13-15, in which the formulation comprises no other bacteria.


25. The composition of any one of paragraphs 1-24, further comprising a prebiotic.


26. The composition of any one of paragraphs 1-11, 23, or 25, further comprising a microbe that supports C. scindens and/or C. bifermentans.

27. The composition of paragraph 26, wherein the microbe that supports C. scindens is Ruminococcus obeum.

28. The composition of any one of paragraphs 1-27, for use in the treatment of C. difficile infection.


29. The composition for use of paragraph 28, wherein the use comprises suppressing the expression of C. difficile toxin.


30. The composition for use of paragraph 28, wherein the use comprises promoting a shift towards use of the proline reductase pathway of Stickland fermentation in C. difficile.

31. The composition for use of paragraph 28, wherein the use comprises inducing CodY activity or expression in C. difficile.

32. The composition for use of paragraph 28, wherein the use comprises promoting ethanolamine utilization by C. difficile.

33. A method comprising administering a composition of any one of paragraphs 1-27 to a subject in need thereof.


34. The method of paragraph 33, wherein the subject has or has been diagnosed with C. difficile infection.


35. The method of paragraph 34, wherein the C. difficile infection is recurrent.


36. The method of paragraph 33, wherein the subject is at risk of C. difficile infection or recurrent C. difficile infection.


37. The method of paragraph 33, wherein the administration is oral.


38. A method comprising administering a composition of any one of paragraphs 2, 3, 14 or 15 directly to the colon of a subject in need thereof.


39. The method of paragraph 38, wherein administration is via colonoscope or enema.


40. The method of any of paragraphs 33-39, wherein the subject is receiving or has recently received antibiotic treatment.


41. A method of treating an infection, the method comprising administering an antibiotic and a composition of any one of paragraphs 1-27.


42. The method of paragraph 41, wherein the composition is administered before or concurrently with the antibiotic.


43. The method of paragraph 41, wherein the composition is administered after a course of an antibiotic.


44. A method of predicting recurrence of C. difficile infection in a subject, the method comprising:

    • (a) determining the relative abundance of all operational taxonomic units (OTUs) in a sample of the subject's stool that are >90% identical to a reference sequence of C. scindens;
    • (b) determining the relative abundance of all operational taxonomic units (OTUs) in a sample of the subject's stool that are >90% identical to a reference sequence of C. hylemonae; and
    • (c) summing the relative abundances determined in steps (a) and (b), wherein a sum of relative abundances less than or equal to 1% indicates an increased risk of C. difficile recurrence relative to a subject in which the sum of relative abundances is greater than 1%.


      45. The method of paragraph 44, wherein the reference sequences for C. scindens and C. hylemonae are 16S rDNA sequences.


      46. The method of paragraph 44 or 45, wherein the determining steps are performed on samples taken before, during or after the subject has been treated with antibiotics for C. difficile infection.


      47. The method of paragraph 44 or 45, wherein the determining steps are performed on samples taken after the subject has been treated with antibiotics for C. difficile infection


      48. The method of paragraph 44, further comprising the step, when the sum of relative abundances is at or below 1%, of administering a composition of any one of paragraphs 1-27.


      49. A method of suppressing expression of a bacterial toxin in a subject, the method comprising administering a defined bacterial microbiota comprising at least one bacterial organism that encodes and secretes a protease and/or performs Stickland fermentation.


      50. A method of treating or preventing a pathology caused by expression of a bacterial toxin, the method comprising administering a defined bacterial microbiota comprising at least one bacterial organism that encodes and secretes a protease and/or performs Stickland fermentation.


      51. A method of promoting CodY expression or activity in a C. difficile bacterium in a subject, the method comprising administering a defined bacterial microbiota comprising a bacterial organism that encodes and secretes a protease and/or performs Stickland fermentation.


      52. A method of promoting ethanolamine utilization by a C. difficile bacterium in a subject, the method comprising administering a defined bacterial microbiota comprising a bacterial organism that encodes and secretes a protease and/or performs Stickland fermentation.


      53. The method of any one of paragraphs 49-52, wherein at least one bacterial organism encodes and secretes at least one protease selected from the group consisting of: a protease of PATRIC ID fig|186802.30.peg.279; a protease of PATRIC ID fig|186802.30.peg.290; a protease of PATRIC ID fig|186802.30.peg.313; a protease of PATRIC ID fig|186802.30.peg.414; a protease of PATRIC ID fig|186802.30.peg.543; a protease of PATRIC ID fig|186802.30.peg.2205; a protease of PATRIC ID fig|186802.30.peg.2313; a protease of PATRIC ID fig|186802.30.peg.2680; a protease of PATRIC ID fig|186802.30.peg.2745; a protease of PATRIC ID fig|186802.30.peg.2746; a protease of PATRIC ID fig|186802.30.peg.830; a protease of PATRIC ID fig|186802.30.peg.921; a protease of PATRIC ID fig|186802.30.peg.936; a protease of PATRIC ID fig|186802.30.peg.3000; a protease of PATRIC ID fig|186802.30.peg.3018; a protease of PATRIC ID fig|186802.30.peg.3019; and a protease of PATRIC ID fig|186802.30.peg.3065.


      54. The method of any of paragraphs 49-52, wherein at least one protease performs the proteolysis reaction of enzymes of Enzyme Commission number (E.C. number) EC 3.4.21.-; EC 3.4.21.53; or EC 3.4.21.92.


      55. The method of any of paragraphs 49-52, wherein at least one bacterial organism encodes and expresses one or more of D-proline reductase, Glycine reductase, Thioredoxin, or Choloylglycine hydrolase.


      56. The method of any of paragraphs 49-52, wherein the at least one bacterial organism falls within Clostridial cluster I, XI, or XIVa, and does not express a pathology-causing bacterial toxin.


      57. The method of paragraph 56, wherein the bacterial organism in Clostridial cluster I is selected from C. sporogenes, and C. histolyticum.

      58. The method of paragraph 56, wherein the bacterial organism in Clostridial cluster XI is selected from C. bifermentans, C. hiranonis, and P. anaerobius.

      59. The method of paragraph 56, wherein the bacterial organism in Clostridial cluster XIVa is selected from C. scindens, C. clostriiforme, and C. nexile.

      60. The method of any of paragraphs 49-52, wherein the at least one bacterial organism inhibits sorbitol/mannitol fermentation by C. difficile.

      61. The method of any of paragraphs 49-52, wherein the at least one bacterial organism promotes Stickland fermentation through the acceptor amino acid proline, or activation of proline reductase.


      62. The method of any of paragraphs 49-52, wherein the at least one bacterial organism promotes 5-aminovalerate production.


      63. The method of any of paragraphs 49-52, wherein the bacterial toxin is a C. difficile toxin.


      64. The method of any of paragraphs 49-52, wherein the bacterial organism is C. bifermentans and/or C. scindens.

      65. The method of any of paragraphs 49-52, wherein suppressing expression of a bacterial toxin compromises by inhibition of butyrate, codY, ccpA, tcdR, and/or tcdA production.


      66. A method of suppressing expression of a bacterial toxin in the gut of a subject, the method comprising administering at least one amino acid that is metabolized by Stickland fermentation.


      67. A method of treating or preventing a pathology caused by expression of a bacterial toxin, comprising administering at least one amino acid that is metabolized by Stickland fermentation.


      68. The method of paragraph 66 or 67, wherein at least one amino acid is a Stickland donor or Stickland acceptor.


      69. The method of paragraph 68, wherein the Stickland donor is selected from the group consisting of: alanine, leucine, valine, isoleucine, tryptophan, tyrosine and phenylalanine.


      70. The method of paragraph 68, wherein the Stickland acceptor is selected from the group consisting of: glycine and proline.


      71. The method of paragraph 66 or 67, wherein the amino acid is a branched-chain amino acid, a branched-keto amino acid, or an aromatic amino acid.


      72. The method of paragraph 66 or 67, wherein the at least one amino acid promotes 5-aminovalerate production.


      73. The method of paragraph 66 or 67, wherein the bacterial toxin is a C. difficile toxin.


      74. The method of paragraph 66 or 67, wherein suppression of the expression of a bacterial toxin comprises inhibition of butyrate, codY, ccpA, tcdR, and/or tcdA activity or production.


      75. A method of determining the therapeutic efficacy of a bacterial organism for treatment of a pathology involving expression of a toxin, produced by a Gram-positive spore-forming bacterium, the method comprising measuring in a biological sample obtained from an individual administered the bacterial organism one or more of:
    • a) the amount and/or activity of a secreted proteolytic enzyme;
    • b) the amount and/or activity of bacterial proline reductase;
    • c) the amount or concentration of one or more branched short-chain fatty acids;
    • d) the amount or concentration of one or more branched keto acids; and
    • e) the amount or concentration of Stickland donor and/or Stickland acceptor amino acids and/or 5-aminovalerate;
    • wherein measurement of an increased amount, concentration or activity of one or more of (a)-(e) relative to the amount measured in a sample taken prior to administration the bacterial organism indicates that the bacterial organism is effective for the treatment.


      76. The method of paragraph 75, wherein the bacterial toxin is produced by a Gram-positive, spore-forming bacterium.


      77. The method of paragraph 75, wherein the bacterial toxin is a C. difficile toxin.


      78. The method of paragraph 75, wherein the pathology comprises expression of a toxin by C. difficile.

      79. The method of paragraph 75, wherein Stickland donor amino acids are selected from leucine, isoleucine, valine, alanine, phenylalanine, tryptophan, tyrosine and Stickland acceptor amino acids are selected from glycine, proline, and hydroxyproline.


      80. The method of paragraph 75, wherein the sample is a stool sample or a sample from within the colon of the individual.


      81. A method to predict the risk of developing a disease involving a toxin produced by a Gram positive, spore-forming bacterium, the method comprising measuring in a biological sample obtained from an individual one or more of the following:
    • a) the amount and/or activity of a secreted proteolytic enzyme;
    • b) the amount and/or activity of bacterial proline reductase;
    • c) the amount or concentration of one or more branched short-chain fatty acids;
    • d) the amount or concentration of one or more branched keto acids; and
    • e) the amount or concentration of Stickland donor and/or Stickland acceptor amino acids; and
    • comparing the amount, concentration or activity measured in one or more of (a)-(d) to a reference, wherein an amount, concentration or activity in one or more of (a)-(d) below the reference indicates increased risk of developing a disease involving a toxin produced by a Gram positive, spore-forming bacterium.


      82. The method of paragraph 81, wherein the disease involves expression of a toxin by C. difficile.

      83. The method of paragraph 81, wherein the reference comprises a biological sample from a healthy individual.


      84. The method of paragraph 81, wherein the biological sample is a stool sample or a sample from within the colon of the individual.


      85. The method of paragraph 81, wherein two or more, of (a)-(e) are measured.


      86. The method of paragraph 81, wherein three or more, of (a)-(e) are measured.


      87. The method of paragraph 81, wherein four or more, of (a)-(e) are measured.


      88. A method of identifying a candidate bacterial organism that is likely to suppress the expression of a toxin by a Gram-positive, spore-forming bacterial pathogen, the method comprising:
    • a) identifying from a database of bacterial genetic information a candidate bacterial organism having in its genome:
    • i) one or more genes encoding a secreted protease enzyme; and/or
    • ii) a gene encoding a proline reductase enzyme; and
    • b) assaying a sample comprising the candidate bacterial organism for the expression of a secreted protease enzyme and/or the proline reductase enzyme;


      wherein the detection of expression of the secreted protease enzyme and/or the expression of the proline reductase enzyme indicates that the candidate bacterial organism is likely to suppress expression of a toxin by a Gram-positive, spore-forming bacterial pathogen.


      89. The method of paragraph 88, wherein the candidate bacterial organism is not an opportunistic gut pathogen in humans.


      90. The method of paragraph 88, wherein the proline reductase enzyme is an enzyme of E.C. 1.21.4.1.


      91. A method to predict the risk of developing a spore-forming, toxin-producing Gram-positive bacterial pathogen in the gut or other location, or its recurrence, comprising measuring in a biological sample
    • (a) amounts or unit activity of proteolytic activity;
    • (b) concentrations of branched short chain fatty acids;
    • (c) concentrations of branched keto acids; and/or
    • (d) concentrations of Stickland donor and Stickland acceptor amino acids,
    • wherein an increase in the amount or activity of at least one of (a)-(d) relative to a biological sample obtained prior to administration identifies a risk of developing a spore-forming, toxin-producing Gram-positive bacterial pathogen in the gut or other location.


      92. The method of paragraph 91, further comprising, prior to measuring, administering the bacterial organism or amino acid to the subject.


      93. The method of paragraph 91, wherein the biological sample is obtained from a subject.


      94. The method of paragraph 91, wherein the biological sample is a stool sample.


      95. The method of paragraph 91, wherein the biological sample is obtained from the gut.


      96. The method of paragraph 91, wherein the gram-positive bacterial pathogen is C. difficile infection.


      97. The composition of any of paragraphs 1-24, further comprising an amount of one or more free Stickland donor and/or Stickland acceptor amino acids effective to promote Stickland fermentation by a species in the composition or by C. difficile after the composition is administered to a subject.


      98. The composition of any of paragraphs 1-24, further comprising an amount of a polypeptide substrate effective for proteolysis by proteolytic activity of a bacterial species in the composition to generate amino acids fermentable by Stickland fermentation.


      99. The composition of paragraph 25, further comprising an amount of one or more free Stickland donor and/or Stickland acceptor amino acids effective to promote Stickland fermentation by a species in the composition or by C. difficile after the composition is administered to a subject.


      100. The composition of paragraph 25, further comprising an amount of a polypeptide substrate effective for proteolysis by proteolytic activity of a bacterial species in the composition to generate amino acids fermentable by Stickland fermentation.


      101. The composition of any one of paragraphs 98 or 100, wherein the polypeptide substrate comprises casein, collagen and/or gelatin.


      102. The composition of any one of claim 98 or 100, wherein the polypeptide substrate comprises a synthetic polymer or copolymer polypeptide hydrolysable by a proteolytic activity of a species in the composition to generate Stickland fermentable amino acids.


      103. The composition of paragraph 102, wherein the synthetic polymer comprises a poly[N] polymer, where N is a Stickland donor amino acid selected from leucine, isoleucine, valine, alanine, phenylalanine, tryptophan, and tyrosine or a Stickland acceptor amino acids selected from glycine and proline.


      104. The composition of paragraph 102, wherein the synthetic copolymer comprises a poly[N,X] copolymer, where N and X are Stickland donor amino acids selected from leucine, isoleucine, valine, alanine, phenylalanine, tryptophan, and tyrosine or Stickland acceptor amino acids selected from glycine and proline.


EXAMPLES
Example 1: Clostridium bifermentans can Provide Complete Protection Against Fatal C. difficile Infection

The technology described herein is related to the surprising discovery that two human commensal species, Clostridium scindens and Clostridium bifermentans, offer protection from an otherwise lethal infection with the Gram-positive, toxigenic bacterium, C. difficile.


The effect of commensal species on the survival of mice infected with C. difficile spores was assessed using a gnotobiotic colonization model. FIG. 1A sets out the experimental approach schematically. Swiss-Webster germfree mice were pre-colonized with a commensal species (C. bifermentans, C. sardiniense or C. scindens) for 7 days, prior to challenge with 1000 spores of the C. difficile strain (ATCC43255). The survival of the mice was monitored for up to 28 days post-challenge with C. difficile (see, e.g., FIG. 1B). Body condition score (BCS) of the mice was monitored daily to assess activity, feeding, grooming and tissue turgor for additional clinical symptoms of infection (FIG. 1C).


As expected, control mice that were challenged with C. difficile alone developed a severe toxin-mediated pathology and died within 5 days of the infection with the Gram-positive toxigenic bacterium C. difficile (FIG. 1B). Surprisingly, mice that were pre-colonized with the commensal species C. bifermentans were completely protected from the otherwise lethal infection with C. difficile. and showed a 100% survival rate compared to the control group infected with C. difficile without any pre-colonization treatment (FIG. 1B). Mice that were precolonized with the commensal species C. scindens showed a survival rate of 80% compared to the control group that was infected with C. difficile without any pre-colonization treatment (FIG. 1B). Precolonization with the commensal species C. sardiniense did not provide any protection against an acute infection with C. difficile (FIG. 1B).


Consistent with the survival data, the body condition scores (BCS) and histopathological assessment of the mice showed that C. bifermentans provided protection against an acute primary C. difficile infection, as shown by a milder acute tissue pathology and lower lymphocytic infiltration as compared to the control group (FIG. 1C and FIG. 1G). C. bifermentans also provided protection against epithelial disruptions and tissue edema (FIG. 1G and FIG. 1J). In contrast, even though 80% of the C. scindens-colonized mice survived the infection with C. difficile, the histopathological assessment showed an increased number of inflammatory infiltrates, including a neutrophilic infiltrate, in addition to rare focal areas of epithelial denudation. Consistent with the short survival data indicating no protection against an acute primary C. difficile infection, C. sardiniense-colonized mice (FIG. 1F) developed a dilation of the colon (megacolon) with rapidly advancing tissue edema, epithelial sloughing and neutrophil efflux into the gut lumen, comparable or worse to the pathology seen in the control mice (FIG. 1E).


The C. difficile biomass and levels of C. difficile toxin were examined in the gnotobiotic mouse model. C. difficile toxin B levels were assessed in cecal samples collected from germfree Swiss-Webster mice infected with 1000 spores of the C. difficile ATCC-43255 strain (FIG. 2). ToxinB was detected using an enzyme-linked immunosorbent assay (ELISA). Germ-free animals infected with C. difficile alone, C. difficile plus C. bifermentans, and C. difficile plus C. sardiniense showed that C. difficile toxin B levels in cecal contents were dramatically lower in animals pre-colonized with C. bifermentans (FIGS. 2A, 2C), but that C. sardiniense actually promoted C. difficile toxin B levels (FIG. 2A). Interestingly, examination of biomass of the individual species showed that the biomass of C. difficile in C. bifermentans pre-colonized mice was considerable despite the complete protection from death (FIG. 2B). This indicates that the protective commensal C. bifermentans involves something other than killing the C. difficile or even suppressing growth of the C. difficile organism, and indicates that C. bifermentans can instead suppress C. difficile toxin production.


The effects of C. bifermentans and C. sardiniense were examined in conventional mice infected with C. difficile. FIG. 3A shows a schematic for the experiments in adult conventional mice. Briefly, adult conventional mice, with a complex gut microflora, were treated with intraperitoneal clindamycin for 24 hours before receiving 1×104 spores of the C. difficile strain ATCC 43255. Approximately 20 hours after dosing, as mice first developed signs of symptomatic infection, animals received 5×107 CFU of C. bifermentans, C. sardiniense or control vehicle alone by gavage, and survival was assessed for an additional 14 days. The results (shown in FIG. 3B) demonstrate that the protective effect of C. bifermentans, and the antagonistic effect of C. sardiniense is recapitulated in conventional mice, and that even in the more complex conventional mouse system, a single species of bacterium can provide complete therapeutic treatment and protection from an otherwise fatal C. difficile infection.


Example 2: Clostridium bifermentans is a Highly Proteolytic Species

An examination of species that provided protective effects against C. difficile was undertaken in order to identify what characteristics of the protective species mediate the protection. Species including C. bifermentans, C. hiranonis, C. sardinense, C. scindens, C. ramosum and C. difficile clinical isolates were assessed for their proteolytic activity using both microbiological and biochemical assays. FIG. 4A shows results of a meat granule microbiological protease assay performed on C. bifermentans, C. hiranonis, C. sardiniense, C. scindens, C. ramosum and two clinical C. difficile isolates. FIG. 4B summarizes results for both microbiological and biochemical protease assays for C. difficile, C. bifermentans, C. sardiniense, and C. scindens. Clostridium bifermentans showed strikingly high proteolytic activity in the meat granule assay (FIG. 4A, 4B) and strong digestion in both gelatin and casein hydrolysis assays (FIG. 4B), while the other species were considerably less active in each of these assays (FIGS. 4A and 4B).


Example 3: Clostridium bifermentans Promotes Stickland Fermentation by the Gram-Positive Toxigenic Bacterium C. difficile

The cecal contents of germfree Swiss-Webster mice were collected 20 hours after infection with C. difficile and untargeted metabolomic analysis was performed. The metabolic profiles of Stickland donor and acceptor amino acids, branched-chain amino acids (FIGS. 5A-5G) and carbohydrates (FIG. 5H) were assessed. A liquid chromatography tandem mass spectrometry (LC-MS) method was used to measure polar metabolites and, amino acids and carbohydrates in each sample. In each of FIGS. 5A-5G, the Y axis shows the Log10 MassSpec units of detected compounds. The X axis shows the experimental condition: GF-germfree controls (no bacteria); Cdiff—challenge with 1000 C. difficile spores of strain ATCC43255; CSAR—7 days mono-association with C. sardiniense; CBI—7 days mono-association with C. bifermentans; Cdiff+CSAR—mice mono-associated with C. sardiniense for 7 days followed by C. difficile challenge; Cdiff+CBI—mice mono-associated with C. bifermentans for 7 days followed by C. difficile challenge. Each group had 8 mice across two experimental replicates. Levels of amino acids/metabolites including 4-hydroxyproline (FIG. 5A), proline (FIG. 5B), 5-aminovalerate (FIG. 5C), glycine (FIG. 5D), leucine (FIG. 5E), isoleucine (FIG. 5E), valine (FIG. 5E), isovalerate (FIG. 5E), phenylalanine (FIG. 5F), tryptophan (FIG. 5F), and tyrosine (FIG. 5F), among others, were examined. The levels of the Stickland donor amino acids proline and glycine were reduced in the cecal contents of animals monoassociated with C. difficile and C. bifermentans and in animals co-colonized with C. bifermentans and C. difficile, while 5-aminovalerate, the product of reduction of proline as a Stickland donor was significantly increased in animals monoassociated with C. difficile, C. bifermentans or co-colonized with these two species. This is in contrast, for example, with the results with animals monoassociated with C. sardiniense, which was not protective, and showed levels of proline similar to those seen in germ free mice. This indicates active Stickland fermentation through the proline reductase pathway in animals colonized with C. difficile and with C. bifermentans or both. Acetate, the product of Stickland fermentation reaction through glycine as donor amino acid is produced by a number of other pathways, making it more difficult to conclude that C. bifermentans promotes Stickland fermentation through glycine. However, levels of glycine were lower than in the gut of germ free mice when mice were monoassociated with the Stickland fermenting species C. difficile and C. bifermentans, consistent with ongoing reduction of glycine through the glycine reductase Stickland pathway. The mice mono-associated with C. sardiniense for 7 days followed by C. difficile challenge also showed a 50% reduction in the levels of 5-aminovalerate (FIG. 5C), while the mice mono-associated with C. bifermentans for 7 days followed by C. difficile challenge had increased 5-aminovalerate production (FIG. 5C). Taken together, these data indicate that C. bifermentans promotes Stickland fermentation by the Gram-positive toxigenic bacterium C. difficile when C. bifermentans suppresses C. difficile toxin production.


Analyses of Stickland donor amino acids including branch chain amino acids (alanine, leucine, valine, and isoleucine) and the aromatic amino acids (phenylalanine, tryptophan and tyrosine) are consistent with these amino acids participating in Stickland fermentation in C. difficile and in C. bifermentans (FIG. 5E, 5F). The products of Stickland fermentation of leucine, isoleucine and valine include the branched short chain fatty acids (bSCFA) isovalerate, isocaproate and isobutyrate, respectively. As shown in FIG. 5I, analyses of C. bifermentans metabolites showed significant production of each of these by C. bifermentans, but negligible production of butyrate, which is not a direct product of Stickland fermentation.



C. difficile Short Chain Fatty Acids (SCFA) produced in in vitro culture was measured (FIG. 6A). The metabolic analysis showed that C. difficile undergoes Stickland fermentations, per branched short-chain fatty acid metabolites, and is able to also use glucose and the sugar alcohols mannitol and sorbitol (FIGS. 6A and 6B).


Example 4: Clostridium bifermentans Inhibits Expression of C. difficile Toxin Expression-Promoting Sigma Factor TcdR

Gene expression was measured by bacterial RNAseq analysis of cecal contents from C. difficile infected gnotobiotic Swiss-Webster mice at 20 hours post-inoculation, compared with mice colonized with C. bifermentans for 7 days prior to challenge with C. difficile for 20 hours. C. bifermentans colonized mice showed a >48× decrease in C. difficile tcdR sigma factor gene expression with concomitant >10× decreases in toxinA (tcdA) and toxinB (tcdB) gene expression (FIG. 7), demonstrating that C. bifermentans inhibits or suppresses expression of C. difficile toxin genes. The strong repression of tcdR strongly infers codY activation and repression of the toxin gene. Similarly, the repression of tcdA and B infer codY and ccpA activation, per a nutritionally and energetically supported state to repress these genes.


Example 5: Clostridium bifermentans Promotes Expression of Genes Permitting Ethanolamine Fermentation by the Gram-Positive Toxigenic Bacterium C. difficile

Gene expression was measured by bacterial RNAseq analysis of cecal contents from C. difficile-infected gnotobiotic Swiss-Webster mice at 20 hours post-inoculation, compared with mice colonized with C. bifermentans for 7 days prior to challenge with C. difficile for 20 hours. It was found that C. difficile structural proteins for the ethanolamine carboxysome (eutH, eutK, eutL, eutN) were up-regulated >10× when C. difficile is inoculated into a C. bifermentans-colonized mouse (FIG. 7). This indicates that Clostridium bifermentans likely promotes ethanolamine fermentation by the gram-positive toxigenic bacterium C. difficile.


The results of the experiments described herein demonstrate that the commensal bacterium C. bifermentans not only suppresses toxin production by a mechanism involving inhibition of the C. difficile TcdR sigma factor required for C. difficile toxin expression, but also promotes Stickland fermentation by Gram-positive toxigenic bacteria such as C. difficile in vivo and thereby can treat and/or prevent the development of a toxin-mediated pathology. These in vivo activities infer specific action of C. bifermentans on the C. difficile metabolic regulators codY (primary repressor of tcdR), ccpA (repressor of the tcdAEB opeon, also of tcdR), prdR (activator of proline reductase and repressor of glycine reductase operons), and rex (sensor of NADH/NAD+ energy state). Suppression of toxin production provides an alternative route of treatment for C. difficile-mediated pathology, in that it can be sufficient for treatment to just suppress production of the pathology-generating toxin without necessarily killing the pathogenic microbe. It is contemplated that at least part of this effect is through the strong extracellular proteolytic activity expressed by C. bifermentans, in that this activity can feed amino acids necessary for the generation of energy by C. difficile through Stickland fermentation into the gut environment, thereby keeping C. difficile energetically satisfied and suppressing its expression of toxin. Another part of the effect is likely through promotion of ethanolamine metabolism for energy by C. difficile; ethanolamine derived from diet, host tissues and other commensals is fairly abundant in the gut, and a shift in C. difficile gene expression induced by C. bifermentans that permits C. difficile to gain energy from ethanolamine is contemplated to further contribute to maintaining C. difficile in an energetically satisfied state that suppresses toxin expression.

Claims
  • 1. A method of suppressing toxin production by C. difficile bacteria in a subject in need thereof, the method comprising orally administering a formulation comprising viable C. scindens and C. bifermentans bacteria in an amount effective to reduce C. difficile toxin by at least 10% as compared to the level of C. difficile toxin prior to treatment onset, wherein one or both of the C. scindens and C. bifermentans bacteria are in spore form or in dried viable form; and wherein the formulation comprises no other bacteria.
  • 2. The method of claim 1, wherein the formulation comprises a capsule or microcapsule, or a composition comprising an enteric coating.
  • 3. The method of claim 1, wherein the formulation further comprises a prebiotic.
  • 4. The method of claim 1, wherein the subject has or has been diagnosed with, or is at risk of C. difficile infection.
  • 5. The method of claim 4, wherein the C. difficile infection is recurrent.
  • 6. The method of claim 1, wherein the formulation is administered before or concurrently with an antibiotic.
  • 7. The method of claim 1, wherein the formulation is administered after a course of an antibiotic.
  • 8. A method of suppressing toxin production by C. difficile bacteria in a subject in need thereof, the method comprising orally administering a formulation comprising viable C. scindens, C. bifermentans and Ruminococcus obeum bacteria in an amount effective to reduce C. difficile toxin by at least 10% as compared to the level of C. difficile toxin prior to treatment onset, wherein the C. scindens, C. bifermentans and R. obeum bacteria are in spore form or in dried viable form.
CROSS-REFERENCE TO RELATED APPLICATION

This application is a 35 U.S.C. § 371 National Phase Entry of International Application No. PCT/US2018/065023 filed Dec. 11, 2018, which designates the U.S., and which claims benefit under 35 U.S.C. § 119(e) of the U.S. Provisional Application Nos 62/597,116 filed Dec. 11, 2017 and 62/665,754 filed May 2, 2018, the contents of each of which are incorporated herein by reference in their entirety.

GOVERNMENT SUPPORT

This invention was made with Government support under Grant Nos. P30DK034854 and T32HL007627 awarded by the National Institutes of Health and under Grant No. HR0011-15-C-0094 awarded by the U.S. Department of Defense. The Government has certain rights in the invention.

PCT Information
Filing Document Filing Date Country Kind
PCT/US2018/065023 12/11/2018 WO
Publishing Document Publishing Date Country Kind
WO2019/118510 6/20/2019 WO A
US Referenced Citations (4)
Number Name Date Kind
10265349 Chatila Apr 2019 B2
10391131 Chatila Aug 2019 B2
10898527 Chatila Jan 2021 B2
20170087196 Pamer et al. May 2017 A1
Foreign Referenced Citations (4)
Number Date Country
2014110493 Jul 2014 WO
2016059350 Apr 2016 WO
2017079450 May 2017 WO
WO-2017079450 May 2017 WO
Non-Patent Literature Citations (1)
Entry
Collins et al., (Microbiol Spectr. Sep. 2017; 5(5): pp. 1-25). (Year: 2017).
Related Publications (1)
Number Date Country
20210069264 A1 Mar 2021 US
Provisional Applications (2)
Number Date Country
62665754 May 2018 US
62597116 Dec 2017 US