WO2017100266A1 - Glycolate oxidase inhibitors and methods of use for the treatment of kidney stones - Google Patents

Glycolate oxidase inhibitors and methods of use for the treatment of kidney stones Download PDF

Info

Publication number
WO2017100266A1
WO2017100266A1 PCT/US2016/065300 US2016065300W WO2017100266A1 WO 2017100266 A1 WO2017100266 A1 WO 2017100266A1 US 2016065300 W US2016065300 W US 2016065300W WO 2017100266 A1 WO2017100266 A1 WO 2017100266A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
pharmaceutically acceptable
acceptable salt
group
prodrug
Prior art date
Application number
PCT/US2016/065300
Other languages
French (fr)
Inventor
W. Todd Lowther
Ross P. HOLMES
Original Assignee
Wake Forest University Health Sciences
Uab Research Foundation
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Wake Forest University Health Sciences, Uab Research Foundation filed Critical Wake Forest University Health Sciences
Priority to EP16873725.2A priority Critical patent/EP3386949A4/en
Priority to US15/781,571 priority patent/US20200262794A1/en
Publication of WO2017100266A1 publication Critical patent/WO2017100266A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/66Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D233/90Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/44Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having three double bonds between ring members or between ring members and non-ring members
    • C07D207/444Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having three double bonds between ring members or between ring members and non-ring members having two doubly-bound oxygen atoms directly attached in positions 2 and 5
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/04Drugs for disorders of the urinary system for urolithiasis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/44Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having three double bonds between ring members or between ring members and non-ring members
    • C07D207/444Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having three double bonds between ring members or between ring members and non-ring members having two doubly-bound oxygen atoms directly attached in positions 2 and 5
    • C07D207/456Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having three double bonds between ring members or between ring members and non-ring members having two doubly-bound oxygen atoms directly attached in positions 2 and 5 with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to other ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/14Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D231/00Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings
    • C07D231/02Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings
    • C07D231/10Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members
    • C07D231/14Heterocyclic compounds containing 1,2-diazole or hydrogenated 1,2-diazole rings not condensed with other rings having two or three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D231/18One oxygen or sulfur atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D233/00Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings
    • C07D233/54Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members
    • C07D233/66Heterocyclic compounds containing 1,3-diazole or hydrogenated 1,3-diazole rings, not condensed with other rings having two double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D237/00Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings
    • C07D237/02Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings not condensed with other rings
    • C07D237/06Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members
    • C07D237/10Heterocyclic compounds containing 1,2-diazine or hydrogenated 1,2-diazine rings not condensed with other rings having three double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D237/14Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D249/00Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms
    • C07D249/02Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms not condensed with other rings
    • C07D249/041,2,3-Triazoles; Hydrogenated 1,2,3-triazoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D249/00Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms
    • C07D249/02Heterocyclic compounds containing five-membered rings having three nitrogen atoms as the only ring hetero atoms not condensed with other rings
    • C07D249/081,2,4-Triazoles; Hydrogenated 1,2,4-triazoles
    • C07D249/101,2,4-Triazoles; Hydrogenated 1,2,4-triazoles with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D249/12Oxygen or sulfur atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D407/00Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00
    • C07D407/02Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00 containing two hetero rings
    • C07D407/06Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/06Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D409/00Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms
    • C07D409/02Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings
    • C07D409/12Heterocyclic compounds containing two or more hetero rings, at least one ring having sulfur atoms as the only ring hetero atoms containing two hetero rings linked by a chain containing hetero atoms as chain links
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration

Definitions

  • Kidney stones are also a significant problem in veterinary medicine. Pets such as dogs and cats can develop stones that lead to painful urination and/or a life-threatening blockage.
  • A is C3 ⁇ 4 or S
  • B is CH or N
  • D is CH or N
  • R 1 is aryl or heteroaryl, wherein said aryl or heteroaryl has two aromatic rings, which rings are fused or directly adjoining,
  • A is CH 2 . In some embodiments, A is S. In some embodiments, B is CH. In some embodiments, B is N. In some embodiments, D is CH. In some embodiments, D is N.
  • R 1 is benzothiophene or biphenyl.
  • R 1 is selected from the group consisting of:
  • R 10 , R 1 ' and R 12 are each independently selected from the group consisting of: H, alkyl. halo and haloalkyl,
  • A is CH 2 or S
  • R 1 is aryl or heteroaryl, wherein said aryl or heteroaryl has two aromatic rings, which rings are fused or directly adjoining;
  • R 2 is H or OH
  • A is CH 2 . In some embodiments, A is S. In some embodiments, R 2 is H. In some embodiments, R 2 is Oi l.
  • R 1 is benzothiophene or biphenyl. In some embodiments, R 1 is selected from the group consisting of:
  • R 10 , R n and R 12 are each independently selected from the group consisting of: H, alkyl, halo and haloalkyl,
  • compositions comprising a compound, pharmaceutically acceptable salt or prodrug as taught herein.
  • the composition is formulated for oral administration.
  • the composition is a food product formulation.
  • kidney stones e.g., inhibiting the formation of oxalate kidney stones; treating primary hyperoxaluria
  • methods of treating kidney stones comprising: administering to a subject in need thereof a therapeutically effective amount of a compound, pharmaceutically acceptable salt or prodrug as taught herein.
  • kidney stones e.g., inhibiting the formation of oxalate kidney stones; treating primary hyperoxaluria
  • inhibiting the production of glyoxylate and/or oxalate e.g., inhibiting glycolate oxidase (GO)
  • GO glycolate oxidase
  • kidney stones e.g., inhibiting the formation of oxalate kidney stones; treating primary hyperoxaluria
  • inhibiting the production of glyoxylate and/or oxalate e.g., inhibiting glycolate oxidase (GO)
  • GO glycolate oxidase
  • FIG. 1 presents a schematic of the metabolism of 4-hydroxyproline. glycolate and glyoxylate within a hepatocyte.
  • mitochondrial enzymes are responsible for Hyp breakdown: hydroxyproline dehydrogenase (HYPDH), A'-pyrroline-S-carboxylate dehydrogenase (1P5CDH), aspartate aminotransferase (AspAT), and 4-hydroxy-2-oxoglutarate aldolase (HOGA).
  • a variety of enzymes including alanine-glyoxylate aminotransferase (AGT), D-amino acid oxidase (DAO), glyoxylate reductase (GR), and lactate dehydrogenase (LDH), can act on the glyoxylate produced from HOG cleavage.
  • AGT. GR, and HOGA are mutated within primary hyperoxaluria patients (PH type 1 , 2, and 3, respectively).
  • Glycolate oxidase (GO) can readily convert glycolate back into glyoxylate within the peroxisome; a feature that is particularly problematic for PH2 patients.
  • GO glycolate oxidase
  • Subjects or “patient” as used herein are generally mammalian subjects, including both human subjects and non-human mammalian subjects (e.g., dog, cat, horse, etc.) for research or veterinary purposes.
  • Subjects may be male or female and may be of any suitable age. including neonate, infant, juvenile, adolescent, adult, and geriatric subjects.
  • Treat refers to any type of treatment that imparts a benefit to a subject, particularly slowing or inhibiting the formation of glyoxylate and/or oxalate, decreasing urinary oxalate, slowing or inhibiting the formation of calcium oxalate stones in the kidneys and/or urinary tract (kidneys, ureters, bladder, and urethra), and/or the deposition of calcium oxalate in other tissues such as the heart.
  • the treatment may reduce the size of and/or decrease the number of such stones, inhibit or slow the growth of such stones or calcium oxalate deposition in tissues such as the heart, alleviate symptoms of such stones or deposition, etc.
  • Treatment may also include prophylactic treatment of a subject deemed to be at risk of kidney stone formation (e.g., after bariatric surgery and recurrent idiopathic "stone formers").
  • Kidney stones are hard deposits of minerals that form a stone or crystal aggregation, which may result in damage or failure of the kidney and/or urinary tract function. Most kidney stones are calcium stones, usually in the form of calcium oxalate.
  • Oxalate or "oxalic acid” is a dianion of the formula C 2 0 4 " produced by the body and also commonly ingested in the diet. Oxalate can combine with calcium in the kidneys or urinary tract to form calcium oxalate, which is the main component of most kidney stones.
  • Glyoxylate is a precursor of oxalate, as shown in FIG. 1.
  • Glycolate oxidase or "GO” is an enzyme that catalyzes the oxidation of glycolate. Multiple GO isoforms exist, such as GOl (predominantly in liver) and G02 (located in kidney and liver) (Jones et al. J Biol Chem 275. 12590-12597, 2000). GOl catalyzes the FMN- dependent oxidation of glycolate to glyoxylate. and glyoxylate to oxalate, although the latter occurs with a 100-fold lower keat/Km value (Murray et al. Biochemistry 47, 2439-2449, 2008).
  • Primary hyperoxaluria is a condition characterized by the overproduction of oxalate and/or defective production or function of one or more enzymes that regulate the levels of oxalate in the body.
  • Sufferers of Type 1 primary hyperoxaluria have a defect or shortage of the alanine:glyoxylate am i notrans ferase enzyme (AGT).
  • Type 2 primary hyperoxaluria sufferers have a defect or shortage of the glyoxylate reductase enzyme (GR).
  • Type 3 primary hyperoxaluria sufferers have a defect or shortage of the 4-hydroxy-2-oxoglutarate aldolase (HOGA).
  • Hydroxyproline is produced in the body primarily from endogenous collagen turnover (Miyata et al., Proc Natl Acad Sci USA. 1 1 1. 14406- 1441 1 , 2014). Using a unique metabolic tracer, 13 C 5 , 15 N-Hyp (all five carbons isotope and nitrogen atom labeled), it was determined that the level of Hyp turnover could be as high as 6-7 g/day (Riedel et al.. Biochim Biophys Acta 1822, 1544-1 552, 2012). Less than 5 mg of free Hyp is excreted in urine each day. indicating that most of the Hyp is metabolized (Belostotsky et al., J Mol Med ( Berl ) 90.
  • Hyp is metabolized primarily in the mitochondria of the liver and renal cortical tissue (Kivirikko, Int Rev Connect Tissue Res 5, 93-163, 1970; Atlante et al., Biochem Biophys Res Commun 202, 58-64, 1994; Monico et al., Clin J Am Soc Nepthrol 6, 2289-2295, 2011 ; Wold et al., J Food Sc 64, 377-383, 1999). Diet can also be a source of collagen. For example, a quarter pound hamburger rich in gristle could contain as much as 6 grams of collagen, yielding 780 mg of Hyp (Khan et al., J Urol 184, 1189-1196, 2010).
  • dietary Hyp can significantly increase oxalate production in humans and lead to hyperoxaluria in mouse and rat models (Khan et al., Kidney Int 70, 914-923, 2006; Valle et al., J Clin Invest 64, 1365-1370, 1979; Adams et al, Annu Rev Biochem 49, 1005-1061, 1980).
  • FIG. 1 presents the Hyp catabolic pathway and the metabolism of glyoxylate and glycolate.
  • the Hyp pathway involves four enzymatic reactions (Miyata et al., Proc Natl Acad Sci USA 11 1, 14406-14411, 2014; Efron et al., New Engl J Med 272, 1299-1309, 1965; Pelkonen et al, New Engl J Med 283, 451-456, 1970).
  • the first step of the pathway is the flavin FAD- dependent oxidation of Hyp to A'-pyrroline-3-hydroxy-5-carboxylate (3-OH-P5C) by HYPDH.
  • the 3-OH-P5C intermediate is converted to 4-hydroxy-gl utamate (4-OH-Glu) by 1P5C dehydrogenase (1P5CDH), an NAD+-dependent enzyme shared with the proline degradation pathway (Efron et al., supra).
  • Aspartate aminotransferase (AspAT) utilizes oxaloacetate to convert 4-OH-Glu to 4-hydroxy-2-oxoglutarate (HOG).
  • HOG is then cleaved by the unique HOG aldolase (HOGA) into two fragments, glyoxylate and pyruvate.
  • the glyoxylate can then be converted to glycolate and glycine via glyoxylate reductase (GR) and alanine: glyoxylate aminotransferase (AGT), respectively.
  • Glycolate can be converted back into glyoyxlate by glycolate oxidase (GO).
  • AGT, GR, and HOGA are mutated within primary hyperoxaluria patients (PH type 1, 2, and 3, respectively).
  • PH type 1, 2, and 3 primary hyperoxaluria patients
  • the glyoxylate produced from Hyp could exacerbate the already high levels of glyoxylate, and increase oxalate production via the lactate dehydrogenase (LDH).
  • LDH lactate dehydrogenase
  • HOGA is inactivated, leading to a buildup of HOG (Riedel et al., Biochim Biophys Acta 1822, 1544-1552, 2012; Belostotsky et al., J Mol Med (Berl) 90, 1497-1504, 2012).
  • inhibition of GO by a small molecule inhibitor that targets the enzyme active site is not expected to lead to any adverse side effects, and will block the formation of glyoxylate and oxalate from glycolate for all PH patient types.
  • Inhibition of GO is also expected to help idiopathic stone formers and other individuals with high urinary oxalate levels, such as those that have undergone gastric bypass surgery. For the latter, there is a significant increase in stone formation that may benefit from prophylactic treatment post surgery. While the exact origins of the oxalate in these patients has not been determined, inhibition of HYPDH will decrease glyoxylate and oxalate levels, which will ultimately reduce the glyoxylate and oxalate burden in them.
  • Active compounds as described herein can be prepared in accordance with known procedures or variations thereof that will be apparent to those skilled in the art.
  • the active compounds of the various formulas disclosed herein may contain chiral centers, e.g. , asymmetric carbon atoms, and the present disclosure is inclusive of both: (i) racemic mixtures of the active compounds, and (ii) enantiomeric forms of the active compounds.
  • the resolution of racemates into enantiomeric forms can be done in accordance with known procedures in the art.
  • the racemate may be converted with an optically active reagent into a diastereomeric pair, and the diastereomeric pair subsequently separated into the enantiomeric forms.
  • tautomers e.g., tautomers of triazole and/or pyrazole
  • rotamers e.g., rotamers
  • substituents such as those illustrated generally herein.
  • substituted refers to the replacement of hydrogen in a given structure with a substituent.
  • a substituted group may have a substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position.
  • Combinations of substituents envisioned by this invention are preferably those that result in the formation of stable compounds.
  • “Stable” as used herein refers to a chemically feasible compound that is not substantially altered when kept at a temperature of 40 °C or less, in the absence of moisture or other chemically reactive conditions, for at least a week.
  • H refers to a hydrogen atom.
  • C refers to a carbon atom.
  • N refers to a nitrogen atom.
  • S refers to a sulfur atom.
  • hydroxy refers to a group -OH.
  • Carboxy as used herein refers to a group -COOH or -COO " .
  • Halo is a halogen group selected from the group consisting of fluoro (-F), choro (-C1), bromo (-Br), and iodo (-1).
  • Haloalkyl is a halogen group connected to the parent compound by an alkyl group.
  • Alkyl refers to a saturated straight or branched chain, or cyclic hydrocarbon containing from 1 to 10 carbon atoms.
  • Representative examples of alkyl include, but are not limited to, methyl, ethyl, n-propyl, iso-propyl. n-butyl, sec-butyl, iso-butyl, tert-butyl. n-pcntyl. isopentyl, neopentyl, n-hexyl, 3 -methyl hexyl, 2,2-dimethylpentyl. 2,3-dimethylpentyl.
  • Lower alkyl as used herein, is a subset of alkyl and refers to a straight or branched chain hydrocarbon group containing from 1 to 4 carbon atoms. Representati ve examples of lower alkyl include, but are not limited to, methyl, ethyl, n-propyl, iso-propyl, n-butyl. iso-butyl. tert-butyl, cyclopropyl, cyclobutyl, and the like.
  • the alkyl groups may be optionally substituted with one or more suitable substituents, such as halo, hydroxy, carboxy, amine, etc.
  • Aryl refers to a monocyclic carbocyclic ring system or a bicyclic carbocyclic fused or directly adjoining ring system having one or more aromatic rings. Examples include, but are not limited to, phenyl, indanyl, indenyl, tetrahydronaphthyl, and the like. As noted, in some embodiments, the aryl has two aromatic rings, which rings are fused or directly adjoining. Examples include, but are not limited to, biphcnyl, napthyl. azulenyl, etc. The aryl may be optionally substituted with one or more suitable substituents, such as alkyl, halo, hydroxy, carboxy, amine, etc.
  • Heteroaryl refers to a monovalent aromatic group having a single ring or two fused or directly adjoining rings and containing in at least one of the rings at least one heteroatom (typically 1 to 3) independently selected from nitrogen, oxygen and sul ur. Examples include, but are not limited to, pyrrole, imidazole, thiazole, oxazole, furan, thiophene, triazole, pyrazole, isoxazole, isothiazole, pyridine, pyrazine, pyridazine, pyrimidine, triazine, and the like. As noted, in some embodiments, the heteroaryl has two aromatic rings, which rings are fused or directly adjoining.
  • Examples include, but are not limited to, benzothiophene, benzofuran, indole, benzoimidazole, benzthiazole, quinoline, isoquinoline, quinazoline, quinoxaline, phenyl-pyrrole, phenyl-thiophene, etc.
  • the heteroaryl may be optionally substituted with one or more suitable substituents, such as alkyl, halo, hydroxy, carboxy, amine, etc.
  • a "pharmaceutically acceptable salt” is a salt that retains the biological effectiveness of the free acids or bases of a specified compound and that is not biologically or otherwise undesirable.
  • pharmaceutically acceptable salts may include sulfates, pyrosulfates, bisulfates, sulfites, bisulfites. phosphates, monohydrogenphosphates, dihydrogenphosphates.
  • metaphosphates pyrophosphates, chlorides, bromides, iodides, acetates, propionates, decanoates, 10 caprylates, acrylates, formates, isobutyrates, caproates, heptanoates, propiolates, oxalates, malonates, succinates, suberates, sebacates, fumarates, maleates, butyne-l ,4-dioates, hexyne-1 ,6- dioates, benzoates, chlorobenzoates, methylbenzoates, dinitrobenzoates, hydroxybenzoates, methoxybenzoates, phthalates, sulfonates, xylenesulfonates, phenylacetates, phenylpropionates, phenylbutyrates, citrates, lactates, gamma-hydroxybutyrates, glycol lates. tartrates, methanol s sulfonates,
  • a “prodrug” is a compound that is converted under physiological conditions or by solvolysis or metabolically to a compound that is pharmaceutically active.
  • a thorough discussion is provided in T. Higuehi and V. Stella, Prodrugs as Novel delivery Systems. Vol. 14 of the 0 A.C.S. Symposium Series and in Edward B. Roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated by reference herein in their entirety. See also Huttunen et al., "Prodrugs - from Serendipity to Rational Design.” Pharmacological Reviews 63(3):750-771 (2011), which is incorporated by reference herein.
  • Example prodrugs include, but are not limited to, the addition 5 of/conversion to phosphate(s), amino acid esters, amino acid amides, sugar derivatives, alkyl or aryl esters, etc., at an -OH, -SH, -NH or -COOH group of the parent active compound.
  • A is CH 2 or S
  • B is CH or N
  • D is CH or N
  • R 1 is aryl or heteroaryl, wherein said aryl or heteroaryl has two aromatic rings, which rings are fused or directly adjoining,
  • A is C3 ⁇ 4. In some embodiments. A is S. In some embodiments, B is CH. In some embodiments. B is N. In some embodiments. D is CH. In some embodiments, D is N.
  • R is benzothiophene or biphenyl.
  • R is selected from the group consisting of:
  • R 10 , R 11 and R 12 are each independently selected from the group consisting of: H, alkyl, halo and haloalkyl.
  • A is CH 2 or S
  • R 1 is aryl or heteroaryl, wherein said aryl or heteroaryl has two aromatic rings, which rings are fused or directly adjoining;
  • R 2 is H or OH
  • A is C I b. In some embodiments. A is S.
  • R is benzothiophcne or biphenyl.
  • R 1 is selected from the group consisting of:
  • R 10 , R 11 and R 12 are each independently selected from the group consisting of: H, alkyl, halo and haloalkyl.
  • the active compounds described herein may be formulated for administration in a pharmaceutical carrier in accordance with known techniques. See, e.g., Remington, The Science and Practice of Pharmacy (9 th Ed. 1995).
  • the active compound (including the physiologically acceptable salts or prodrugs thereof) is typically admixed with, inter alia, an acceptable carrier.
  • the carrier must, of course, be acceptable in the sense of being compatible with any other ingredients in the formulation and must not be deleterious to the patient.
  • the carrier may be a solid or a liquid, or both, and is preferably formulated with the compound as a unit-dose formulation, for example, a tablet, which may contain from 0.01 or 0.5% to 95% or 99% by weight of the active agent.
  • One or more active agents may be incorporated in the formulations of the invention, which may be prepared by any of the well-known techniques of pharmacy comprising admixing the components, optionally including one or more accessory ingredients.
  • the pharmaceutical compositions may also contain other additives, such as pH-adj listing additives.
  • useful pH-adjusting agents include acids, such as hydrochloric acid, bases and/or buffers, such as sodium lactate, sodium acetate, sodium phosphate, sodium citrate, sodium borate, or sodium gluconate.
  • the compositions may contain preservatives.
  • Useful preservatives include methyl paraben, propylparaben, benzoic acid and benzyl alcohol.
  • the formulations may comprise nanoparticles, such as biodegradable polymers and/or liposome-forming material, for encapsulation and/or delivery of the active agent(s).
  • nanoparticles such as biodegradable polymers and/or liposome-forming material
  • WO 2014/201312 to Wang et al; Cho and Jung.
  • Molecules 20:19620-19646 2015
  • Nogueira et al. “Design of liposomal formulations for cell targeting," Colloids Surf B Biointerfaces 136:514-526, 2015.
  • liver-targeting nanoparticles may be used for specific delivery of active agent(s) acting at the liver.
  • kidney-targeting nanoparticles may be used for specific delivery of active agent(s) acting at the kidney. See, e.g., U.S. Patent No. 8,318,199 to Kim et al.; U.S. 2012/0196807 to Nakamura et al.
  • the active agent(s) may be provided in a controlled-release or sustained-release formulation. See, e.g., Grinyo and Petruzzelli, "Once-daily LCP-Tacro MeltDose tacrolimus for the prophylaxis of organ rejection in kidney and liver transplantations," Expert Review of Clinical Immunology 10( 12): 1 567-1579, 2014 (Erratum: Expert Review of Clinical Immunology 1 1(4):547, 2015).
  • Formulations of the invention may include those suitable for oral, buccal (sub-lingual), parenteral (e.g. , subcutaneous, intramuscular, intradermal, or intravenous), topical (i.e., both skin and mucosal surfaces, including airway surfaces) and transdermal administration, although the most suitable route in any given case will depend on the nature and severity of the condition being treated and on the nature of the particular active compound being used.
  • Formulations suitable for oral administration may be presented in discrete units, such as capsules, cachets, lozenges, or tablets, each containing a predetermined amount of the active compound(s); as a powder or granules; as a solution or a suspension in an aqueous or nonaqueous liquid; or as an oil -in- water or water-in-oil emulsion.
  • Such formulations may be prepared by any suitable method of pharmacy which includes the step of bringing into association the active compound and a suitable carrier (which may contain one or more accessory ingredients as noted above).
  • the formulations of the invention are prepared by uniformly and intimately admixing the active compound with a liquid or finely divided solid carrier, or both, and then, if necessary, shaping the resulting mixture.
  • a tablet may be prepared by compressing or molding a powder or granules containing the active compound, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared by compressing, in a suitable machine, the compound in a free-flowing form, such as a powder or granules optionally mixed with a binder, lubricant, inert diluent, and/or surface active/dispersing agent(s).
  • Molded tablets may be made by molding, in a suitable machine, the powdered compound moistened with an inert liquid binder.
  • Formulations suitable for oral administration also include food product formulations, such as a nutritional bar or an animal feed (e.g., pet food such as dog or cat food).
  • Food product formulations may include one or more of carbohydrates such as wheat, corn rice, barley or oats, dairy products such as milk, oils such as canola oil or soybean oil. flavorants such as sugar or syrup, coloring, chocolate, preservatives, etc.
  • Pet food formulations in particular, may include meat, poultry, fish or other animal-derived components such as eggs.
  • Formulations suitable for buccal (sub-lingual) administration include lozenges comprising the active compound in a flavored base, usually sucrose and acacia or tragacanth; and pastilles comprising the compound in an inert base such as gelatin and glycerin or sucrose and acacia.
  • Formulations of the present invention suitable for parenteral administration comprise sterile aqueous and non-aqueous injection solutions, which preparations are preferably isotonic with the blood of the intended recipient. These preparations may contain anti-oxidants. buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient.
  • Aqueous and non-aqueous sterile suspensions may include suspending agents and thickening agents.
  • the formulations may be presented in unit ⁇ dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dricd (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example, saline or water-for-injection immediately prior to use.
  • Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described.
  • an injectable, stable, sterile composition comprising an active compound(s) in a unit dosage form in a sealed container.
  • the active eompound(s) may be provided in the form of a lyophilizate which is capable of being reconstituted with a suitable pharmaceutically acceptable carrier to form a liquid composition suitable for injection thereof into a subject.
  • emulsifying agent which is physiologically acceptable may be employed in sufficient quantity to emulsify the compound or salt in an aqueous carrier.
  • emulsifying agent is phosphatidyl choline.
  • Formulations suitable for topical application to the skin preferably take the form of an ointment, cream, lotion, paste, gel, spray, aerosol, or oil.
  • Carriers which may be used include petroleum jelly, lanoline, polyethylene glycols, alcohols, transdermal enhancers, and combinations of two or more thereof.
  • Formulations suitable for transdermal administration may be presented as discrete patches adapted to remain in intimate contact with the epidermis of the recipient for a prolonged period of time. Formulations suitable for transdermal administration may also be delivered by iontophoresis (see, for example. Pharmaceutical Research 3 (6):318 (1986)) and typically take the form of an optionally buffered aqueous solution of the active compound. Suitable formulations comprise citrate or bisXtris buffer (pH 6) or ethanol/water and contain from 0.1 to 0.2M active ingredient.
  • the unit dosage form typically comprises from about 1 mg, 5 mg, 10 mg, 100 mg, 250 mg, 500 mg, 1 gram, 5 grams, 10 grams, or any ranges therein, of the active compound(s), depending on the subject being treated (e.g., human or non-human mammalian subject). In some embodiments, the unit dosage form is in the range of 500 mg to 10 grams, keeping in mind that a good portion of the active compound(s) may not be absorbed upon administration (e.g., oral adminstration).
  • GO inhibitor design Based on crystal structures of human GO 1 with CCPST and CDST as well as other biochemical data, GO inhibitors are designed to exploit one or more of the following interactions:
  • Example 2 Example GO inhibitors. With the above considerations in mind, the followin compounds are designed as GO inhibitors.
  • Example 3 Testing of inhibitors of GO.
  • the inhibition of recombinant, human liver GO (the H AO 1 gene product) is readily determined by a coupled assay that contains 2,6- dichloroindophenol (DCIP) (Murray et al, Biochemistry 47, 2439-2449. 2008). Briefly. GO is pre-incubated at 37 °C with or without inhibitor in 100 mM potassium phosphate pH 7.5 (0.1% DMSO final) for 5 min. An aliquot of pre-warmed DCIP and glycolate is added to start the reaction (final concentration 75 ⁇ DCIP, 3 mM glycolate). The reaction rate is determined by monitoring the decrease at 600 nm (extinction coefficient of 21 mM "1 cm "1 ). COST inhibits GO with an apparent Ki of - 15 nM.
  • DCIP 2,6- dichloroindophenol
  • Example 4 Therapy with GO inhibitor. Subjects are administered a GO inhibitor to treat kidney stones. Literature Cited.
  • Example 5 Mouse model. Mice that do not express GO have been generated. The Haol (GO) deficient animals developed normally and exhibited similar behavior to wild-type litter mates. The genotype of each mouse was confirmed by PGR analysis from a tail snip. Liver was analyzed by western analysis. These tests confirmed that the Haol homozygous mouse did not contain GO in any of the samples. As expected, GO is not present in the kidney of all mouse strains.
  • mice lacking GO appear normal apart from an increased urinary glycolate excretion and elevated plasma glycolate level.
  • the heterozygous (Htz) Haol mouse strain showed reduced expression of protein as measured by western Blot.
  • AGT alanine-glyoxylate aminotransferase

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Urology & Nephrology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

Provided herein are compounds of Formula I and Formula II, and compositions comprising the same, as well as methods of use thereof for treating kidney stones (e.g., inhibiting the formation of oxalate kidney stones; treating primary hyperoxaluria), inhibiting the production of glyoxylate and/or oxalate, and/or inhibiting glycolate oxidase (GO).

Description

Glycolate Oxidase Inhibitors and Methods of Use
for the Treatment of Kidney Stones
RELATED APPLICATIONS
This application claims the benefit of United States Provisional Patent Application Serial No. 62/263,938, filed December 7, 2015, the disclosure of which is incorporated by reference herein in its entirety.
GOVERNMENT FUNDING
This invention was made with government support under grant numbers DK083527 and DK073732 awarded by National Institutes of Health. The United States government has certain rights in the invention.
BACKGROUND
Kidney stones affect approximately 1 in 11 individuals in the United States. The 2012 National Health and Nutrition and Examination Survey (NHANES). part of the Urological Diseases in America Project, reported that the overall prevalence of kidney stones was 8.8% (10.6 % and 7.1 % for men and women, respectively) (Jiang et al., Am .1 Physiol Gastrointest Liver Physiol 302. G637-643, 2012). This study and others attest to the significant increase in stone cases in general, but especially in individuals with obesity, diabetes, and following bariatric surgery (Jiang et al., supra; Knight et al., Am J Nephrol 25, 171 -175, 2005). The direct and indirect costs associated with kidney stone treatment (i.e., nephrocalcinosis) are significant (Knight et al., Kidney Int 70. 1929-1934, 2006).
Individuals with Primary Hyperoxaluria (PH) have mutations in a variety of genes involved in glyoxylate and hydroxyproline (Hyp) metabolism that result in a significant increase in oxalate production and deposition of calci um oxalate stones, the most common type of stones for all stone formers. The treatments for these individuals range from a combined kidney-liver transplant to a life-long use of potassium citrate, increased fluid intake and dietary restriction of oxalate (Riedel et al., PLoS One 6, e26021, 201 1 ; Knight et al.. Am J Physiol-Rcnal 302, Γ688- 693, 2012). Treatments for the removal of stones currently include shock-wave lithotripsy, ureteroscopic stone removal, and percutaneous nephrolithotomy (Riedel et al.. supra). However, the recurrence of stones following the available procedures is over 50%. Kidney stones are also a significant problem in veterinary medicine. Pets such as dogs and cats can develop stones that lead to painful urination and/or a life-threatening blockage.
Considering that the current treatments only address symptoms, novel treatments to prevent the formation of stones in PH and other idiopathic stone formers are greatly needed.
SUMMARY
Provided herein are compounds of Formula I :
Figure imgf000003_0001
I
wherein:
A is C¾ or S;
B is CH or N;
D is CH or N; and
R1 is aryl or heteroaryl, wherein said aryl or heteroaryl has two aromatic rings, which rings are fused or directly adjoining,
or a pharmaceutically acceptable salt or prodrug thereof.
In some embodiments, A is CH2. In some embodiments, A is S. In some embodiments, B is CH. In some embodiments, B is N. In some embodiments, D is CH. In some embodiments, D is N.
In some embodiments, R1 is benzothiophene or biphenyl.
In some embodiments, R1 is selected from the group consisting of:
Figure imgf000004_0001
wherein R10, R1 ' and R12 are each independently selected from the group consisting of: H, alkyl. halo and haloalkyl,
or a pharmaceutically acceptable salt or prodrug thereof.
Also provided arc compounds of Formula II:
Figure imgf000004_0002
II
wherein:
A is CH2 or S;
R1 is aryl or heteroaryl, wherein said aryl or heteroaryl has two aromatic rings, which rings are fused or directly adjoining; and
R2 is H or OH,
or a pharmaceutically acceptable salt or prodrug thereof.
In some embodiments. A is CH2. In some embodiments, A is S. In some embodiments, R2 is H. In some embodiments, R2 is Oi l.
In some embodiments, R1 is benzothiophene or biphenyl. In some embodiments, R1 is selected from the group consisting of:
Figure imgf000005_0001
wherein R10, Rn and R12 are each independently selected from the group consisting of: H, alkyl, halo and haloalkyl,
or a pharmaceutically acceptable salt or prodrug thereof.
Also provided are pharmaceutical compositions comprising a compound, pharmaceutically acceptable salt or prodrug as taught herein. In some embodiments, the composition is formulated for oral administration. In some embodiments, the composition is a food product formulation.
Further provided are methods of treating kidney stones (e.g., inhibiting the formation of oxalate kidney stones; treating primary hyperoxaluria), comprising: administering to a subject in need thereof a therapeutically effective amount of a compound, pharmaceutically acceptable salt or prodrug as taught herein.
Still further provided are methods of inhibiting the production of glyoxylate and/or oxalate, and/or inhibiting glycolate oxidase (GO), in a subject in need thereof, comprising: administering to said subject a therapeutically effective amount of a compound, pharmaceutically acceptable salt or prodrug as taught herein.
Also provided is the use of the compound, pharmaceutically acceptable salt or prodrug as taught herein, or a pharmaceutical composition as taught herein, for treating kidney stones (e.g., inhibiting the formation of oxalate kidney stones; treating primary hyperoxaluria), inhibiting the production of glyoxylate and/or oxalate, and/or inhibiting glycolate oxidase (GO), in a human or non-human animal subject in need thereof.
Further provided is the use of a compound, pharmaceutically acceptable salt or prodrug as taught herein in the preparation of a medicament for treating kidney stones (e.g., inhibiting the formation of oxalate kidney stones; treating primary hyperoxaluria), inhibiting the production of glyoxylate and/or oxalate, and/or inhibiting glycolate oxidase (GO), in a human or non-human animal subject in need thereof.
BRIEF DESCRIPTION OF THE DRAWINGS FIG. 1 presents a schematic of the metabolism of 4-hydroxyproline. glycolate and glyoxylate within a hepatocyte. Four mitochondrial enzymes are responsible for Hyp breakdown: hydroxyproline dehydrogenase (HYPDH), A'-pyrroline-S-carboxylate dehydrogenase (1P5CDH), aspartate aminotransferase (AspAT), and 4-hydroxy-2-oxoglutarate aldolase (HOGA). A variety of enzymes, including alanine-glyoxylate aminotransferase (AGT), D-amino acid oxidase (DAO), glyoxylate reductase (GR), and lactate dehydrogenase (LDH), can act on the glyoxylate produced from HOG cleavage. AGT. GR, and HOGA are mutated within primary hyperoxaluria patients (PH type 1 , 2, and 3, respectively). Glycolate oxidase (GO) can readily convert glycolate back into glyoxylate within the peroxisome; a feature that is particularly problematic for PH2 patients.
DETAILED DESCRIPTION
Provided herein are methods of treatment for controlling or inhibiting the formation of kidney stones comprising administering to a subject in need thereof an inhibitor of glycolate oxidase (GO), as well as compounds and compositions useful for the same.
The disclosures of all patent references cited herein arc hereby incorporated by reference to the extent they are consistent with the disclosure set forth herein. As used herein in the description of the invention and the appended claims, the singular forms "a." "an" and "the" are intended to include the plural forms as well, unless the context clearly indicates otherwise.
"Subject" or "patient" as used herein are generally mammalian subjects, including both human subjects and non-human mammalian subjects (e.g., dog, cat, horse, etc.) for research or veterinary purposes. Subjects may be male or female and may be of any suitable age. including neonate, infant, juvenile, adolescent, adult, and geriatric subjects.
"Treat" as used herein refers to any type of treatment that imparts a benefit to a subject, particularly slowing or inhibiting the formation of glyoxylate and/or oxalate, decreasing urinary oxalate, slowing or inhibiting the formation of calcium oxalate stones in the kidneys and/or urinary tract (kidneys, ureters, bladder, and urethra), and/or the deposition of calcium oxalate in other tissues such as the heart. For example, the treatment may reduce the size of and/or decrease the number of such stones, inhibit or slow the growth of such stones or calcium oxalate deposition in tissues such as the heart, alleviate symptoms of such stones or deposition, etc.
Treatment may also include prophylactic treatment of a subject deemed to be at risk of kidney stone formation (e.g., after bariatric surgery and recurrent idiopathic "stone formers").
"Kidney stones" are hard deposits of minerals that form a stone or crystal aggregation, which may result in damage or failure of the kidney and/or urinary tract function. Most kidney stones are calcium stones, usually in the form of calcium oxalate.
"Oxalate" or "oxalic acid" is a dianion of the formula C204 " produced by the body and also commonly ingested in the diet. Oxalate can combine with calcium in the kidneys or urinary tract to form calcium oxalate, which is the main component of most kidney stones.
"Glyoxylate" is a precursor of oxalate, as shown in FIG. 1.
"Glycolate oxidase" or "GO" is an enzyme that catalyzes the oxidation of glycolate. Multiple GO isoforms exist, such as GOl (predominantly in liver) and G02 (located in kidney and liver) (Jones et al. J Biol Chem 275. 12590-12597, 2000). GOl catalyzes the FMN- dependent oxidation of glycolate to glyoxylate. and glyoxylate to oxalate, although the latter occurs with a 100-fold lower keat/Km value (Murray et al. Biochemistry 47, 2439-2449, 2008).
"Primary hyperoxaluria" is a condition characterized by the overproduction of oxalate and/or defective production or function of one or more enzymes that regulate the levels of oxalate in the body. Sufferers of Type 1 primary hyperoxaluria have a defect or shortage of the alanine:glyoxylate am i notrans ferase enzyme (AGT). Type 2 primary hyperoxaluria sufferers have a defect or shortage of the glyoxylate reductase enzyme (GR). Type 3 primary hyperoxaluria sufferers have a defect or shortage of the 4-hydroxy-2-oxoglutarate aldolase (HOGA).
"I lydroxyproline" or "Hyp" has the structure:
Figure imgf000007_0001
Hydroxyproline is produced in the body primarily from endogenous collagen turnover (Miyata et al., Proc Natl Acad Sci USA. 1 1 1. 14406- 1441 1 , 2014). Using a unique metabolic tracer, 13C5,15N-Hyp (all five carbons isotope and nitrogen atom labeled), it was determined that the level of Hyp turnover could be as high as 6-7 g/day (Riedel et al.. Biochim Biophys Acta 1822, 1544-1 552, 2012). Less than 5 mg of free Hyp is excreted in urine each day. indicating that most of the Hyp is metabolized (Belostotsky et al., J Mol Med ( Berl ) 90. 1497- 1504, 2012). This significant metabolic load could contribute up to 25% of the endogenous oxalate produced (Phang et al, (2001) Disorders of proline and hydroxyproline metabolism, in The Metabolic and Molecular Bases of Inherited Disease (Scriver, C. R., Beaudet, A. L., Sly, W. S., Vallee, D., Childs, B., Kinzler, K. W., and Vogelstein, B. eds.), McGraw-Hill, New York, pp 1821-1838). The biological reason why Hyp metabolism occurs is not clear, although it does enable some pyruvate to feed back into other pathways.
Hyp is metabolized primarily in the mitochondria of the liver and renal cortical tissue (Kivirikko, Int Rev Connect Tissue Res 5, 93-163, 1970; Atlante et al., Biochem Biophys Res Commun 202, 58-64, 1994; Monico et al., Clin J Am Soc Nepthrol 6, 2289-2295, 2011 ; Wold et al., J Food Sc 64, 377-383, 1999). Diet can also be a source of collagen. For example, a quarter pound hamburger rich in gristle could contain as much as 6 grams of collagen, yielding 780 mg of Hyp (Khan et al., J Urol 184, 1189-1196, 2010). In fact, dietary Hyp can significantly increase oxalate production in humans and lead to hyperoxaluria in mouse and rat models (Khan et al., Kidney Int 70, 914-923, 2006; Valle et al., J Clin Invest 64, 1365-1370, 1979; Adams et al, Annu Rev Biochem 49, 1005-1061, 1980).
FIG. 1 presents the Hyp catabolic pathway and the metabolism of glyoxylate and glycolate. The Hyp pathway involves four enzymatic reactions (Miyata et al., Proc Natl Acad Sci USA 11 1, 14406-14411, 2014; Efron et al., New Engl J Med 272, 1299-1309, 1965; Pelkonen et al, New Engl J Med 283, 451-456, 1970). The first step of the pathway is the flavin FAD- dependent oxidation of Hyp to A'-pyrroline-3-hydroxy-5-carboxylate (3-OH-P5C) by HYPDH. The 3-OH-P5C intermediate is converted to 4-hydroxy-gl utamate (4-OH-Glu) by 1P5C dehydrogenase (1P5CDH), an NAD+-dependent enzyme shared with the proline degradation pathway (Efron et al., supra). Aspartate aminotransferase (AspAT) utilizes oxaloacetate to convert 4-OH-Glu to 4-hydroxy-2-oxoglutarate (HOG). HOG is then cleaved by the unique HOG aldolase (HOGA) into two fragments, glyoxylate and pyruvate. The glyoxylate can then be converted to glycolate and glycine via glyoxylate reductase (GR) and alanine: glyoxylate aminotransferase (AGT), respectively. Glycolate can be converted back into glyoyxlate by glycolate oxidase (GO).
AGT, GR, and HOGA are mutated within primary hyperoxaluria patients (PH type 1, 2, and 3, respectively). For PHI and PH2 patients, the glyoxylate produced from Hyp could exacerbate the already high levels of glyoxylate, and increase oxalate production via the lactate dehydrogenase (LDH). For PH3 patients, HOGA is inactivated, leading to a buildup of HOG (Riedel et al., Biochim Biophys Acta 1822, 1544-1552, 2012; Belostotsky et al., J Mol Med (Berl) 90, 1497-1504, 2012). Recent studies identified that HOG can inhibit GR, potentially leading to a PH2-like phenotype (Riedel et al., Biochim Biophys Acta 1822, 1544-1552, 2012). In contrast, glycolic aciduria, caused by deficiencies in GO, is not associated with any overt consequences, and glycolate can be excreted (Frishberg et al. J Med Genet 51, 526-529, 2014).
Thus, and without wishing to be bound by theory, inhibition of GO by a small molecule inhibitor that targets the enzyme active site is not expected to lead to any adverse side effects, and will block the formation of glyoxylate and oxalate from glycolate for all PH patient types. Inhibition of GO is also expected to help idiopathic stone formers and other individuals with high urinary oxalate levels, such as those that have undergone gastric bypass surgery. For the latter, there is a significant increase in stone formation that may benefit from prophylactic treatment post surgery. While the exact origins of the oxalate in these patients has not been determined, inhibition of HYPDH will decrease glyoxylate and oxalate levels, which will ultimately reduce the glyoxylate and oxalate burden in them.
1. Active compounds.
Active compounds as described herein can be prepared in accordance with known procedures or variations thereof that will be apparent to those skilled in the art.
As will be appreciated by those of skill in the art, the active compounds of the various formulas disclosed herein may contain chiral centers, e.g. , asymmetric carbon atoms, and the present disclosure is inclusive of both: (i) racemic mixtures of the active compounds, and (ii) enantiomeric forms of the active compounds. The resolution of racemates into enantiomeric forms can be done in accordance with known procedures in the art. For example, the racemate may be converted with an optically active reagent into a diastereomeric pair, and the diastereomeric pair subsequently separated into the enantiomeric forms.
Also included in active compounds disclosed herein are tautomers (e.g., tautomers of triazole and/or pyrazole) and rotamers.
As described herein, certain groups or portions of the compounds of the invention may optionally be substituted with one or more substituents, such as those illustrated generally herein. In general, the term "substituted" refers to the replacement of hydrogen in a given structure with a substituent. Unless otherwise indicated, a substituted group may have a substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position. Combinations of substituents envisioned by this invention are preferably those that result in the formation of stable compounds. "Stable" as used herein refers to a chemically feasible compound that is not substantially altered when kept at a temperature of 40 °C or less, in the absence of moisture or other chemically reactive conditions, for at least a week.
As used herein in the accompanying chemical structures, "H" refers to a hydrogen atom. "C" refers to a carbon atom. "N" refers to a nitrogen atom. "S" refers to a sulfur atom.
The term "hydroxy," as used herein, refers to a group -OH.
"Carbonyl" is a group having a carbon atom double-bonded to an oxygen atom (C=0).
"Carboxy" as used herein refers to a group -COOH or -COO ".
"Amine" or "amino" refers to a group— NH2.
"Halo" is a halogen group selected from the group consisting of fluoro (-F), choro (-C1), bromo (-Br), and iodo (-1). "Haloalkyl" is a halogen group connected to the parent compound by an alkyl group.
"Alkyl," as used herein, refers to a saturated straight or branched chain, or cyclic hydrocarbon containing from 1 to 10 carbon atoms. Representative examples of alkyl include, but are not limited to, methyl, ethyl, n-propyl, iso-propyl. n-butyl, sec-butyl, iso-butyl, tert-butyl. n-pcntyl. isopentyl, neopentyl, n-hexyl, 3 -methyl hexyl, 2,2-dimethylpentyl. 2,3-dimethylpentyl. n-heptyl, n-octyl, n-nonyl, n-decyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like. "Lower alkyl" as used herein, is a subset of alkyl and refers to a straight or branched chain hydrocarbon group containing from 1 to 4 carbon atoms. Representati ve examples of lower alkyl include, but are not limited to, methyl, ethyl, n-propyl, iso-propyl, n-butyl. iso-butyl. tert-butyl, cyclopropyl, cyclobutyl, and the like. The alkyl groups may be optionally substituted with one or more suitable substituents, such as halo, hydroxy, carboxy, amine, etc.
"Aryl," as used herein, refers to a monocyclic carbocyclic ring system or a bicyclic carbocyclic fused or directly adjoining ring system having one or more aromatic rings. Examples include, but are not limited to, phenyl, indanyl, indenyl, tetrahydronaphthyl, and the like. As noted, in some embodiments, the aryl has two aromatic rings, which rings are fused or directly adjoining. Examples include, but are not limited to, biphcnyl, napthyl. azulenyl, etc. The aryl may be optionally substituted with one or more suitable substituents, such as alkyl, halo, hydroxy, carboxy, amine, etc.
"Heteroaryl," as used herein, refers to a monovalent aromatic group having a single ring or two fused or directly adjoining rings and containing in at least one of the rings at least one heteroatom (typically 1 to 3) independently selected from nitrogen, oxygen and sul ur. Examples include, but are not limited to, pyrrole, imidazole, thiazole, oxazole, furan, thiophene, triazole, pyrazole, isoxazole, isothiazole, pyridine, pyrazine, pyridazine, pyrimidine, triazine, and the like. As noted, in some embodiments, the heteroaryl has two aromatic rings, which rings are fused or directly adjoining. Examples include, but are not limited to, benzothiophene, benzofuran, indole, benzoimidazole, benzthiazole, quinoline, isoquinoline, quinazoline, quinoxaline, phenyl-pyrrole, phenyl-thiophene, etc. The heteroaryl may be optionally substituted with one or more suitable substituents, such as alkyl, halo, hydroxy, carboxy, amine, etc.
5 A "pharmaceutically acceptable salt" is a salt that retains the biological effectiveness of the free acids or bases of a specified compound and that is not biologically or otherwise undesirable. Examples of pharmaceutically acceptable salts may include sulfates, pyrosulfates, bisulfates, sulfites, bisulfites. phosphates, monohydrogenphosphates, dihydrogenphosphates. metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, propionates, decanoates, 10 caprylates, acrylates, formates, isobutyrates, caproates, heptanoates, propiolates, oxalates, malonates, succinates, suberates, sebacates, fumarates, maleates, butyne-l ,4-dioates, hexyne-1 ,6- dioates, benzoates, chlorobenzoates, methylbenzoates, dinitrobenzoates, hydroxybenzoates, methoxybenzoates, phthalates, sulfonates, xylenesulfonates, phenylacetates, phenylpropionates, phenylbutyrates, citrates, lactates, gamma-hydroxybutyrates, glycol lates. tartrates, methanol s sulfonates, propanesulfonates, naphthalene- 1 -sulfonates, naphthalene-2-sulfonates, and mandelates.
A "prodrug" is a compound that is converted under physiological conditions or by solvolysis or metabolically to a compound that is pharmaceutically active. A thorough discussion is provided in T. Higuehi and V. Stella, Prodrugs as Novel delivery Systems. Vol. 14 of the 0 A.C.S. Symposium Series and in Edward B. Roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated by reference herein in their entirety. See also Huttunen et al., "Prodrugs - from Serendipity to Rational Design." Pharmacological Reviews 63(3):750-771 (2011), which is incorporated by reference herein. Example prodrugs include, but are not limited to, the addition 5 of/conversion to phosphate(s), amino acid esters, amino acid amides, sugar derivatives, alkyl or aryl esters, etc., at an -OH, -SH, -NH or -COOH group of the parent active compound.
Provided herein as active compounds according to some embodiments are compounds of
Formula I:
0 I
wherein:
A is CH2 or S;
B is CH or N;
D is CH or N; and
R1 is aryl or heteroaryl, wherein said aryl or heteroaryl has two aromatic rings, which rings are fused or directly adjoining,
or a pharmaceutically acceptable salt or prodrug thereof.
In some embodiments of Formula I, A is C¾. In some embodiments. A is S. In some embodiments, B is CH. In some embodiments. B is N. In some embodiments. D is CH. In some embodiments, D is N.
In some embodiments of Formula I, R is benzothiophene or biphenyl.
In some embodiments of Formula I, R is selected from the group consisting of:
Figure imgf000012_0001
wherein R10, R11 and R12 are each independently selected from the group consisting of: H, alkyl, halo and haloalkyl.
Also provided are GO inhibitor compounds of Formula II:
Figure imgf000013_0001
II
wherein:
A is CH2 or S;
R1 is aryl or heteroaryl, wherein said aryl or heteroaryl has two aromatic rings, which rings are fused or directly adjoining; and
R2 is H or OH,
or a pharmaceutically acceptable salt or prodrug thereof.
In some embodiments of Formula II. A is C I b. In some embodiments. A is S.
In some embodiments of Formula II. R is benzothiophcne or biphenyl.
In ome embodiments of Formula II, R1 is selected from the group consisting
Figure imgf000013_0002
wherein R10, R11 and R12 are each independently selected from the group consisting of: H, alkyl, halo and haloalkyl.
2. Formulations.
The active compounds described herein may be formulated for administration in a pharmaceutical carrier in accordance with known techniques. See, e.g., Remington, The Science and Practice of Pharmacy (9th Ed. 1995). In the manufacture of a pharmaceutical formulation according to the invention, the active compound (including the physiologically acceptable salts or prodrugs thereof) is typically admixed with, inter alia, an acceptable carrier. The carrier must, of course, be acceptable in the sense of being compatible with any other ingredients in the formulation and must not be deleterious to the patient. The carrier may be a solid or a liquid, or both, and is preferably formulated with the compound as a unit-dose formulation, for example, a tablet, which may contain from 0.01 or 0.5% to 95% or 99% by weight of the active agent. One or more active agents may be incorporated in the formulations of the invention, which may be prepared by any of the well-known techniques of pharmacy comprising admixing the components, optionally including one or more accessory ingredients.
The pharmaceutical compositions may also contain other additives, such as pH-adj listing additives. In particular, useful pH-adjusting agents include acids, such as hydrochloric acid, bases and/or buffers, such as sodium lactate, sodium acetate, sodium phosphate, sodium citrate, sodium borate, or sodium gluconate. Further, the compositions may contain preservatives. Useful preservatives include methyl paraben, propylparaben, benzoic acid and benzyl alcohol.
The formulations may comprise nanoparticles, such as biodegradable polymers and/or liposome-forming material, for encapsulation and/or delivery of the active agent(s). See, e.g. , WO 2014/201312 to Wang et al; Cho and Jung. "Supramolecular Complexation f Carbohydrates for the Bioavailability Enhancement of Poorly Soluble Drugs," Molecules 20:19620-19646, 2015; Nogueira et al., "Design of liposomal formulations for cell targeting," Colloids Surf B Biointerfaces 136:514-526, 2015. In some embodiments, liver-targeting nanoparticles may be used for specific delivery of active agent(s) acting at the liver. See, e.g., U.S. 2015/0150994 to Hahn et al; U.S. 2008/0138394 to Kim et al. In some embodiments, kidney-targeting nanoparticles may be used for specific delivery of active agent(s) acting at the kidney. See, e.g., U.S. Patent No. 8,318,199 to Kim et al.; U.S. 2012/0196807 to Nakamura et al.
In some embodiments, the active agent(s) may be provided in a controlled-release or sustained-release formulation. See, e.g., Grinyo and Petruzzelli, "Once-daily LCP-Tacro MeltDose tacrolimus for the prophylaxis of organ rejection in kidney and liver transplantations," Expert Review of Clinical Immunology 10( 12): 1 567-1579, 2014 (Erratum: Expert Review of Clinical Immunology 1 1(4):547, 2015).
Formulations of the invention may include those suitable for oral, buccal (sub-lingual), parenteral (e.g. , subcutaneous, intramuscular, intradermal, or intravenous), topical (i.e., both skin and mucosal surfaces, including airway surfaces) and transdermal administration, although the most suitable route in any given case will depend on the nature and severity of the condition being treated and on the nature of the particular active compound being used.
Formulations suitable for oral administration may be presented in discrete units, such as capsules, cachets, lozenges, or tablets, each containing a predetermined amount of the active compound(s); as a powder or granules; as a solution or a suspension in an aqueous or nonaqueous liquid; or as an oil -in- water or water-in-oil emulsion. Such formulations may be prepared by any suitable method of pharmacy which includes the step of bringing into association the active compound and a suitable carrier (which may contain one or more accessory ingredients as noted above). In general, the formulations of the invention are prepared by uniformly and intimately admixing the active compound with a liquid or finely divided solid carrier, or both, and then, if necessary, shaping the resulting mixture. For example, a tablet may be prepared by compressing or molding a powder or granules containing the active compound, optionally with one or more accessory ingredients. Compressed tablets may be prepared by compressing, in a suitable machine, the compound in a free-flowing form, such as a powder or granules optionally mixed with a binder, lubricant, inert diluent, and/or surface active/dispersing agent(s). Molded tablets may be made by molding, in a suitable machine, the powdered compound moistened with an inert liquid binder.
Formulations suitable for oral administration also include food product formulations, such as a nutritional bar or an animal feed (e.g., pet food such as dog or cat food). Food product formulations may include one or more of carbohydrates such as wheat, corn rice, barley or oats, dairy products such as milk, oils such as canola oil or soybean oil. flavorants such as sugar or syrup, coloring, chocolate, preservatives, etc. Pet food formulations, in particular, may include meat, poultry, fish or other animal-derived components such as eggs.
Formulations suitable for buccal (sub-lingual) administration include lozenges comprising the active compound in a flavored base, usually sucrose and acacia or tragacanth; and pastilles comprising the compound in an inert base such as gelatin and glycerin or sucrose and acacia.
Formulations of the present invention suitable for parenteral administration comprise sterile aqueous and non-aqueous injection solutions, which preparations are preferably isotonic with the blood of the intended recipient. These preparations may contain anti-oxidants. buffers, bacteriostats and solutes which render the formulation isotonic with the blood of the intended recipient. Aqueous and non-aqueous sterile suspensions may include suspending agents and thickening agents. The formulations may be presented in unit\dose or multi-dose containers, for example sealed ampoules and vials, and may be stored in a freeze-dricd (lyophilized) condition requiring only the addition of the sterile liquid carrier, for example, saline or water-for-injection immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the kind previously described. For example, in one aspect of the present invention, there is provided an injectable, stable, sterile composition comprising an active compound(s) in a unit dosage form in a sealed container. The active eompound(s) may be provided in the form of a lyophilizate which is capable of being reconstituted with a suitable pharmaceutically acceptable carrier to form a liquid composition suitable for injection thereof into a subject.
When the active compound(s') is substantially water-insoluble, a sufficient amount of emulsifying agent which is physiologically acceptable may be employed in sufficient quantity to emulsify the compound or salt in an aqueous carrier. One such useful emulsifying agent is phosphatidyl choline.
Formulations suitable for topical application to the skin preferably take the form of an ointment, cream, lotion, paste, gel, spray, aerosol, or oil. Carriers which may be used include petroleum jelly, lanoline, polyethylene glycols, alcohols, transdermal enhancers, and combinations of two or more thereof.
Formulations suitable for transdermal administration may be presented as discrete patches adapted to remain in intimate contact with the epidermis of the recipient for a prolonged period of time. Formulations suitable for transdermal administration may also be delivered by iontophoresis (see, for example. Pharmaceutical Research 3 (6):318 (1986)) and typically take the form of an optionally buffered aqueous solution of the active compound. Suitable formulations comprise citrate or bisXtris buffer (pH 6) or ethanol/water and contain from 0.1 to 0.2M active ingredient.
The unit dosage form typically comprises from about 1 mg, 5 mg, 10 mg, 100 mg, 250 mg, 500 mg, 1 gram, 5 grams, 10 grams, or any ranges therein, of the active compound(s), depending on the subject being treated (e.g., human or non-human mammalian subject). In some embodiments, the unit dosage form is in the range of 500 mg to 10 grams, keeping in mind that a good portion of the active compound(s) may not be absorbed upon administration (e.g., oral adminstration).
The present invention is explained in greater detail in the following non-limiting examples.
EXAMPLES Example 1. Glycolate oxidase (GO) inhibitor design. Based on crystal structures of human GO 1 with CCPST and CDST as well as other biochemical data, GO inhibitors are designed to exploit one or more of the following interactions:
(1) force Wl 10 to "flip" out of the active site, causing loop 4 to become disordered;
(2) protonated nitrogen at position 3 directly interacts with the catalytic residue His260;
(3) carboxylate interaction with one or both of two conserved Arg residues.
Example 2. Example GO inhibitors. With the above considerations in mind, the followin compounds are designed as GO inhibitors.
Figure imgf000017_0001
Figure imgf000018_0001
a. R = OH b. R = H
Figure imgf000019_0001

Figure imgf000020_0001
Example 3. Testing of inhibitors of GO. The inhibition of recombinant, human liver GO (the H AO 1 gene product) is readily determined by a coupled assay that contains 2,6- dichloroindophenol (DCIP) (Murray et al, Biochemistry 47, 2439-2449. 2008). Briefly. GO is pre-incubated at 37 °C with or without inhibitor in 100 mM potassium phosphate pH 7.5 (0.1% DMSO final) for 5 min. An aliquot of pre-warmed DCIP and glycolate is added to start the reaction (final concentration 75 μΜ DCIP, 3 mM glycolate). The reaction rate is determined by monitoring the decrease at 600 nm (extinction coefficient of 21 mM"1 cm"1). COST inhibits GO with an apparent Ki of - 15 nM.
Example 4. Therapy with GO inhibitor. Subjects are administered a GO inhibitor to treat kidney stones. Literature Cited.
Adams, E., and Frank. L. (1980) Metabolism of proline and the hydroxyprolines. Annu Rev Biochem 49. 1005-1061
Belostotsky. R., Pitt, J. J., and Frishberg. Y. (2012) Primary hyperoxaluria type III— a model for studying perturbations in glyoxylate metabolism. J Mol Med (Berl) 90, 1497- 1504 Atlantc, A., Passarella, S., and Quagliariello. E. ( 1994) Spectroscopic study of hydroxyproline transport in rat kidney mitochondria. Biochem Biophys Res Commun 202, 58-64
Curhan. G. C, and Taylor, E. N. (2008) 24-h uric acid excretion and the risk of kidney stones. Kidney Int 73, 489-496
Efron, M. L., Bixby, E. M., and Pryles, C. V. ( 1965) Hydroxyprolinemia. li. A Rare Metabolic Disease Due to a Deficiency of the Enzyme "Hydroxyproline Oxidase". New Engl J Med 272, 1299- 1309 Frishberg, Y., Zeharia, A., Lyakhovetsky. R. Bargal, R. and Belostotsky, R. (2014) Mutations in Haol encoding glycolate oxidase cause isolated glycoli aciduria. J Med Genet 51, 526-529
Murray, M.S., Holmes, R.P. and Lowther, W.T. (2008) Active site loop 4 movements with human glycoalte oxidase: implications for substrate specificity and drug design.
Biochemistry 47, 2439-2449.
Jiang, J., Johnson, L. C, Knight, J., Callahan, M. F., Riedel, T. J., Holmes, R. P., and Lowther. W. T. (2012) Metabolism of [13C5 Jhydroxyproline in vitro and in vivo: implications for primary hyperoxaluria. Am J Physiol Gastrointest Liver Physiol 302, G637-643
Jones, J.M, Morrell, J.C. and Gould, S.J. (2000) Indentification and characterization of HAOXl . HOAX2. and HOAX3. three human peroxisome 2-hydorxy acid oxidases. J Biol Chem 275. 12590-12597
Khan, S. R., and Glenton, P. A. (2010) Experimental induction of calcium oxalate nephrolithiasis in mice. J Urol 184, 1189- 1 196
Khan, S. R., Glenton. P. A., and Byer, K. J. (2006) Modeling of hyperoxaluria calcium oxalate nephrolithiasis: experimental induction of hyperoxaluria by hydroxy-L-proline. Kidney Int 70, 914-923
Kivirikko. K. I. ( 1970) Urinary excretion of hydroxyproline in health and disease. Int Rev Connect Tissue Res 5, 93-163
Knight, J., and Holmes, R. P. (2005) Mitochondrial hydroxyproline metabolism:
implications for primary hyperoxaluria. Am J "Nephrol 25, 171 -175
Knight, J.. Holmes. R. P., Cramer, S. D., Takayama, T., and Salido. E. (2012)
Hydroxyproline metabolism in mouse models of primary hyperoxaluria. Am J Physiol-Renal 302, F688-693
Knight. J., Jiang, J., Assimos, D. G., and Holmes, R. P. (2006) Hydroxyproline ingestion and urinary oxalate and glycolate excretion. Kidney Int 70, 1929-1934
Lipinski, C. A., Lombardo, P., Dominy, B. W., and Feeney, P. J. (2001) Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Deliv Rev 46, 3-26
Miyata, N., Steffen, J., Johnson, M. E., Fargue, S., Danpure, C. J., and Koehler, C. M. (2014) Pharmacologic rescue of an enzyme -trafficking defect in primary hyperoxaluria 1. Proc Natl Acad Sci USA 111, 14406-14411
Monico, C. G., Rossetti, S., Belostotsky, R., Cogal, A. G., Herges, R. M., Seide, B. M., Olson, J. B.. Bergstrahl. E. J., Williams. 11. .!., Haley. W. E., Frishberg. Y.. and Milliner, D. S. (201 1) Primary hyperoxaluria type III gene HOGA1 (formerly DHDPSL) as a possible risk factor for idiopathic calcium oxalate urolithiasis. Clin J Am Soc Nepthrol 6, 2289-2295
Moxley, M. A., and Becker. D. F. (2012) Rapid reaction kinetics of proline
dehydrogenase in the multifunctional proline utilization A protein. Biochemistry 51, 511-520 Moxley, M. A., Tanner, J. J., and Becker, D. F. (2011) Steady-state kinetic mechanism of the proline:ubiquinone oxidoreductase activity of proline utilization A (Put A) from Escherichia coli. Arch Biochem Biophys 516, 1 13-120
Ostrander. E. L., Larson, J. D.. Schuermann, J. P.. and Tanner. .1. .1. (2009) A Conserved Active Site Tyrosine Residue of Proline Dehydrogenase Helps Enforce the Preference for Proline over Hydroxyproline as the Substrate. Biochemistry 48, 951-959
Pelkonen, R.. and Kivirikko, K. I. ( 1970) I hdroxyprolinemia: an apparently harmless familial metabolic disorder. New Engl J Med 283, 451-456
Pemberton, T. A., and Tanner, J. J. (2013) Structural basis of substrate selectivity of Delta(l)-pyrroline-5-carboxylate dehydrogenase (ALDH4A1): semialdehyde chain length. Arch Biochem Biophys 538, 34-40
Phang, J. M., Hu, C. A., and Valle, D. (2001) Disorders of proline and hydroxyproline metabolism, in The Metabolic and Molecular Bases of Inherited Disease (Scriver, C. R., Beaudet, A. I,, Sly, W. S., Vallee, D., Childs, B., Kinzler, K. W., and Vogelstein, B. eds.), McGraw-Hill, New York, pp 1821-1838
Riedel. T. J., Johnson, L. C, Knight. J., Hantgan, R. R.. Holmes, R. P.. and Lowther, W.
T. (2011) Structural and Biochemical Studies of Human 4-hydroxy-2-oxoglutarate Aldolase: Implications for Hydroxyproline Metabolism in Primary Hyperoxaluria. PLoS One 6, e26021
Riedel, T. J., Knight, J., Murray, M. S., Milliner, D. S., Holmes, R. P., and Lowther, W. T. (2012) 4-11 ydroxy-2-oxogl utarate aldolase inactivity in primary hyperoxaluria type 3 and glyoxylate reductase inhibition. Biochim Biophys Acta 1822, 1544-1552
Roy, A., Kucukural, A., and Zhang, Y. (2010) I-TASSER: a unified platform for automated protein structure and function prediction. Nature Protoc 5, 725-738
Srivastava, D., Schuermann, J. P., White. T. A., Krishnan. N., Sanyal. N., Hura, G. I ... Tan. A.. Henzl, M. T., Becker, D. F., and Tanner, J. J. (2010) Crystal structure of the
bifunctional proline utilization A flavoenzyme from Bradyrhizobium japonicum. Proceedings of the National Academy of Sciences of the United States of America 107, 2878-2883
Tallarita, E., Pollegioni, L., Servi, S., and Molla, G. (2012) Expression in Escherichia coli of the catalytic domain of human proline oxidase. Protein Expres Purif 82, 345-351 Vallc, D., Goodman, S. I., Harris, S. C, and Phang, J. M. (1979) Genetic evidence for a common enzyme catalyzing the second step in the degradation of proline and hydroxyproline. J Clin Invest 64, 1365-1370
White, T. A.. Krishnan, N., Becker. D. F., and Tanner. J. J. (2007) Structure and kinetics of monofunctional proline dehydrogenase from Therm us thermophilus. J Biol Chem 282, 14316- 14327
Williams, I., and Frank, L. (1975) Improved chemical synthesis and enzymatic assay of delta- l-pyrroline-5-carboxylic acid. Anal Biochem 64, 85-97
Williams, H. J., Williams, N., Spurlock, G., Norton, N., Zammit, S., Kirov. G., Owen, M. J., and O'Donovan, M. C. (2003) Detailed analysis of PRODH and PsPRODH reveals no association with schizophrenia. Am J Med Genet B Neuropsychiatr Genet 120B, 42-46
Willis. A., Bender. H. U., Steel, G., and Valle, D. (2008) PRODH variants and risk for schizophrenia. Amino Acids 35, 673-679
Wold, J. P., Lundby, F., and Egelandsdel, B. (1999) Quantification of connective tissue (hydroxyproline) in ground beef by autofluorcscence spectroscopy. J Food Sc 64, 377-383
Zhang, Y. (2008) I-TASSER server for protein 3D structure prediction. BMC
Bioinformatics 9, 40
Example 5. Mouse model. Mice that do not express GO have been generated. The Haol (GO) deficient animals developed normally and exhibited similar behavior to wild-type litter mates. The genotype of each mouse was confirmed by PGR analysis from a tail snip. Liver was analyzed by western analysis. These tests confirmed that the Haol homozygous mouse did not contain GO in any of the samples. As expected, GO is not present in the kidney of all mouse strains.
Mice lacking GO appear normal apart from an increased urinary glycolate excretion and elevated plasma glycolate level. Male mice lacking GO excreted -1.4 fold more urinary oxalate than wild type litter mates; however, female Haol deficient mice show no significant difference in urinary oxalate excretion compared to Wt litter mates. It is noted that this finding with male Haol deficient mice is not consistent with data recently published by Dr. Salido's group that showed no difference in urinary oxalate excretion between Wt and Haol deficient male mice. However, the diet used by Dr. Salido's group was different from that used in this study.
The heterozygous (Htz) Haol mouse strain showed reduced expression of protein as measured by western Blot. Given the cycle of glycolate-glyoxylate interconversion that will occur via GO and glyoxylate reductase activities in hepatocytes lacking alanine-glyoxylate aminotransferase (AGT), inhibition of GO is likely to reduce oxalate synthesis in PHI patients. The contribution of glycolate to oxalate synthesis in humans with functional AGT activity is not known; however, individuals lacking GO appear normal. Frishberg et al., J Med Genet 51(8):526-9 (2014). Therefore, strategies to reduce GO activity may provide benefit for reducing urinary oxalate excretion in patients with calcium oxalate kidney stone disease.
The foregoing is illustrative of the present invention, and is not to be construed as limiting thereof. The invention is defined by the following claims, with equivalents of the claims to be included therein.

Claims

That which is claimed is:
1. A compound of Formula I:
Figure imgf000025_0001
I
wherein:
A is CH2 or S;
B is CH or N;
D is CH or N; and
R1 is aryl or heteroaryl, wherein said aryl or heteroaryl has two aromatic rings, which rings are fused or directly adjoining,
or a pharmaceutically acceptable salt or prodrug thereof.
2. The compound of claim 1 , wherein A. is C¾.
3. The compound of claim 1, wherein A is S.
The compound of claim 1, claim 2 or claim 3, wherein B is CH. The compound of claim 1, claim 2 or claim 3, wherein B is N.
6. The compound of any one of claims 1-5, wherein D is CH.
7. The compound of any one of claims 1-5, wherein D is N.
8. The compound of any one of claims 1 -7, wherein R1 is benzothiophene or biphenyl
9. The compound of any one of claims 1 -7. wherein R is selected from the group consistin of:
Figure imgf000026_0001
alkyl, halo and haloalkyl,
or a pharmaceutically acceptable salt or prodrug thereof.
10. The compound of claim 1, wherein said compound is selected from the group consisting of:
Figure imgf000026_0002
or a pharmaceutically acceptable salt thereof.
11. The compound of claim 1, wherein said compound is selected from the group consisting of:
Figure imgf000027_0001
or a pharmaceutically acceptable salt thereof.
12. The compound of claim 1, wherein said compound is selected from the group consisting of:
Figure imgf000027_0002
or a pharmaceutically acceptable salt thereof.
13. The compound of claim 1, wherein said compound is selected from the group consisting of:
Figure imgf000028_0001
or a pharmaceutically acceptable salt thereof.
14. The compound of claim 1, wherein said compound is selected from the group consisting of:
Figure imgf000028_0002
or a pharmaceutically acceptable salt thereof.
15. The compound of claim 1, wherein said compound is selected from the group consisting of:
Figure imgf000029_0001
or a pharmaceutically acceptable salt thereof.
16. The compound of claim 1, wherein said compound is selected from the group consisting of:
Figure imgf000029_0002
or a pharmaceutically acceptable salt thereof.
17. The compound of claim 1, wherein said compound is selected from the group consisting of:
Figure imgf000030_0001
or a pharmaceutically acceptable salt thereof.
18. The compound of claim 1, wherein said compound is selected from the group consisting of:
Figure imgf000030_0002
pharmaceutically acceptable salt thereof.
A compound of Formula II:
Figure imgf000030_0003
II
wherein:
A is C¾ or S; R is aryl or heteroaryl, wherein said aryl or heteroaryl has two aromatic rings, which rings are fused or directly adjoining; and
R2 is H or OH,
or a pharmaceutically acceptable salt or prodrug thereof.
The compound of claim 19, wherein A is C¾.
The compound of claim 19, wherein A is S.
The compound of claim 19, claim 20 or claim 21, wherein R2 is H.
The compound of claim 19. claim 20 or claim 21. wherein R2 is Oi l.
The compound of any one of claims 19-23, wherein R1 is benzothiophene or
The compound of any one of claims 19-23, wherein R1 is selected from the group
Figure imgf000031_0001
wherein R10, Rn and R12 are each independently selected from the group consisting of: H, alkyl, halo and haloalkyl,
or a pharmaceutically acceptable salt or prodrug thereof.
26. The compound of claim 19, wherein said compound is selected from the group consisting of:
Figure imgf000032_0001
or a pharmaceutically acceptable salt thereof.
27. The compound of claim 19, wherein said compound is selected from the g consisting of:
Figure imgf000032_0002
or a pharmaceutically acceptable salt thereof.
28. The compound of claim 19, wherein said compound is selected from the group consisting of:
Figure imgf000033_0001
or a pharmaceutically acceptable salt thereof.
29. The compound of claim 19, wherein said compound is selected from the group consisting of:
Figure imgf000034_0001
or a pharmaceutically acceptable salt thereof.
30. A pharmaceutical composition comprising a compound, pharmaceutically acceptable salt or prodrug of any one of claims 1-29.
31. The pharmaceutical composition of claim 30, wherein said composition is formulated for oral administration.
32. The pharaiaceutical composition of claim 30, wherein said composition is a food product formulation.
33. The pharmaceutical composition of claim 30, wherein said composition is a capsule, cachet, lozenge, or tablet.
34. The pharmaceutical composition of any one of claims 30-33, wherein said formulation is provided in unit dosage form of from 1 mg to 10 grams of the compound, pharmaceutically acceptable salt or prodrug.
35. A method of treating kidney stones (e.g., inhibiting the formation of oxalate kidney stones; treating primary hyperoxaluria), comprising: administering to a subject in need thereof a therapeutically effective amount of the compound, pharmaceutically acceptable salt or prodrug of any one of claims 1-29, or the pharmaceutical composition of any one of claims 30- 34.
36. A method of inhibiting the production of glyoxylate and/or oxalate, and/or inhibiting glycolate oxidase (GO), in a subject in need thereof, comprising: administering to said subject a therapeutically effective amount of the compound, pharmaceutically acceptable salt or prodrug of any one of claims 1-29, or the pharmaceutical composition of any one of claims 30- 34.
37. The method of claim 35 or claim 36, wherein said subject is a human subject.
38. The method of claim 35 or claim 36, wherein said subject is a non-human animal subject.
39. The use of the compound, pharmaceutically acceptable salt or prodrug of any one of claims 1-29, or the pharmaceutical composition of any one of claims 30-33, for controlling or inhibiting the formation of calcium oxalate kidney stones, inhibiting the production of glyoxylate and/or oxalate, and/or inhibiting glycolate oxidase (GO), in a human or non-human animal subject in need thereof.
40. The use of the compound, pharmaceutically acceptable salt or prodrug of any one of claims 1-29, in the preparation of a medicament for controlling or inhibiting the formation of calcium oxalate kidney stones, inhibiting the production of glyoxylate and/or oxalate, and/or inhibiting glycolate oxidase (GO), in a human or non-human animal subject in need thereof.
PCT/US2016/065300 2015-12-07 2016-12-07 Glycolate oxidase inhibitors and methods of use for the treatment of kidney stones WO2017100266A1 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP16873725.2A EP3386949A4 (en) 2015-12-07 2016-12-07 Glycolate oxidase inhibitors and methods of use for the treatment of kidney stones
US15/781,571 US20200262794A1 (en) 2015-12-07 2016-12-07 Glycolate oxidase inhibitors and methods of use for the treatment of kidney stones

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201562263938P 2015-12-07 2015-12-07
US62/263,938 2015-12-07

Publications (1)

Publication Number Publication Date
WO2017100266A1 true WO2017100266A1 (en) 2017-06-15

Family

ID=59013248

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2016/065300 WO2017100266A1 (en) 2015-12-07 2016-12-07 Glycolate oxidase inhibitors and methods of use for the treatment of kidney stones

Country Status (3)

Country Link
US (1) US20200262794A1 (en)
EP (1) EP3386949A4 (en)
WO (1) WO2017100266A1 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019133770A3 (en) * 2017-12-29 2019-08-15 Biomarin Pharmaceutical Inc. Glycolate oxidase inhibitors for the treatment of disease
WO2019165159A1 (en) * 2018-02-23 2019-08-29 Oxalurx, Inc. Compounds and methods for treating oxalate-related diseases
WO2020010309A1 (en) * 2018-07-06 2020-01-09 Orfan Biotech Inc. Triazole glycolate oxidase inhibitors
CN110934867A (en) * 2019-12-25 2020-03-31 南方医科大学 Application of Hao1 inhibitor in preparation of drugs for inhibiting microenvironment formation before tumor lung metastasis and preventing and treating tumor lung metastasis
WO2020257487A1 (en) * 2019-06-19 2020-12-24 Biomarin Pharmaceutical Inc. Glycolate oxidase inhibitors for the treatment of disease
WO2021035196A1 (en) * 2019-08-22 2021-02-25 Oxalurx, Inc. Compounds and methods for treating oxalate-related diseases
EP3731840A4 (en) * 2017-12-29 2021-09-15 Orfan Biotech Inc. Glycolate oxidase inhibitors and use thereof
RU2805308C2 (en) * 2017-12-29 2023-10-13 Байомарин Фармасьютикал Инк. Glycolatoxidase inhibitors for the treatment of diseases
US11806335B2 (en) 2019-11-01 2023-11-07 Lilac Therapeutics, Inc. Heterocyclic carboxylate compounds as glycolate oxidase inhibitors

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5041554A (en) * 1989-01-31 1991-08-20 Merrell Dow Pharmaceuticals Inc. Novel aryl-or heteroaryl-1-alkyl-pyrrole-2-carboxylic acid compounds useful in treating interleukin-1 mediated conditions
US20070142452A1 (en) * 2005-12-15 2007-06-21 David Banner Fused pyrrole derivatives
US20090298905A1 (en) * 2005-04-15 2009-12-03 Fernando Pedro Cossio Nitrogenatd trans-stilbene analogs, method for the obtention and medical applications thereof
US20150038545A1 (en) * 2011-04-06 2015-02-05 Universidad Del País Vasco Hydroxyphenyl pyrrole compounds containing an hydroxamic acid as hdac inhibitors and medicinal applications thereof

Patent Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5041554A (en) * 1989-01-31 1991-08-20 Merrell Dow Pharmaceuticals Inc. Novel aryl-or heteroaryl-1-alkyl-pyrrole-2-carboxylic acid compounds useful in treating interleukin-1 mediated conditions
US20090298905A1 (en) * 2005-04-15 2009-12-03 Fernando Pedro Cossio Nitrogenatd trans-stilbene analogs, method for the obtention and medical applications thereof
US20070142452A1 (en) * 2005-12-15 2007-06-21 David Banner Fused pyrrole derivatives
US20150038545A1 (en) * 2011-04-06 2015-02-05 Universidad Del País Vasco Hydroxyphenyl pyrrole compounds containing an hydroxamic acid as hdac inhibitors and medicinal applications thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
DATABASE Pubchem [O] 12 February 2015 (2015-02-12), XP055391017, Database accession no. 234035183 *
See also references of EP3386949A4 *

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP7344878B2 (en) 2017-12-29 2023-09-14 ビオマリン プハルマセウトイカル インコーポレイテッド Glycolate oxidase inhibitors for the treatment of diseases
TWI811282B (en) * 2017-12-29 2023-08-11 美商拜奧馬林製藥公司 Glycolate oxidase inhibitors for the treatment of disease
US11384055B2 (en) 2017-12-29 2022-07-12 Biomarin Pharmaceutical Inc. Glycolate oxidase inhibitors for the treatment of disease
RU2805308C2 (en) * 2017-12-29 2023-10-13 Байомарин Фармасьютикал Инк. Glycolatoxidase inhibitors for the treatment of diseases
CN111788187A (en) * 2017-12-29 2020-10-16 生物马林药物股份有限公司 Glycolate oxidase inhibitors for disease treatment
EP3731840A4 (en) * 2017-12-29 2021-09-15 Orfan Biotech Inc. Glycolate oxidase inhibitors and use thereof
WO2019133770A3 (en) * 2017-12-29 2019-08-15 Biomarin Pharmaceutical Inc. Glycolate oxidase inhibitors for the treatment of disease
JP2021509401A (en) * 2017-12-29 2021-03-25 ビオマリン プハルマセウトイカル インコーポレイテッド Glucolic acid oxidase inhibitor for the treatment of disease
WO2019165159A1 (en) * 2018-02-23 2019-08-29 Oxalurx, Inc. Compounds and methods for treating oxalate-related diseases
US11389456B2 (en) 2018-02-23 2022-07-19 Oxalurx, Inc. Compounds and methods for treating oxalate-related diseases
JP2021530560A (en) * 2018-07-06 2021-11-11 カンテロ セラピューティクス インコーポレイティド Triazole Glycolic Acid Oxidase Inhibitor
JP7348705B2 (en) 2018-07-06 2023-09-21 カンテロ セラピューティクス,インコーポレイティド Triazole glycolate oxidase inhibitor
WO2020010309A1 (en) * 2018-07-06 2020-01-09 Orfan Biotech Inc. Triazole glycolate oxidase inhibitors
US11401246B2 (en) 2018-07-06 2022-08-02 Cantero Therapeutics, Inc. Triazole glycolate oxidase inhibitors
CN114258390A (en) * 2019-06-19 2022-03-29 生物马林药物股份有限公司 Glycolate oxidase inhibitors for disease treatment
WO2020257487A1 (en) * 2019-06-19 2020-12-24 Biomarin Pharmaceutical Inc. Glycolate oxidase inhibitors for the treatment of disease
US11504367B2 (en) 2019-08-22 2022-11-22 Oxalurx, Inc. Compounds and methods for treating oxalate-related diseases
CN114980886A (en) * 2019-08-22 2022-08-30 奥沙鲁克斯公司 Compounds and methods for treating oxalate-related diseases
WO2021035196A1 (en) * 2019-08-22 2021-02-25 Oxalurx, Inc. Compounds and methods for treating oxalate-related diseases
US11806335B2 (en) 2019-11-01 2023-11-07 Lilac Therapeutics, Inc. Heterocyclic carboxylate compounds as glycolate oxidase inhibitors
CN110934867B (en) * 2019-12-25 2022-10-21 南方医科大学 Application of Hao1 inhibitor in preparation of medicine for inhibiting microenvironment formation before tumor lung metastasis and preventing and treating tumor lung metastasis
CN110934867A (en) * 2019-12-25 2020-03-31 南方医科大学 Application of Hao1 inhibitor in preparation of drugs for inhibiting microenvironment formation before tumor lung metastasis and preventing and treating tumor lung metastasis

Also Published As

Publication number Publication date
EP3386949A1 (en) 2018-10-17
US20200262794A1 (en) 2020-08-20
EP3386949A4 (en) 2019-11-20

Similar Documents

Publication Publication Date Title
US20200262794A1 (en) Glycolate oxidase inhibitors and methods of use for the treatment of kidney stones
US20200261419A1 (en) Combinations for the treatment of kidney stones
US10092532B2 (en) Methods of modulation of branched chain acids and uses thereof
Víteček et al. Arginine-based inhibitors of nitric oxide synthase: therapeutic potential and challenges
JP5787911B2 (en) Methods for extending lifespan and delaying the onset of aging-related diseases
EP1146788B1 (en) Indomethacin amide derivatives for antiangiogenic and/or antitumorigenic use
BR112020001285A2 (en) beta-hydroxybutyrate and butanediol s-enantiomers and methods for their use
AU2016269491B2 (en) Systems, methods, and formulations for treating cancer
US10029956B2 (en) Therapies for cancer using isotopically substituted lysine
US20130150346A1 (en) Use of FAAH Inhibitors for Treating Parkinson's Disease and Restless Legs Syndrome
US20220249535A1 (en) Compositions comprising nicotinamide adenine dinucleotide-related compounds and use thereof
US20190382336A1 (en) HYPDH Inhibitors and Methods of Use for the Treatment of Kidney Stones
JP6052736B2 (en) Preventive and / or therapeutic agent for sepsis
JP2002537325A (en) Use of R-arylpropionic acids for the manufacture of a medicament for treating diseases in humans and animals, which can have a therapeutic effect by inhibiting the activation of NF-κB
EP1148783B1 (en) Converting cox inhibition compounds that are not cox-2 selective inhibitors to derivatives that are cox-2 selective inhibitors
TW201320995A (en) Anemia of cancer improving / prophylactic agent
WO2008100980A1 (en) Methods for treating or preventing inflammation using a glycerophosphate salt
JPWO2003068215A1 (en) Mitochondrial drugs
WO2020080398A1 (en) Agent for suppressing alcohol sickness or hangover caused by alcoholic beverage intake
RU2435574C1 (en) Method of treating anaemia, associated with failure of porphyrins synthesis
JP2022523308A (en) Cannabidiol and / or its derivatives for use in the treatment of mitochondrial disease
JPWO2020090570A1 (en) Light damage reducing agent
WO2014108812A2 (en) Glycine and folate as treatment for sideroblastic anemia

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 16873725

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2016873725

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2016873725

Country of ref document: EP

Effective date: 20180709