EP4178560A1 - Wasserlösliche ptba-prodrugs zur verwendung bei der hdac-hemmung und zur verbesserung der nierenerholung nach akuter nierenläsion - Google Patents

Wasserlösliche ptba-prodrugs zur verwendung bei der hdac-hemmung und zur verbesserung der nierenerholung nach akuter nierenläsion

Info

Publication number
EP4178560A1
EP4178560A1 EP21838077.2A EP21838077A EP4178560A1 EP 4178560 A1 EP4178560 A1 EP 4178560A1 EP 21838077 A EP21838077 A EP 21838077A EP 4178560 A1 EP4178560 A1 EP 4178560A1
Authority
EP
European Patent Office
Prior art keywords
substituted
acid
still
unsubstituted
amine
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
EP21838077.2A
Other languages
English (en)
French (fr)
Inventor
James Plante
William Ramage
Christopher Smith
Michael Bishop
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Klotho Therapeutics Inc
Original Assignee
Klotho Therapeutics Inc
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 Klotho Therapeutics Inc filed Critical Klotho Therapeutics Inc
Publication of EP4178560A1 publication Critical patent/EP4178560A1/de
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/06Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D211/36Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no 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
    • C07D211/40Oxygen atoms
    • C07D211/44Oxygen atoms attached in position 4
    • C07D211/46Oxygen atoms attached in position 4 having a hydrogen atom as the second substituent in position 4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C323/00Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups
    • C07C323/50Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton
    • C07C323/51Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton
    • C07C323/52Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton the carbon skeleton being acyclic and saturated
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C323/00Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups
    • C07C323/50Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton
    • C07C323/51Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton
    • C07C323/60Thiols, sulfides, hydropolysulfides or polysulfides substituted by halogen, oxygen or nitrogen atoms, or by sulfur atoms not being part of thio groups containing thio groups and carboxyl groups bound to the same carbon skeleton having the sulfur atoms of the thio groups bound to acyclic carbon atoms of the carbon skeleton with the carbon atom of at least one of the carboxyl groups bound to nitrogen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C53/00Saturated compounds having only one carboxyl group bound to an acyclic carbon atom or hydrogen
    • C07C53/15Saturated compounds having only one carboxyl group bound to an acyclic carbon atom or hydrogen containing halogen
    • C07C53/16Halogenated acetic acids
    • C07C53/18Halogenated acetic acids containing fluorine
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D205/00Heterocyclic compounds containing four-membered rings with one nitrogen atom as the only ring hetero atom
    • C07D205/12Heterocyclic compounds containing four-membered rings with one nitrogen atom as the only ring hetero atom condensed with carbocyclic rings or ring systems
    • 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
    • C07D207/04Heterocyclic 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 no double bonds between ring members or between ring members and non-ring members
    • C07D207/10Heterocyclic 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 no 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
    • C07D207/12Oxygen or sulfur atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D211/00Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings
    • C07D211/04Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D211/06Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D211/36Heterocyclic compounds containing hydrogenated pyridine rings, not condensed with other rings with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no 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
    • C07D211/40Oxygen atoms
    • C07D211/44Oxygen atoms attached in position 4
    • C07D211/48Oxygen atoms attached in position 4 having an acyclic carbon atom attached in position 4

Definitions

  • the present disclosure relates to novel prodrugs of 4-(phenylthio)butanoic acid (PTBA) and compositions comprising the same, and to methods of manufacturing and using the same, particularly for use in inhibiting histone deacetylase (HDAC) activity and/or for enhancing renal recovery following acute kidney injury (AKI), preferably through HDAC inhibition.
  • HDAC histone deacetylase
  • AKI acute kidney injury
  • AKI Severe acute kidney injury
  • AKI therapies that have been developed in experimental models when administered prior to the onset of injury have failed to show therapeutic benefit in humans.
  • AKI is a multi-factorial disorder that occurs in approximately 7% of in-patients hospital admissions. It is an independent predictor of in-hospital mortality.
  • Severe AKI requiring renal replacement therapy occurs in 4% of critically ill patients and has 50% in-patient mortality. Long term follow up studies in survivors of severe AKI indicate that approximately 12.5% become dialysis-dependent.
  • kidney disease CKD
  • WO 2012109527 the entirety of which is incorporated herein by reference, provides examples of certain useful therapies.
  • AKI is also associated with changes in genome-wide and gene-specific DNA methylation; however, the role and regulation of DNA methylation in kidney injury and repair remains largely elusive.
  • HDACi histone deacetylase inhibitor
  • PTBA 4-(phenylthio)butanoic acid
  • PTBA was shown to increase renal tubular cell proliferation, increase survival, and increase renal functional recovery in fish and various models of acute kidney injury. Immunohistological analyses suggested increased cell proliferation to be accompanied by increased epithelial-to-mesenchymal transition in the RTECs. Later, Cosentino et al. published Histone Deacetylase Inhibitor Enhances Recovery after AKI (Journal of the American Society of Nephrology, Vol. 24, Issue 6, June 2013), Skrypnyk et al. published Delayed treatment with PTBA analogs reduces postinjury renal fibrosis after kidney injury (Am J Physiol Renal Physiol.
  • Inhibitors Expands the Renal Progenitor Cells Population and improves the Rate of Recovery from acute Kidney Injury.
  • Each of the foregoing references is incorporated by herein by reference, in its entirety.
  • Developing strategies and health intervention methods based on the production and purification of compounds that inhibit HDAC (activity), and the administration of such compounds to subjects may help to ameliorate this situation and the problems associated therewith.
  • Developing strategies and health intervention methods based on the administration of small molecule prodrugs of PTBA (that inhibit HDAC activity and, thereby, improve renal recovery and/or function), especially humans and/or within an increasing aging population, may help to ameliorate this situation.
  • Embodiments of the present disclosure solve one or more of the foregoing or other problems in the art with novel compounds (e.g., so-called small molecules) that serve as (water soluble and/or non-toxic) prodrugs of PTBA, and compositions or medicaments including the same, preferably for use in inhibiting HDAC activity and/or for enhancing renal recovery following AKI or in response to (or for treating) chronic kidney disease, preferably through HDAC inhibition, and methods of manufacturing and using the same.
  • novel compounds e.g., so-called small molecules
  • compositions or medicaments including the same preferably for use in inhibiting HDAC activity and/or for enhancing renal recovery following AKI or in response to (or for treating) chronic kidney disease, preferably through HDAC inhibition, and methods of manufacturing and using the same.
  • the novel compounds are, or function as, therapeutic agents that accelerate recovery from AKI by enhancing the innate regenerative capacity of the kidney, illustratively through HDAC inhibition.
  • the present disclosure provides a number of compounds with direct application in human (and other mammal) AKI and, optionally, for CKD.
  • Embodiments of the present disclosure include compounds, compositions comprising said compounds, medicaments comprising said compounds or compositions, methods of manufacturing said medicaments, compounds, and compositions, and uses of said medicaments, compounds, and compositions, or methods of using said medicaments, compounds, and compositions.
  • Some embodiments include a method of treating kidney injury or disease or improving kidney function in patients is provided.
  • the method comprising administering to a patient an amount of a compound, composition, or medicament as described herein.
  • the administered amount is effective to treat kidney injury or disease or to improve kidney function in a patient.
  • the compound or pharmaceutically acceptable salt thereof can be administered in an amount and in a dosage regimen effective to improve kidney function in a patient, to inhibit a histone deacetylase (activity) in a cell (e.g., renal cell) or systemically, to expand renal progenitor cells, and/or to stimulate kidney repair in cells (in vitro, ex vivo or in vivo).
  • the manufacture and/or use of any compound, composition, and/or medicament described herein also is provided, preferably for treating kidney injury or disease or improving kidney function in a patient.
  • Embodiments of the present disclosure are designed to be effective for use as prodrugs of 4-(phenylthio)butanoic acid (PTBA) and/or for use in inhibiting histone deacetylase (HD AC) activity. Inhibition of HD AC activity has been shown to have several downstream and/or therapeutic effects. Those skilled in the art will appreciate that any disease or condition that is caused, worsened, or exacerbated, in whole or in part, by (or associated with) high or excessive HDAC activity, may be addressed and/or treated (post diagnosis or prophylactically) by administration of the novel compound(s), or composition(s) comprising the same, disclosed herein. [0011] Embodiments of the present disclosure include prodrugs of PTBA.
  • Illustrative embodiments of the present disclosure include a compound according to Formula I: or a stereoisomer, tautomer or pharmaceutically acceptable salt thereof.
  • Ri is O or NH. In some embodiments, Ri is O. In some embodiments, Ri is NH.
  • R2 is not H. In some embodiments, R2 is not CH3. In some embodiments, R2 is not, one or more of, alkyl, ethyl, propyl, isopropyl, or butyl. In some embodiments, R2 is not aryl, phenyl or benzyl. In some embodiments, R2 is not carboxyl (COOH). In some embodiments, R2 is not, one or more of, methanol, ethanol, propanol, or isopropanol. In some embodiments, when Ri is O, then R2 is not H. In some embodiments, when Ri is O, then R2 is not CH3.
  • R2 when Ri is O, then R2 is not, one or more of, alkyl, ethyl, propyl, isopropyl, or butyl. In some embodiments, when Ri is O, then R2 is not aryl, phenyl or benzyl. In some embodiments, when Ri is O, then R2 is not carboxyl (COOH). In some embodiments, when Ri is NH, then R2 is not H. In some embodiments, when Ri is NH, then R2 is not CH3. In some embodiments, when Ri is NH, then R2 is not, one or more of, alkyl, ethyl, propyl, isopropyl, or butyl. In some embodiments, when Ri is NH, then R2 is not aryl, phenyl or benzyl. In some embodiments, when Ri is NH, then R2 is not carboxyl (COOH).
  • R2 is selected from the group consisting of:
  • substituted or unsubstituted amine preferably substituted or unsubstituted tertiary amine or quaternary amine (trisubstituted ammonium or quaternary ammonium), more preferably l-(N,N-dimethyl)ethanamine, l-(l-methyl-N,N-dimethyl)ethanamine, or 1- (N,N,N-trimethyl)ethanamine,
  • substituted or unsubstituted heterocyclic amine or azaspiroalkyl or hetero- dualcycloalkyl, preferably 5-7 member heterocyclic amine or azaspiroalkyl, optionally substituted at one or more ring positions with C1-C2 alkyl, or 4-7 member substituted or unsubstituted heterocycloalkyl or hetero-dualcycloalkyl (a.k.a.
  • azaspiroalkyl optionally substituted at 1-4 (ring) positions and each (ring) substituent is selected from the group consisting of branched or unbranched C1-C3 substituted or unsubstituted alkyl or cycloalkyl, more preferably substituted or unsubstituted pyrrolidine, piperidine, or piperazine, still more preferably, substituted or unsubstituted 3-N-pyrrolidine, 4-N-piperidine, or 4-N-piperazine, still more preferably, 4-N-piperidine or 4-N-piperazine substituted at C-l, N-4, or C-l and N- 4, still more preferably, 4-N-methylpiperidine, 4-N-ethylpiperidine, 4-N-l,4- methylpiperidine, 4-N-methylpiperazine, or substituted or unsubstituted azaspiroheptyl, preferably 2-azaspiroheptyl or azaspiro[3.3]heptyl,
  • alkoxyalcohol preferably alkoxy ethanol or ethoxy alcohol, more preferably 2- ethoxy ethanol
  • polyol preferably diol, more preferably propanediol, still more preferably 2,3- propanediol
  • carboxylic acid preferably dicarboxylic acid, more preferably butanedioic acid, still more preferably, butanedioic acid,
  • amide preferably substituted amide, more preferably N-ethyl-N-amide, N-propyl- N-amide, or 2-amino-propanamide, still more preferably 2,6-diamino-N-ethyl-N-hexanamide, 2-amino-3-methyl-N-propyl-N-butanamide, or N-(l,3-dicarboyl)-2-amino-propanamide, or wherein the amide comprises:
  • R3 is substituted alkyl, preferably substituted ethyl, more preferably amino ethyl, still more preferably 2-amino ethyl
  • R4 is carboxylic acid, preferably dicarboxylic acid, more preferably pentanedioic acid, still more preferably, pentanedioic acid or pentanedioic acid, or
  • R5 is substituted alkyl, ethyl or propyl
  • R6 is substituted alkyl, preferably l-amino-2-methylpropane or 1,5-diaminopentane
  • azaspiroalkyl optionally substituted at 1-4 (ring) positions and each (ring) substituent is selected from the group consisting of branched or unbranched C1-C3 substituted or unsubstituted alkyl or cycloalkyl, preferably substituted or unsubstituted pyrrolidine, piperidine, or piperazine, more preferably, substituted or unsubstituted 3-N-pyrrolidine, 4-N- piperidine, or 4-N -piperazine, still more preferably, 4-N-piperidine or 4-N-piperazine substituted at C-l, N-4, or C-l and N-4, still more preferably, 4-N-methylpiperidine, 4-N- ethylpiperidine, 4-N- 1,4-methylpiperi dine, 4-N-methylpiperazine, most preferably, 4-N- methylpiperazine, preferably when X is (CH2)2, or tertiary amine or quaternary amine (trisubstituted
  • each Rx is, independently, selected from the group consisting of H or C3 ⁇ 4, each R9 is, independently, selected from: H, hydroxyl (OH), carboxyl (COOH), amino (NH2), or together with Rio, forms substituted or unsubstituted heterocyclic amine or azaspiroalkyl (or hetero-dualcycloalkyl), preferably 5-7 member heterocyclic amine or azaspiroalkyl, optionally substituted at one or more ring positions with C1-C2 alkyl, more preferably 5-6 member heterocyclic amine or 7 member azaspiroalkyl, optionally substituted at one or more ring positions with C1-C2 alkyl, substituted or unsubstituted pyrrolidine, piperidine, or piperazine, still more preferably, substituted or unsubstituted 3-N-pyrrolidine, 4-N-piperidine, or 4-N
  • heterocyclic amine preferably 6 member heterocyclic amine, still more preferably 6 member heterocyclic amine, still more preferably piperazine, still more preferably, 4-N-piperazine, still more preferably 4-N-methylpiperazine or 4-N- methy 1- 1 -piperazine,
  • alkanolamine preferably ethanolamine, N-ethanolamine or 1-aminoethanol
  • alkoxyalcohol preferably alkoxy ethanol or ethoxy alcohol
  • polyol preferably diol, more preferably ethanediol
  • carboxyl preferably COOH
  • amide preferably substituted amide, more preferably substituted N-pentanamide, N-hexanamide, or 1 -amide, more preferably 2,6-diamino-N-hexanamide, 2-amino-3-methyl- N-butanamide, or N-(l,3-dicarboyl)-2-amino-l-amide, or wherein the amide comprises:
  • R9 together with R9, forms substituted or unsubstituted heterocyclic amine or azaspiroalkyl (or hetero-dualcycloalkyl), preferably 5-7 member heterocyclic amine or azaspiroalkyl, optionally substituted at one or more ring positions with C1-C2 alkyl, more preferably 5-6 member heterocyclic amine or 7 member azaspiroalkyl, optionally substituted at one or more ring positions with C1-C2 alkyl, substituted or unsubstituted pyrrolidine, piperidine, or piperazine, still more preferably, substituted or unsubstituted 3-N-pyrrolidine, 4-N-piperidine, or 4-N-piperazine, still more preferably unsubstituted 3-N-pyrrolidine or substituted 4-N-piperidine or 4-N-piperazine, substituted at C-l, N-4, or C-l and N-4, still more preferably, 4-N-methylpiperidine, 4-N
  • R2 is selected from the group consisting of: [0039] substituted or unsubstituted amine, preferably substituted or unsubstituted tertiary amine or quaternary amine (trisubstituted ammonium or quaternary ammonium), more preferably l-(N,N-dimethyl)ethanamine, l-(l-methyl-N,N-dimethyl)ethanamine, or 1- (N,N,N-trimethyl)ethanamine, [0040] substituted or unsubstituted heterocyclic amine or azaspiroalkyl (or hetero- dualcycloalkyl), preferably 5-7 member heterocyclic amine or azaspiroalkyl, optionally substituted at one or more ring positions with C1-C2 alkyl, or 4-7 member substituted or unsubstituted heterocycloalkyl or hetero-dualcycloalkyl (a.k.a) substituted or unsubstituted amine,
  • azaspiroalkyl optionally substituted at 1-4 (ring) positions and each (ring) substituent is selected from the group consisting of branched or unbranched C1-C3 substituted or unsubstituted alkyl or cycloalkyl, more preferably substituted or unsubstituted pyrrolidine, piperidine, or piperazine, still more preferably, substituted or unsubstituted 3-N-pyrrolidine, 4-N-piperidine, or 4-N-piperazine, still more preferably, 4-N-piperidine or 4-N-piperazine substituted at C-l, N-4, or C-l and N- 4, still more preferably, 4-N-methylpiperidine, 4-N-ethylpiperidine, 4-N-l,4- methylpiperidine, 4-N-methylpiperazine, or substituted or unsubstituted azaspiroheptyl, preferably 2-azaspiroheptyl or azaspiro[3.3]heptyl,
  • alkanolamine preferably ethanolamine, more preferably N-ethylethanolamine or 2- (ethylamino)ethanol,
  • alkoxyalcohol preferably alkoxy ethanol or ethoxy alcohol, more preferably 2- ethoxy ethanol,
  • polyol preferably diol, more preferably propanediol, still more preferably 2,3- propanediol,
  • carboxylic acid preferably dicarboxylic acid, more preferably butanedioic acid, still more preferably, butanedioic acid or butanedioic acid,
  • amide preferably substituted amide, more preferably N-ethyl-N-amide, N-propyl- N-amide, or 2-amino-propanamide, still more preferably 2,6-diamino-N-ethyl-N-hexanamide, 2-amino-3-methyl-N-propyl-N-butanamide, or N-(l,3-dicarboyl)-2-amino-propanamide, or wherein the amide comprises:
  • R3 is substituted alkyl, preferably substituted ethyl, more preferably amino ethyl, still more preferably 2-amino ethyl
  • R4 is carboxylic acid, preferably dicarboxylic acid, more preferably pentanedioic acid, still more preferably, pentanedioic acid or pentanedioic acid,
  • R5 is substituted alkyl, ethyl or propyl
  • R6 is substituted alkyl, preferably l-amino-2-methylpropane or 1,5-diaminopentane
  • X-R.7 wherein X is C1-C3 branched or unbranched alkyl, and wherein R is a 4-7 member substituted or unsubstituted heterocycloalkyl or hetero-dualcycloalkyl (a.k.a.
  • azaspiroalkyl optionally substituted at 1-4 (ring) positions and each (ring) substituent is selected from the group consisting of branched or unbranched C1-C3 substituted or unsubstituted alkyl or cycloalkyl, preferably substituted or unsubstituted pyrrolidine, piperidine, or piperazine, more preferably, substituted or unsubstituted 3-N-pyrrolidine, 4-N- piperidine, or 4-N -piperazine, still more preferably, 4-N-piperidine or 4-N-piperazine substituted at C-l, N-4, or C-l and N-4, still more preferably, 4-N-methylpiperidine, 4-N- ethylpiperidine, 4-N- 1,4-methylpiperi dine, 4-N-methylpiperazine, most preferably, 4-N- methylpiperazine, preferably when X is (CH2)2, or tertiary amine or quaternary amine (trisubstituted
  • R2 is selected from the group consisting of: [0051] substituted or unsubstituted amine, preferably substituted or unsubstituted tertiary amine or quaternary amine (trisubstituted ammonium or quaternary ammonium), more preferably l-(N,N-dimethyl)ethanamine, l-(l-methyl-N,N-dimethyl)ethanamine, or 1- (N,N,N-trimethyl)ethanamine,
  • substituted or unsubstituted heterocyclic amine or azaspiroalkyl preferably 5-7 member heterocyclic amine or azaspiroalkyl, optionally substituted at one or more ring positions with C1-C2 alkyl
  • alkanolamine preferably ethanolamine, more preferably N-ethylethanolamine or 2-
  • alkoxyalcohol preferably alkoxy ethanol or ethoxy alcohol, more preferably 2- ethoxy ethanol
  • polyol preferably diol, more preferably propanediol, still more preferably 2,3- propanediol
  • carboxylic acid preferably dicarboxylic acid, more preferably butanedioic acid, still more preferably, butanedioic acid or butanedioic acid, [0057] amide, preferably substituted amide, more preferably N-ethyl-N-amide, N-propyl-
  • N-amide, or 2-amino-propanamide still more preferably 2,6-diamino-N-ethyl-N-hexanamide, 2-amino-3-methyl-N-propyl-N-butanamide, or N-(l,3-dicarboyl)-2-amino-propanamide, or wherein the amide comprises:
  • R3 is substituted alkyl, preferably substituted ethyl, more preferably amino ethyl, still more preferably 2-amino ethyl
  • R4 is carboxylic acid, preferably dicarboxylic acid, more preferably pentanedioic acid, still more preferably, pentanedioic acid or pentanedioic acid, or
  • R5 is selected from the group consisting of substituted alkyl, ethyl or propyl, and R6 is substituted alkyl, preferably l-amino-2-methylpropane or 1,5-diaminopentane.
  • R2 is (CRxR ⁇ )z-Rio.
  • Z is an integer from 1-3
  • each Rx is, independently, H or CH3,
  • each R9 is, independently, selected from the group consisting of: H, hydroxyl (OH), carboxyl (COOH), amino (NH2), or together with Rio, forms substituted or unsubstituted heterocyclic amine or azaspiroalkyl (or hetero-dualcycloalkyl), preferably 5-7 member heterocyclic amine or azaspiroalkyl, optionally substituted at one or more ring positions with C1-C2 alkyl, more preferably 5-6 member heterocyclic amine or 7 member azaspiroalkyl, optionally substituted at one or more ring positions with C1-C2 alkyl, substituted or unsubstituted pyrrolidine, piperidine, or piperazine, still more preferably, substituted or unsubstituted 3-N-pyrrolidine, 4-N-piperidine, or 4-N-piperazine, still more preferably unsubstituted 3-N-pyrrolidine or substituted 4-N-piperidine or 4-N-piperazine, still
  • heterocyclic amine preferably 6 member heterocyclic amine, still more preferably 6 member heterocyclic amine, still more preferably piperazine, still more preferably, 4-N-piperazine, still more preferably 4-N-methylpiperazine or 4-N- methy 1- 1 -piperazine,
  • alkanolamine preferably ethanolamine, N-ethanolamine or 1-aminoethanol
  • alkoxyalcohol preferably alkoxy ethanol or ethoxy alcohol
  • polyol preferably diol, more preferably ethanediol
  • amide preferably substituted amide, more preferably substituted N-pentanamide, N-hexanamide, or 1 -amide, more preferably 2,6-diamino-N-hexanamide, 2-amino-3-methyl- N-butanamide, or N-(l,3-dicarboyl)-2-amino-l-amide, or wherein the amide comprises:
  • R9 together with R9, forms substituted or unsubstituted heterocyclic amine or azaspiroalkyl (or hetero-dualcycloalkyl), preferably 5-7 member heterocyclic amine or azaspiroalkyl, optionally substituted at one or more ring positions with C1-C2 alkyl, more preferably 5-6 member heterocyclic amine or 7 member azaspiroalkyl, optionally substituted at one or more ring positions with C1-C2 alkyl, substituted or unsubstituted pyrrolidine, piperidine, or piperazine, still more preferably, substituted or unsubstituted 3-N-pyrrolidine, 4-N-piperidine, or 4-N-piperazine, still more preferably unsubstituted 3-N-pyrrolidine or substituted 4-N-piperidine or 4-N-piperazine, substituted at C-l, N-4, or C-l and N-4, still more preferably, 4-N-methylpiperidine, 4-N
  • R2 is CRnRi8)2-Ri9, wherein each R17 is H and each Rix is, independently, H or CH3; and R19 is N(CH3)Y, wherein Y is an integer from 2-3.
  • R2 is (CH2)2-Ri9, and R19 is + N(03 ⁇ 4)3, or, in other words, is:
  • the compound or composition can or may include a suitable (negatively- charged) counterion (W).
  • W can be any suitable (negatively-charged, or single negative charged) counterions, optionally or preferably Cl .
  • Suitable (negatively -charged, or single negative charged) counterion(s), such as Cl , etc., will be readily apparent to those skilled in the art.
  • R2 is selected from the group consisting of:
  • R2 is selected from the group consisting of:
  • carboxylic acid or dicarboxylic acid preferably pentanoic acid, butanedioic acid, or pentanedioic acid, still more preferably, 2-(6-amino)-pentanoic acid, 2-butanedioic acid or 2- butanedioic acid, or 2-pentanedioic acid or 2-pentanedioic acid, and
  • amide preferably substituted amide, more preferably substituted ethanamide, still more preferably substituted 2-ethanamide, still more preferably 2-(3-aminopropane)-N-2- butanedioic-2-ethanamide, 2-(3-aminopropane)-N-2-butanedioic-2-ethanamide, 2-(3- aminopropane)-N-2-pentanedioic-2-ethanamide, 2-(3-aminopropane)-N-2-pentanedioic-2- ethanamide, 2-(2-carboxyethyl)-N-(2-(3-aminopropane)-carboxymethyl)-2-ethanamide, 2- carboxymethyl-N-(2-(3-aminopropane)-carboxymethyl)-2-ethanamide, or 2-
  • R2 is selected from the group consisting of:
  • R2 is (CRnRu/z-Ri wherein [0087] each R13 is H;
  • each RM is, independently, selected from the group consisting of H, CH3, carboxyl (COOH), ethanol (CH2OH), preferably 2-ethanol, amine or alkylamine, preferably propylamine, carboxylic acid, preferably ethanoic acid or propanoic acid,
  • Z is and integer from 1-3, and
  • R15 is selected from the group consisting of carboxyl (-COOH), amino (-NH2), or amide, preferably 1 -amide, more preferably N-substituted-1 -amide (or N-substituted carboxamide) (-CONH-R16, where R1 ⁇ 2 is substituted alkyl, preferably 1,3-dicarboxypropane, l-(l-carboxy)butyric acid or butanedioic acid, l-carboxy-2-pentanoic acid or pentanedioic acid, preferably 2-pentanedioic acid, or 2-(3-aminopropyl)ethanoic acid.
  • R1 ⁇ 2 is substituted alkyl, preferably 1,3-dicarboxypropane, l-(l-carboxy)butyric acid or butanedioic acid, l-carboxy-2-pentanoic acid or pentanedioic acid, preferably 2-p
  • R2 is selected from the group consisting of Formulas Iq-Ix:
  • the compound according to Formula I can be one of Compounds 1-24 of Table 1. [0093] Table 1. Nonlimiting examples of illustrative compounds:
  • Some embodiments are directed to a (pharmaceutical) composition
  • a pharmaceutically acceptable carrier or excipient comprising a pharmaceutically acceptable carrier or excipient and a compound of Formula I.
  • Some embodiments are directed to a (pharmaceutical) medicament comprising a pharmaceutically acceptable carrier or excipient and a compound of Formula I or composition comprising the same.
  • Some embodiments are directed to a composition or medicament including a compound of Formula I for use in (i) inhibiting histone deacetylase (HDAC) activity, (ii) enhancing renal recovery following acute kidney injury (AKI), preferably through HDAC inhibition, (iii) mitigating renal damage following AKI, (iv) treatment of AKI, and/or treating other related conditions or affecting other related molecular mechanisms, in a mammal or mammalian subject (in need thereof).
  • the mammal or mammalian subject is human.
  • Some embodiments are directed to use of the compound of Formula I, a pharmaceutical compositions comprising the same, or a medicament comprising the same, for (i) inhibiting histone deacetylase (HDAC) activity, (ii) enhancing renal recovery following acute kidney injury (AKI), preferably through HDAC inhibition, (iii) mitigating renal damage following AKI, (iv) treatment of AKI, and/or treating other related conditions or affecting other related molecular mechanisms, in a mammal or mammalian subject (in need thereof).
  • the mammal or mammalian subject is human.
  • Some embodiments are directed to a method of (i) inhibiting histone deacetylase (HDAC) activity, (ii) enhancing renal recovery following acute kidney injury (AKI), preferably through HDAC inhibition, (iii) mitigating renal damage following AKI, (iv) treatment of AKI, and/or treating other related conditions or affecting other related molecular mechanisms, in a mammal or mammalian subject (in need thereof), the method comprising administering a compound according to Formula I, a pharmaceutical compositions comprising the same, or a medicament comprising the same, to the mammal or mammalian subject (in need thereof).
  • the mammal or mammalian subject is human.
  • Some embodiments may include any of the features, options, and/or possibilities set out elsewhere in the present disclosure, including in other aspects or embodiments of the present disclosure. It is also noted that each of the foregoing, following, and/or other features described herein represent a distinct embodiment of the present disclosure. Moreover, combinations of any two or more of such features represent distinct embodiments of the present disclosure. Such features or embodiments can also be combined in any suitable combination and/or order without departing from the scope of this disclosure. Thus, each of the features described herein can be combinable with any one or more other features described herein in any suitable combination and/or order. Accordingly, the present disclosure is not limited to the specific combinations of exemplary embodiments described in detail herein.
  • Figure 1 depicts 24 illustrative compounds and information associated with the same.
  • Figure 2 illustrates stability of illustrative Compound 22 in Rat plasma.
  • Figure 3 illustrates stability of illustrative Compound 22 in Dog plasma.
  • Figure 4 illustrates stability of illustrative Compound 22 in Human plasma.
  • Figure 5 illustrates the percentage of illustrative Compound 22 remaining in
  • FIG. 6 illustrates PTBA levels in Monkey plasma over time following administration of Compound 22.
  • Figure 7A illustrates plasma concentration of pro-drug (Compound 22) and PTBA over time in rats following i.v. administration of pro-drug (Compound 22).
  • Figure 7B illustrates plasma concentration of pro-drug (Compound 17) and
  • Figure 7C illustrates plasma concentration of pro-drug (Compound 1) and PTBA over time in Rats.
  • Figure 7D illustrates plasma concentration of pro-drug (Compound 15) and PTBA over time in Rats.
  • Figure 8 illustrates urea levels in in cisplatin-induced AKI mice treated with illustrative Compound 22.
  • Figures 9A illustrates body weight in cisplatin-induced AKI mice treated with illustrative Compound 22 once or twice a day on days 2-8.
  • Figure 9B illustrates body weight in cisplatin-induced AKI mice treated with illustrative Compound 22 once or twice a day on days 0-6.
  • Figure 10 illustrates creatinine levels in the serum of rats treated with illustrative compound 22.
  • Figure 11 illustrates urea levels in in cisplatin-induced AKI mice treated with illustrative Compound 3.
  • Figure 12 illustrates creatinine levels in the serum of rats treated with illustrative compound 3.
  • Figure 13 illustrates body weight in cisplatin-induced AKI mice treated with illustrative Compound 3.
  • the words “can” and “may” are used in a permissive sense (i.e., meaning having the potential to), rather than the mandatory sense (i.e., meaning must).
  • the terms “including,” “having,” “involving,” “containing,” “characterized by,” variants thereof (e.g., “includes,” “has,” and “involves,” “contains,” etc.), and similar terms as used herein, including the claims, shall be inclusive and/or open-ended, shall have the same meaning as the word “comprising” and variants thereof (e.g, “comprise” and “comprises”), and do not exclude additional, un-recited elements or method steps, illustratively.
  • condition refers to any disorder, disease, injury, or illness, as understood by those skilled in the art, that is manifested or anticipated in a patient. Manifestation of such a condition can be an early, middle, or late stage manifestation, as known in the art, including pre-condition symptoms, signs, or markers. Anticipation of such a condition can be or include the predicted, expected, envisioned, presumed, supposed, and/or speculated occurrence of the same, whether founded in scientific or medical evidence, risk assessment, or mere apprehension or trepidation.
  • patient is synonymous with the term “subject” and generally refers to any animal under the care of a medical professional, as that term is defined herein, with particular reference to (i) humans (under the care of a doctor, nurse, or medical assistant or volunteer) and (ii) non-human animals, such as non-human mammals (under the care of a veterinarian or other veterinary professional, assistant, or volunteer).
  • prodrug is meant to indicate a compound that may be converted under physiological conditions or by solvolysis to a biologically active compound described herein (e.g., compound according to Formula I).
  • prodrug refers to a precursor of a biologically active compound that is pharmaceutically acceptable.
  • a prodrug is or may be inactive/less-active when administered to a subject, but is converted in vivo to an active compound, for example, by hydrolysis.
  • the prodrug compound often offers advantages of solubility, tissue compatibility or delayed release in a mammalian organism (see, e.g., Bundgard, H., Design of Prodrugs (1985), pp.
  • prodrugs are provided in Higuchi, T., et al., “Pro-drugs as Novel Delivery Systems,” A.C.S. Symposium Series, Vol. 14, and in Bioreversible Carriers in Drug Design, ed. Edward B. Roche, American Pharmaceutical Association and Pergamon Press, 1987, the entirety of each of which is incorporated herein by specific reference.
  • Embodiments of the present disclosure are also meant to encompass all pharmaceutically acceptable compounds according to Formula I that are isotopically-labelled by having one or more atoms replaced by an atom having a different atomic mass or mass number.
  • isotopes that can be incorporated into the disclosed compounds include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, fluorine, chlorine, and iodine, such as 2 H, 3 ⁇ 4, n C, 13 C, 14 C, 13 N, 15 N, 15 0, 17 0, 18 0, 31 P, 32 P, 35 S, 18 F, 36 C1, 123 I, and 125 I, respectively.
  • radiolabeled compounds may be useful to help determine or measure the effectiveness of the compounds, by characterizing, for example, the site or mode of action, or binding affinity to pharmacologically important site of action.
  • Certain isotopically-labelled compounds according to Formula I for example, those incorporating a radioactive isotope, are useful in drug and/or substrate tissue distribution studies.
  • the radioactive isotopes tritium, i.e. 3 H, and carbon-14, i.e. 14 C, are particularly useful for this purpose in view of their ease of incorporation and ready means of detection.
  • substitution with heavier isotopes such as deuterium, i.e. 2 H may afford certain therapeutic advantages resulting from greater metabolic stability, for example, increased in vivo half-life or reduced dosage requirements, and hence may be preferred in some circumstances.
  • Substitution with positron emitting isotopes, such as n C, 18 F, 15 0 and 13 N can be useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy.
  • PET Positron Emission Topography
  • Isotopically-labeled compounds of Formula I can generally be prepared by conventional techniques known to those skilled in the art or by processes analogous to those described in the Preparations and Examples as set out below using an appropriate isotopically-labeled reagent in place of the non-labeled reagent previously employed.
  • Embodiments of the present disclosure may also encompass the in vivo metabolic products of the disclosed compounds. Such products may result from, for example, the oxidation, reduction, hydrolysis, amidation, esterification, and the like of the administered compound, primarily due to enzymatic processes. Accordingly, embodiments of the present disclosure include compounds produced by a process comprising administering a compound of this present disclosure to a mammal for a period of time sufficient to yield a metabolic product thereof.
  • Such products are typically identified by administering a radiolabeled compound of the present disclosure in a detectable dose to an animal, such as rat, mouse, guinea pig, monkey, or to human, allowing sufficient time for metabolism to occur, and isolating its conversion products from the urine, blood or other biological samples.
  • carrier, diluent and/or excipient include, without limitation, any adjuvant, carrier, excipient, glidant, sweetening agent, diluent, preservative, dye/colorant, flavor enhancer, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, solvent, or emulsifier which has been approved by the United States Food and Drug Administration as being acceptable for use in humans or domestic animals.
  • salt or “pharmaceutically acceptable salt” includes both acid and base addition salts.
  • Salts may include “acid addition salt” or “pharmaceutically acceptable acid addition salt”, which refer to those salts which retain the biological effectiveness and properties of the free bases, which are not biologically or otherwise undesirable, and which are formed with inorganic acids such as, but are not limited to, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid and the like, and organic acids such as, but not limited to, acetic acid, 2,2-dichloroacetic acid, adipic acid, alginic acid, ascorbic acid, aspartic acid, benzenesulfonic acid, benzoic acid, 4-acetamidobenzoic acid, camphoric acid, camphor- 10-sulfonic acid, capric acid, caproic acid, caprylic acid, carbonic acid, cinnamic acid, citric acid, cyclamic acid, dodecylsulfuric acid, ethane- 1,2-disulfonic acid, ethanesul
  • Salts may include “base addition salt” or “pharmaceutically acceptable base addition salt”, which refer to those salts which retain the biological effectiveness and properties of the free acids, which are not biologically or otherwise undesirable. These salts are prepared from addition of an inorganic base or an organic base to the free acid. Salts derived from inorganic bases include, but are not limited to, the sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Preferred inorganic salts are the ammonium, sodium, potassium, calcium, and magnesium salts.
  • Salts derived from organic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins, such as ammonia, isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, diethanolamine, ethanolamine, deanol, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, benethamine, benzathine, ethylenediamine, glucosamine, methylglucamine, theobromine, triethanolamine, tromethamine, purines, piperazine, piperidine, N-ethylpiperidine, polyamine resins and the like.
  • Particularly preferred organic bases are isoprop
  • solvate refers to an aggregate that comprises one or more molecules of a compound of the present disclosure with one or more molecules of solvent.
  • the solvent may be water, in which case the solvate may be a hydrate.
  • the solvent may be an organic solvent.
  • embodiments of the compounds of the present disclosure may exist as a hydrate, including a monohydrate, dihydrate, hemihydrate, sesquihydrate, trihydrate, tetrahydrate and the like, as well as the corresponding solvated forms.
  • Embodiments of the compound of the present disclosure may be true solvates, while in other cases, the compound of the present disclosure may merely retain adventitious water or be a mixture of water plus some adventitious solvent.
  • a “pharmaceutical composition” refers to a formulation of a compound of the present disclosure and a medium generally accepted in the art for the delivery of the biologically active compound to mammals, e.g., humans.
  • a medium includes all pharmaceutically acceptable carriers, diluents or excipients therefor.
  • “Mammal” includes humans and both domestic animals such as laboratory animals and household pets (e.g., cats, dogs, swine, cattle, sheep, goats, horses, rabbits), and non domestic animals such as wildlife and the like.
  • Effective amount refers to that amount of a compound of the present disclosure which, when administered to a mammal, preferably a human, is sufficient to effect treatment, as defined below, of a disease associated with overexpression of a cyclin-dependent kinase (CDK) in the mammal, preferably a human.
  • CDK cyclin-dependent kinase
  • the amount of a compound of the present disclosure which constitutes a “therapeutically effective amount” will vary depending on the compound, the condition and its severity, the manner of administration, and the age of the mammal to be treated, but can be determined routinely by one of ordinary skill in the art having regard to his own knowledge and to this disclosure.
  • Treating covers the treatment of the disease or condition of interest in a mammal, preferably a human, having the disease or condition of interest, and includes:
  • disease and “condition” may be used interchangeably or may be different in that the particular malady or condition may not have a known causative agent (so that etiology has not yet been worked out) and it is therefore not yet recognized as a disease but only as an undesirable condition or syndrome, wherein a more or less specific set of symptoms have been identified by clinicians.
  • Compounds of the present disclosure, or their pharmaceutically acceptable salts may contain one or more asymmetric centers and may thus give rise to enantiomers, diastereomers, and other stereoisomeric forms that may be defined, in terms of absolute stereochemistry, as (R)- or (S)- or, as (D)- or (L)- for amino acids.
  • the present disclosure is meant to include all such possible isomers, as well as their racemic and optically pure forms.
  • Optically active (+) and (-), (R)- and (S)-, or (D)- and (L)- isomers may be prepared using chiral synthons or chiral reagents, or resolved using conventional techniques, for example, chromatography and fractional crystallization.
  • a “stereoisomer” refers to a compound made up of the same atoms bonded by the same bonds but having different three-dimensional structures, which are not interchangeable.
  • Embodiments of the present disclosure contemplate various stereoisomers and mixtures thereof and includes “enantiomers”, which refers to two stereoisomers whose molecules are nonsuperimposeable mirror images of one another.
  • a “tautomer” refers to a proton shift from one atom of a molecule to another atom of the same molecule.
  • Embodiments of the present disclosure include tautomers of any said compounds.
  • the present disclosure may recite a list or range of numerical values. It will be appreciated, however, that where such a list or range of numerical values (e.g., greater than, less than, up to, at least, and/or about a certain value, and/or between two recited values) is disclosed or recited, any specific value or range of values falling within the disclosed values or list or range of values is likewise specifically disclosed and contemplated herein.
  • a list or range of numerical values e.g., greater than, less than, up to, at least, and/or about a certain value, and/or between two recited values
  • compositions and medicaments are also provided for delivery of the compounds to a patient.
  • Also provided are methods for improving kidney function, inhibiting a histone deacetylase in a cell, expanding renal progenitor cells and/or stimulating kidney repair in cells in vitro, ex vivo or in vivo (in a patient) comprising contacting the cells with, or administering to a patient an amount of one or more of the compounds effective to improve kidney function in a patient, inhibit a histone deacetylase in a cell, expand renal progenitor cells and/or stimulate kidney repair in cells. Therefore provided are in vitro (including ex vivo) or in vivo (in a patient) methods. Efficacy of the compounds is demonstrated below.
  • Embodiments of the present disclosure include prodrugs of PTBA.
  • Illustrative embodiments of the present disclosure include a compound according to Formula I:
  • Ri is O or NH. In some embodiments, Ri is O. In some embodiments, Ri is NH.
  • R2 is not H. In some embodiments, R2 is not CH3. In some embodiments, R2 is not, one or more of, alkyl, ethyl, propyl, isopropyl, or butyl. In some embodiments, R2 is not aryl, phenyl or benzyl. In some embodiments, R2 is not carboxyl (COOH). In some embodiments, R2 is not, one or more of, methanol, ethanol, propanol, or isopropanol.
  • R2 is selected from the group consisting of:
  • substituted or unsubstituted amine preferably substituted or unsubstituted tertiary amine or quaternary amine (trisubstituted ammonium or quaternary ammonium), more preferably l-(N,N-dimethyl)ethanamine, l-(l-methyl-N,N-dimethyl)ethanamine, or 1- (N,N,N-trimethyl)ethanamine,
  • substituted or unsubstituted heterocyclic amine or azaspiroalkyl or hetero- dualcycloalkyl, preferably 5-7 member heterocyclic amine or azaspiroalkyl, optionally substituted at one or more ring positions with C1-C2 alkyl, or 4-7 member substituted or unsubstituted heterocycloalkyl or hetero-dualcycloalkyl (a.k.a.
  • azaspiroalkyl optionally substituted at 1-4 (ring) positions and each (ring) substituent is selected from the group consisting of branched or unbranched C1-C3 substituted or unsubstituted alkyl or cycloalkyl, more preferably substituted or unsubstituted pyrrolidine, piperidine, or piperazine, still more preferably, substituted or unsubstituted 3-N-pyrrolidine, 4-N-piperidine, or 4-N-piperazine, still more preferably, 4-N-piperidine or 4-N-piperazine substituted at C-l, N-4, or C-l and N- 4, still more preferably, 4-N-methylpiperidine, 4-N-ethylpiperidine, 4-N-l,4- methylpiperidine, 4-N-methylpiperazine, or substituted or unsubstituted azaspiroheptyl, preferably 2-azaspiroheptyl or azaspiro[3.3]heptyl,
  • alkanolamine preferably ethanolamine, more preferably N-ethylethanolamine or 2-(ethylamino)ethanol,
  • alkoxyalcohol preferably alkoxy ethanol or ethoxy alcohol, more preferably 2- ethoxy ethanol,
  • polyol preferably diol, more preferably propanediol, still more preferably 2,3- propanediol,
  • carboxylic acid preferably dicarboxylic acid, more preferably butanedioic acid, still more preferably, butanedioic acid,
  • amide preferably substituted amide, more preferably N-ethyl-N-amide, N-propyl- N-amide, or 2-amino-propanamide, still more preferably 2,6-diamino-N-ethyl-N-hexanamide, 2-amino-3-methyl-N-propyl-N-butanamide, or N-(l,3-dicarboyl)-2-amino-propanamide, or wherein the amide comprises:
  • R3 is substituted alkyl, preferably substituted ethyl, more preferably amino ethyl, still more preferably 2-amino ethyl, and R4 is carboxylic acid, preferably dicarboxybc acid, more preferably pentanedioic acid, still more preferably, pentanedioic acid or pentanedioic acid, or
  • R5 is substituted alkyl, ethyl or propyl
  • R6 is substituted alkyl, preferably l-amino-2-methylpropane or 1,5-diaminopentane
  • azaspiroalkyl optionally substituted at 1-4 (ring) positions and each (ring) substituent is selected from the group consisting of branched or unbranched C1-C3 substituted or unsubstituted alkyl or cycloalkyl, preferably substituted or unsubstituted pyrrolidine, piperidine, or piperazine, more preferably, substituted or unsubstituted 3-N-pyrrobdine, 4-N- piperidine, or 4-N -piperazine, still more preferably, 4-N-piperidine or 4-N-piperazine substituted at C-l, N-4, or C-l and N-4, still more preferably, 4-N-methylpiperidine, 4-N- ethylpiperidine, 4-N- 1,4-methylpiperi dine, 4-N-methylpiperazine, most preferably, 4-N- methylpiperazine, preferably when X is (CH2)2, or tertiary amine or quaternary amine (trisubstitute
  • each Rx is, independently, selected from the group consisting of H or CH3
  • each R9 is, independently, selected from: H, hydroxyl (OH), carboxyl (COOH), amino (NH2), or together with Rio, forms substituted or unsubstituted heterocyclic amine or azaspiroalkyl (or hetero-dualcycloalkyl), preferably 5-7 member heterocyclic amine or azaspiroalkyl, optionally substituted at one or more ring positions with C1-C2 alkyl, more preferably 5-6 member heterocyclic amine or 7 member azaspiroalkyl, optionally substituted at one or more ring positions with C1-C2 alkyl, substituted or unsubstituted pyrrolidine, piperidine, or piperazine, still more preferably, substituted or unsubstituted 3-N-pyrrolidine, 4-N-piperidine, or 4-N-
  • amine preferably tertiary amine or quaternary amine (trisubstituted ammonium or quaternary ammonium), more preferably l-(N,N-dimethyl)amine, l-(l-methyl-N,N- dimethyl)amine, or l-(N,N,N-trimethyl)amine,
  • heterocyclic amine preferably 6 member heterocyclic amine, still more preferably 6 member heterocyclic amine, still more preferably piperazine, still more preferably, 4-N-piperazine, still more preferably 4-N-methylpiperazine or 4-N- methy 1- 1 -piperazine,
  • alkanolamine preferably ethanolamine, N-ethanolamine or 1 -aminoethanol
  • alkoxyalcohol preferably alkoxy ethanol or ethoxy alcohol
  • polyol preferably diol, more preferably ethanediol
  • R12 is substituted alkyl, preferably l-amino-2- methylpropane or 1,5-diaminopentane
  • R9 forms substituted or unsubstituted heterocyclic amine or azaspiroalkyl (or hetero-dualcycloalkyl), preferably 5-7 member heterocyclic amine or azaspiroalkyl, optionally substituted at one or more ring positions with C1-C2 alkyl, more preferably 5-6 member heterocyclic amine or 7 member azaspiroalkyl, optionally substituted at one or more ring positions with C1-C2 alkyl, substituted or unsubstituted pyrrolidine, piperidine, or piperazine, still more preferably, substituted or unsubstituted 3-N-pyrrobdine, 4-N-piperidine, or 4-N-piperazine, still more preferably unsubstituted 3-
  • R2 is selected from the group consisting of:
  • substituted or unsubstituted amine preferably substituted or unsubstituted tertiary amine or quaternary amine (trisubstituted ammonium or quaternary ammonium), more preferably l-(N,N-dimethyl)ethanamine, l-(l-methyl-N,N-dimethyl)ethanamine, or 1- (N,N,N-trimethyl)ethanamine,
  • substituted or unsubstituted heterocyclic amine or azaspiroalkyl or hetero- dualcycloalkyl, preferably 5-7 member heterocyclic amine or azaspiroalkyl, optionally substituted at one or more ring positions with C1-C2 alkyl, or 4-7 member substituted or unsubstituted heterocycloalkyl or hetero-dualcycloalkyl (a.k.a.
  • azaspiroalkyl optionally substituted at 1-4 (ring) positions and each (ring) substituent is selected from the group consisting of branched or unbranched C1-C3 substituted or unsubstituted alkyl or cycloalkyl, more preferably substituted or unsubstituted pyrrolidine, piperidine, or piperazine, still more preferably, substituted or unsubstituted 3-N-pyrrolidine, 4-N-piperidine, or 4-N-piperazine, still more preferably, 4-N-piperidine or 4-N-piperazine substituted at C-l, N-4, or C-l and N- 4, still more preferably, 4-N-methylpiperidine, 4-N-ethylpiperidine, 4-N-l,4- methylpiperidine, 4-N-methylpiperazine, or substituted or unsubstituted azaspiroheptyl, preferably 2-azaspiroheptyl or azaspiro[3.3]heptyl,
  • alkanolamine preferably ethanolamine, more preferably N-ethylethanolamine or 2-(ethylamino)ethanol,
  • alkoxyalcohol preferably alkoxy ethanol or ethoxy alcohol, more preferably 2- ethoxy ethanol
  • polyol preferably diol, more preferably propanediol, still more preferably 2,3- propanediol
  • carboxylic acid preferably dicarboxylic acid, more preferably butanedioic acid, still more preferably, butanedioic acid or butanedioic acid,
  • amide preferably substituted amide, more preferably N-ethyl-N-amide, N-propyl- N-amide, or 2-amino-propanamide, still more preferably 2,6-diamino-N-ethyl-N-hexanamide, 2-amino-3-methyl-N-propyl-N-butanamide, or N-(l,3-dicarboyl)-2-amino-propanamide, or wherein the amide comprises:
  • R3 is substituted alkyl, preferably substituted ethyl, more preferably amino ethyl, still more preferably 2-amino ethyl
  • R4 is carboxylic acid, preferably dicarboxylic acid, more preferably pentanedioic acid, still more preferably, pentanedioic acid or pentanedioic acid,
  • R5 is substituted alkyl, ethyl or propyl
  • R6 is substituted alkyl, preferably l-amino-2-methylpropane or 1,5-diaminopentane
  • azaspiroalkyl optionally substituted at 1-4 (ring) positions and each (ring) substituent is selected from the group consisting of branched or unbranched C1-C3 substituted or unsubstituted alkyl or cycloalkyl, preferably substituted or unsubstituted pyrrolidine, piperidine, or piperazine, more preferably, substituted or unsubstituted 3-N-pyrrolidine, 4-N- piperidine, or 4-N -piperazine, still more preferably, 4-N-piperidine or 4-N-piperazine substituted at C-l, N-4, or C-l and N-4, still more preferably, 4-N-methylpiperidine, 4-N- ethylpiperidine, 4-N- 1,4-methylpiperi dine, 4-N-methylpiperazine, most preferably, 4-N- methylpiperazine, preferably when X is (CH2)2, or tertiary amine or quaternary amine (trisubstituted
  • R2 is selected from the group consisting of:
  • substituted or unsubstituted amine preferably substituted or unsubstituted tertiary amine or quaternary amine (trisubstituted ammonium or quaternary ammonium), more preferably l-(N,N-dimethyl)ethanamine, l-(l-methyl-N,N-dimethyl)ethanamine, or 1- (N,N,N-trimethyl)ethanamine, [00188] substituted or unsubstituted heterocyclic amine or azaspiroalkyl, preferably 5-7 member heterocyclic amine or azaspiroalkyl, optionally substituted at one or more ring positions with C1-C2 alkyl,
  • alkanolamine preferably ethanolamine, more preferably N-ethylethanolamine or 2-(ethylamino)ethanol
  • alkoxyalcohol preferably alkoxy ethanol or ethoxy alcohol, more preferably 2- ethoxy ethanol
  • polyol preferably diol, more preferably propanediol, still more preferably 2,3- propanediol,
  • carboxylic acid preferably dicarboxylic acid, more preferably butanedioic acid, still more preferably, butanedioic acid or butanedioic acid,
  • R2 is (CRsRi z-Rio, wherein:
  • Z is an integer from 1-3, [00198] each Rx is, independently, H or CH3,
  • each R9 is, independently, selected from the group consisting of: H, hydroxyl (OH), carboxyl (COOH), amino (NH2), or together with Rio, forms substituted or unsubstituted heterocyclic amine or azaspiroalkyl (or hetero-dualcycloalkyl), preferably 5-7 member heterocyclic amine or azaspiroalkyl, optionally substituted at one or more ring positions with C1-C2 alkyl, more preferably 5-6 member heterocyclic amine or 7 member azaspiroalkyl, optionally substituted at one or more ring positions with C1-C2 alkyl, substituted or unsubstituted pyrrolidine, piperidine, or piperazine, still more preferably, substituted or unsubstituted 3-N-pyrrolidine, 4-N-piperidine, or 4-N-piperazine, still more preferably unsubstituted 3-N-pyrrolidine or substituted 4-N-piperidine or 4-N-piperazine, still
  • amine preferably tertiary amine or quaternary amine (trisubstituted ammonium or quaternary ammonium), more preferably l-(N,N-dimethyl)amine, l-(l-methyl-N,N- dimethyl)amine, or l-(N,N,N-trimethyl)amine,
  • heterocyclic amine preferably 6 member heterocyclic amine, still more preferably 6 member heterocyclic amine, still more preferably piperazine, still more preferably, 4-N-piperazine, still more preferably 4-N-methylpiperazine or 4-N- methy 1- 1 -piperazine,
  • alkanolamine preferably ethanolamine, N-ethanolamine or 1 -aminoethanol
  • alkoxyalcohol preferably alkoxy ethanol or ethoxy alcohol
  • polyol preferably diol, more preferably ethanediol
  • amide preferably substituted amide, more preferably substituted N-pentanamide, N-hexanamide, or 1 -amide, more preferably 2,6-diamino-N-hexanamide, 2-amino-3-methyl- N-butanamide, or N-(l,3-dicarboyl)-2-amino-l-amide, or wherein the amide comprises:
  • R.4 is carboxylic acid, preferably dicarboxylic acid, more preferably pentanedioic acid, still more preferably, pentanedioic acid or pentanedioic acid, or
  • R12 is substituted alkyl, preferably 1 -amino-2- methylpropane or 1,5-diaminopentane, or
  • R9 together with R9, forms substituted or unsubstituted heterocyclic amine or azaspiroalkyl (or hetero-dualcycloalkyl), preferably 5-7 member heterocyclic amine or azaspiroalkyl, optionally substituted at one or more ring positions with C1-C2 alkyl, more preferably 5-6 member heterocyclic amine or 7 member azaspiroalkyl, optionally substituted at one or more ring positions with C1-C2 alkyl, substituted or unsubstituted pyrrolidine, piperidine, or piperazine, still more preferably, substituted or unsubstituted 3-N-pyrrolidine, 4-N-piperidine, or 4-N-piperazine, still more preferably unsubstituted 3-N-pyrrolidine or substituted 4-N-piperidine or 4-N-piperazine, substituted at C-l, N-4, or C-l and N-4, still more preferably, 4-N-methylpiperidine, 4-N
  • R2 is CRnRi8)2-Ri9, wherein each R17 is
  • each Rix is, independently, H or CH3; and R19 is N(CH3)Y, wherein Y is an integer from 2-3.
  • R2 when Ri is O, then R2 is (CH2)2-Ri9, and R19 is N(CH3)3.
  • R2 is selected from the group consisting of:
  • R2 is selected from the group consisting of: [00217] carboxylic acid or dicarboxylic acid, preferably pentanoic acid, butanedioic acid, or pentanedioic acid, still more preferably, 2-(6-amino)-pentanoic acid, 2-butanedioic acid or 2-butanedioic acid, or 2-pentanedioic acid or 2-pentanedioic acid, and [00218] amide, preferably substituted amide, more preferably substituted ethanamide, still more preferably substituted 2-ethanamide, still more preferably 2-(3-aminopropane)-N-2- butanedioic-2-ethanamide, 2-(3-aminopropane)-N-2-butanedioic-2-ethanamide, 2-(3- aminopropane)-N-2-pentanedioic
  • R2 is selected from the group consisting of:
  • R2 is (CRi3Ri4)z-Ri5, wherein [00223] each R13 is H;
  • each R14 is, independently, selected from the group consisting of H, CH3, carboxyl (COOH), ethanol (CH2OH), preferably 2-ethanol, amine or alkylamine, preferably propylamine, carboxylic acid, preferably ethanoic acid or propanoic acid,
  • Ri5 is selected from the group consisting of carboxyl (-COOH), amino (-NH2), or amide, preferably 1 -amide, more preferably N-substituted-1 -amide (or N-substituted carboxamide) (-CONH-R16, where R1 ⁇ 2 is substituted alkyl, preferably 1,3-dicarboxypropane, l-(l-carboxy)butyric acid or butanedioic acid, l-carboxy-2-pentanoic acid or pentanedioic acid, preferably 2-pentanedioic acid, or 2-(3-aminopropyl)ethanoic acid.
  • R2 is selected from the group consisting of Formulas Iq-Ix: Formula Iq Formula Ir Formula Is Formula It
  • the compound according to Formula I can be one of Compounds 1-24 of Table 1.
  • all compounds of the present disclosure which exist in free base or acid form can be converted to their pharmaceutically acceptable salts by treatment with the appropriate inorganic or organic base or acid by methods known to one skilled in the art. Salts of the compounds of the present disclosure can be converted to their free base or acid form by standard techniques.
  • compositions or medicaments of embodiments of the present disclosure can be prepared by combining a compound of the present disclosure with an appropriate pharmaceutically acceptable carrier, diluent or excipient, and may be formulated into preparations in solid, semi-solid, liquid or gaseous forms, such as tablets, capsules, powders, granules, ointments, solutions, suppositories, injections, inhalants, gels, microspheres, and aerosols.
  • compositions or medicaments of the present disclosure are formulated so as to allow the active ingredients contained therein to be bioavailable upon administration of the composition or medicament to a patient.
  • compositions or medicaments that will be administered to a subject or patient take the form of one or more dosage units, where for example, a tablet may be a single dosage unit, and a container of a compound of the present disclosure in aerosol form may hold a plurality of dosage units.
  • Actual methods of preparing such dosage forms are known, or will be apparent, to those skilled in this art; for example, see Remington: The Science and Practice of Pharmacy, 20th Edition (Philadelphia College of Pharmacy and Science, 2000).
  • the composition or medicament to be administered will, in any event, contain a therapeutically effective amount of a compound of the present disclosure, or a pharmaceutically acceptable salt thereof, for treatment of a disease or condition of interest in accordance with the teachings of this disclosure.
  • a pharmaceutical composition or medicament of some embodiments of the present disclosure may be in the form of a solid or liquid.
  • the carrier(s) are particulate, so that the compositions or medicaments are, for example, in tablet or powder form.
  • the carrier(s) may be liquid, with the compositions being, for example, an oral syrup, injectable liquid or an aerosol, which is useful in, for example, inhalation or inhalatory administration.
  • the pharmaceutical composition or medicament is preferably in either solid or liquid form, where semi-solid, semi-liquid, suspension and gel forms are included within the forms considered herein as either solid or liquid.
  • the pharmaceutical composition or medicament may be formulated into a powder, granule, compressed tablet, pill, capsule, chewing gum, wafer or the like form.
  • a solid composition or medicament will typically contain one or more inert diluents or edible carriers.
  • binders such as carboxymethylcellulose, ethyl cellulose, microcrystalline cellulose, gum tragacanth or gelatin; excipients such as starch, lactose or dextrins, disintegrating agents such as alginic acid, sodium alginate, Primogel, com starch and the like; lubricants such as magnesium stearate or Sterotex; glidants such as colloidal silicon dioxide; sweetening agents such as sucrose or saccharin; a flavoring agent such as peppermint, methyl salicylate or orange flavoring; and a coloring agent.
  • excipients such as starch, lactose or dextrins, disintegrating agents such as alginic acid, sodium alginate, Primogel, com starch and the like
  • lubricants such as magnesium stearate or Sterotex
  • glidants such as colloidal silicon dioxide
  • sweetening agents such as sucrose or saccharin
  • a flavoring agent such as peppermint,
  • the pharmaceutical composition or medicament when it is in the form of a capsule, for example, a gelatin capsule, it may contain, in addition to materials of the above type, a liquid carrier such as polyethylene glycol or oil.
  • a liquid carrier such as polyethylene glycol or oil.
  • the pharmaceutical composition or medicament may be in the form of a liquid, for example, an elixir, syrup, solution, emulsion or suspension.
  • the liquid may be for oral administration or for delivery by injection, as two examples.
  • preferred composition or medicament contain, in addition to the present compounds, one or more of a sweetening agent, preservatives, dye/colorant and flavor enhancer.
  • a surfactant, preservative, wetting agent, dispersing agent, suspending agent, buffer, stabilizer and isotonic agent may be included.
  • the liquid pharmaceutical compositions and medicaments of some embodiments of the present disclosure may include one or more of the following adjuvants: sterile diluents such as water for injection, saline solution, preferably physiological saline, Ringer's solution, isotonic sodium chloride, fixed oils such as synthetic mono or diglycerides which may serve as the solvent or suspending medium, polyethylene glycols, glycerin, propylene glycol or other solvents; antibacterial agents such as benzyl alcohol or methyl paraben; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents such as ethylenediaminetetraacetic acid; buffers such as acetates, citrates or phosphates and agents for the adjustment of tonicity such as sodium chloride or dextrose.
  • sterile diluents such as water for injection, saline solution, preferably physiological saline, Ringer's solution, isotonic sodium chloride
  • fixed oils such as synthetic
  • parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.
  • Physiological saline is a preferred adjuvant.
  • An injectable pharmaceutical composition or medicament is preferably sterile.
  • a liquid pharmaceutical composition or medicament of certain embodiments of the present disclosure intended for either parenteral or oral administration should contain an amount of a compound of the present disclosure such that a suitable dosage will be obtained.
  • the pharmaceutical composition or medicament of the present disclosure may be intended for topical administration, in which case the carrier may suitably comprise a solution, emulsion, ointment or gel base.
  • the base may comprise one or more of the following: petrolatum, lanolin, polyethylene glycols, bee wax, mineral oil, diluents such as water and alcohol, and emulsifiers and stabilizers.
  • Thickening agents may be present in a pharmaceutical composition or medicament for topical administration. If intended for transdermal administration, the composition or medicament may include a transdermal patch or iontophoresis device.
  • composition or medicament of various embodiments of the present disclosure may be intended for rectal administration, in the form, for example, of a suppository, which will melt in the rectum and release the drug.
  • the composition or medicament for rectal administration may contain an oleaginous base as a suitable nonirritating excipient.
  • bases include, without limitation, lanolin, cocoa butter and polyethylene glycol.
  • Embodiments of the pharmaceutical composition or medicament of the present disclosure may include various materials, which modify the physical form of a solid or liquid dosage unit.
  • the composition or medicament may include materials that form a coating shell around the active ingredients.
  • the materials that form the coating shell are typically inert, and may be selected from, for example, sugar, shellac, and other enteric coating agents.
  • the active ingredients may be encased in a gelatin capsule.
  • the pharmaceutical composition or medicament of some embodiments of the present disclosure in solid or liquid form may include an agent that binds to the compound of the present disclosure and thereby assists in the delivery of the compound.
  • Suitable agents that may act in this capacity include a monoclonal or polyclonal antibody, a protein or a liposome.
  • the pharmaceutical composition or medicament of other embodiments of the present disclosure may consist of dosage units that can be administered as an aerosol.
  • aerosol is used to denote a variety of systems ranging from those of colloidal nature to systems consisting of pressurized packages. Delivery may be by a liquefied or compressed gas or by a suitable pump system that dispenses the active ingredients. Aerosols of compounds of the present disclosure may be delivered in single phase, bi-phasic, or tri-phasic systems in order to deliver the active ingredient(s).
  • the pharmaceutical compositions or medicaments of the present disclosure may be prepared by methodology well known in the pharmaceutical art.
  • a pharmaceutical composition or medicament intended to be administered by injection can be prepared by combining a compound of the present disclosure with sterile, distilled water so as to form a solution.
  • a surfactant may be added to facilitate the formation of a homogeneous solution or suspension.
  • Surfactants are compounds that non-covalently interact with the compound of the present disclosure so as to facilitate dissolution or homogeneous suspension of the compound in the aqueous delivery system.
  • the compounds of the present disclosure, or their pharmaceutically acceptable salts are administered in a therapeutically effective amount, which will vary depending upon a variety of factors including the activity of the specific compound employed; the metabolic stability and length of action of the compound; the age, body weight, general health, sex, and diet of the patient; the mode and time of administration; the rate of excretion; the drug combination; the severity of the particular disorder or condition; and the subject undergoing therapy.
  • Compounds of the present disclosure, or pharmaceutically acceptable derivatives thereof may also be administered simultaneously with, prior to, or after administration of one or more other therapeutic agents.
  • Such combination therapy includes administration of a single pharmaceutical dosage formulation which contains a compound of the present disclosure and one or more additional active agents, as well as administration of the compound of the present disclosure and each active agent in its own separate pharmaceutical dosage formulation.
  • a compound of the present disclosure and the other active agent can be administered to the patient together in a single oral dosage composition such as a tablet or capsule, or each agent administered in separate oral dosage formulations.
  • the compounds of the present disclosure and one or more additional active agents can be administered at essentially the same time, i.e., concurrently, or at separately staggered times, i.e., sequentially; combination therapy is understood to include all these regimens.
  • the concentration of the compound of Formula I provided in the pharmaceutical compositions or medicaments of the present disclosure is less than 100%, 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 19%, 18%, 17%, 16%, 15%, 14%, 13%, 12%,
  • the concentration of the compound of Formula I provided in the pharmaceutical compositions or medicaments of the present disclosure is greater than 90%, 80%, 70%, 60%, 50%, 40%, 30%, 20%, 19.75%, 19.50%, 19.25% 19%, 18.75%, 18.50%, 18.25% 18%, 17.75%, 17.50%, 17.25% 17%, 16.75%, 16.50%, 16.25% 16%, 15.75%, 15.50%, 15.25% 15%, 14.75%, 14.50%, 14.25% 14%, 13.75%, 13.50%, 13.25%
  • the concentration of the compound of the Formula I provided in the pharmaceutical compositions or medicaments of the present disclosure is in the range from approximately 0.0001% to approximately 50%, approximately 0.001% to approximately 40%, approximately 0.01% to approximately 30%, approximately 0.02% to approximately
  • the concentration of the compound of Formula I provided in the pharmaceutical compositions or medicaments of the present disclosure is in the range from approximately 0.001% to approximately 10%, approximately 0.01% to approximately 5%, approximately 0.02% to approximately 4.5%, approximately 0.03% to approximately 4%, approximately 0.04% to approximately 3.5%, approximately 0.05% to approximately 3%, approximately 0.06% to approximately 2.5%, approximately 0.07% to approximately 2%, approximately 0.08% to approximately 1.5%, approximately 0.09% to approximately 1%, approximately 0.1% to approximately 0.9% w/w, w/v or v/v, of the pharmaceutical composition or medicament.
  • the amount the compound of Formula I provided in the pharmaceutical compositions or medicaments of the present disclosure is equal to or less than 10 g, 9.5 g, 9.0 g, 8.5 g, 8.0 g, 7.5 g, 7.0 g, 6.5 g, 6.0 g, 5.5 g, 5.0 g, 4.5 g, 4.0 g, 3.5 g, 3.0 g, 2.5 g, 2.0 g, 1.5 g, 1.0 g, 0.95 g, 0.9 g, 0.85 g, 0.8 g, 0.75 g, 0.7 g, 0.65 g, 0.6 g, 0.55 g, 0.5 g, 0.45 g, 0.4 g, 0.35 g, 0.3 g, 0.25 g, 0.2 g, 0.15 g, 0.1 g, 0.09 g, 0.08 g, 0.07 g, 0.06 g, 0.05 g, 0.04 g, 0.03 g, 0.02 g
  • the amount of the compound of Formula I provided in the pharmaceutical compositions or medicaments of the present disclosure is more than 0.0001 g, 0.0002 g, 0.0003 g, 0.0004 g, 0.0005 g, 0.0006 g, 0.0007 g, 0.0008 g, 0.0009 g, 0.001 g, 0.0015 g, 0.002 g, 0.0025 g, 0.003 g, 0.0035 g, 0.004 g, 0.0045 g, 0.005 g, 0.0055 g, 0.006 g, 0.0065 g, 0.007 g, 0.0075 g, 0.008 g, 0.0085 g, 0.009 g, 0.0095 g, 0.01 g, 0.015 g, 0.02 g, 0.025 g, 0.03 g, 0.035 g, 0.04 g, 0.045 g, 0.05 g, 0.055 g, 0.06 g
  • the amount of the compound of Formula I provided in the pharmaceutical compositions or medicaments of the present disclosure is in the range of 0.0001-10 g, 0.0005-9 g, 0.001-8 g, 0.005-7 g, 0.01-6 g, 0.05-5 g, 0.1-4 g, 0.5-4 g, or 1-3 g of the pharmaceutical composition or medicament.
  • Phenylthiobutanoic acid (30 g, 152.8 mmol) and N,N-dimethylethanolamine (15 g, 168 mmol) were dissolved in CH2CI2 (300 mL).
  • the stirred solution was cooled with an ice- bath and 4-dimethylaminopyridine (1.86 g, 15.28 mmol) and /V,/V'-dicyclohexylcarbodiimide (31.52 g, 152.8 mmol) were sequentially added.
  • the temperature was slowly raised to r.t. and the reaction mixture was stirred overnight. After the consumption of the starting material, water (300 mL) was added to the solution.
  • Example 4 /V V,/V-trimethyl-2- ⁇ [4-(phenylsulfanyl)butanoyl]oxy ⁇ ethan-l-aminium chloride
  • Phenylthiobutanoic acid 24 g, 122.28 mmol, 1 eq.
  • choline chloride 18.78 g, 134.5 mmol, 1.1 eq.
  • the solution was cooled with ice- bath, and with stirring, 4-dimethylaminopyridine (1.494 g, 12.22 mmol, 0.1 eq.) and N,N'- dicyclohexylcarbodiimide (25.23 g, 122.28 mmol, 1 eq.) were sequentially added.
  • the temperature was slowly raised to r.t. and the solution was stirred overnight.
  • a 20 mM stock solution of the test compound in 100% DMSO dilutions were prepared to a theoretical concentration of 400 mM in duplicates in phosphate-buffered saline pH 7.4 (138 mM NaCl, 2.7 mM KC1, 10 mM K-phosphate) with 2% final DMSO.
  • the experimental compound dilutions in PBS were further allowed to equilibrate at 25°C on a thermostatic shaker for two hours and then filtered through HTS filter plates using a vacuum manifold.
  • the filtrates of test compounds were diluted 2-fold with acetonitrile with 2% DMSO before measuring.
  • concentrations of compounds in PBS filtrate were calculated using a dedicated Microsoft Excel calculation script. Proper absorbance wavelengths for calculations were selected for each compound manually based on absorbance maximums (absolute absorbance unit values for the minimum and maximum concentration points within 0 - 3 OD range). Each of the final datasets was additionally visually evaluated by the operator and goodness of fit (R2) was calculated for each calibration curve.
  • the effective range of this assay was approximately 2-400 pM and the compounds returning values close to the upper limit of the range may have higher actual solubility.
  • Compounds (pro-drugs) of the present disclosure are designed to release the active carboxylic acid, PTBA, in vivo.
  • Compounds of the present disclosure have been tested in vitro for plasma stability and in vivo to demonstrate loss of pro-drug over time and the appearance of PTBA.
  • Example 7 Plasma Stability Studies
  • Plasma stability studies were carried out in plasma from various species. For example, the stability of Compound 22 was evaluated in rat, dog, and human plasma at five time points over 120 minutes using HPLC-MS/MS.
  • All measurements were performed using Shimadzu HPLC system including vacuum degasser, gradient pumps, reverse phase column, column oven and autosampler.
  • the HPLC system was coupled with tandem mass spectrometer API 3000 (PE Sciex). Both the positive and negative ion modes of the TurboIonSpray ion source were used. Acquisition and analysis of the data were performed using Analyst 1.5.2 software (PE Sciex). Incubations were carried out in 5 aliquots of 70 pL each (one for each time point), in duplicates.
  • Test compounds (1 mM, final DMSO concentration 1%) were incubated at 37 °C with shaking at 100 rpm. Five time points over 120 minutes have been analyzed. The reactions were stopped by adding 420 pL of acetonitrile-water mixture (90:10) with subsequent plasma proteins sedimentation by centrifuging at 5500 rpm for 5 minutes. Supernatants were analyzed by the HPLC system coupled with tandem mass spectrometer. The percentage of the test compounds remaining after incubation in plasma and their half-lives (T1/2) were calculated.
  • Figures 2-4 illustrate the plasma stability of illustrative Compound 22 over time in Rat, Dog, and Human, respectively.
  • Incubations were carried out in multiple aliquots of 70 pL each (one for each time point), in duplicates.
  • Test compounds (1 mM, final DMSO concentration 1%) were incubated at 37 °C with shaking at 100 rpm. 3-5 time points over 120 minutes were analyzed. The reactions were stopped by adding 420 pL of acetonitrile- water mixture (90:10; stop-solution) with subsequent plasma proteins sedimentation by centrifuging at 5500 rpm for 5 minutes. Supernatants were analyzed by the HPLC system coupled with tandem mass spectrometer.
  • a “true zero timepoint” was established by adding the compound to a mixture of plasma with the stop- solution (no direct contact of the compound with plasma prior to stop solution), due to the rapid degradation of some pro-drugs in plasma.
  • Compound 22 was incubated in monkey plasma, and disappearance of Compound 22 and appearance of PTBA was evaluated.
  • Figure 5 illustrates the percentage of illustrative Compound 22 remaining in Monkey plasma over time.
  • Figure 6 illustrates PTBA levels in Monkey plasma over time following administration of Compound 22.
  • Example 8 Rat Pharmacokinetic Studies
  • Compounds of the present disclosure were evaluated in rat PK studies to determine release of PTBA in vivo after i.v. injection (see Figure 7).
  • the vehicle was Captisol-Saline-Water for injections (20%:40%:40%, v/v/v).
  • the compound was dissolved in the necessary volume of 20% Captisol solution (Captisol was previously dissolved in a mixture of Saline-Water for injections, 1:1, v/v), vortexed for 1 min and sonicated for 1 min at 40°C. The resulting solution was clear.
  • the batch of working formulation was prepared 30 min prior to the in vivo study. Study design, animal selection, handling and treatment were all in accordance with the CRO’s PK study protocols and Institutional Animal Care and Use Guidelines. Animal treatment and plasma samples preparation were conducted by the Animal Laboratory personnel at the CRO.
  • the compound-treated group included 3 animals. Solution of Lidocaine (7 mg/kg) was administered subcutaneously five minutes prior to catheter placement in the left lateral tail vein. The catheter was flushed with 500-IU/ml heparin. The test compound formulation was injected into the right lateral vein. Blood collection was performed from the tail vein in tubes containing K3EDTA. Plasma samples were immediately prepared, flash-frozen and stored at -70°C until subsequent analysis.
  • Elution rate 400 pL/min.
  • a divert valve directed the flow to the detector from 1.3 to 1.8 min.
  • Scan type Positive MRM
  • Ion source Turbo spray
  • Ionization mode ESI
  • Nebulize gas 15 L/min
  • Curtain gas 8 L/min
  • Collision gas 4 L/min
  • Ionspray voltage 5000 V
  • Temperature 400°C
  • Chromatographic Conditions for PTBA
  • Scan type Negative MRM
  • Ion source Turbo spray
  • Ionization mode ESI
  • Nebulize gas 15 L/min
  • Curtain gas 8 L/min
  • Collision gas 4 L/min
  • Ionspray voltage -4200 V
  • Temperature 400°C
  • Illustrative Compound 22 was dosed at 2 mg/kg i.v to 3 rats.
  • Figure 7A shows the concentration time profile of the pro-drug (Compound 22) and PTBA.
  • Illustrative Compound 17 was dosed at 2 mg/kg i.v to 3 rats.
  • Figure 7B shows the concentration time profile of the pro-drug (Compound 17) and PTBA.
  • Illustrative Compound 1 was dosed at 2 mg/kg i.v to 3 rats.
  • Figure 7C shows the concentration time profile of the pro-drug (Compound 1) and PTBA.
  • Illustrative Compound 15 was dosed at 6 mg/kg i.v to 5 rats.
  • Figure 7D shows the concentration time profile of the pro-drug (Compound 15) and PTBA.
  • Example 9 In vivo Efficacy in a Murine AKI Model
  • the aim of the study was to investigate the efficacy of test compound(s) in a (mouse) model for acute kidney injury.
  • the study investigated the efficacy of test compound(s) in cisplatin-induced acute kidney injury (CI-AKI) mouse model.
  • CI-AKI cisplatin-induced acute kidney injury
  • IP intraperitoneal
  • Serum analysis for urea (BUN) was performed on the 5th day and 12th days after cisplatin administration.
  • the mean body weight loss per group was calculated every day of the study as percentage loss of initial (day 0) weight for each individual animal.
  • mice in Group 2 and 4 were repeatedly treated with Compound 22 at the dose of 50 mg/kg with an interval of 24h for seven consecutive days starting from Day 2 or Day 0 of the study, respectively.
  • the mice in Groups 3 and 5 were repeatedly treated with compound 22 at the dose of 50 mg/kg twice a day (daily 9AM and 6PM injections) for seven consecutive days starting from the Day 2 or Day 0 of the study, respectively.
  • the mice in Group 1 remained untreated after the cisplatin injection until the terminal sacrifice.
  • Cisplatin was administered without dilution as a factory -made 1 mg/ml solution for infusion at the injection volume of 11 ml/kg for the dose of 11 mg/kg.
  • the formulation vehicle for compound 22 was Captisol - physiological saline - water for injections (20%:40%:40%, w/v/v). Only freshly prepared formulation was used for each dosing; each working formulation batch was prepared immediately prior to the administration.
  • the working formulations were sterile filtered using 0.2 pm cellulose acetate syringe filters before injections.
  • the dosing volume for the test compound was 5 ml/kg of body weight.
  • the working formulations were administered as transparent clear solutions.
  • IP Intraperitoneal
  • BUN Urea
  • the principle of the method is the ability of urease to catalyze the transfer of in- between urea and H2O.
  • the product of this reaction is NH4+ and C032-.
  • Reproducibility: CV 5.75 %.
  • FIG. 8 Urea level (mean per group ⁇ SE, %) in serum of C57B1/6J female mice on the 5th and 12th day of the study. Cisplatin at a dose of 11 mg/kg was IP administered at Day 0 to all mice. The mice in G2, G3, G4 and G5 groups were repeatedly treated with Compound 22 once or twice a day on days 2-8 (D2-8) or days 0-6 (DO-6) of the study. Difference significance level compared to G1 (ANOVA): * p ⁇ 0.05, ** pO.Ol.
  • FIGS. 9A-9B Body weight loss (mean per group ⁇ SE, %) of the mice survived during the study. Body weight loss was measured on different days compared to the initial weight on Day 0. Cisplatin at dose of 11 mg/kg was IP administered at Day 0 to all mice. The mice in G2, G3, G4 and G5 groups were repeatedly treated with Compound 22 once or twice a day on ( Figure 9A) days 2-8 (D2-8) of the study or ( Figure 9B) days 0-6 (DO-6) of the study. Statistically significant differences were determined by two-way ANOVA and Tukey post-hok analysis, denoted by * for G2 or G4, or ⁇ for G3 or G5 respectively, compared to Gl.
  • Example 10 In vivo Efficacy in a Rat AKI Model
  • IRI-AKI Ischemia-Reperfusion Induced Acute Kidney Injury
  • IRI-AKI model was developed in experimental animals by applying bilateral renal occlusion. The duration of the occlusion was equal 60 minutes. For instance, rhe effects of seven days repeated intravenous (IV) administration of compound 22 of 10 mg/kg on pathology dynamics in rats exposed to IRI-AKI was assessed. Animals were observed for mortality and clinical signs of toxicity daily for eight consecutive days during the study. Serum analysis for creatinine was performed one day prior to ischemia-reperfusion surgery, 24 hours after, and on the 3rd, 5th, and 7th day after IRI-AKI intervention.
  • the formulation for compound 22 was Captisol - physiological saline - water for injections (20%:40%:40%, w/v/v).
  • the formulation vehicle for the compounds was Captisol - physiological saline - water for injections (20%:40%:40%, w/v/v).
  • the working formulations were sterile filtered using 0.2 pm cellulose acetate syringe filters before injections.
  • IV route of administration was used in the study for all compounds. Formulation was injected slow to each animals (during 5 minutes). Each working formulation batch was prepared immediately prior to the administration. The working formulations were administered as transparent clear solutions for all doses for all compounds. The dosing volume for the test compounds and Vehicle was 5 ml/kg of body weight.
  • Creatinine level of the experimental animals exposed to 60 minutes to bilateral renal occlusion and treated with compound 22 was significantly decreased 3 days after IRI with Vehicle treated group.
  • Figure 10 Creatinine values in the serum of Sprague Dawley male rats following renal ischemia and repeated 7 days IV dosing with compound 22. Values are expressed as means ⁇ SEM of 8-14 rats per group. Significant differences were determined by two-way ANOVA, individual comparison was performed by Bonferroni's analysis, denoted by **. [00319] Example 11 : In vivo Efficacy in a Murine AKI Model
  • the aim of the study was to investigate the efficacy of test compounds in the model of cisplatin induced acute kidney injury (CI-AKI) in mice.
  • CI-AKI cisplatin induced acute kidney injury
  • IP intraperitoneal
  • BUN Serum analysis for urea
  • the mean body weight loss per group was calculated every day of the study as percentage loss of initial (day 0) weight for each individual animal.
  • Cisplatin was administered without dilution as a factory-made 1 mg/ml solution for infusion at the injection volume of 11 ml/kg for the dose of 11 mg/kg.
  • the formulation vehicle for compound 3 was Captisol - physiological saline - water for injections (20%:40%:40%, w/v/v). Only freshly prepared formulation was used for each dosing; each working formulation batch was prepared immediately prior to the administration.
  • the working formulations were sterile filtered using 0.2 pm cellulose acetate syringe filters before injections.
  • the dosing volume for the test compound was 5 ml/kg of body weight.
  • the working formulations were administered as transparent clear solutions. Intraperitoneal (IP) route of administration was used in the study.
  • IP Intraperitoneal
  • Urea (BUN) was determined in the serum using a commercial kit according to the manufacturer’s instructions.
  • the principle of the method is the ability of urease to catalyze the transfer of H+ between urea and H20.
  • the product of this reaction is NH4+ and C032-.
  • Reproducibility: CV 5.75 %.
  • Creatinine was measured using mModified Jaffe’s method, without deproteinization. In alkaline solution picrate reacts with creatinine to form a yellow-red 2,4,6- trinitrocyclohexadienate. The color intensity is proportional to the creatinine concentration.
  • Reproducibility: CV 6.1 %.
  • FIG. 11 Urea level (mean per group ⁇ SE, %) in serum of C57B1/6J female mice during the study. Cisplatin at a dose of 11 mg/kg was IP administered at Day 0 to all mice. The mice in G1 or G2 were repeatedly treated with Vehicle or Compound 3, respectively, with an interval of 24h on days 2-9. Statistical significance was determined by ANOVA (p ⁇ 0.05) followed by Bonferroni's multiple comparisons test. ** p ⁇ 0.01.
  • FIG. 12 Creatinine level (mean per group ⁇ SE, %) in serum of C57B1/6J female mice during the study. Cisplatin at a dose of 11 mg/kg was IP administered at Day 0 to all mice. The mice in G1 or G2 were repeatedly treated with Vehicle or Compound 3, respectively, with an interval of 24h on days 2-9. Statistical significance was determined by ANOVA ( p ⁇ 0.05) followed by Bonferroni's multiple comparisons test. * p ⁇ 0.05.
  • FIG. 13 Body weight loss (mean per group ⁇ SE, %) of mice in study. Body weight loss was measured on different days compared to the initial weight on Day 0. Cisplatin at a dose of 11 mg/kg was IP administered at Day 0 to all mice. The mice in G1 or G2 were repeatedly treated with Vehicle or Compound 3, respectively, with an interval of 24h on days 2-9. Statistical significance was determined by ANOVA ( p ⁇ 0.05) followed by Bonferroni's multiple comparisons test. * p ⁇ 0.05. Conclusion
  • any feature herein may be combined with any other feature of a same or different embodiment disclosed herein. It will be appreciated that while features may be optional in certain embodiments, when features are included in such embodiments, they can be required to have a specific configuration as described in the present disclosure.
  • any steps recited in any method or process described herein and/or recited in the claims can be executed in any suitable order and are not necessarily limited to the order described and/or recited, unless otherwise stated (explicitly or implicitly). Such steps can, however, also be required to be performed in a specific order or any suitable order in certain embodiments of the present disclosure.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Urology & Nephrology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Hydrogenated Pyridines (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Polysaccharides And Polysaccharide Derivatives (AREA)
  • Emergency Medicine (AREA)
  • Medicinal Preparation (AREA)
  • Organic Low-Molecular-Weight Compounds And Preparation Thereof (AREA)
EP21838077.2A 2020-07-08 2021-07-08 Wasserlösliche ptba-prodrugs zur verwendung bei der hdac-hemmung und zur verbesserung der nierenerholung nach akuter nierenläsion Pending EP4178560A1 (de)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US202063049615P 2020-07-08 2020-07-08
US202063112444P 2020-11-11 2020-11-11
PCT/US2021/040940 WO2022011174A1 (en) 2020-07-08 2021-07-08 Water soluble prodrugs of ptba for use in hdac inhibition and enhancing renal recovery following acute kidney injury

Publications (1)

Publication Number Publication Date
EP4178560A1 true EP4178560A1 (de) 2023-05-17

Family

ID=79552101

Family Applications (1)

Application Number Title Priority Date Filing Date
EP21838077.2A Pending EP4178560A1 (de) 2020-07-08 2021-07-08 Wasserlösliche ptba-prodrugs zur verwendung bei der hdac-hemmung und zur verbesserung der nierenerholung nach akuter nierenläsion

Country Status (6)

Country Link
EP (1) EP4178560A1 (de)
JP (1) JP2023533986A (de)
KR (1) KR20230039680A (de)
AU (1) AU2021305330A1 (de)
CA (1) CA3185103A1 (de)
WO (1) WO2022011174A1 (de)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP4366716A1 (de) * 2021-07-08 2024-05-15 Klotho Therapeutics, Inc. Neue kombinatorische antitumor-behandlungsverfahren und zugehörige zusammensetzungen, kits und systeme

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2009079375A1 (en) * 2007-12-14 2009-06-25 Georgetown University Histone deacetylase inhibitors
JP2015504056A (ja) * 2011-12-29 2015-02-05 ファーマサイクリックス,インク. ヒストンデアセチラーゼ8の阻害剤としての珪皮酸ヒドロキシアミド
US9670236B2 (en) * 2012-10-31 2017-06-06 University of Pittsburgh—of the Commonwealth System of Higher Education Class of HDAC inhibitors expands the renal progenitor cells population and improves the rate of recovery from acute kidney injury

Also Published As

Publication number Publication date
AU2021305330A1 (en) 2023-02-09
CA3185103A1 (en) 2022-01-13
KR20230039680A (ko) 2023-03-21
WO2022011174A1 (en) 2022-01-13
JP2023533986A (ja) 2023-08-07

Similar Documents

Publication Publication Date Title
JP7133680B2 (ja) バイオアベイラビリティが高いアルボシジブプロドラッグ
US8735454B2 (en) Deuterium-enriched bupropion
US10940143B2 (en) Deuterium-enriched pioglitazone
WO2018096504A1 (en) Cbd prodrugs, compositions, and methods of administering cbd and cbd prodrugs
JP7409719B2 (ja) Erk阻害剤としてのチアゾロラクタム系化合物およびその使用
EP4178560A1 (de) Wasserlösliche ptba-prodrugs zur verwendung bei der hdac-hemmung und zur verbesserung der nierenerholung nach akuter nierenläsion
US20230278953A1 (en) Water soluble prodrugs of ptba for use in hdac inhibition and enhancing renal recovery following acute kidney injury
US20090082312A1 (en) Deuterium-enriched zoledronic acid
US20080161395A1 (en) Derivatives of Aminobutanoic Acid Inhibiting Cpt
US20090076162A1 (en) Deuterium-enriched desvenlafaxine
WO2023283648A1 (en) Novel combinatorial antitumor treatment methods and related compositions, kits, and systems
US20130090357A1 (en) Deuterium-enriched donepezil
US20090069431A1 (en) Deuterium-enriched milnacipran
US7915309B2 (en) Deuterium-enriched oseltamivir
EP3934702B1 (de) [18f]-markiertes benzothiazolderivat als pet-radiotracer
US20090082363A1 (en) Deuterium-enriched posaconazole
US20210121422A1 (en) Glucagon receptor antagonists
US20090062312A1 (en) Deuterium-enriched indiplon
US20100081720A1 (en) Deuterium-enriched atomoxetine
US20090069339A1 (en) Deuterium-enriched ciprofloxacin
US20090076164A1 (en) Deuterium-enriched tapentadol
TW202302118A (zh) 一種選擇性nav抑制劑的可藥用鹽、結晶形式及其製備方法
US20090306215A1 (en) Deuterium-enriched arformoterol
WO2022011171A1 (en) Novel compounds and methods for increasing klotho gene expression
US20090075947A1 (en) Deuterium-enriched fospropofol

Legal Events

Date Code Title Description
STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE INTERNATIONAL PUBLICATION HAS BEEN MADE

PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: REQUEST FOR EXAMINATION WAS MADE

17P Request for examination filed

Effective date: 20230206

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AL AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO RS SE SI SK SM TR

DAV Request for validation of the european patent (deleted)
DAX Request for extension of the european patent (deleted)