Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
SECRETAGOGUES DERIVED FROM OXALOBACTER FORMIGENES
Document Type and Number:
WIPO Patent Application WO/2015/002604
Kind Code:
A1
Abstract:
The present invention relates to a secretagogue compound derived from oxalate degrading bacteria, for use in the treatment of an oxalate related disease and/or oxalate related imbalance in a subject, wherein the administration of the secretagogue results in a reduction of urinary oxalate and/or plasma oxalate in the subject. The invention further relates to a pharmaceutical composition comprising such a secretagogue compound, a method for treating a subject suffering from an oxalate related disease, and to a method for preparing a secretagogue.

Inventors:
LINDNER ELISABETH (SE)
COWLEY HELENA (US)
COWLEY AARON (US)
Application Number:
PCT/SE2014/050848
Publication Date:
January 08, 2015
Filing Date:
July 03, 2014
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
OXTHERA INTELLECTUAL PROPERTY AB (SE)
International Classes:
A61K38/00; A61K35/74; A61P3/00; A61P3/10
Domestic Patent References:
WO2005097176A22005-10-20
WO2007070052A22007-06-21
WO2005097176A22005-10-20
WO2005123116A22005-12-29
Foreign References:
US20050232901A12005-10-20
US20070178070A12007-08-02
US20040234514A12004-11-25
US20080038246A12008-02-14
US6200562B12001-03-13
US6355242B12002-03-12
US20050232901A12005-10-20
Other References:
B. HOPPE ET AL: "Efficacy and safety of Oxalobacter formigenes to reduce urinary oxalate in primary hyperoxaluria", NEPHROLOGY DIALYSIS TRANSPLANTATION, vol. 26, no. 11, 1 November 2011 (2011-11-01), pages 3609 - 3615, XP055076654, ISSN: 0931-0509, DOI: 10.1093/ndt/gfr107
MOUNT, D.B. ET AL., PFLUGERS ARCH, vol. 447, no. 5, 2004, pages 710 - 721
SOLEIMANI, M.; XU, J., SEMINARS IN NEPHROLOGY, vol. 26, no. 5, 2006, pages 375 - 385
HATCH, M. ET AL., NIH OXALOSIS AND HYPEROXALURIA WORKSHOP, 2003
MOROZUMI, M. ET AL., KIDNEY STONES: INSIDE & OUT. HONG KONG, 2004, pages 170 - 180
HASSAN, H.A, AM J PHYSIOL CELL PHYSIOL, vol. 302, 2012, pages C46 - C58
ARONSON, P.S., JNEPHROL, vol. 23, no. S16, 2010, pages S158 - S164
CHERNOVA, M.N. ET AL., J PHYSIOL, vol. 549, 2003, pages 3
BYEON, M.K. ET AL., PROTEIN EXPR PURIF, vol. 12, 1998, pages 67
XIE Q. ET AL., AM J PHYSIOL RENAL PHYSIOL, vol. 283, 2002, pages F826
MOROZUMI, M. ET AL.: "Kidney stones: inside and out. Hong Kong", pages: 170
VENKATASUBRAMANIAN, J. ET AL., CURR OPIN GASTROENTEROL, vol. 26, 2010, pages 123 - 128
HOPPE ET AL., NEPHROL DIAL TRANSPLANT, vol. 26, 2011, pages 3609 - 3615
KELLER, A. ET AL., ANAL CHEM, vol. 74, 2002, pages 5383 - 92
NESVIZHSKII, A.I. ET AL., ANAL CHEM, vol. 75, 2003, pages 4646 - 58
JANNICK DYRLOV BENDTSEN ET AL., BMC BIOINFORMATICS, vol. 6, 2005, pages 167
THOMAS C. EVANS JR.; MING-QUN XU: "Methods in Molecular Biology", 2011, HUMANA PRESS
NICOLA CASALI; ANDREW PRESTON: "Methods in Molecular Biology"
J. BACTERIOL, vol. 178, pages 4794 - 4800
J. BIOL. CHEM., vol. 267, pages 5751 - 5754
J. BACTERIOL., vol. 174, pages 3818 - 3821
J. BIOL. CHEM., vol. 254, pages 9778 - 9785
HATCH; FREEL, AM J NEPHROL, vol. 23, 2003, pages 18 - 26
FREE ET AL., AM J PHYSIOL GASTROINTEST LIVER PHYSIOL, vol. 290, 2006, pages G719 - G728
SALIDO ET AL., PNAS USA, vol. 103, 2006, pages 18249 - 18254
WANG ET AL., AM J PHYSIOL CELL PHYSIOL, vol. 288, 2005, pages C957 - C965
Attorney, Agent or Firm:
BRANN AB (S- Stockholm, SE)
Download PDF:
Claims:
Claims

1 . A secretagogue derived from oxalate degrading bacteria, e.g. from Oxalobacter formigenes, for use in the treatment of an oxalate related disease and/or oxalate related imbalance in a subject, wherein the administration of the secretagogue results in a reduction of urinary oxalate and/or plasma oxalate in the subject.

2. The secretagogue for use according to claim 1 , wherein the oxalate related disease is selected from the group consisting of primary hyperoxaluria, hyperoxaluria, absorptive hyperoxaluria, enteric hyperoxaluria, idiopathic calcium oxalate kidney stone disease (urolithiasis), vulvodynia, oxalosis associated with end- stage renal disease, cardiac conductance disorders, inflammatory bowel disease, Crohn's disease, ulcerative colitis, and disorders/conditions caused by/associated with gastrointestinal surgery, bariatric surgery (surgery for obesity), and/or antibiotic treatment.

3. The secretagogue for use according to claim 1 or 2, comprising an amino acid sequence, which has a secretagogue activity and at least 85% identity with an amino acid sequence according to any one of SEQ ID NO: 1 -19.

4. The secretagogue for use according to claim 3, comprising an amino acid sequence which has a secretagogue activity and at least 85% identity, with an amino acid sequence according to any one of SEQ ID NO: 3, 4, 6, 13 and 19.

5. The secretagogue for use according to any one of claims 1 -4, which has been recombinantly expressed in a suitable organism or extracted from conditioned media. 6. A pharmaceutical composition comprising one or more secretagogues as defined in any one of claims 1 -5, optionally further comprising one or more oxalate degrading bacteria, oxalate degrading enzymes, enzymes involved in oxalate metabolism, cofactors, substrates, or combinations thereof.

7. The composition of claim 6, wherein the composition is formulated to have an enteral, parenteral or topical route of administration. 8. A method for treating a subject suffering from an oxalate related disease, comprising administering to said subject a secretagogue derived from oxalate degrading bacteria, wherein administration of the secretagogue results in a reduction of urinary and/or plasma oxalate in the subject. 9. The method according to claim 8, wherein the secretagogue comprises an amino acid sequence, which has a secretagogue activity and at least 85% identity with an amino acid sequence according to any one of SEQ ID NO: 1 -19.

10. The method according to claim 9, wherein the secretagogue comprises an amino acid sequence, which has a secretagogue activity and at least 85% identity with an amino acid sequence according to any one of SEQ ID NO: 3, 4, 6, 13 and 19.

1 1 . The method according to any one of claims 8-10, further comprising administering to said subject one or more oxalate degrading bacteria, oxalate degrading enzymes, enzymes involved in oxalate metabolism, cofactors, substrates, or combinations thereof.

12. A method according to any one of claims 8-1 1 , wherein the oxalate related disease is selected from the group consisting of primary hyperoxaluria, hyperoxaluria, absorptive hyperoxaluria, enteric hyperoxaluria, idiopathic calcium oxalate kidney stone disease (urolithiasis), vulvodynia, oxalosis associated with end- stage renal disease, cardiac conductance disorders, inflammatory bowel disease, Crohn's disease, ulcerative colitis, and disorders/conditions caused by/associated with gastrointestinal surgery, bariatric surgery (surgery for obesity), and/or antibiotic treatment.

13. A method for preparing a secretagogue as defined in any one of claims 1 -5, comprising: i) inoculating oxalate degrading bacteria, e.g. Oxalobacter formigenes, into a temperature equilibrated selective growth medium, specific for microbes utilizing oxalate as sole carbon source, said growth medium including oxalate as carbon source, necessary trace metals and amino acids under anaerobic conditions;

ii) transferring cell culture between containers under anaerobic conditions, such as between anaerobic bottles or between an anaerobic bottle and a fermenter, for example by using bottle ports and a sterile gas pressure to displace liquid through a non air-permeable tubing;

iii) harvesting a cell suspension, and processing of said cell suspension by using for example tangential flow filtration to separate whole cells and debris from the suspension, thereby obtaining a cell-free permeate;

iv) filtering the cell-free permeate to obtain a retentate, for example by using hollow fiber which selectively removes compounds of < 10k Da and peptides of reduced interest.

v) isolating one or more secretagogues from the retentate by using for

example liquid chromatography, electrophoresis, precipitation,

ultracentrifugation and/or concentrating spin columns.

Description:
SECRETAGOGUES DERIVED FROM OXALOBACTER FORMIGENES Field of the invention

The present invention relates to an identified secretagogue compound derived from oxalate degrading bacteria. The invention further relates to a method of treating a subject in need, wherein the method comprises administering a pharmaceutical composition comprising one or more secretagogues derived from oxalate degrading bacteria together with or without one or more oxalate degrading bacteria, oxalate degrading enzyme(s), enzyme(s) involved in oxalate metabolism, cofactors, substrates, or combinations thereof, to reduce the presence of oxalate in a subject. The invention also encompasses the process of manufacturing the secretagogue in sufficient quantities for identification and proposed use. Background of the invention

Oxalate is the salt of a weak acid and is a metabolic end product that is excreted through the kidneys. A disrupted oxalate homeostasis in humans and animals can cause severe conditions due to the tendency of oxalate to damage the renal parenchymal cells both as free oxalate and as calcium-oxalate crystals; thus, in many cases causing irreversible damage. Oxalate homeostasis is severely disrupted due to inborn errors such as Primary Hyperoxaluria (PH).

Primary Hyperoxaluria (PH) is a rare autosomal recessive inborn error of the glyoxylate metabolism, with an incidence rate of 0.1 -0.2 per million. PH type I is caused by deficient or absent activity of liver specific peroxisomal alanine/glyoxylate aminotransferase (AGT). In some patients, enzyme is present but mis-targeted to mitochondria where it is metabolically inactive.

PH type II occurs as a result of deficient glyoxylate reductase / hydroxy pyruvate reductase (GRHPR) enzyme activity. Both types of PH are characterized by severe hyperoxaluria that is present from birth. Patients experience recurrent calcium-oxalate urolithiasis, nephrocalcinosis and progressive renal failure. There are no approved therapies to treat the enzyme deficiency or enzyme dysfunction in either PH type I or PH type II. Current therapies for PH are directed to decrease oxalate production or to increase the urinary solubility of calcium oxalate in order to preserve renal function. Patients are given treatment with magnesium, citrate, and orthophosphate supplementation to increase the urinary solubility of calcium oxalate. Pyridoxine is a co-factor of the deficient AGT and pharmacological doses of pyridoxine may reduce urinary oxalate levels in a minority of patients with PH I. Eventually the only curative therapy is a combined kidney and liver transplantation.

Secondary Hyperoxaluria includes oxalate-related conditions such as, but not limited to, hyperoxaluria, absorptive hyperoxaluria, enteric hyperoxaluria, idiopathic calcium oxalate kidney stone disease (urolithiasis), vulvodynia, oxalosis associated with end-stage renal disease, cardiac conductance disorders, inflammatory bowel disease, Crohn's disease, ulcerative colitis, and disorders/conditions caused by/associated with gastrointestinal surgery, bariatric surgery (surgery for obesity), and/or antibiotic treatment. Kidney/urinary tract stone disease (urolithiasis) is a major health problem throughout the world. Most of the stones associated with urolithiasis are composed of calcium oxalate alone or calcium oxalate plus calcium phosphate.

Kidney or urinary tract stone disease occurs in as many as 12 % of the population in Western countries and about 70% of these stones are composed of calcium oxalate or of calcium oxalate plus calcium phosphate. Some individuals (e.g. patients with intestinal disease such as Crohn's disease, inflammatory bowel disease, or steatorrhea and also patients that have undergone jejunoileal or Roux-en-Y bypass surgery) absorb more of the oxalate in their diets than do others. For these individuals, the incidence of oxalate urolithiasis is markedly increased.

Oxalate homeostasis is a complex process, not yet completely resolved, including many different bodily organs and mechanisms. The mammalian kidneys as well as intestinal tract act as excretion avenues, consequently lowering the oxalate concentrations within the body. However, oxalate excretion through the kidneys pose risk of toxicological effects on the renal parenchymal cells and of crystal formation in the form of kidney stones. The mammalian intestinal tract thus plays a major role in oxalate control both due to passive and active oxalate transport (Hatch and Freel, 2004; Freel et al., 2006) as well as symbiotic relationships with colonizing oxalate-degrading bacteria.

It has previously been reported that a highly concentrated lyophilized powder containing Oxalobacter formigenes (O. formigenes), a non-pathogenic, obligate anaerobic bacterium that utilizes oxalic acid as its sole source of energy, can be used to decrease oxalate in plasma and urine (US Patent No.: 6,200,562 B1 ; US Patent No.: 6,355,242 B1 ). The mechanism for oxalate degradation has been characterized and involves three unique proteins, an oxalate:formate membrane transporter, oxalyl-CoA decarboxylase (OXC), and formyl-CoA transferase (FRC). A very high expression of oxalate degrading proteins and their unique kinetic properties makes O. formigenes one of the most efficient oxalate degrading systems known.

O. formigenes is part of a healthy intestinal microbiota in vertebrates and as such partakes significantly in the oxalate homeostasis process. Mechanistically, O. formigenes increases degradation of oxalate in the gastrointestinal tract, creates a suitable transepithelial gradient, and promotes passive enteric elimination of oxalate. For the skilled artisan the importance of a balanced intestinal microbiota is well known, further, it is also known that developments in the field of microbiology have demonstrated numerous cases of bacterial secretagogue influences in the human intestine. To name a few examples; Vibrio Cholerae and enterotoxigenic Escherichia coli produce secretagogue compounds, which, briefly described, cause an efflux of ions and subsequently fluids into the intestinal lumen with the result of diarrheas (Flores, J., Sharp, G.W., 1976; Field, M., Graf, L.H., et al., 1978). Other intestinal inhabitants induce a hyper-secretion of mucin by secretagogue action (e.g. Navarro-Garcia, F., et al., 2010, Caballero-Franco, C, et al., 2006). Recently the SLC26 (solute-linked carrier) gene family was identified (Mount, D.B., et al., Pflugers Arch. 2004; 447 (5):710-721 ; Soleimani, M., Xu, J., Seminars in nephrology. 2006; 26 (5):375-385). This gene family encodes for structurally related anion transporters that have a measurable oxalate affinity and are found in the Gl tract: SLC26A1 (SAT1 ), SCL26A2 (DTDST), SLC26A3 (DRA), SLC26A6 (PAT1 or CFEX), SLC26A7, and SCL26A9. In addition, SLC26A1 and SCL26A2 have been observed in post-confluent and confluent Caco-2 monolayers, respectively (Hatch, M., et al., NIH Oxalosis and Hyperoxaluria Workshop, 2003; Morozumi, M., et al., Kidney Stones: Inside & Out. Hong Kong: 2004, Pp. 170-180). Despite these recent advancement many questions remain on the complex balance of counter ions over the epithelial membrane, the role of different oxalate transporters and what impact they have on hyperoxaluric conditions (Hassan, H.A., Am J Physiol Cell Physiol, 302: C46- C58, 2012; Aronson, P.S., JNephrol 2010; 23 (S16): S158-S164).

Many of the transporters with oxalate affinity also demonstrate affinity for other substrates and often are linked to acid-base balances within the cells; to name a few examples: DRA expressed in Xenopus oocytes is a CI " base exchanger (Chernova, M.N. et al., J Physiol, 549:3, 2003) and studies suggests S0 4 2" is another substrate (Byeon, M.K. et al., Protein Expr Purif, 12: 67, 1998); mouse PAT-1 expressed in Xenopus exhibits a variety of affinities to CI HCO 3 " , CIOx 2" , S0 2" Ox 2" (Xie Q. et al., Am J Physiol Renal Physiol 283: F826, 2002); and the SLC26A4 gene encodes a CI " formate exchanger (Morozumi, M. et al. In: Gohel MDI, Au DWT (eds) Kidney stones: inside and out. Hong Kong, p. 170). Many other transporters, without affinity for oxalate, have substrates in common with the oxalate transporting proteins, for example: Na + /K + ATPase, Na + /H + exchangers (NHEs), Na + -K + -2CI " cotransporters (NKCC), and basolateral K + channels and apical CI " channels, such as the cystic fibrosis transmembrane conductance regulator (CFTR) (Venkatasubramanian, J. et al., Curr Opin Gastroenterol, 26: 123-128, 2010). The regulation of Na + , CI " , K + , HC0 3 " is highly coordinated in the intestinal tract, as it also dictates movement of water, through interactions and regulations of this multitude of transporters outlined. Several oxalate-reducing pharmaceutical formulations have been described in the art, such as in WO2007070052 A2, Hoppe et al. (Nephrol Dial Transplant, 201 1 , 26: 3609-3615), and US 20050232901 A1 . Among these are enteric coated or other compositions comprising oxalate degrading bacteria or enzymes that have been suggested as a means for reducing oxalate concentrations. An objective with such a treatment is for the patients to get lowered or normalized urinary oxalate levels.

Further, WO2005097176 A2 describes compositions comprising Oxalobacter formigenes, which can be viable and/or a lysate thereof. The compositions are useful for treating an animal subject suffering from renal failure.

Summary of the invention

The invention described herein pertains to a secretagogue component, or a secretagogue compound, derived from an oxalate degrading bacteria, which exerts a direct or indirect effect in the intestinal tract of a mammal. The effect relates to an exchange of oxalate across the intestinal epithelia in a mammal via passive flux of oxalate and/or a promotion of active transport. This invention further describes the manufacturing process for producing sufficient amounts of this secretagogue component for identification, analysis and proposed use in hyperoxaluria therapy.

Due to the complex multi-component interactions of ions and transporters in the intestinal tract, the recognition of multiple substrates per transporter, and passive and active flux dependence on different ion equilibria, it is important to point out that a secretagogue effect may be indirect in the sense that an identified secretagogue compound may exert its effect through the shift of nominal ion balances and equilibria and thus altering the flux of the oxalate ion. O. formigenes is a naturally occurring oxalate degrading bacterium, present in the Gl-tracts of vertebrates, where it takes part in a complex symbiosis with the mammal host. Studies with O. formigenes support a promotion of enteric elimination of oxalate by inducing the trans epithelium flux of oxalate from a parenteral site to the intestinal lumen, when inhabiting the intestinal tract of rats and mice. Further, 0. formigenes administration to human subjects with end- stage renal disease demonstrates a significant lowering of plasma oxalate (WO20051231 16A2). These findings supported the inventors in their theorized concept of secreted compounds affecting the intestinal epithelia cell in a way favorable for O. formigenes survival in the intestinal tract of humans as well as rodents.

A large-scale process of manufacturing and using a potential secretagogue was explored, which led to the identification of protein compounds with a proposed secretagogue effect and the invention as described here. As many of the proteins are present at low amounts, the large-scale manufacturing process and subsequent purification and isolation made identification possible. To the best of our (present inventors) knowledge, there are currently no identified secretagogue compounds described for O. formigenes in the art and this is the first attempt of isolating and identifying them for use in hyperoxaluria therapy.

Based on investigations by the present inventors, compounds produced by 0. formigenes, proposed to cause an induced secretory flux, have been manufactured at sufficient scale to be identified. The compounds (secretagogues) have been purified and characterized.

Thus, the present invention further provides a method of isolating a secretagogue in sufficient quantities for identification and proposed use in hyperoxaluria therapy. The isolation and enrichment of the secretagogue can be combined with the isolation of oxalate degrading bacteria; hence, resulting in one process pertaining to both harvest of oxalate degrading bacteria as well as an enrichment of compounds secreted by the oxalate degrading bacteria during fermentation.

The secretagogue compounds from 0. formigenes, which should be produced in sufficient quantities for identification and use in hyperoxaluria therapy, are identified as one or more secretagogue proteins and peptides that individually or together exert a direct or indirect effect on oxalate flux in the intestinal tract of a mammal. The identified compounds are secreted out of the bacterial cell, or maintained intra-cellularly and released into culture media upon cell death. Cell death is a continuous process during cell culture growth; hence, proteins or other compounds, identified from cell-free culture media, with a proposed secretagogue effect, may not inherently be secreted compounds.

The secretagogue compounds may exert their combined or individual effects in the intestinal tract in a reaction necessitating a co-factor. Therefore, a combination of one or more co-factors (for example: NAD + , NADP + , FAD, CoA, ATP, ADP among others) and one or more secretagogues, if necessary to commence the secretagogue positive effect in the intestinal tract of a mammal, is also encompassed by this invention. The secretagogue compounds may exert their combined or individual secretagogue effects indirectly by altering the nominal fluxes of other compounds for example, ions and small organic compounds and thereby cause a change in oxalate flux. Therefore, an indirect effect of the secretagogue compounds mentioned herein, which indirectly alters the oxalate flux through a change in flux of ions or small organic compounds, for example, is also encompassed by this invention.

The effect of the secretagogue compounds may be improved in combination with oxalate degrading bacteria, oxalate degrading enzyme(s), enzyme(s) involved in oxalate metabolism, cofactors, substrates, or combinations thereof. Therefore, a combination of one or more oxalate degrading bacteria, oxalate degrading enzyme(s), enzyme(s) involved in oxalate metabolism, cofactors, substrates, and one or more secretagogues, if necessary to commence the secretagogue positive effect in the intestinal tract of a mammal, is also encompassed by this invention.

Disclosed herein is also a method of removing exogenous and/or endogenous oxalate through enteric elimination by administering the secretagogue itself, or together with oxalate degrading enzymes, oxalate degrading bacteria, enzyme(s) involved in oxalate metabolism, cofactor(s), substrate(s), to a mammal subject in need of oxalate removal. The combined administration of one or more secretagogues and oxalate degrading bacteria, oxalate degrading enzyme(s), enzyme(s) involved in oxalate metabolism, cofactor(s), substrate(s), ensures both a stimuli of the oxalate flux, to increase oxalate transport in the direction of the intestinal lumen, as well as increased degradation of oxalate throughout the gastrointestinal tract.

Consequently, the present invention relates to pharmaceutical compositions comprising one or more identified components or secretagogues, derived from oxalate degrading bacteria, recombinantly expressed or extracted from conditioned media, administered to patients with or without one or more oxalate degrading bacteria, oxalate degrading enzymes, enzymes involved in oxalate metabolism, cofactors, substrates, or combinations thereof.

The pharmaceutical composition comprises an effective amount of a secretagogue. An effective amount of a secretagogue comprises an amount that will reduce a portion of the oxalate present systemically by secretion into the intestinal tract. The amount that can be used in a single dose composition, alone or in combination with oxalate-degrading bacteria and/or enzymes, can range from about 10pg to 1000,000Mg, from about l OOpg to 100,000Mg, from about 1 mg to 100mg, and all ranges encompassed therein.

This pharmaceutical composition may be resistant to degradation by gastric acid. The composition can be, for example, in the form of a tablet, capsule or bead, optionally provided with an enteric coating or other means for providing resistance to degradation in the stomach and the upper small intestine.

The invention further relates to a method of treating a subject in need, wherein the method comprises administering an effective amount of a secretagogue compound, derived from or produced by oxalate degrading bacteria, or a pharmaceutical composition comprising such a secretagogue compound administered with or without one or more oxalate degrading bacteria, oxalate degrading enzymes, enzymes involved in oxalate metabolism, cofactors, substrates, or combinations thereof, to thereby reduce the presence of oxalate in the subject. An effective amount of a secretagogue comprises an amount that will reduce a portion of the oxalate present systemically by secretion into the intestinal tract. The amount that can be used in a single dose composition, alone or in combination with oxalate-degrading bacteria and/or enzymes, can range from about 10pg to 1000,000Mg, from about l OOpg to 100,000Mg, from about 1 mg to 100mg, and all ranges encompassed therein.

Thus, the present disclosure provides a secretagogue (defined more in detail below) derived from oxalate degrading bacteria, e.g. from Oxalobacter formigenes, for use in the treatment of an oxalate related disease and/or oxalate related imbalance in a subject (defined more in detail below), wherein the administration of the secretagogue results in a reduction of urinary oxalate and/or plasma oxalate in the subject.

According to the present disclosure, the oxalate related disease may be selected from the group consisting of primary hyperoxaluria, hyperoxaluria, absorptive hyperoxaluria, enteric hyperoxaluria, idiopathic calcium oxalate kidney stone disease (urolithiasis), vulvodynia, oxalosis associated with end- stage renal disease, cardiac conductance disorders, inflammatory bowel disease, Crohn's disease, ulcerative colitis, and disorders/conditions caused by/associated with gastrointestinal surgery, bariatric surgery (surgery for obesity), and/or antibiotic treatment.

In accordance with the present disclosure, the secretagogue may comprise an amino acid sequence, which has a secretagogue activity and at least 85%, such as 90%, 95%, 96%, 97%, 98%, 99%, or 100% identity with an amino acid sequence according to any one of SEQ ID NO: 1 -19. The secretagogue may be recombinantly expressed in a suitable organism or extracted from conditioned media.

The present disclosure further provides a pharmaceutical composition comprising one or more secretagogues as defined above, optionally further comprising one or more oxalate degrading bacteria, oxalate degrading enzymes (defined more in detail below), enzymes involved in oxalate metabolism (defined more in detail below), cofactors (defined more in detail below), substrates (defined more in detail below), or combinations thereof.

The pharmaceutical composition may be formulated to have an enteral, parenteral or topical route of administration.

The present disclosure further provides a method for treating a subject suffering from an oxalate related disease, comprising administering to said subject a secretagogue derived from oxalate degrading bacteria, wherein administration of the secretagogue results in a reduction of urinary and/or plasma oxalate in the subject. In said method, the secretagogue may comprise an amino acid sequence, which has a secretagogue activity and at least 85%, such as 90%, 95%, 96%, 97%, 98%, 99%, or 100% identity with an amino acid sequence according to any one of SEQ ID NO: 1 -19. The method may further comprise administering to said subject one or more oxalate degrading bacteria, oxalate degrading enzymes, enzymes involved in oxalate metabolism, cofactors, substrates, or combinations thereof.

In said method, the oxalate related disease is selected from the group consisting of primary hyperoxaluria, hyperoxaluria, absorptive hyperoxaluria, enteric hyperoxaluria, idiopathic calcium oxalate kidney stone disease (urolithiasis), vulvodynia, oxalosis associated with end-stage renal disease, cardiac conductance disorders, inflammatory bowel disease, Crohn's disease, ulcerative colitis, and disorders/conditions caused by/associated with gastrointestinal surgery, bariatric surgery (surgery for obesity), and/or antibiotic treatment.

The present disclosure also provides a method for preparing a secretagogue as defined above, comprising:

i) inoculating oxalate degrading bacteria, e.g. Oxalobacter formigenes, into a temperature equilibrated selective growth medium, specific for microbes utilizing oxalate as sole carbon source, said growth medium including oxalate as carbon source, necessary trace metals and amino acids under anaerobic conditions;

ii) transferring cell culture between containers under anaerobic

conditions, such as between anaerobic bottles or between an anaerobic bottle and a fermenter, for example by using bottle ports and a sterile gas pressure to displace liquid through a non air- permeable tubing;

iii) harvesting a cell suspension, and processing of said cell suspension by using for example tangential flow filtration to separate whole cells and debris from the suspension, thereby obtaining a cell-free permeate;

iv) filtering the cell-free permeate to obtain a retentate, for example by using hollow fiber which selectively removes compounds of < 10k Da and peptides of reduced interest;

v) isolating one or more secretagogues from the retentate by using for example liquid chromatography, electrophoresis, precipitation, ultracentrifugation and/or concentrating spin columns.

Particularly, the present disclosure provides a secretagogue, derived from oxalate degrading bacteria, e.g. from Oxalobacter formigenes, for use in the treatment of an oxalate related disease and/or oxalate related imbalance in a subject, wherein the administration of the secretagogue results in a reduction of urinary oxalate and/or plasma oxalate in the subject, wherein the secretagogue compound comprises an amino acid sequence which has a secretagogue activity and at least 85%, such as 90%, 95%, 96%, 97%, 98%, 99%, or 100% identity, with an amino acid sequence according to any one of SEQ ID NO: 3, 4, 6, 13 and 19. Pharmaceutical compositions comprising such a secretagogue are also provided. Further, the present disclosure provides a method for treating a subject suffering from an oxalate related disease, comprising administering to said subject a secretagogue as defined above, as well as a method for preparing such a secretagogue.

Brief description of the drawings

Fig. 1 A-E. SDS-PAGE Gels (left) and Western Blots (right) for recombinantly expressed Oxalobacter formigenes potential secretagogue proteins. Bovine Serum Albumin (2ug) was run as control (Lane 1 ). Target protein on SDS- PAGE gel is presented in Lane 2, and target protein on Western blot is presented in Lane 3. Ladder weights are shown to the right of respective analysis image. M1 = protein marker Genscript Cat. No. M00505. M2 = protein marker Genscript Cat. No. MM0908.

Fig. 1A: SEQ ID No. 3 expressed protein analysis.

Fig. 1 B: SEQ ID No. 4 expressed protein analysis.

Fig. 1 C: SEQ ID No. 6 expressed protein analysis.

Fig. 1 D: SEQ ID No. 13 expressed protein analysis.

Fig. 1 E: SEQ ID No. 19 expressed protein analysis.

Figure 2. Bar chart showing flux of labeled oxalate (C14-oxalate) during first flux period, 0-30 minutes. Striped bars depict mucosal-to-serosal flux (M-S flux), and filled bars depict serosal-to-mucosal flux (S-M flux). On the horizontal axis, G1 -G3 denote the three groups of treatment, G = group. Error bars = ± SEM (standard error of mean). The vertical axis shows 14C-oxalate in umol.cm2.h.

Figure 3. Bar chart showing flux of labeled oxalate (C14-oxalate) during second flux period, 30-60 minutes. Striped bars depict mucosal-to-serosal flux (M-S flux), and filled bars depict serosal-to-mucosal flux (S-M flux). On the horizontal axis, G1 -G3 denote the three groups of treatment, G = group. Error bars = ± SEM (standard error of mean). The vertical axis shows 14C-oxalate in umol.cm2.h.

Definitions

All terms used in the present text are intended to have the meaning usually given to them in the art. For the sake of clarity, som terms are also defined below.

Secretagogue

A secretagogue is a substance, component or compound that directly or indirectly promotes secretion of another substance, component or compound. Secretagogues can be peptides, hormones, proteins or small molecules. Herein the word secretagogue will be used for identified protein compounds, which have a proposed secretagogue effect in the body of a mammal.

The secretagogues described herein were isolated from the supernatant of an O. formigenes cell culture suspension and as such can originate from a secretion process or a cell death and lysis promoted release. The secreted compounds and proposed secretagogues were characterised as proteins with a molecular weight ranging from 8.8 - 98.8 kDa, with > 50% of compounds being in the span of 30 - 50 kDa. Of the nineteen proteins, thirteen were enzymes and the remaining six were proteins without known catalysing function. Of these six proteins, one was defined as "conserved hypothetical protein" i.e. this protein is found in organisms from several phylogenetic lineages but have not been functionally characterized. Another of the six proteins was defined as "predicted protein". This is used for entries without evidence at protein, transcript, or homology levels. The amino acid sequences of the identified proposed secretagogues are presented in Example 4, with sequence IDs: SEQ ID No: 1 , SEQ ID No: 2, SEQ ID No:3, SEQ ID No:4, etc., to the last one, i.e. SEQ ID No: 19. Five proteins, corresponding to amino acid sequence IDs: SEQ ID No: 3, 4, 6, 13 and 19, were recombinantly expressed in E.coli and screened for secretagogue activity of labeled oxalate over rat intestinal tissue in Ussing chambers. All recombinant proteins expressed demonstrated a reduced overall oxalate flux as compared to control. One protein in particular, of SEQ ID No. 3, had an effect that increased net secretion of labeled oxalate over rat distal intestinal tissue.

The secreted compounds are also described herein in terms of stability (Instability index II), thermostability (for globular proteins: aliphatic index) and solubility (GRAVY, Grand Average of Hydropathy), see Example 5. Three proteins were predicted as instable according to index (score > 40): serine hydroxymethyltransferase, acyl carrier protein, riboflavin synthase subunit beta. In addition, the amino acid sequences of the secretagogues were investigated for potential signal peptide cleavage sites (see Example 6).

Conditioned medium

A conditioned medium is a growth medium that has been used to grow a particular organism. The term conditioned refers to the fact that the organism has utilized components of the original medium composition for growth and metabolism, as well as released products of its metabolism and gene expression. A conditioned medium also contains and refers to products of metabolism and gene expression released upon the continuous cell death and lysis inherent to a culture growth. Conditioned media may also be referred to as cell-free culture suspensions. The conditioned medium described herein originates from an unconditioned medium with the composition described in Example 1. This unconditioned medium is a specialty medium developed specifically for the unique purpose of O. formigenes growth, and thus the concomitant secretagogue preparation as well, and contains oxalate as main source of carbon.

Oxalate-degrading enzyme The term "oxalate-degrading enzyme" shall be construed as any enzyme that is capable of reducing oxalate, and includes oxalate decarboxylase, oxalate oxidase, formyl-CoA transferase and oxalyl-CoA decarboxylase. It may reduce oxalate per se and/or it may function in an oxalate reduction pathway. In this context the term "oxalate" encompasses oxalic acid and/or any salts thereof.

Enzyme involved in oxalate metabolism

The term "enzyme involved in oxalate metabolism" shall be construed as any enzyme functioning in a pathway relating to oxalate metabolism, and includes alanine-glyoxylate aminotransferase (AGT), glyoxylate reductase (GR) and 4- hydroxy-2-oxoglutarate aldolase (HOGA).

Co-factor

The term "co-factor" shall be construed as a non-enzymatic compound necessary for the activity of an enzyme, and includes vitamin B 6 , NAD + , NADP + , FAD, CoA, ATP and AD P.

Substrate

The term "substrate" shall be construed as the ingoing compound of an enzyme catalyzed reaction, and includes oxalate, glyoxylate, and 4-hydroxy-2- oxoglutarate, among others.

Oxalate-related disease and/or oxalate related imbalance

The term "oxalate-related disease and/or oxalate related imbalance" shall be construed as diseases that are caused or realized by an imbalance in systemic oxalate levels., and includes primary hyperoxaluria, hyperoxaluria, absorptive hyperoxaluria, enteric hyperoxaluria, idiopathic calcium oxalate kidney stone disease (urolithiasis), vulvodynia, oxalosis associated with end-stage renal disease, cardiac conductance disorders, inflammatory bowel disease, Crohn's disease, ulcerative colitis, and disorders/conditions caused by/associated with gastrointestinal surgery, bariatric surgery (surgery for obesity), and/or antibiotic treatment. Detailed disclosure of the invention

The invention described herein relates to one or more protein secretagogues identified from a cell-free culture suspension originating from an 0. formigenes culture. The identified protein secretagogues exert an effect on the oxalate transport in the intestinal tract of an animal or human. The proposed effect, described above, exerted by the identified protein secretagogues is a result of an impact from one identified protein secretagogue or from a combination of two identified secretagogues, three identified secretagogues, four identified secretagogues, five identified secretagogues or six or more identified secretagogues. The identified compounds may exert a positive effect in the intestine of animals or humans, which will positively influence critical conditions as described herein, when administered as described herein.

The effect exerted in the intestinal tract of an animal or human by the action of one or more O. formigenes derived secretagogues may require a co-factor of sorts. Therefore, a potential co-factor, which ensures a positive effect on oxalate flux in the intestine in combination with one or more secretagogues, should also be considered encompassed by this invention. The co-factor which may be necessary for the secretagogue to exert a positive action within the intestinal tract of an animal or human, as described above, includes but is not limited to: NAD + , NADP + , FAD, CoA, ATP, ADP.

The effect exerted in the intestinal tract of an animal or human by the action of one or more O. formigenes derived secretagogues may be improved with the inclusion of one or more oxalate degrading bacteria, oxalate degrading enzyme(s), enzyme(s) involved in oxalate metabolism, cofactors, substrates, or combinations thereof. Therefore, one or more oxalate degrading bacteria, oxalate degrading enzyme(s), enzyme(s) involved in oxalate metabolism, cofactor(s), substrate(s), or combinations thereof, which ensures a positive effect on oxalate flux in the intestine in combination with one or more secretagogues, should also be considered encompassed by this invention. The oxalate degrading bacterium that may improve the effect of the secretagogue(s) is preferably 0. formigenes.

The oxalate-degrading enzyme(s) that may improve the overall effect in combination with one or more secretagogues include but is not limited to oxalate decarboxylase, oxalate oxidase, formyl-CoA transferase and oxalyl-CoA decarboxylase.

The enzyme(s) involved in oxalate metabolism that may improve the overall effect in combination with one or more secretagogues include but are not limited to alanine-glyoxylate aminotransferase (AGT), glyoxylate reductase (GR) and 4- hydroxy-2-oxoglutarate aldolase (HOGA).

The co-factors that are referred to above include but are not limited to vitamin B 6 , NAD + , NADP + , FAD, CoA, ATP and ADP. Substrates referred to in the pharmaceutical composition include but is not limited to oxalate, glyoxylate, 4- hydroxy-2-oxoglutarate, among others.

The secretagogue compounds may exert their combined or individual secretagogue effects indirectly by altering the nominal fluxes of other compounds for example, ions and small organic compounds and thereby cause a change in oxalate flux. For example, it is known in the field that the transporters with oxalate affinity also demonstrate affinity for other small compounds or ions. For sake of description and without limiting the scope of this invention, those ions and small compounds include but are not limited to; carbonate, sulphate, chloride, sodium and formate to mention a few. Therefore, by altering the balances of these ions and small compounds, the flux of oxalate could be altered. Thus, an indirect effect of the secretagogue compounds mentioned herein, that indirectly alters the oxalate flux through a change in flux of ions or small organic compounds, for example, is also encompassed by this invention. The identified protein secretagogue that exerts a direct or indirect effect, individually or in combination with other identified secretagogue(s), oxalate degrading bacteria, enzyme(s), co-factor(s), and/or substrates, on the oxalate transport in the intestinal tract of animals or humans includes: tartronic semialdehyde reductase, cysteine synthase A, acyl carrier protein, predicted protein (accession number: gi|237749499), methionine adenosyltransferase 1 , YgiW protein, riboflavin synthase subunit beta, alkyl hydroperoxide reductase/thiol specific antioxidant/mal allergen, phospho-2-dehydro-3- deoxyheptonate aldolase, elongation factor Tu, s-adenosylhomocysteine hydrolase, conserved hypothetical protein (accession number: gi|237747886), diaminopimelate dehydrogenase, serine hydroxymethyltransferase, aspartate- semialdehyde dehydrogenase, malic enzyme, aconitate hydratase 1 , hsp70-like protein. The secretagogue according to the present invention is a polypeptide comprising an amino acid sequence, which has a secretagogue activity and at least 85% identity, such as 90%, 95%, 96%, 97%, 98%, 99%, or 100% identity, with an amino acid sequence according to any one of SEQ ID NO: 1 -19.

Without limiting the scope of this invention, it is noted that after analysis of the amino acid sequence, the YgiW protein (accession number: gi|237748090) was predicted to have a signal peptide cleavage site between amino acid number 28 and 29 (ASA-QY). A signal peptide on a protein has the purpose of targeting the protein to a particular location inside (a certain organell) or outside of the cell. Secreted proteins and membrane proteins are also localized using signal peptides.

In addition, without affecting the scope of this invention, it has been noted that the calculated pi of one of the proteins identified (YgiW: accession number: gi|237748090) was as high as 9.33. This pi means that the protein has a net positive charge at physiological pH. It is well known that most bio surfaces carry a net negative charge; hence, a high net positive charge of a protein supports a function, which is interactive with either a bio surface or a bio membrane. It is likely that anything supporting interaction of a potential secretagogue compount with a bio surface facilitates the process of promoting secretion by the same.

The proposed effect exerted by the identified protein secretagogues, individually or in combination with other identified secretagogue(s), bacteria, enzyme(s) co- factors), and/or substrates, on the oxalate transport in the intestinal tract of animals or humans, includes but is not limited to: a direct or indirect interaction with a membrane protein or membrane transporter, an indirect interaction with an expression factor within the epithelial cells or the genome of the epithelial cells. The membrane transporters referred to above may include but are not limited to SLC26A1 (SAT1 ), SCL26A2 (DTDST), SLC26A3 (DRA), SLC26A6 (PAT1 or CFEX), SLC26A7, and SCL26A9. In addition, the interaction may be any modulation i.e. any activity including but not limited to: increase, enhance, agonize, promote, decrease, reduce, suppress, block, or antagonize.

Method of isolation

The present invention provides a method of producing secretagogues. The invention further provides a method of isolating the secretagogues with a low product loss. The production of the secretagogues involves a seed train using vials, anaerobic bottles or fermenters in any combination, in one, two, three, four or more steps. The medium composition, growth conditions, fermentation time, harvest and filtration steps used are all described in Examples 1 and 2 and are covered under this present invention for the production of secretagogues. Any other addition, subtraction or general alteration as reasonable for anyone skilled in the art also falls under this invention.

The fermentation process for producing a secretagogue may involve modifications to induce the secretagogue production by O. formigenes cells by using different means of induction, for example, varied or lower concentrations of oxalate, varied exposure to or concentration of formate, and/or other forms of stress induction. Such an induction modification to the process, or other means of induction to promote secretion of an active secretagogue, as reasonable to those skilled in the art, are considered to be encompassed by this invention. The isolation of secretagogues preferably uses Tangential Flow Filtration (TFF) and hollow fiber filtration steps but may include other commonly used methods of isolation of proteinacious species as reasonable for those skilled in the art.

The isolation and enrichment of the secretagogue may be combined with the isolation of oxalate degrading bacteria; hence, resulting in one process pertaining to both harvest of oxalate degrading bacteria as well as an enrichment of compounds secreted by the oxalate degrading bacteria during fermentation. This process includes but is not limited to methods such as centrifugation, TFF, hollow fiber and other methods of cell collection, filtration and concentration as reasonable for those skilled in the art.

The identified secretagogue may also be expressed recombinantly. The recombinantly expressed proteins may originate from a naturally extracted or synthesized gene sequence having at least 85% sequence identity to the sequences of SEQ ID No:s 1 -19. Protein homologs and variants include but are not limited to: polymorphic variants and natural or artificial mutants, modified polypeptides in which one or more residue is modified, and mutants comprising one or more modified residues. Mutations include but are not limited to truncation, deletion, substitution or addition mutations of nucleic acids.

The recombinant enzymes may be expressed in a wide variety of hosts, known to those skilled in the art of protein expression, including but not limited to: E. coli, Lactobacillus spp, Bacillus spp etc.

For a recombinant production of the enzyme or protein the host should comprise a construct in the form of a plasm id, vector, phagemid, or transcription or expression cassette that comprises the enzyme or protein or a functional fragment thereof. A variety of constructs are available, including constructs, which are maintained in single copy or multiple copy. Many recombinant expression systems, components, and reagents for recombinant expression are commercially available, for example from Invitrogen Corporation (Carlsbad, Calif.); U.S. Biological (Swampscott, Mass.); BD Biosciences Pharmingen (San Diego, Calif.): Novagen (Madison, Wis.); Stratagene (La Jolla, Calif.); and Deutsche Sammlung von Mikroorganismen and Zellkulturen GmbH (DSMZ), (Braunschweigh, Germany).

A heterologous promotor, including a constitutive and/or inducible promotor, optionally controls recombinant expression of the proteins. Promotors such as, for example, T7 or other promotors, as suitable for the host, and which are well- known for those skilled in the art. The promotor may also originate from the Oxalobacter genus.

The enzyme's or protein's recombinant nucleic acid sequence may include nucleic acids for purposes additional to the expression of the protein, including but not limited to for purification purposes, folding purposes etc. Examples of those are: secretion sequences, signal sequences, linkers, expression control elements, affinity tags, to name a few. The amino acids resulting from these nucleic acid sequences may or may not be removed after expression of the protein. All the constructs mentioned above may be used for expression of the enzymes and proteins, which will be used in methods described herein.

The host cells will be transform ed/transfected with the chosen expression system, outlined above. The cells will be cultured using methods known to those skilled in the art, this include liquid cultures in shake flasks and fermenters as well as solid cultures in plates etc.

The proteins may be purified from the source, such as a natural or recombinant source, prior to being used in methods outlined herein. Purification may comprise extraction from the host cells by means of sonication, French press, glass beads or other mean of physical lysis, or chemical cell lysis, and separation by chromatographic steps or other means as known to those skilled in the art. Optionally, a concentration step may be used, e.g., by dialysis, chromatofocusing chromatography, and/or associated with buffer exchange. Compounds and compositions

The compounds and compositions of the present invention are suitable for use in reducing oxalate levels in humans or animals. An identified protein species, with a secretagogue effect, and an oxalate- degrading particle or cell and/or necessary co-factor(s), in a composition of the current invention is administered in an effective amount to an individual. An effective amount comprises an amount of one or several components or secretagogues, derived from or produced by oxalate degrading bacteria, together with one or more oxalate degrading enzymes or bacteria. The effective amount is sufficient, optionally in combination with oxalate-degrading activity from bacteria or enzyme(s), to reduce systemic oxalate for a clinical effect in a disease state. The amount that can be used in a single dose composition, alone or in combination with oxalate-degrading bacteria and/or enzymes, can range from about 10pg to 1000,000Mg, from about l OOpg to 100,000Mg, from about 1 mg to 100mg, and all ranges encompassed therein. The component or secretagogue will actively promote flux of oxalate to the intestine, where one or more oxalate degrading enzymes or bacteria will degrade the oxalate. The present invention also relates to pharmaceutical compositions comprising one or more identified components or secretagogues, derived from oxalate degrading bacteria, recombinantly expressed or extracted from conditioned media, which pharmaceutical compositions may be administered with or without oxalate degrading enzyme(s), enzyme(s) involved in oxalate metabolism, cofactor(s), oxalate degrading bacteria and/or substrate(s). In a preferred embodiment, this pharmaceutical composition is resistant to degradation by gastrointestinal acids and degrading enzymes in the stomach and the upper small intestine. The composition can be, for example, in the form of a tablet or capsule.

The oxalate degrading bacteria that may be administered in combination with (i.e. that may additionally be present in a composition comprising) one or more identified compounds or secretagogues are O. formigenes. The oxalate-degrading enzyme administered in combination with (i.e. that may additionally be present in a composition comprising) one or several identified secretagogues include but is not limited to oxalate decarboxylase, oxalate oxidase, formyl-CoA transferase and oxalyl-CoA decarboxylase.

The enzyme(s) administered in combination with (i.e. that may additionally be present in a composition comprising) one or several identified secretagogues include but are not limited to alanine-glyoxylate aminotransferase (AGT), glyoxylate reductase (GR) and 4-hydroxy-2-oxoglutarate aldolase (HOGA).

The co-factors administered in combination with (i.e. that may additionally be present in a composition comprising) one or several identified secretagogues include but are not limited to vitamin B 6 , NAD + , NADP + , FAD, CoA, ATP and ADP. Substrates referred to in the pharmaceutical composition include but is not limited to oxalate, glyoxylate, 4-hydroxy-2-oxoglutarate, among others.

The oxalate degrading enzyme(s), enzyme(s) involved in oxalate metabolism, cofactor(s), bacteria and/or substrate(s) administered in combination with one or several identified secretagogues may be administered in the same or different fashion in accordance with the most suitable method for optimal bioavailability and activity, as known in the art. The administration methods related include but are not limited to enteral, parenteral and/or topical. The combined administration of one or more secretagogues and O. formigenes, oxalate degrading enzyme(s), enzyme(s) involved in oxalate metabolism, cofactor(s), and/or substrate(s), ensures both a stimuli of the oxalate flux, to increase oxalate transport in the direction of the intestinal lumen, as well as increased degradation of oxalate throughout the gastrointestinal tract.

Use of particles and compositions - method for treatment

Methods of the present invention comprise providing a composition according to the invention comprising one or several identified secretagogues produced by oxalate degrading bacteria administered with or without oxalate degrading enzyme(s), enzyme(s) involved in oxalate metabolism, cofactor(s), oxalate degrading bacteria and/or substrate(s). The identified components or secretagogues of this composition include, but are not limited to; tartronic semialdehyde reductase, cysteine synthase A, acyl carrier protein, predicted protein (accession number: gi|237749499), methionine adenosyltransferase 1 , YgiW protein, riboflavin synthase subunit beta, alkyl hydroperoxide reductase/thiol specific antioxidant/mal allergen, phospho-2-dehydro-3- deoxyheptonate aldolase, elongation factor Tu, s-adenosylhomocysteine hydrolase, conserved hypothetical protein (accession number: gi|237747886), diaminopimelate dehydrogenase, serine hydroxymethyltransferase, aspartate- semialdehyde dehydrogenase, malic enzyme, aconitate hydratase 1 , hsp70-like protein, and comprise a sequence having at least 85% sequence identity, such as 90%, or 95%, sequence identity, to the sequences outlined in Example 4, with sequence IDs: SEQ ID No: 1 , SEQ ID No: 2, SEQ ID No:3, SEQ ID No:4, etc., to the last one, i.e. SEQ ID No: 19.

The compositions of the present invention are suitable in methods of reducing oxalate levels in the animal or human body and are used in the treatment or prevention of oxalate-related conditions including, but not limited to, hyperoxaluria, absorptive hyperoxaluria, enteric hyperoxaluria, primary hyperoxaluria, idiopathic calcium oxalate kidney stone disease (urolithiasis), vulvodynia, oxalosis associated with end-stage renal disease, cardiac conductance disorders, inflammatory bowel disease, Crohn's disease, ulcerative colitis, and disorders/conditions caused by/associated with gastrointestinal surgery, bariatric surgery (surgery for obesity), and/or antibiotic treatment.

Methods of the present invention comprise administering a composition that enables increased net flux of oxalate from plasma to the intestine for degradation by one or more oxalate degrading enzymes or bacteria. A method of providing one or several identified components or secretagogues produced from one or more oxalate degrading bacteria, or derived from oxalate degrading bacteria and recombinantly expressed, to the stomach and/or small intestines by providing a formulation according to the invention.

A reduction in oxalate may be achieved by providing one or several identified components or secretagogues with oxalate degrading enzyme(s), enzyme(s) involved in oxalate metabolism, cofactor(s), bacteria and/or substrate(s) to the gastrointestinal tract. The compositions of the present invention are useful in degrading the oxalate secreted into the intestine from the circulatory system, and thus the methods of the present invention contemplate an overall reduction of the oxalate load in an individual.

The reduction may be measured in any tissue or body fluid environment of the subject. Body fluids include secretions of the body such as nasal or gastric secretions, saliva, blood, serum, plasma, urine, chyme or digestive matter, tissue fluid, and other fluid or semi-solid materials made by humans or animals. For example, oxalate reducing enzyme particle compositions and/or bacteria can be administered to a human or animal to induce a net flux of oxalate to the intestines where the oxalate-reducing enzyme activity degrades the oxalate present in the subject.

Methods for reducing oxalate levels in a human or animal and treating and preventing oxalate-related conditions comprise administering one or several identified components or secretagogues derived from one or more oxalate degrading bacteria, recombinantly expressed or extracted from conditioned media, together with or without one or more oxalate degrading enzyme(s), enzyme(s) involved in oxalate metabolism, cofactor(s), bacteria and/or substrate(s). An effective amount comprises an amount of one or several components or secretagogues, derived from or produced from one or more oxalate degrading bacteria, together with or without one or more oxalate degrading enzymes and/or bacteria and/or co-factor(s) that will reduce the oxalate present in the intestines, tissues or bodily fluids of the subject or maintain a lowered amount of oxalate in the subject compared to the amount of oxalate present before administration of the composition. Such an effective amount can range from about 10pg to 1000,000pg, such as from about 100pg to 100,000pg, or from about 1 mg to 100mg, of the secretagogue, alone or in combination with oxalate-degrading bacteria and enzymes. In a treatment method, an effective amount of a composition/ compound/secretagogue is administered orally to be ingested by a subject at least once a day, at least twice a day, at least three times a day, at least four times a day or more if necessary, and such administration can last for one day, two days, three days, four days, five days, or a week, two weeks, three weeks, or a month, two months, three months, four months, five months, six months, more than six months, one year, two years, or for years or continuously throughout the life of the patient. Such treatment may be continued to maintain the desired oxalate levels in a subject. All patents, patent applications and references included herein are specifically incorporated by reference in their entireties.

It should be understood, of course, that the foregoing relates only to exemplary examples of the present invention and that numerous modifications or alterations may be made therein without departing from the spirit and the scope of the invention as set forth in this disclosure.

Although the exemplary examples of the present invention are provided herein, the present invention is not limited there. There are numerous modifications or alterations that may suggest themselves to those skilled in the art.

The present invention is further illustrated by way of the examples contained herein, which are provided for clarity of understanding. The examples should not to be construed in any way as imposing limitations upon the scope thereof. On the contrary, it is to be clearly understood that changes can be made to various other embodiments, modifications, and equivalents thereof which, after reading the description herein, may suggest themselves to those skilled in the art without departing from the spirit of the present invention and/or the scope of the appended claims.

Examples

Example 1

Preparation of proposed secretagogues of 0. formigenes.

To prepare sufficient amounts for identification, the proposed secretagogues were isolated from a 400 L fermentation. This culture suspension was produced in a four-step seed train (500 ml_ to 400 L) inoculated from a glycerol master cell bank. All steps of the seed train used sterile anaerobic specialized oxalate media (100 mM oxalate) composed of trace metals and the following compounds in gm/L: 0.25 potassium phosphate monohydrate, 0.25 potassium phosphate dihydrate, 0.5 ammonium sulfate, 0.025 magnesium sulfate, 0.02 EDTA (disodium salt), 1 .36 sodium acetate trihydrate, 0.001 resazurin sodium salt, 1 yeast extract, 13.4 sodium oxalate, 4 sodium carbonate (anhydrous), 0.5 L-Cystein HCI. The fermenter media (40 L and 320 L) was prepared without resazurin. All transfers during the fermentation process were performed when the seed had reached late log phase growth. The fermentation progressed as follows: an anaerobic bottle containing 500 ml of oxalate media (37°C) was inoculated with 1 % O. formigenes MCB and grown at 37°C, 75 rpm until reaching late exponential phase. Subsequently the culture was anaerobically transferred, using sterile syringes, needles and CO 2 gas, into an anaerobic bottle containing 3.5 L of oxalate media and incubated at 37°C, 75 rpm until reaching late exponential phase. The grown seed culture (total volume 4 L) was inoculated into a 100 L seed fermenter containing 40 L of sterile anaerobic specialized oxalate media. The seed fermenter culture was transferred to a 500 L fermenter containing 320 L oxalate media after reaching late exponential phase. Harvest of the total 400 L of culture suspension was performed when the culture had reached late exponential phase. Example 2

Isolation of proposed secretagogues from 400 L culture suspension.

Harvest of the cells from the final culture suspension was performed using Tangential Flow Filtration (TFF) with a 500 kDa nominal molecular weight cut- off filter. The resulting cell-free filtrate was further processed through a 10 kDa hollow fiber. The retentate was aseptically collected and intermittently stored at 4°C before being sterile filtered using 0.2pm bottle top filters into 250 mL aliquots. The bottles were subsequently frozen for long-term storage at -80°C. In a subsequent step, the retentate was further processed using concentrator centrifuge tubes with a nominal molecular weight cut-off of 3 kDa for an additional concentration of approximately 90-fold. Protein concentration was determined on the final concentrate using Bradford assay and Coomassie reagent. Bovine Serum Albumin (BSA) was used for standard curve preparation. The final concentrate was a clear yellow liquid with 1 mg of total protein per mL.

Example 3

Identification of proposed secretagogues using LC- MS/MS.

Sample preparation (trypsin digest)

The protein concentrate was re-dissolved 100 μΙ_ 50 mM NH4HC03 under agitation and 5 μΙ_ 200 mM DTT was added. Sample was heated to 95 ° C for 5 minutes. Protein was alkylated by adding 4μΙ_ 1 M iodoacetamide in 100 mM NH4HCO3 and let to react (45 min, 25 ° C, dark). Alkylation was stopped by adding 20μΙ_ of 200 mM DTT and incubating (25 ° C, 45 min). Trypsin in NH4HCO3 was added to the sample in a ratio of trypsin to protein 1 :50 and incubated (37 ° C, 16-18 h).

LC-MS/MS analysis

The enzymatically digested samples were injected onto a capillary trap and desalted (3 μΙ/min 0.1 % v/v formic acid for 5 min). Samples were then loaded onto a nanoflow HPLC column with elution gradient (Solvent A 97-60 Solvent B 3-40%) over 95 min with a 25 min re-equilibration for protein identification. Solvent A consisted of 0.1 % v/v formic acid, 3% v/v ACN, and 96.9% v/v H20. Solvent B consisted of 0.1 % v/v formic acid, 96.9% v/v ACN, and 3% v/v H20. LC-MS/MS analysis was carried out on a hybrid quadrupole-TOF mass spectrometer using 225 V focusing potential and 2400 V ion spray voltage. Information-dependent acquisition (IDA) mode of operation was employed with a survey scan (m/z 400-1800) followed by collision-induced dissociation (CID) of the four most intense ions. Survey and MS/MS spectra for each IDA cycle were accumulated for 1 and 3 s, respectively.

Protein Search Algorithm

Tandem mass spectra were extracted by ABI Analyst version 2.0. All samples were analyzed using Mascot (Matrix Science, London, UK; version 2.2.2) set up to search NCBI (taxonomy bacteria database assuming digestion enzyme trypsin). Mascot was searched with 0.50 Da fragment ion mass tolerance and 0.50 Da parent ion tolerance, lodoacetamide derivative of Cys, deamidation of Asn and Gin, oxidation of Met, was specified in Mascot as variable modifications. Scaffold (version Scaffold-3.3.2, Proteome Software Inc., Portland, OR) was used to validate MS/MS based peptide and protein identifications. Peptide identifications are accepted if they could be established at greater than 95.0% probability as specified by the Peptide Prophet algorithm (Keller, A., et al., Anal Chem 74, 5383-92 (2002)). Protein identifications are accepted if they can be established at greater than 99.0% probability and contain at least 2 identified unique peptides. Protein probabilities were assigned by the Protein Prophet algorithm (Nesvizhskii, A. I., et al., Anal Chem 75, 4646-58 (2003)).

Results

The secreted compounds were characterised as proteins with a molecular weight ranging from 8.8 - 98.8 kDa, with > 50% of compounds being in the span of 30 - 50 kDa. Table 1 below lists the proteins identified and the sequence origin (human strain O. formigenes OXCC13). O. formigenes OXCC13 is of the same sub-grouping (group 1 ) as O. formigenes HC-1 ; its complete genome has been sequenced and is thus searchable. Of the nineteen proteins, thirteen were enzymes with a known catalysing function and the remaining six were proteins. Of the six proteins, one was defined as "conserved hypothetical protein" i.e. this protein is found in organisms from several phylogenetic lineages but have not been functionally characterized. Another of the six proteins was defined as "predicted protein". This is used for entries without evidence at protein, transcript, or homology levels.

Example 4

Amino acid sequences of proteins identified from O. formigenes OXCC13.

The 19 amino acid sequences presented herein (SEQ ID NO:s 1 -19) are the results from the protein identification described in Example 3 and the respective identification numbers (gi number and RefSeq accession number (zp). The sequences are searchable using any of these identifying numbers.

SEQ ID NO: 1

tartronic semialdehyde reductase [Oxalobacter formigenes OXCC13] gi|237749254|ref|ZP_04579734.11

MANLGFIGLGIMGVPMAVNLQNGGNKLFVNDKKAPPAALTDGGAKACATGAEVAKNA DIIFIMV PDTPDVEAVLFGENGVAQGLSAGKIWDMSSISPIATKEFAKKINDLGCEYLDAPVSGGEV GAK AGSLTIMVGGKEETFNKVKPLFELMGKNITLVGGNGDGQTTKVANQIIVALNIQAVAEAL LFASK AGADPAKVRQALMGGFASSRILEVHGERMINRTFNPGFRINLHQKDLNLALQGAKALGIS LPNT ATAQELMNACAANGLSGMDHSALCRAVEIMSAHEIAKA

SEQ ID NO: 2

cysteine synthase A [Oxalobacter formigenes OXCC13]

gi|237748046|ref|ZP_04578526.11

MSNIAKSATDLIGNTPLLEISRFGKHVDADATILAKLEYFNPAGSAKDRIAKAMIDA AEADGKLIP GSTI

IEPTSGNTGIALAAVGAARGYRVIITMPETMSVERQKLIRGYGADLVLTEGAKGMKG AIEKAQEL AATIPGGFVPLQFANPANPAIHKATTGPEIWRDTDGKVDIFVAGVGTGGTLTGVGKYLKE QNPA VQVVAIEPAASPVLAGGKPGPHKIQGIGAGFIPEALDVNVFDEVIGVPNDAAFATAKTLA HAEGV LVGISSGAALWAASELAKRPENRGKTIVVLLADNGERYLSTDLFSN

SEQ ID NO: 3

acyl carrier protein [Oxalobacter formigenes OXCC13]

gi|237747699|ref|ZP_04578179.11

MSDIEQRVKKIVAEQLGVAEDEIKLESSFVDDLGADSLDTVELVMALEDEFEIEIPDEQA EKITTV QQAV DYATANMQS

SEQ ID NO: 4 predicted protein [Oxalobacter formigenes OXCC13]

gi|237749499|ref|ZP_04579979.11

MKLRPLQDRIIVKRVDQEKTTASGIVIPDNAAEKPDQGEVIAVGNGKVLEDGKVLPLDVK VGDIV LFGKYSGQTVKVEGEELLVMHESDVMAIVQN

SEQ ID NO: 5

methionine adenosyltransferase 1 [Oxalobacter formigenes OXCC13] gi|237747590|ref|ZP_04578070.11

MSQDYLFTSESVSEGHPDKVADQISDAILDAILEQDPHARVAAETLCNTGLVVLAGEITT TANID YISVA

RDTIKRIGYDNNDYGIDHRGCAVLVGYDKQSPDIAQGVDEGRGIDLDQGAGDQGLMF GYACD ETPELMPAAIYYAHRLVERQSQLRKDGRIAWLRPDAKSQVTLRYVDGKPVAAETIVLSTQ HAPE VEHKQIEEAVIEEIIKPVMPAEWLKESKYLVNPTGRFVIGGPQGDCGLTGRKIIVDTYGG SCPHG GGAFSGKDPSKVDRSAAYAARYVAKNIVAAGLAKKCQIQVSYAIGVARPINITINTEGTG VIADSK IAALVNDHFDLRPKGIVQMLDLLHPRYVKTAAYGHFGRDEPEFTWEKTDKAADLRQAAGL

SEQ ID NO: 6

YgiW protein [Oxalobacter formigenes OXCC13]

gi|237748090|ref|ZP_04578570.11

MMSKKLLTGLFAAGLSIGMLAVSSTASAQYIGPTRIAPTTVNKVLKSPVDDQYVLLSGKI TSQVS KDKYTFTDKTGSIWEIDNEVFAGRQVGPDTQVEIWGKVDKDMFKEPKIDVKRLGIPNQTK

SEQ ID NO: 7

riboflavin synthase subunit beta [Oxalobacter formigenes OXCC13]

gi|237749243|ref|ZP_04579723.11

MMTVNTFEIDLQGQDLRIGIVQSRFNEEICRGLLGACLEELKRLGVADEDILVATVP GALEIPTAL QKMAESQQFDALIAVGGIIKGETYHFELVSNESAAGISRVALDFDMPIANAILTTYTDEQ AEARM VEKGTEAARVAVEMANLVMAIDELEPPEEDE

SEQ ID NO: 8

alkyl hydroperoxide reductase/Thiol specific antioxidant/Mal allergen

[Oxalobacter formigenes OXCC13]

gi|237747586|ref|ZP_04578066.11

MSSLINTEIIPFKAQAYHNGQFVKVTDADLKGKWSVVFFYPADFSFVCPTELGDLADHYE EFKK LGVEIYSVSTDTHFVHKGWHDASDTIKKIQFPMVGDPSGQISRNFNVLIDDDGVALRGTF VINPE GVIKLCEIHDLGIGRSATELLRKVQAAQYVATHKGQVCPASWQPGAETLAPSLDLVGKI

SEQ ID NO: 9

Chain A, Formyl-Coa Transferase With Aspartyl-Coa Thioester Intermediate Derived From Oxalyl-Coa

gi|163931 105|pdb|2VJK|A

MTKPLDGINVLDFTHVQAGPACTQMMGFLGANVIKIERRGSGDMTRGWLQDKPNVDSLYF TM FNCNKRSIELDMKTPEGKELLEQMIKKADVMVENFGPGALDRMGFTWEYIQELNPRVILA SVKG YAEGHANEHLKVYENVAQCSGGAAATTGFWDGPPTVSGAALGDSNSGMHLMIGILAALEI RHK TGRGQKVAVAMQDAVLNLVRIKLRDQQRLERTGILAEYPQAQPNFAFDRDGNPLSFDNIT SVP RGGNAGGGGQPGWMLKCKGWETDADSYVYFTIAANMWPQICDMIDKPEWKDDPAYNTFEG RVDKLMDIFSFIETKFADKDKFEVTEWAAQYGIPCGPVMSMKELAHDPSLQKVGTWEVVD EIR GNHLTVGAPFKFSGFQPEITRAPLLGEHTDEVLKELGLDDAKIKELHAKQVV

SEQ ID NO: 10

phospho-2-dehydro-3-deoxyheptonate aldolase [Oxalobacter formigenes OXCC13]

gi|237749194|ref|ZP_04579674.11

MDTTDDLRILAMKELTPPAHLIREFPCEEKAAETVSGCRKAIQRVLHNQDDRLVVIIGPC SIHDPK AAMEYAHRLAEEKERYGDELVWMRVYFEKPRTTIGWKGLINDPFMDHSYRINEGLHIARE LLR DVNELGLPAATEYLDMISPQYVADMISWGAIGARTTESQVHRELSSGLSCPVGFKNGTDG NIKI AIDAIKAASHPHHFLSVTKGGHTAIFETEGNQDCHIILRGGYKPNYDAASVNEAARAVEA AGLAP KIMIDASHGNSSKKAENQVPVSLEVGENIAKGDDRIIGLMIESNLVGGRQDHEVGKKLVY GQSV TDACIGWEDSSKLLGQLAETVKRRRDVLKK

SEQ ID NO: 1 1

elongation factor Tu [Oxalobacter formigenes OXCC13]

gi|237747517|ref|ZP_04577997.11

MSKKFGGEAKDYDQIDAAPEEKARGITINTSHVEYETAARHYAHVDCPGHADYIKNM ITGAAQM DGAILVVSAADGPMPQTREHILLARQVGVPYIIVFLNKCDMVDDAELLELVEMEVRELLS RYEFP GDDIPIIKGSAKLALEGDAGELGETAILALADALDSYIPTPERAVDGAFLMPVEDVFSIS GRGTVV TGRVERGIIKVGEEIEIVGIKETAKTTCTGVEMFRKLLDQGQAGDNIGVLLRGTKREEVE RGQVL AKPGSIKPHLNFEGEVYVLSKEEGGRHTPFFNNYRPQFYFRTTDVTGAIELPKDKEMVMP GDN VSISVKLISPIAMEEGLRFAIREGGRTVGAGVVAKITE

SEQ ID NO: 12

S-adenosylhomocysteine hydrolase [Oxalobacter formigenes OXCC13] gi|237749247|ref|ZP_04579727.11

MNAVSKTEQDFYIADPDLTAWGNKEIRIAETEMPGLMAIREEYAASKPLSGARISGSLHM TIQTA VLIQTLEALGAKVRWASCNIYSTQDHAAAAIASNGTPVFAFKGESLDDYWEFTHRIFEWP DGGY SNMILDDGGDATLLLHLGSRAEKDATVLDNPGSEEEVCLFNAIKRHLKTDPNWYSKRIKE IKGVT EETTTGVHRLYQMHEEGKLKFPAINVNDSVTKSKFDNLYGCRESLVDGIKRATDVMIAGK VAVV CGYGDVGKGCAQALKALSAQVWVTEVDPICALQAAMEGYRWTMDYAAEMADIFVTCTGNY H VITHDHMVKMKDQAIVCNIGHFDNEIDVASMKKYTWDNIKPQVDHIILPNGNRIILLAEG RLVNLG CGTGHPSYVMSSSFANQTIAQIELFTNTEAYPVGVYTLPKHLDEKVARLQLKKLNAVLTE LSDE QAAYIGVKKEGPYKPNHYRY SEQ ID NO: 13

conserved hypothetical protein [Oxalobacter formigenes OXCC13]

gi|237747886|ref|ZP_04578366.11

MANQEEPIKVTDDFKQCWQSAGRHLQSQVEGGLTWMRASLDEPFMEHLSFRLGNQLFFVR VI DVDNELKVPGTDENLIKIAEGCKGHACIMPMRFSFGNWMPVEKGWGLLSAVDKKPVNPPD LVT DEKIEMTDWELHDFAVQVVRQNLMHEGEHVAGWVSNPELQPSIWISTEEFPQWVVVQAVR W PAEAKIPDNIKEIEDAYASKEAKGTFAYVTFANENQNVKEPLKEGEKPLPIYRGDKAYIS YSGLLS TEN

SEQ ID NO: 14

diaminopimelate dehydrogenase [Oxalobacter formigenes OXCC13]

gi|237749390|ref|ZP_04579870.11 MTTIKAAVHGLGNIGRHVIDCLTCAPDFECLGVIRRESSLGTQTLERRNIPDYASIDKLI AEKGKP DVVI

LCGPSRSVPEDAKFYLSRGIRTVDSFDIHTDIAELVEKLDVVAKENNSACITAAGWD PGTDSVF RTLFEAMAPTGTTFTNFGRGRSMGHSVAARAIKGVADATSITIPIGGGRHARLVYVLAEK GASF EQIKKDLASDPYFSHDPLDVREVKTPEEMEAVADNSHGVLMERIGASGRTSNQNLTFTMK IDN PALTSQVLVSCARAVTRMGAGCHTLIDVPPVMLLAGERMQHIARLV

SEQ ID NO: 15

serine hydroxymethyltransferase [Oxalobacter formigenes OXCC13]

gi|237749274|ref|ZP_04579754.11

MFAKDYSLAQVDSELWDAILRENTRQEEHIELIASENYCSPAVMQAQGSQLTNKYAEGYP GKR YYGGCEYVDIAEQLALDRVKKLFGAEAANVQPNSGSQANQAIFLAMLNPGDTIMGMSLAE GGH LTHGMALNMSGKWFNVVSYGLNEKEEIDYDRMEQLAHEHKPKLIIAGASAYSLRIDFERF AKVA RDVGAFFMVDMAHYAGLIAAGVYPSPVPYADFVTSTTHKSLRGPRGGFILMKPEFERKIN SAVF PGLQGGPLMHVIAGKAVAFKEALQPEFKTYQEQVLKNASVLAKTLVDRGFRIISGRTESH VMLV DLQSKNITGRQAETILNSGHITCNKNAIPNDPQTPFVTSGVRLGSPAMTTRGFKETESAI VGNLL ADVIENPNDQATIERVRAEVKKLTTAFPVYQH SEQ ID NO: 16

aspartate-semialdehyde dehydrogenase [Oxalobacter formigenes OXCC13] gi|237748872|ref|ZP_04579352.11

MKLVGLIGWRGMVGSVLMQRMQEENDFDLFEPVFFTTSNVGGKAPAMAKNETVLKDAFNI DE LKKCDILISCQGGDYTVDVFPKLRAAGWDGYWIDAASKLRMNDDALIILDPVNRKVIEDG LSKGI KNYIGGNCTVSCMLMGLGGLFENDLVEWMTSMTYQAASGGGAQHMRELLTQFGSIHTEVR M NLENPASAILEIDRQVLARQRGMTADETKQFGVPLAGNLIPWIDTDLGNGMSREEWKGGA ETN KILGKNDGNKVIVDGLCVRVGAMRCHSQALTIKLKKDVPLDEITDILKSHNQWAKVVPNT KEDS VRDLTPAAVSGSLTIPVGRLRKLEMGNDYLSAFTVGDQLLWGAAEPLRRMLRIILE

SEQ ID NO: 17

malic enzyme [Oxalobacter formigenes OXCC13]

gi|237749327|ref|ZP_04579807.11

MNSQDQKKELLKKNALAFHRFPIPGKISVNPTKEVRDQNELALAYTPGVACACEEIH ANPENAYI YTTKGNLVAVISNGTAVLGLGNIGAQASKPVMEGKGVLFKKFADINVFDLEINELDPDKL CDIIAS LEPTFGGINLEDIRAPECFYIERKLREKMNIPVFHDDQHGTAVIVGAAVLNALKWGKNIK NCKM VVSGAGAGAMGCLELLVDLGFPVENIWVTDIKGVVYKGRKELMDPEKEKYAQETDARTLM DVI SDADIFLGLSAGNVLKPEMVLKMAKDPVIFAMANPIPEILPEVAHATRDDVIMGTGRSDY PNQIN NSMCFPYLFRGALDCRAKTINREMELAAVRAIASLAEMECPEEIVAMYGKKYTFGRDYLL PFQF DPRLLWWAPAVAQAAMDSGVARVQIADMDAYRAKLKEFVG

SEQ ID NO: 18

aconitate hydratase 1 [Oxalobacter formigenes OXCC13]

gi|237747686|ref|ZP_04578166.11

MSCFTQNTYKEFPVTHEKKGHFYSIPALGKELGLDLSRLPVSIRIVLESVLRNCDGKKIT EEHVR

QLANWKPNEERSNEIPFVVARVILQDFTGIPLLVDLAAMRNVAVKTGKNPKKIEPLV PVDLVVDH

SVQIDYFRQDNALDLNMKLEFDRNRERYQFMKWGMQAFDTFGVVPPGFGIVHQVNME YLAR GVHKRNDAEAGDVYYPDTLVGTDSHTTMINGVGVVGWGVGGIEAEAGMLGQPVYFLTPDV IG MNLTGKLREGCTATDLVLTITELLRKEKVVGKFVEFFGEGAASLSATDRATIANMAPEYG ATIGF FTVDEATISYFKNTGRTDEEVSALESYFRAQGMFGIPKAGQIDYTRVVNLDLGSVTASVS GPRR PQDRIELGNLKKRFTELFSAPVKDGGFNKKPADMEATYVNSDNVELKNGDILIAAITSCT NTSNP AVLLAAGLLAKKAVEAGLQVSPRIKTSLAPGSRIVTNYLEKAGLLPYLEKLGFNVAAYGC TTCIG NAGDLTPAMNEAIVKNDWAAAVLSGNRNFEARIHPNIRANFLASPPLVVAYAIAGNVTRD LTTE PLGKGKDGKDIYLSDIWPTSHEVAALVPLALDAPSFRKNYSDIKTAPGELWQKIAGFATG DVYD WPQSTYIAEPPFFSDFGMEPNAASANISGARALALFGDSITTDHISPAGSIQEKSPAGQW LMEH GISKANFNSFGSRRGNHEVMMRGTFGNVRIKNQMLPVGPDGSRREGGYTLYQPGGEETSI FD AAMRYQKENVPTIVIGGEEYGTGSSRDWAAKGTQLLGVKAVIARSFERIHRSNLVGMAVL PLQF TGNDSAESLGLKGDETFDLTGLDDITPLQDVTLWHRADGTTQNVPLLLRIDTPIEVDYYR HGGI LPFVLRQLLSN

SEQ ID NO: 19

hsp70-like protein [Oxalobacter formigenes OXCC13]

gi|237749571 |ref|ZP_04580051 .11

MSKIIGIDLGTTNSCVAIIEGSQPRVVENSEGNRTTPSVIAYLDDGEILVGAPAKRQAVT NPKNTL YAIK

RLIGRKFDDKEVQRDIPIMPFSIIKAENNDAWVSVLNDKKLAPPQVSAEVLRKMKKT AEDYLGEE VTEAV

ITVPAYFNDAQRQATKDAGRIAGLDVKRIINEPTAAALAFGLDKAGKGDKKIAVYDL GGGTFDISII EIA

DLDGDKQFEVLSTNGDTFLGGEDFDQRIIDFIIDEFNKINGIDLKKDPIALQRIKAS AERAKIELSS SQQ TEINEPYIAMANGAPVHLNMKLTRAKLESLAEGLIDQTIEPCRIALKDAGLSVSDIDDVI LVGGMT RMPA

VQDKVKAFFGKEPRKDINPDEAVAVGAALQGAVLSGDRKDLLLLDVTPLSLGIETLG GVMTKMI QKNTTIPTKFSQIFSTAEDNQPAVTIKVYQGEREMAAGNKALGEFNLEGIPASPRGMPQI EVTFD IDANGILHVSAKDKATGKENKITIKANSGLSEDEIQRMIKDAEVNAAEDHKVRELTEARN QGDAL VHTTKKSMEEYGDKLDAPAKESIESAIKDLEESLKGDDKADIDSKMSALSAAAQKLGEKM YADQ APEGAAAGAAGAGASAGAAPEPELEDDVVDADFKEVKDKD

Example 5

Description of physico-chemical characteristics of identified secretagogues. Methods

Calculation of protein extinction coefficient of native protein in water:

Protein extinction coefficient calculations used were as described by Edelhoch (Edelhoch, H., (1967) [PubMed: 6049437] using extinction coefficients for Tyr and Trp determined by Pace (Pace, C.N., et al., (1995) [PubMed: 8563639].

Calculation and interpretation of instability index:

The instability index was determined as described by Guruprasad, K., et al., (1990) [PubMed: 2075190] and provides and estimate of the stability of the protein in a test tube. A stability index lower than 40 is predicted as stable while a value above 40 predicts that the protein may be unstable.

Calculation of aliphatic index:

Aliphatic index is defined as the relative volume occupied by aliphatic side chains (alanine, valine, isoleucine, and leucine) and is calculated according to the formula described by Ikai (Ikai, A.J. (1980) [PubMed: 7462208]. It may be regarded as a positive factor for the increase of thermostability of globular proteins. Calculation of Grand Average of Hydropathy (GRAVY)

GRAVY is calculated as the sum of amino acid hydropathy values, divided by the number of residues in the sequence (Kyte, J., Doolittle, R.F. (1982) [PubMed: 7108955]. See Table 2 below. Example 6

Identification of putative signal peptide cleavage sites in the amino acid sequence of the identified secretagogues.

Methods

Identification of putative presence and location of signal peptide cleavage sites was performed using the online prediction services provided by The Center for Biological Sequence Analysis at the Technical University of Denmark

(http://www.cbs.dtu.dk/services/).

Prediction of presence and location of Twin-arginine signal peptide cleavage sites in bacteria:

The prediction of twin-arginine signal peptides was performed as described by Jannick Dyrl0v Bendtsen et al (BMC bioinformatics, 6: 167, 2005).

Results:

One protein of the nineteen proposed secretagogues had a sequence that was positive for a twin-arginine signal peptide cleavage site: YgiW protein (accession number: gi|237748090). The max. C, max. Y, max. S, mean S and max. D all gave a positive (YES) response for a signal peptide cleavage site with a value of 0.872, 0.500, 0.797, 0.314 and 0.407, respectively. The likely cleavage site is between position 28 and 29: ASA-QY.

Two additional proteins had a sequence that was positive for a signal peptide cleavage site in max. C but which was not confirmed in max. Y: phospho-2- dehydro-3-deoxyheptonate aldolase and cysteine synthase A. Example 7.1

Recombinant expression of the identified potential secretagogue compounds Methods

The enzymes and proteins identified and outlined herein may be recombinantly expressed in the native host or a host of choice, suitable for recombinant overexpression as known by those skilled in the art. The recombinantly expressed enzymes and proteins will comprise a sequence having at least 85% sequence identity to the sequences outlined (SEQ No 1 -19). Protein homologs and variants include but are not limited to: polymorphic variants and natural or artificial mutants, modified polypeptides in which one or more residue is modified, and mutants comprising one or more modified residues. Mutations include but are not limited to truncation, deletion, substitution or addition mutations of nucleic acids.

The recombinant enzymes may be expressed in a wide variety of hosts, known to those skilled in the art of protein expression, including but not limited to: E. coli, Lactobacillus spp, Bacillus spp etc. For a recombinant production of the enzyme or protein the host should comprise a construct in the form of a plasm id, vector, phagemid, or transcription or expression cassette that comprises the enzyme or protein or a functional fragment thereof. A variety of constructs are available, including constructs, which are maintained in single copy or multiple copy. Many recombinant expression systems, components, and reagents for recombinant expression are commercially available, for example from Invitrogen Corporation (Carlsbad, Calif.); U.S. Biological (Swampscott, Mass.); BD Biosciences Pharmingen (San Diego, Calif.): Novagen (Madison, Wis.); Stratagene (La Jolla, Calif.); and Deutsche Sammlung von Mikroorganismen and Zellkulturen GmbH (DSMZ), (Braunschweigh, Germany).

A heterologous promotor, including a constitutive and/or inducible promotor, optionally controls recombinant expression of the proteins. Promotors such as, for example, T7 or other promotors, as suitable for the host, and which are well- known for those skilled in the art. The promotor may also originate from the Oxalobacter genus. The enzyme or proteins recombinant nucleic acid sequence may include nucleic acids for purposes additional to the expression of the protein, including but not limited to for purification purposes, folding purposes etc. Examples of those are: secretion sequences, signal sequences, linkers, expression control elements, affinity tags, to name a few. The amino acids resulting from these nucleic acid sequences may or may not be removed after expression of the protein. All the constructs mentioned above may be used for expression of the enzymes and proteins, which will be used in methods described herein.

The host cells will be transform ed/transfected with the chosen expression system, outlined above. The cells will be cultured using methods known to those skilled in the art, this include liquid cultures in shake flasks and fermenters as well as solid cultures in plates etc.

The proteins may be purified from the source, such as a natural or recombinant source, prior to being used in methods outlined herein. Purification may comprise extraction from the host cells by means of sonication, French press, glass beads or other mean of physical lysis, or chemical cell lysis, and separation by chromatographic steps or other means as known for those skilled in the art. Optionally, a concentration step may be used, e.g., by dialysis, chromatofocusing chromatography, and/or associated with buffer exchange.

Example 7.2

As outlined above in Example 7.1 , of the enzymes and proteins identified and outlined herein, SEQ ID No:s 3, 4, 6, 13 and 19 were recombinantly over- expressed in E.coli, according to commonly used methods and techniques (Methods in Molecular Biology, Book 705, Thomas C. Evans Jr., Ming-Qun Xu, Humana Press; 201 1 ; Methods in Molecular Biology, Book 235, Nicola Casali, Andrew Preston). The recombinantly expressed proteins originated from a synthesized gene sequence having at least 99% sequence identity to the sequences of SEQ ID No:s 3, 4, 6, 13 and 19 of Example 4. No mutations were performed to the sequences presented in Example 4 but short sequence tags were added to facilitate purification (Histidine tag). Purified protein molecular weight and identity were confirmed by way of SDS-PAGE and Western blot. SDS-PAGE was run on 4%-20% gradient gel, followed by Coomassie Blue staining. The Western blot analysis utilized Anti-His Antibody from Genscript, Cat. No. A00612 or A00186. Results

The proteins were successfully overexpressed and purified from E.coli culture supernatant by use of Histidine tag affinity purification, to near homogeneity (approximately 85% purity). The final storage buffer for all proteins was 50mM Tris-HCI, 150mM NaCI, 10% Glycerol, pH 8. Theoretical molecular weight was confirmed with SDS-PAGE gel for all proteins except SEQ ID No. 3 and SEQ ID No. 4, for which migration on the gel insinuated a larger molecular weight than expected. However, for both proteins the theoretical molecular weight was confirmed by Maldi-TOF. Further, such anomalous behaviour on SDS-PAGE gel is consistent with observations from in particular acyl carrier proteins of other bacteria (J. Bacteriol. 178:4794-4800, J. Biol. Chem. 267:5751 -5754, J. Bacteriol. 174:3818-3821 ) and has been attributed to the high charge-to-mass ratio as well as its low hydrophobic amino acid content, two factors that have a considerable influence on SDS binding (J. Biol. Chem. 254:9778-9785), and thus can be the cause of this anomality for both SEQ ID No. 3 and SEQ ID No. 4.

Purity of all proteins was determined by densitometric analysis of Coomassie Blue-stained SDS-PAGE gel to approximately 85% (Fig. 1 A-E). Bovine Serum Albumin (2ug) was run as control (Lane 1 ). Target protein on SDS-PAGE gel is presented in Lane 2, and target protein on Western blot is presented in Lane 3.

Fig. 1A: SEQ ID No. 3 expressed protein analysis.

Anti-His antibody used: Genscript Cat. No. A00612. Fig. 1 B: SEQ ID No. 4 expressed protein analysis.

Anti-His antibody used: Genscript Cat. No. A00612.

Fig. 1 C: SEQ ID No. 6 expressed protein analysis.

Anti-His antibody used: Genscript Cat. No. A00612.

Fig. 1 D: SEQ ID No. 13 expressed protein analysis.

Anti-His antibody used: Genscript Cat. No. A00186. Fig. 1 E: SEQ ID No. 19 expressed protein analysis.

Anti-His antibody used: Genscript Cat. No. A00186.

Example 8

Screen of potential secretagogues using intestinal cell model

Methods

Using human intestinal Caco2-BBE cells, T84 or another cell line suitable for simulating transport over intestinal epithelia, the effect of Oxalobacter formigenes conditioned medium and/or recombinantly expressed secretagogues may be screened for altered net oxalate transport over the cell layer. In the case of evaluation of conditioned media the cells will be pre- incubated with a suitable dilution (e.g. 1 :50) of O. formigenes conditioned media and unconditioned media, as well as Lactobacillus Acidophilus conditioned media, for 24 h. The apical [ 1 C] oxalate influx will be measured in the presence of an outward CI gradient. In the case of evaluation of recombinantly expressed proteins the cells will be pre-incubated with either a suitable concentration of purified protein or a cell lysate from the expression host. Several dilutions will be included to evaluate dose-response. As controls the protein buffer or the untransformed host cell lysate will be used, respectively.

Example 9.1

Screen of potential secretagogues using intestinal tissue from animal

Methods As a method to screen potential secretagogue compounds intestinal tissue can be isolated from mice or rats. The animals are euthanized using 100% CO2 and the intestine is immediately removed. The tissue is thoroughly cleansed using ice-cold 0.9% NaCI, connective tissue is removed, and the segments are opened along the mesenteric border. Flat sheets of tissue are mounted in a modified Ussing chamber and tissue is bathed on both sides by buffered saline (pH 7.4) containing 1 .25uM oxalate at 37°C, while bubbling with 95% 0 2 , 5% CO2. The two unidirectional fluxes of [ 1 C]oxalate will be measured at suitable time intervals under short-circuit conditions using an automatic voltage clamp. The electrical parameters of the tissue will be measured (open-circuit potential, short-circuit current etc.) throughout the experiment. Details to planned experiments are described in the art (Hatch and Freel Am J Nephrol 23: 18-26, 2003, Freel et al Am J Physiol Gastrointest Liver Physiol 290: G719-G728, 2006).

The animals that may be suitable to use include but are not limited to Sprague- Dawley rats with or without hyperoxaluria induction using ethylene glycol, AGT knock-out mice (C57BL/6 background strain as described by Salido et al (PNAS USA 103: 18249-18254, 2006), or transport protein knock-out mice e.g. Slc26a67 " (Wang et al., Am J Physiol Cell Physiol 288: C957-C965, 2005). ,

The intestinal tissues used for transport studies will be derived from the above described strains of mice or rats, raised using an oxalate containing diet or not. The animals may also be repeatedly gavaged with live cultures of O. formigenes to induce colonization, while fed an oxalate containing diet or not.

To confirm colonization and state of hyperoxaluria, urine and fecal samples will be collected during the housing of the animals and analyzed for oxalate and O. formigenes as thoroughly described in the art. Animals will be confirmed non- colonized before any attempts to colonize using O. formigenes.

To confirm potential secretagogue action on an anion exchange transport protein, suitable inhibitors may be added to the Ussing chamber compartments, these include but are not limited to DIDS (4,4'-diisothiocyano-2,2'-stilbene disulfonate), and SITS (4-acetamido-4'-isothiocyanostilbene 2'-disulfonate).

Example 9.2

Screen of recombinantly expressed potential protein secretagogues from Oxalobacter formigenes using intestinal tissue from rat

Methods

Ussing Chamber Experiment

As outlined above in Example 9.1 , intestinal segments were stripped of muscle layers and mounted in Ussing chambers, each side bathed in 5 ml of glucose- Ringer solution containing 1 .5μΜ (unlabelled) sodium oxalate (Ringer composition in mmol/L: Na + 140, CI " 1 19.8, K + 5.2, HC0 3 " 25, Ca 2+ 1 .2, Mg ++ 1 .2, HP0 4 2" 2.4, H2PO4 " 0.4, glucose 10, pH 7.4 when gassed with 95%0 2 5%C0 2 at 37°C). Direct recording of trans mural electrical potential difference (PD) in mV and determination of short-circuit current (l S c) in μΑ/cm 2 was performed using two sets of electrodes connecting the chamber solutions to voltage clamps. Total electrical resistance (R T ) in ohms.cm 2 was calculated using Ohm's Law, where PD = l S c x R T - All experiments were conducted under short circuit conditions except when periodically switched to the open circuit state for recording of PD. At T = 0 min, treatments were added to the mucosal solution of the pair. Tissues were short- circuited and equilibrated for 20 min while reading l S c and R T . At T=20 min 10 Ci of labeled C14-oxalate was added to either the mucosal or serosal bath, of one of the paired tissues. At T=30 min flux sampling of 100μΙ every 15 min, from the unlabelled side, was started, and continued for a 60 min period. The 10ΟμΙ of solution from the 'unlabelled' side was replaced with 10ΟμΙ of unlabelled buffer. Aliquots (1 ΟΟμΙ) of the 'hot' side solution were taken at the beginning and end of experimental periods and their values averaged to obtain the specific activity for the period. Isotope activity was measured with a liquid scintillation counter. Unidirectional fluxes were computed (J ox =cpm/time. specific

activity. area) and expressed as umol/min.cm 2 . Net fluxes were calculated as the difference in the unidirectional fluxes between paired specimens. l S c in μΑ (converted to Eq/cm 2 .hr) and PD in mV were recorded at the start and end of each sampling interval and averaged for that interval.

The control (only buffer) was evaluted identically to the test article, using

Sprague-Dawley rat distal colon tissue. The test articles entailed the

recombinantly expressed proteins of Example 7.2 divided into three groups: Group 1 : Protein of SEQ ID No. 3, Group 2: Proteins of SEQ ID No. 4 and 19, Group 3: Proteins of SEQ ID No. 6 and 13. Tissues from six rats were used per treatment (n=6). Differences between basal mucosal-to-serosal (m-s) and serosal-to-mucosal (s-m) fluxes of C14-oxalate for paired tissues is used to establish net absorption (m-s > s-m) or secretion (s-m > m-s) of oxalate for a specific treatment. Calculation of the mean +/- SEM of the net flux for all treatments determines if there is an effect from a treatment over the respective control.

Results

The buffer control demonstrated net absorptive flux for this distal intestinal segment of rat tissue, confirming prior literature studies with similar intestinal rat segments. The flux was measured in two periods, 0-30 minutes and 30-60 minutes. Similar behaviour of the control was seen in both periods. In both periods, the treatments of the three groups all reduce the total flux of labeled oxalate. However, in the later period Group 1 demonstrate a larger change in serosal-to-mucosal (s-m) flux, resulting in a larger net secretion over the intestinal tissue, of labeled oxalate, in this period. Due to one outlier replicate, only five replicates of the experiment were included in this data set.

Figure 2 depicts labeled oxalate flux during first flux period, 0-30 minutes (T=30 to T=60, as per "Ussing Chamber Experiment" denotations). M-S flux = mucosal-to-serosal flux, S-M flux = serosal-to-mucosal flux. G1 -G3 denote the three groups of treatment, G = group.

Figure 3 shows labeled oxalate flux during second flux period, 30-60 minutes (T=60 to T=90, as per "Ussing Chamber Experiment" denotations). M-S flux = mucosal-to-serosal flux, S-M flux = serosal-to-mucosal flux. G1 -G3 denote the three groups of treatment, G = group.

Example 10

Evaluation of expression pattern of transport proteins in vivo

To confirm presence of transport proteins in vivo, in particular proteins belonging to the class of SLC26A (solute-linked carrrier), mucosal scrapings can be analyzed using suitable immunoblotting and probing, methods which are well described in the art and described herein in brief.

Methods

The mucosal scraping would be analyzed using SDS-PAGE and subsequently transferred onto a nitrocellulose membrane. The membrane would be blocked with for example a 1 % casein solution, washed with phosphate buffered saline containing detergent and then probed using a specific primary antibody and subsequently a suitable secondary antibody linked to a enzyme for detection e.g. HRP (horse radish peroxidase) enzyme.

The primary antibodies may be raised against recombinantiy expressed transport proteins and used to detect for specific transport proteins e.g. SLC26a6. Methods for preparation of antibodies is common place in the art and will not be outlined herein.

Example 11

Evaluation of binding interactions between recombinantiy expressed transport proteins and potential secretagogue compounds

As a complementary or alternative way to screen the identified potential secretagogue compounds for the compound/s with the desired characteristics the interaction between the transporter/s and the proposed secretagogue compounds can be analyzed.

Methods There are several suitable methods to characterize protein-protein interactions, known to those skilled in the art. For example various indirect methods i.e. ELISA, RIA and surface plasmon resonance may be used to characterize the interaction between the oxalate transporter membrane protein and the expressed and purified potential secretagogue. Also, Isothermal Titration Calorimetry (ITC) can be used to quantify the interactions by determining the heat evolved on protein-protein association.

Example 12

Evaluation of potential secretagogues in hyperoxaluria animal models

Following a screen of the recombinantly expressed potential secretagogues in vitro, the promising compounds will be included in a study using suitable animal models (see Example 9 for examples of animal models that may be used as described herein), to confirm the effect in vivo. This study may be performed using a respective arm for the concurrent colonization with 0. formigenes, or the lack of colonization.

Methods

Several animal models have been used to simulate hyperoxaluric conditions, among one is the AGT knock-out mouse model (see Example 9). This mouse model may be used to evaluate the potential secretagogue compound. The execution for these animal models is well described in the literature and will only generally be described herein. Further, methods for plasma and urinary oxalate and calcium analysis are commonly used in the field and will not be outlined.

Both female and male mice will be used. In the beginning of the study (approx. 5 days) the mice will be given free access to water and standard mouse chow (e.g. diet 2018S, Harlan teklad) to establish a base line value for the parameters to be monitored, including but not limited to plasma and urinary oxalate and calcium.

In order to increase contrast in oxalate levels and/or facilitate O. formigenes colonization a period (approx. 5 days) in which the mice are primed with a high oxalate diet may be incorporated (e.g. 1 .5% oxalate supplemented diet (0.5% calcium; diet 89000, Harlan Teklad). If desired, a select arm will be colonized with O. formigenes by esophageal gavage of a 0.5-mL inoculum containing wet O. formigenes cell pellet at two occasions, with 48 hours separation. The control group will be gavaged with an inactivated O. formigenes culture or similar. After five days the colonization is confirmed in the respective animals using detection in fecal samples by PCR and the administration of the secretagogue compound can ensue for both groups. Each group may be divided into separate arms in which the secretagogue is administrated or not, secretagogue administration may continue for 3-5 days. Throughout the study plasma and urinary oxalate and calcium will be monitored. Subsequently of ending administration of product a period of continued high- oxalate diet may be performed in order to see the parameter values return to pre-secretagogue levels. If desired, a separate arm of the colonized animals given product may be kept on product but returned to regular oxalate level diets (0.5%) in order to evaluate if the knock-out mice can sustain colonization without exogenous addition of oxalate.

Table 1 1dentified proposed secretagogues

Table 2 Description of physico-chemical characteristics of identified secretagogues