EP1525185A1 - Composes d'acide acetylamino benzoique et leur utilisation pour la suppression de non-sens et le traitement de maladie - Google Patents

Composes d'acide acetylamino benzoique et leur utilisation pour la suppression de non-sens et le traitement de maladie

Info

Publication number
EP1525185A1
EP1525185A1 EP03766013A EP03766013A EP1525185A1 EP 1525185 A1 EP1525185 A1 EP 1525185A1 EP 03766013 A EP03766013 A EP 03766013A EP 03766013 A EP03766013 A EP 03766013A EP 1525185 A1 EP1525185 A1 EP 1525185A1
Authority
EP
European Patent Office
Prior art keywords
substituted
unsubstituted
benzoic acid
phenoxy
heteroaryl
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.)
Withdrawn
Application number
EP03766013A
Other languages
German (de)
English (en)
Inventor
Richard G. Wilde
M. Welch. Ellen
James Jan Takasugi
Neil G. Almstead
Steven Marc Rubenstein
Holger Beckmann
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.)
Tularik Inc
PTC Therapeutics Inc
Original Assignee
Tularik Inc
PTC 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 Tularik Inc, PTC Therapeutics Inc filed Critical Tularik Inc
Publication of EP1525185A1 publication Critical patent/EP1525185A1/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • 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
    • C07D207/18Heterocyclic 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 one double bond between ring members or between a ring member and a non-ring member
    • C07D207/22Heterocyclic 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 one double bond between ring members or between a ring member and a non-ring member 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
    • C07D207/24Oxygen or sulfur atoms
    • C07D207/262-Pyrrolidones
    • C07D207/2732-Pyrrolidones with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to other ring carbon atoms
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C235/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms
    • C07C235/02Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton
    • C07C235/04Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton the carbon skeleton being acyclic and saturated
    • C07C235/18Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton the carbon skeleton being acyclic and saturated having at least one of the singly-bound oxygen atoms further bound to a carbon atom of a six-membered aromatic ring, e.g. phenoxyacetamides
    • C07C235/24Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by oxygen atoms having carbon atoms of carboxamide groups bound to acyclic carbon atoms and singly-bound oxygen atoms bound to the same carbon skeleton the carbon skeleton being acyclic and saturated having at least one of the singly-bound oxygen atoms further bound to a carbon atom of a six-membered aromatic ring, e.g. phenoxyacetamides having the nitrogen atom of at least one of the carboxamide groups bound to a carbon atom of a six-membered aromatic ring
    • 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
    • C07C323/62Thiols, 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 atom of at least one of the thio groups bound to a carbon atom of a six-membered aromatic ring of the carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C327/00Thiocarboxylic acids
    • C07C327/38Amides of thiocarboxylic acids
    • C07C327/40Amides of thiocarboxylic acids having carbon atoms of thiocarboxamide groups bound to hydrogen atoms or to acyclic carbon atoms
    • C07C327/42Amides of thiocarboxylic acids having carbon atoms of thiocarboxamide groups bound to hydrogen atoms or to acyclic carbon atoms to hydrogen atoms or to carbon atoms of a saturated carbon skeleton
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D209/00Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D209/02Heterocyclic compounds containing five-membered rings, condensed with other rings, with one nitrogen atom as the only ring hetero atom condensed with one carbocyclic ring
    • C07D209/44Iso-indoles; Hydrogenated iso-indoles
    • C07D209/48Iso-indoles; Hydrogenated iso-indoles with oxygen atoms in positions 1 and 3, e.g. phthalimide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom 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
    • C07D213/62Oxygen or sulfur atoms
    • C07D213/63One oxygen atom
    • C07D213/64One oxygen atom attached in position 2 or 6
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D239/00Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings
    • C07D239/02Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings
    • C07D239/24Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more double bonds between ring members or between ring members and non-ring members
    • C07D239/28Heterocyclic compounds containing 1,3-diazine or hydrogenated 1,3-diazine rings not condensed with other rings having three or more 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, directly attached to ring carbon atoms
    • C07D239/32One oxygen, sulfur or nitrogen atom
    • C07D239/38One sulfur atom
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D257/00Heterocyclic compounds containing rings having four nitrogen atoms as the only ring hetero atoms
    • C07D257/02Heterocyclic compounds containing rings having four nitrogen atoms as the only ring hetero atoms not condensed with other rings
    • C07D257/04Five-membered rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D271/00Heterocyclic compounds containing five-membered rings having two nitrogen atoms and one oxygen atom as the only ring hetero atoms
    • C07D271/02Heterocyclic compounds containing five-membered rings having two nitrogen atoms and one oxygen atom as the only ring hetero atoms not condensed with other rings
    • C07D271/061,2,4-Oxadiazoles; Hydrogenated 1,2,4-oxadiazoles
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D317/00Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms
    • C07D317/08Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3
    • C07D317/44Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3 ortho- or peri-condensed with carbocyclic rings or ring systems
    • C07D317/46Heterocyclic compounds containing five-membered rings having two oxygen atoms as the only ring hetero atoms having the hetero atoms in positions 1 and 3 ortho- or peri-condensed with carbocyclic rings or ring systems condensed with one six-membered ring
    • C07D317/48Methylenedioxybenzenes or hydrogenated methylenedioxybenzenes, unsubstituted on the hetero ring
    • C07D317/62Methylenedioxybenzenes or hydrogenated methylenedioxybenzenes, unsubstituted on the hetero ring 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 atoms of the carbocyclic ring
    • C07D317/64Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D405/00Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom
    • C07D405/02Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings
    • C07D405/12Heterocyclic compounds containing both one or more hetero rings having oxygen atoms as the only ring hetero atoms, and one or more rings having nitrogen as the only ring hetero atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2601/00Systems containing only non-condensed rings
    • C07C2601/06Systems containing only non-condensed rings with a five-membered ring
    • C07C2601/08Systems containing only non-condensed rings with a five-membered ring the ring being saturated
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2602/00Systems containing two condensed rings
    • C07C2602/02Systems containing two condensed rings the rings having only two atoms in common
    • C07C2602/04One of the condensed rings being a six-membered aromatic ring
    • C07C2602/08One of the condensed rings being a six-membered aromatic ring the other ring being five-membered, e.g. indane
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2602/00Systems containing two condensed rings
    • C07C2602/02Systems containing two condensed rings the rings having only two atoms in common
    • C07C2602/04One of the condensed rings being a six-membered aromatic ring
    • C07C2602/10One of the condensed rings being a six-membered aromatic ring the other ring being six-membered, e.g. tetraline

Definitions

  • the invention relates to acetylamino benzoic acid compounds, compositions comprising the compounds and methods for treating or preventing diseases associated with nonsense mutations of mRNA by administering these compounds or compositions.
  • Gene expression in cells depends upon the sequential processes of transcription and translation. Together, these processes produce a protein from the nucleotide sequence of its corresponding gene.
  • Transcription involves the synthesis of mRNA from DNA by RNA polymerase. Transcription begins at a promoter region of the gene and continues until termination is induced, such as by the formation of a stem-loop structure in the nascent RNA or the binding of the rho gene product.
  • Protein is then produced from mRNA by the process of translation, occurring on the ribosome with the aid of tRNA, tRNA synthetases and various other protein and RNA species.
  • Translation comprises the three phases of initiation, elongation and termination. Translation is initiated by the formation of an initiation complex consisting of protein factors, mRNA, tRNA, cofactors and the ribosomal subunits that recognize signals on the mRNA that direct the translation machinery to begin translation on the mRNA. Once the initiation complex is formed, growth of the polypeptide chain occurs by the repetitive addition of amino acids by the peptidyl transferase activity of the ribosome as well as tRNA and tRNA synthetases.
  • the presence of one of the three termination codons (UAA, UAG, UGA) in the A site of the ribosome signals the polypeptide chain release factors (RFs) to bind and recognize the termination signal. Subsequently, the ester bond between the 3' nucleotide of the tRNA located in the ribosome' s P site and the nascent polypeptide chain is hydrolyzed, the completed polypeptide chain is released, and the ribosome subunits are recycled for another round of translation.
  • RFs polypeptide chain release factors
  • Mutations of the DNA sequence in which the number of bases is altered are categorized as insertion or deletion mutations (frameshift mutations) and can result in major disruptions of the genome. Mutations of the DNA that change one base into another are labeled missense mutations and are subdivided into the classes of transitions (one purine to another purine, or one pyrimidine to another pyrimidine) and transversions (a purine to a pyrimidine, or a pyrimidine to a purine).
  • Insertions, deletions, transition and transversion mutations can all result in a nonsense mutation, or chain termination mutation, in which the base mutation or frameshift mutation changes an amino acid codon into one of the three stop codons. These premature stop codons can produce aberrant proteins in cells as a result of premature translation termination.
  • a nonsense mutation in an essential gene can be lethal and can also result in a number of diseases, such as, cancers, lysosomal storage disorders, the muscular dystrophies, cystic fibrosis and hemophilia, to name a few.
  • suppression of the nonsense mutation can arise as a result of a mutation in one of the tRNA molecules so that the mutant tRNA can recognize the nonsense codon, as a result of mutations in proteins that are involved in the translation process, as a result of mutations in the ribosome (either the ribosomal RNA or ribosomal proteins), or by the addition of compounds known to alter the translation process (for example, cycloheximide or the aminoglycoside antibiotics) .
  • LINCL classical late infantile neuronal ceroid lipofuscinosis
  • LINCL a fatal childhood neurodegenerative disease with currently no effective treatment.
  • Premature stop codon mutations in the gene CLN2, encoding the lysosomal tripeptidyl-peptidase 1 (TPP-I) are associated with disease in approximately half of children diagnosed with LLNCL.
  • the ability of the aminoglycoside gentamicin to restore TPP-I activity in LINCL cell lines has been examined.
  • CFTR Transmembrane Conductance Regulator
  • Small molecule therapeutics or prophylactics that suppress premature translation termination by mediating the misreading of the nonsense codon would be useful for the treatment of a number of diseases.
  • the discovery of small molecule drugs, particularly orally bioavailable drugs, may lead to the introduction of a broad spectrum of selective therapeutics which can be used against disease caused by nonsense mutations
  • the invention encompasses novel compounds, novel pharmaceutical compositions and novel methods of treatment.
  • the compounds, compositions, and methods are, in part, based upon the modulation of premature translation termination and/or nonsense-mediated mRNA decay which play a role in a variety of diseases. Such diseases can occur due to the decreased amount of active protein produced as a result of premature termination of translation.
  • the compounds of the invention allow the translation of mRNA to continue past the nonsense mutation resulting in the production of full length protein.
  • the invention encompasses compounds, compositions, and methods for treating and preventing a variety of diseases, in particular genetic diseases.
  • This invention encompasses acetylamino benzoic acid compounds of formula I:
  • X is oxygen, sulfur, CO, SO or S(O) 2 ;
  • Y is oxygen or sulfur
  • Z is substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted cycloalkyl; n is an integer from 0 to 4; R 1 is hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl; substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, SO 2 R 7 , CF 3 , CN, COOH, COR 7 , or COOR 7 ;
  • R is hydrogen, or taken together with R and the atoms to which they are attached form an optionally substituted 5-7 membered heterocyclic, or heteroaryl ring;
  • R 2 , R 3 , R 4 and R 5 are independently hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl; substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, alkoxy, aryloxy, heteroaryloxy, halogen, CF 3 , OCF 3 , OCHF 2 , CN, COOH, COOR 7 , SO 2 R 7 , NO 2 , NH 2 , or N(R 7 ) 2 ;
  • R 6 is hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, or any biohydrolyzable group; and each occurrence of R is independently hydrogen, substituted or unsubsituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl; substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, alkoxy, aryloxy, heteroaryloxy, halogen or CF 3 ; with the proviso that when X is O, Y is O, n is 0 and R 1 is hydrogen, then Z is not 4- chloroph
  • the invention encompasses acetylamino benzoic acid compounds of the formula IV:
  • the invention further encompasses a method for modulation of premature translation termination or nonsense-mediated mRNA decay in a cell comprising contacting __ _ _ a cell exhibiting premature translation termination or nonsense-mediated mRNA decay with an effective amount of a compound of the formula V:
  • V or pharmaceutically acceptable salts, hydrates, clathrates, prodrugs, polymorphs, stereoisomers, including enantiomers, diastereomers, racemates or mixtures of stereoisomers thereof wherein:
  • X is oxygen, sulfur, CO, SO or S(O) 2 ;
  • Y is oxygen or sulfur;
  • Z is substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted cycloalkyl;
  • n is an integer from 0 to 4;
  • R 1 is hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl; substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, SO 2 R 7 , CF 3 , CN, COOH, COR 7 , or COOR 7 ;
  • is hydrogen, or taken together with R 1 and the atoms to which they are attached form an optionally substituted 5-7 membered heterocyclic, or heteroaryl ring;
  • R 2 , R 3 , R 4 and R 5 are independently hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl; substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, alkoxy, aryloxy, heteroaryloxy, halogen, CF 3 , OCF 3 , OCHF 2 , CN, COOH, COOR 7 , SO 2 R 7 , NO 2 , NH 2 , orN(R 7 ) 2 ;
  • R 6 is hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl; substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, or any biohydrolyzable group; and each occurrence of R 7 is independently hydrogen, substituted or unsubsituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl; substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, alkoxy, aryloxy, heteroaryloxy, halogen or CF 3 .
  • the compounds of formulas I, II, III, IV and V are pharmaceutically acceptable salts, hydrates, clathrates, prodrugs, polymorphs, bio-hydrolyzable esters, racemates, or purified stereoisomers including, but not limited to, optically pure enantiomers and diastereomers.
  • the invention further encompasses methods of treating or preventing a disease ameliorated by modulation of premature translation termination or nonsense-mediated mRNA decay, or ameliorating one or more symptoms associated therewith comprising administering to a patient in need thereof a therapeutically or prophylactically effective amount of a compound of the formula V and pharmaceutically acceptable salts, hydrates, solvates, clathrates, prodrugs or polymorphs thereof.
  • the disease is a genetic disease; a CNS disease; an inflammatory disease; a neurodegenerative disease; an autoimmune disease; a proliferative disease, in particular cancer; a cardiovascular disease; or a pulmonary disease; more preferably the disease includes, but is not limited to, amyloidosis, LINCL, hemophilia, Alzheimer's disease, atherosclerosis, giantism, dwarfism, hypothyroidism, hyperthyroidism, cystic fibrosis, aging, obesity, Parkinson's disease, Niemann Pick's disease, cystic fibrosis, familial hypercholesterolemia, retinitis pigmentosa, Duchenne muscular dystrophy, or Marfan syndrome.
  • the invention further encompasses methods of treating or preventing, or ameliorating a genetic disease one or more symptoms associated with or manifestations of a genetic disease comprising administering to a patient in need thereof a therapeutically or prophylactically effective amount of a compound of the formula V and pharmaceutically acceptable salts, hydrates, solvates, clathrates, prodrugs or polymorphs thereof, hi a preferred embodiment, the disease is a CNS disease; an inflammatory disease; a neurodegenerative disease; a cardiovascular disease; an autoimmune disease; cancer; more preferably, the genetic disease includes, but is not limited to, amyloidosis, LINCL, hemophilia, Alzheimer's disease, atherosclerosis, giantism, dwarfism, hypothyroidism, hyperthyroidism, cystic fibrosis, aging, obesity, Parkinson's disease, Niemann Pick's disease, cystic fibrosis, familial hypercholesterolemia, retinitis pigmentosa, Duchenne muscular dystrophy, or Marfan
  • the invention further relates to methods of treating, preventing, or ameliorating cancer or one or more symptoms associated with or manifestations of cancer comprising administering to a patient in need thereof a therapeutically or prophylactically effective amount of a compound of the formula V and pharmaceutically acceptable salts, hydrates, solvates, clathrates, prodrugs or polymo ⁇ hs thereof.
  • the patient is a mammal, more preferably a human susceptible to or at risk of acquiring a genetic disease
  • the patient has undergone a screening process to determine the presence of a nonsense mutation comprising the steps of screening a subject or cells extracted therefrom by an acceptable nonsense mutation screening assay
  • the therapy is personalized in that the patient is screened for a nonsense mutation screening assay and treated by the administration of one or more compounds of the invention; particularly, the patient may be treated with a compound particularly suited for the mutations in question; e.g., depending upon the disease type, cell type, and the gene in question.
  • the patient is an infant or child.
  • the invention encompassing the treatment of pregnant woman or the fetus directly.
  • the compound is administered parenterally, transdermally, mucosally, nasally, buccally, sublingually, or orally; more preferably the compound is administered orally, most preferably the compound is administered orally in the form of a tablet or capsule.
  • the invention encompasses methods for modulating premature translation termination and/or nonsense-mediated mRNA decay.
  • the invention further encompasses a method for suppressing premature translation termination and/or nonsense-mediated mRNA decay in a cell comprising contacting a cell exhibiting premature translation termination and/or nonsense-mediated mRNA decay with an effective amount of a compound of formula I, II, III, IV or V.
  • the invention further encompasses a method for inducing nonsense suppression in a cell comprising contacting a cell exhibiting a nonsense mutation with an effective amount of a compound of formula I, II, III, IV or V.
  • a nonsense codon can be present in the DNA or RNA of any type of cell and can arise naturally or result from mutagenesis.
  • cells encompassed by the present methods include animal cells, mammalian cells, bacterial cells, plant cells and virally infected cells.
  • the nonsense codon was present in the progenitor DNA.
  • the nonsense codon resulted from mutagenesis.
  • the ability of the compounds of formula I, II, III, IV or V to promote readthrough of stop codons makes them useful in the treatment or prevention of any disease which is caused in whole or in part by a nonsense mutation. Such diseases can occur due to the decreased amount of active protein produced as a result of premature termination of translation.
  • the compounds of formula I, II, III, IV or V allow the translation of mRNA to continue past the nonsense mutation resulting in the production of full length protein.
  • a powerful aspect of the invention is that the therapeutic activity of compounds of formula I, II, III, IV or V are not necessarily disease specific, instead are effective at treating of preventing any disease associated with a nonsense mutation. Further, the methods of the invention may be patient specific.
  • a patient may be screened to determine if this disease is associated with a nonsense mutation. If so, they can be treated with a compound of the invention.
  • the compounds of formula I, II, III, IV or V are useful for treating or preventing genetic diseases. Genetic diseases that can be treated or prevented by compounds of formula I, II, III, IV or V include cancer, autoimmune disease, blood disease, collagen disease, diabetes, inflammatory diseases or a central nervous system disease.
  • premature translation termination refers to the result of a mutation that changes a codon corresponding to an amino acid to a stop codon.
  • nonsense-mediated mRNA decay refers to any mechanism that mediates the decay of mRNAs containing a premature translation termination codon.
  • a "premature termination codon” or “premature stop codon” refers to the occurrence of a stop codon where a codon corresponding to an amino acid should be.
  • a "nonsense mutation” is a point mutation changing a codon corresponding to an amino acid to a stop codon.
  • nonsense suppression refers to the inhibition or suppression of premature translation and/or nonsense-mediated mRNA decay.
  • modulation of premature translation termination and/or nonsense- mediated mRNA decay refers to the regulation of gene expression by altering the level of nonsense suppression. For example, if it is desirable to increase production of a defective protein encoded by a gene with a premature stop codon, i.e., to permit readthrough of the premature stop codon of the disease gene so translation of the gene can occur, then modulation of premature translation termination and/or nonsense-mediated mRNA decay entails up-regulation of nonsense suppression. Conversely, if it is desirable to promote the degradation of an mRNA with a premature stop codon, then modulation of premature translation termination and/or nonsense-mediated mRNA decays entails down-regulation of nonsense suppression.
  • the term "patient” means an animal (e.g., cow, horse, sheep, pig, chicken, turkey, quail, cat, dog, mouse, rat, rabbit, guinea pig, etc.), preferably a mammal such as a non-primate and a primate (e.g. , monkey and human), most preferably a human.
  • the patient is an infant, child, adolescent or adult.
  • it has been determined through pre-screening that the patient possesses a nonsense mutation, hi another embodiment, it has been determined through pre-screening which non-sense mutation the patient has (i.e., UAA, UGA, or UAG).
  • the patient is infected with bacterial cells (e.g., Pseudomonas aeruginosa).
  • the cells of the patient are virally infected.
  • substituted means a group substituted by one to four or more substituents, such as, alkyl, alkenyl, alkynyl, aroyl, cycloalkyl, halo, haloalkyl (e.g., trifluoromethyl, difluoromethyl, fluoromethyl, trifluoromethoxy, difluoromethoxy, fluoromethoxy), haloalkoxy, hydroxy, alkoxy, cycloalkyoxy, heterocylooxy, oxo, alkanoyl, aryl, aryloxy, arylalkyl, alkylaryl, heteroaryl, heteroarylalkyl, alkylheteroaryl, heterocyclo, alkanoyloxy, amino, alkylamino, arylamino, aralkylamino, cycloalkylamino, heterocycloamino, mono and disubstituted
  • CONH 2 substituted carbamyl (e.g., CONH alkyl, CONH aryl, CONH aralkyl or instances where there are two substituents on the nitrogen selected from alkyl, aryl or aralkyl), alkoxycarbonyl, aryl, substituted aryl, guanidino,heterocycloalkyl, substituted heterocycloalkyl, heterocycloaryl and substituted heterocycloaryl (such as, indolyl, imidazolyl, furyl, thienyl, thiazolyl, pyrrolidyl, pyridyl, pyrimidyl and the like).
  • substituted carbamyl e.g., CONH alkyl, CONH aryl, CONH aralkyl or instances where there are two substituents on the nitrogen selected from alkyl, aryl or aralkyl
  • alkoxycarbonyl aryl, substituted aryl, guanidin
  • substituents themselves are further substituted, such further substituents are selected from the group consisting of halogen, alkyl, alkoxy, aryl, heteroaryl, heterocyclo, cycloalkyl, and arylalkyl.
  • alkyl means a saturated straight chain or branched non-cyclic hydrocarbon having from 1 to 20 carbon atoms, preferably 1-10 carbon atoms and most preferably 1-4 carbon atoms.
  • saturated straight chain alkyls include -methyl, -ethyl, -n-propyl, -n-butyl, -n-pentyl, -n- hexyl, -n-heptyl, -n-octyl, -n-nonyl and -n-decyl; while saturated branched alkyls include - isopropyl, -sec-butyl, -isobutyl, -tert-butyl, -isopentyl, 2-methylbutyl, 3-methylbutyl, 2- methylpentyl, 3-methylpentyl, 4-methylpentyl, 2-methylhexyl, 3-methylhexyl, 4- methylhexyl, 5-methylhexyl, 2,3-dimethylbutyl, 2,3-dimethylpentyl, 2,4-dimethylpentyl, 2,3-dimethylhexyl, 2,4-dimethyl
  • alkenyl group means a straight chain or branched non-cyclic hydrocarbon having from 2 to 20 carbon atoms, more preferably 2-10 carbon atoms, most preferably 2-6 carbon atoms, and including at least one carbon-carbon double bond.
  • Representative straight chain and branched (C 2 -C 10 )alkenyls include -vinyl, -allyl, -1-butenyl, -2-butenyl, -isobutylenyl, -1-pentenyl, -2-pentenyl, -3- methyl-1-butenyl, -2-methyl-2-butenyl, -2,3-dimethyl-2-butenyl, -1-hexenyl, -2-hexenyl, -3- hexenyl, -1-heptenyl, -2-heptenyl, -3-heptenyl, -1-octenyl, -2-octenyl, -3-octenyl, -1- nonenyl, -2-nonenyl, -3-nonenyl, -1-decenyl, -2-decenyl, -3-decenyl and the like.
  • alkynyl group means a straight chain or branched non-cyclic hydrocarbon having from 2 to 20 carbon atoms, more preferably 2-10 carbon atoms, most preferably 2-6 carbon atoms, and including at lease one carbon-carbon triple bond.
  • Representative straight chain and branched -(C 2 -C 10 )alkynyls include -acetylenyl, -propynyl, -1-butynyl, -2-butynyl.
  • the triple bond of an alkynyl group can be unconjugated or conjugated to another unsaturated group.
  • An alkynyl group can be unsubstituted or substituted.
  • halogen'Or “halo” means fluorine, chlorine, bromine, or iodine.
  • haloalkyl means -alkyl substituted with one or more halogens, wherein alkyl and halogen are defined as above, including -CF3, -CHF2, -CH2F, -CC13, -CHC12, -CBr3, -CHBr2, -CH2CF3, - CH2CHF2, - CH2CH2F, and the like.
  • alkyl sulfonyl means -SO 3 - alkyl, wherein alkyl is defined as above, including -SO 2 -CH 3 , -SO 2 -CH 2 CH 3 , -SO 2 - (CH 2 ) 2 CH 3 , -SO 2 -(CH 2 ) 3 CH 3, -SO 2 -(CH 2 ) 4 CH 3 , -SO 2 -(CH 2 ) 5 CH 3 , and the like.
  • sulfonyl alkyl means -Alkyl- SO H, wherein alkyl is defined as above.
  • carboxyl and “carboxy” mean -COOH or a salt thereof (e.g, -COO " Na + ).
  • alkoxy means -O-(alkyl), wherein alkyl is defined above, including -OCH 3 , -OCH 2 CH 3 , -O(CH 2 ) 2 CH 3) -O(CH 2 ) 3 CH 3, -O(CH 2 ) 4 CH 3 , -O(CH 2 ) 5 CH 3 , and the like.
  • the esters are biohydrolyzable (i.e., the ester is hydrolyzed to a carboxylic acid in vitro or in vivo).
  • haloalkoxy means -alkoxy substituted with one or more halogens, wherein alkoxy and halogen are defined as above, including -OCF3, -OCHF2, - OCH2F, -OCC13, -OCHC12, -OCBr3, -OCHBr2, -OCH2CF3, -OCH2CHF2, -OCH2CH2F, and the like.
  • the esters are biohydrolyzable (i.e., the ester is hydrolyzed to a carboxylic acid in vitro or in vivo).
  • carboxyalky means -(alkyl)- carboxy, wherein alkyl and carboxy are defined above, including -CH2-COOH, -(CH2)2- COOH, -(CH2)3-COOH, -(CH2)4-COOH, and the like.
  • aryl means a carbocychc aromatic ring containing from 5 to 14 ring atoms.
  • the ring atoms of a carbocychc aryl group are all carbon atoms.
  • Aryl ring structures include compounds having one or more ring structures such as mono-, bi-, or tricyclic compounds as well as benzo-fused carbocychc moieties such as 5,6,7,8-tefrahydronaphthyl and the like.
  • the aryl group is a monocyclic ring or bicyclic ring.
  • aryl groups include phenyl, tolyl, anthracenyl, fluorenyl, indenyl, azulenyl, phenanthrenyl and naphthyl.
  • a carbocyclic aryl group can be unsubstituted or substituted.
  • heteroaryl means a carbocyclic aromatic ring containing from 5 to 14 ring atoms and the ring atoms contain at least one heteroatom, preferably 1 to 3 heteroatoms, independently selected from nitrogen, oxygen, or sulfur.
  • Heteroaryl ring structures include compounds having one or more ring structures such as mono-, bi-, or tricyclic compounds as well as fused heterocyclic moities.
  • heteroaryls are triazolyl, tetrazolyl, oxadiazolyl, pyridyl, furyl, benzofuranyl, thiophenyl, benzothiophenyl, benzoisoxazolyl, benzoisothiazolyl, quinolinyl, pyrrolyl, indolyl, oxazolyl, benzoxazolyl, imidazolyl, benzimidazolyl, thiazolyl, benzothiazolyl, isoxazolyl, pyrazolyl, isothiazolyl, pyridazinyl, pyrimidinyl, pyrazinyl, triazinyl, cimiolinyl, phthalazinyl, quinazolinyl, benzoquinazolinyl, acridinyl, pyrimidyl, oxazolyl, benzo[l,3]dioxole and 2,3-d
  • aryloxy means -O-aryl group, wherein aryl is as defined above.
  • An aryloxy group can be unsubstituted or substituted.
  • arylalkyl means -(alkyl)-(aryl), wherein alkyl and aryl are defined above, including, but not limited to -(CH 2 )phenyl, - (CH 2 ) 2 phenyl, -(CH 2 ) 3 phenyl, -CH(phenyl) 2 , -CH(phenyl) 3 , -(CH 2 )tolyl, -(CH 2 )anthracenyl, -(CH 2 )fluorenyl, -(CH 2 )indenyl, -(CH 2 )azulenyl, -(CH 2 )naphthyl, and the like.
  • alkylaryl means -(aryl)-(alkyl), wherein alkyl and aryl are defined above, including, but not limited to 2-methyl-phenyl, 3- methyl-phenyl, 4-methyl-phenyl, 5-methyl-phenyl, 2,3-dimethylphenyl, 2,4-dimethyl- phenyl, 2,5-dimethylphenyl, 3,4-dimethyl-phenyl, 3,5-dimethyl-phenyl, 2-ethyl-phenyl, 3- ethyl-phenyl, 4-ethyl-phenyl, 5-ethyl-phenyl, 2-isopropyl-phenyl, 3-isopropyl-phenyl, 4- isopropyl-phenyl, 5-isopropyl-phenyl, 4-isopropyl-3-methyl-phenyl, 3-isopropyl-5-methyl- phenyl and the like.
  • alkyl and aryl are defined above, including, but not limited to
  • alkyl (alkyl)-(heteroaryl), wherein alkyl and heteroaryl are defined above, including, but not limited to -(CH 2 )pyridyl, -(CH 2 ) 2 pyridyl, -(CH 2 ) 3 pyridyl, -CH(pyridyl) 2 , -C(pyridyl) 3 , - (CH 2 )triazolyl, -(CH 2 )tetrazolyl, -(CH 2 )oxadiazolyl, -(CH 2 )furyl, -(CH 2 )benzofuranyl, - (CH 2 )thiophenyl, -(CH 2 )benzothiophenyl, and the like.
  • alkylheteroaryl means -( heteroaryl)-( alkyl), wherein heteroaryl and alkyl are defined above, including, but not limited to, -pyridyl-(CH3), -triazolyl-(CH3), -thiazolyl-(CH3), -tetrazolyl-(CH3), - oxadiazolyl-(CH3), -furyl-(CH3), -benzofuranyl-(CH3), -thiophenyl-(CH3), - benzothiophenyl-(CH3), and the like wherein each alkyl group can be further substituted.
  • arylalkyloxy means -O-
  • alkyl (alkyl)-(aryl), wherein alkyl and aryl are defined above, including, but not limited to -O- (CH 2 ) 2 phenyl, -O-(CH 2 ) 3 phenyl, -O-CH(phenyl) 2 , -O-CH(phenyl) 3 , -O-(CH 2 )tolyl, -O- (CH 2 )anthracenyl, -O-(CH 2 )fluorenyl, -O-(CH 2 )indenyl, -O-(CH 2 )azulenyl, -O- (CH 2 )naphthyl, and the like.
  • cycloalkyl means a monocyclic or polycyclic saturated ring comprising carbon and hydrogen atoms and having no carbon-carbon multiple bonds.
  • a cycloalkyl group can be unsubstituted or substituted.
  • Examples of cycloalkyl groups include, but are not limited to, (C 3 _C )cycloalkyl groups, including cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and cycloheptyl, and saturated cyclic and bicyclic terpenes.
  • a cycloalkyl group can be unsubstituted or substituted.
  • the cycloalkyl group is a monocyclic ring or bicyclic ring.
  • heterocyclyl and “heterocyclo” means a monocyclic or polycyclic ring comprising carbon and hydrogen atoms, optionally having 1 or 2 multiple bonds, and the ring atoms contain at least one heteroatom, preferably 1 to 3 heteroatoms, independently selected from nitrogen, oxygen, and sulfur.
  • Heterocyclyl ring structures include compounds having one or more ring structures such as mono-, bi-, or tricylic compounds.
  • the heterocyclyl group is a monocyclic ring or bicyclic ring.
  • heterocycles include, but are not limited to morpholinyl, pyrrolidinonyl, pyrrolidinyl, piperidinyl, hydantoinyl, valerolactamyl, t _ ⁇ ⁇ _ _ oxiranyl, oxetanyl, tetrahydrofuranyl, tetraliydropyranyl, tetrahydropyridinyl, tefrahydroprimidinyl, tetrahydrothiophenyl, tetrahydrothiopyranyl, and the like.
  • a heterocyclyl ring can be unsubstituted or substituted.
  • cycloalkyloxy means -O- (cycloalkyl), wherein cycloalkyl is defined above, including -O-cyclopropyl, -O-cyclobutyl, -O-cyclopentyl, -O-cyclohexyl, -O-cycloheptyl and the like.
  • cycloalkylalkyloxy means -O- (alkyl)-(cycloalkyl), wherein cycloalkyl and alkyl are defined above, including, but not limited to -O-cyclopropyl, -O-cyclobutyl, -O-cyclopentyl, -O-cyclohexyl, -O-cycloheptyl and the like.
  • heterocycloalkyl means - (alkyl)-(heterocyclo), wherein heterocyclo and alkyl are defined above, including, but not limited to -CH2-morpholinyl, -CH2-pyrrolidinonyl, -CH2-pyrrolidinyl, -(CH2)2- piperidinyl, -(CH2)3 -piperidinyl, -(CH2)4-piperidinyl, -CH2-hydantoinyl and the like.
  • aminoalkoxy means -O-
  • alkyl (alkyl)-NH2, wherein alkyl is defined above, including, but not limited to -O-CH 2 -NH 2 , -O- (CH 2 ) 2 -NH 2 , -O-(CH 2 ) 3 -NH 2 , -O-(CH 2 ) 4 -NH 2 , -O-(CH 2 ) 5 -NH 2 , and the like.
  • alkylamino means -NH(alkyl) or -N(alkyl)(alkyl), wherein alkyl is defined above, including, but not limited to NHCH 3 , - NHCH 2 CH 3 , -NH(CH 2 ) 2 CH 3 , -NH(CH 2 ) 3 CH 3 , -NH(CH 2 ) CH 3 , -NH(CH 2 ) 5 CH 3 , -N(CH 3 ) 2 , - N(CH 2 CH 3 ) 2 , -N((CH 2 ) 2 CH 3 ) 2 , -N(CH 3 )(CH 2 CH 3 ), and the like.
  • arylamino means -NH(aryl), wherein aryl is defined above, including, but not limited to -NH(phenyl), -NH(tolyl), - NH(anthracenyl), -NH(fluorenyl), -NH(indenyl), -NH(azulenyl), -NH(pyridinyl), - NH(naphthyl), and the like.
  • arylalkylamino means -NH- (alkyl)-(aryl), wherein alkyl and aryl are defined above, including -NH-CH 2 -(phenyl), -NH- CH 2 -(tolyl), -NH-CH 2 -(anthracenyl), -NH-CH 2 -(fluorenyl), -NH-CH 2 -(indenyl), -NH-CH 2 - (azulenyl), -NH-CH 2 -(pyridinyl), -NH-CH 2 -(naphthyl), -NH-(CH 2 ) 2 -( ⁇ henyl) and the like.
  • cycloalkylamino means -NH-NH-NH-
  • cycloalkyl wherein cycloalkyl is defined above, including -NH-cyclopropyl, -NH- cyclobutyl, -NH-cyclopentyl, -NH-cyclohexyl, -NH-cycloheptyl, and the like.
  • aminoalkyl means -(alkyl)- NH 2 , wherein alkyl is defined above, including -CH 2 -NH 2 , -(CH 2 )2-NH 2 , -(CH 2 ) 3 -NH 2 , - (CH 2 ) 4 -NH 2 , -(CH 2 ) 5 -NH 2 and the like.
  • alkylaminoalkyi means - (alkyl)-NH(alkyl) or -(alkyl)-N(alkyl)(alkyl), wherein each "alkyl” is independently an alkyl group defined above, including -CH 2 -NH-CH 3 , -CH 2 -NHCH 2 CH 3 , -CH 2 - NH(CH 2 ) 2 CH 3 , -CH 2 -NH(CH 2 ) 3 CH 3 , -CH 2 -NH(CH 2 ) 4 CH 3 , -CH 2 -NH(CH 2 ) 5 CH 3 , -(CH 2 ) 2 - NH-CH 3 , -CH 2 -N(CH 3 ) 2 , -CH2-N(CH 2 CH 3 )2, -CH 2 -N((CH 2 )2CH 3 ) 2 , -CH 2 - N(CH 3 )(CH 2 CH 3 ),
  • a "therapeutically effective amount” refers to that amount of the compound of the invention or other active ingredient sufficient to provide a therapeutic benefit in the treatment or management of the disease or to delay or minimize symptoms associated with the disease.
  • a therapeutically effective amount with respect to a compound of the invention means that amount of therapeutic agent alone, or in combination with other therapies, that provides a therapeutic benefit in the treatment or management of the disease. Used in connection with an amount of a compound of the invention, the term can encompass an amount that improves overall therapy, reduces or avoids symptoms or causes of disease, or enhances the therapeutic efficacy of or synergies with another therapeutic agent.
  • a prophylactically effective amount refers to that amount of a compound of the invention or other active ingredient sufficient to result in the prevention, recurrence or spread of the disease.
  • a prophylactically effective amount may refer to the amount sufficient to prevent initial disease or the recurrence or spread of the disease or the occurrence of the disease in a patient, including but not limited to those predisposed to the disease.
  • a prophylactically effective amount may also refer to the amount that provides a prophylactic benefit in the prevention of the disease.
  • a prophylactically effective amount with respect to a compound of the invention means that amount alone, or in combination with other agents, that provides a prophylactic benefit in the prevention of the disease. Used in connection with an amount of a compound of the invention, the term can encompass an amount that improves overall prophylaxis or enhances the prophylactic efficacy of or synergies with another prophylactic agent.
  • a "therapeutic protocol” refers to a regimen of timing and dosing of one or more therapeutic agents.
  • a “prophylactic protocol” refers to a regimen of timing and dosing of one or more prophylactic agents.
  • a “protocol” includes dosing schedules and dosing regimens.
  • in combination refers to the use of more than one prophylactic and/or therapeutic agents.
  • a subject is administered one or more prophylactic or therapeutic agents to "manage” a disease so as to prevent the progression or worsening of the disease.
  • the terms "prevent”, “preventing” and “prevention” refer to the prevention of the onset, recurrence, or spread of the disease in a subject resulting from the administration of a prophylactic or therapeutic agent.
  • the terms “treat”, “treating” and “treatment” refer to the eradication or amelioration of the disease or symptoms associated with the disease. In certain embodiments, such terms refer to minimizing the spread or worsening of the disease resulting from the administration of one or more prophylactic or therapeutic agents to a subject with such a disease.
  • pharmaceutically acceptable salts refer to salts prepared from pharmaceutically acceptable non-toxic acids or bases including inorganic acids and bases and organic acids and bases.
  • suitable pharmaceutically acceptable base addition salts for the compound of the present invention include, but are not limited to, metallic salts made from aluminum, calcium, lithium, magnesium, potassium, sodium and zinc or organic salts made from lysine, N,N'-dibenzylethylenediamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine) and procaine.
  • Suitable non-toxic acids include, but are not limited to, inorganic and organic acids such as acetic, alginic, anthranilic, benzenesulfonic, benzoic, camphorsulfonic, citric, ethenesulfonic, formic, fumaric, furoic, galacturonic, gluconic, glucuronic, glutamic, glycolic, hydrobromic, hydrochloric, isethionic, lactic, maleic, malic, mandelic, methanesulfonic, mucic, nitric, pamoic, pantothenic, phenylacetic, phosphoric, propionic, salicylic, stearic, succinic, sulfanilic, sulfuric, tartaric acid, and p-toluenesulfonic acid.
  • inorganic and organic acids such as acetic, alginic, anthranilic, benzenesulfonic, benzoic, camphorsulfonic
  • Specific non-toxic acids include hydrochloric, hydrobromic, phosphoric, sulfuric, and methanesulfonic acids.
  • Examples of specific salts thus include hydrochloride and mesylate salts.
  • Other examples of salts are well known in the art, see, e.g., Remington 's Pharmaceutical Sciences, 18th ed., Mack Publishing, Easton PA (1990).
  • polymorph refers to solid crystalline forms of a compound of the present invention or complex thereof. Different polymo ⁇ hs of the same compound can exhibit different physical, chemical and/or spectroscopic properties. Different physical properties include, but are not limited to stability (e.g., to heat or light), compressibility and density (important in formulation and product manufacturing), and dissolution rates (which can affect bioavailability).
  • Differences in stability can result from changes in chemical reactivity (e.g., differential oxidation, such that a dosage form discolors more rapidly when comprised of one polymo ⁇ h than when comprised of another polymo ⁇ h) or mechanical characteristics (e.g., tablets crumble on storage as a kinetically favored polymo ⁇ h converts to thennodynamically more stable polymo ⁇ h) or both (e.g., tablets of one polymo ⁇ h are more susceptible to breakdown at high humidity).
  • Different physical properties of polymo ⁇ hs can affect their processing. For example, one polymo ⁇ h might be more likely to form solvates or might be more difficult to filter or wash free of impurities than another due to, for example, the shape or size distribution of particles of it.
  • hydrate means a compound of the present invention or a salt thereof, that further includes a stoichiometric or non-stoichiometric amount of water bound by non-covalent intermolecular forces.
  • clathrate means a compound of the present invention or a salt thereof in the fonn of a crystal lattice that contains spaces (e.g., channels) that have a guest molecule (e.g., a solvent or water) trapped within.
  • spaces e.g., channels
  • guest molecule e.g., a solvent or water
  • prodrug means a derivative of a compound that can hydrolyze, oxidize, or otherwise react under biological conditions (in vitro or in vivo) to provide an active compound, particularly a compound of the invention.
  • prodrugs include, but are not limited to, derivatives and metabolites of a compound of the invention that include biohydrolyzable moieties such as biohydrolyzable amides, biohydrolyzable esters, biohydrolyzable carbamates, biohydrolyzable carbonates, biohydrolyzable ureides, and biohydrolyzable phosphate analogues.
  • prodrugs of compounds with carboxyl functional groups are the lower alkyl esters of the carboxylic acid.
  • the carboxylate esters are conveniently formed by esterifying any of the carboxylic acid moieties present on the molecule.
  • Prodrugs can typically be prepared using well-known methods, such as those described by Burger 's Medicinal Chemistry and Drug Discovery 6th ed. (Donald J. Abraham ed., 2001, Wiley) and£>e5?g77 and Application of Prodrugs (H. Bundgaard erf., 1985, Harwood Academic Publishers Gmfh).
  • biohydrolyzable amide means an amide, ester, carbamate, carbonate, ureide, or phosphate, respectively, of a compound that either: 1) does not interfere with the biological activity of the compound but can confer upon that compound advantageous properties in vivo, such as uptake, duration of action, or onset of action; or 2) is biologically inactive but is converted in vivo to the biologically active compound.
  • biohydrolyzable esters include, but are not limited to, lower alkyl esters, alkoxyacyloxy esters, alkyl acylamino alkyl esters, and choline esters.
  • biohydrolyzable amides include, but are not limited to, lower alkyl amides, ⁇ -amino acid amides, alkoxyacyl amides, and alkylaminoalkylcarbonyl amides.
  • biohydrolyzable carbamates include, but are not limited to, lower alkylamines, substituted ethylenediamines, aminoacids, hydroxyalkylamines, heterocyclic and heteroaromatic amines, and polyether amines.
  • optically pure or “stereomerically pure” means a the stereoisomer of a compound is substantially free of the other stereoisomers of that compound.
  • a stereomerically pure compound having one chiral center will be substantially free of the opposite enantiomer of the compound.
  • a stereomerically pure compound having two chiral centers will be substantially free of other diastereomers of the compound.
  • a typical stereomerically pure compound comprises greater than about 80% by weight of one stereoisomer of the compound and less than about 20% by weight of other stereoisomers of the compound, more preferably greater than about 90% by weight of one stereoisomer of the compound and less than about 10% by weight of the other stereoisomers of the compound, even more preferably greater than about 95% by weight of one stereoisomer of the compound and less than about 5% by weight of the other stereoisomers of the compound, and most preferably greater than about 97% by weight of one stereoisomer of the compound and less than about 3% by weight of the other stereoisomers of the compound.
  • enantiomerically pure means a stereomerically pure composition of a compound having one chiral center.
  • the depicted structure is to be accorded more weight.
  • the stereochemistry of a structure or a portion of a structure is not indicated with, for example, bold or dashed lines, the structure or portion of the structure is to be inte ⁇ reted as encompassing all stereoisomers of it.
  • This invention includes acetylamino benzoic acid compounds of formula I:
  • X is oxygen, sulfur, CO, SO or S(O) 2 ;
  • Y is oxygen or sulfur;
  • Z is substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or xmsubstituted cycloalkyl; n is an integer from 0 to 4;
  • R 1 is hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl; substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, SO 2 R 7 , CF 3 , CN, COOH, COR 7 , or COOR 7 ;
  • is hydrogen, or taken together with R 1 and the atoms to which they are attached form an optionally substituted 5-7 membered heterocyclic, or heteroaryl ring;
  • R 6 is hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, or any biohydrolyzable group; and each occurrence of R 7 is independently hydrogen, substituted or unsubsituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl; substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, alkoxy, aryloxy, heteroaryloxy, halogen or CF 3 ; with the proviso that when X is O, Y is O, n is 0 and R 1 is hydrogen, then Z is not 4- chlor
  • This invention further encompasses acetylamino benzoic acid compounds of the formula II:
  • X is oxygen or sulfur
  • Z is substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted cycloalkyl;
  • R 1 is hydrogen, substituted or unsubsituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl; substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, halogen, SO 2 R 7 , CF 3 , COOH, COR 7 , or COOR 7 ;
  • R 2 , R 3 , R 4 and R 5 are independently hydrogen, substituted or unsubsituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl; substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, alkoxy, aryloxy, heteroaryloxy, halogen, CF 3 , OCF 3 , OCHF 2 , CN, COOH, COOR 7 , SO 2 R 7 , NO 2 , NH 2 , or NR 7 2 ; and
  • R 6 is hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, or any biohydrolyzable group; and each occurrence of R 7 is independently hydrogen, substituted or unsubsituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl; substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, alkoxy, aryloxy, heteroaryloxy, halogen or CF 3 ; with the proviso that when X is O and R 1 is hydrogen, then Z is not 4-chlorophenyl, 4-methylphenyl
  • This invention further encompasses acetylamino benzoic acid compounds of the formula III:
  • Z is substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted cycloalkyl;
  • R 1 is hydrogen, substituted or unsubsituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl; substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, SO 2 R 2 ,CF 3 , CN, COOH, COR 2 , or COOR 2 ; R is hydrogen, substituted or unsubsituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl; substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, alkoxy, aryloxy, heteroaryloxy, halogen,
  • R 6 is hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, or any biohydrolyzable group; and each occurrence of R 2 is independently hydrogen, substituted or unsubsituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl; substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, alkoxy, aryloxy, heteroaryloxy, halogen or CF 3 ; with the proviso that when R 1 is hydrogen, then Z is not 4-chlorophenyl, 4- methylphenyl, 3-chlor
  • R is not hydrogen.
  • Z is 4-isopropyl-3 -methylphenyl, 3 -isopropyl-5 -methylphenyl, 2'-methyl-biphenyl-4-yl, 3'-methyl-biphenyl-4-yl, 4-propyl- phenyl, 4-(l,l-dimethyl-propyl)-phenyl, 5,6,7,8,-tetrahydronaphthalen-2-yl, 4-tert-butyl- phenyl, 4-cyclopentyl-phenyl, or 4-trifluoromethoxyphenyl, more preferably, Z is 4- isopropyl-3 -methylphenyl, 3-isopropyl-5-methylphenyl, 2'-methyl-biphenyl-4-yl, 3'- methyl-biphenyl-4-yl, 4-propyl-phenyl, 4-(l,l-dimethyl-propyl)
  • Z is substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted cycloalkyl;
  • R° and R 1 and the atoms to which they are attached form an optionally substituted 5- 7 membered heterocyclicor heteroaryl ring;
  • R is hydrogen, substituted or unsubsituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl; substituted or unsubstituted cycloalkyl, ⁇ _ _ substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, alkoxy, aryloxy, heteroaryloxy, halogen, CF 3 , OCF 3 , OCHF 2 , CN, COOR 2 , SO 2 , NO 2 , NH 2 , or NR 2 2 ;
  • R 6 is hydrogen, substituted or unsubstituted alkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, or any biohydrolyzable group; and each occunence of R 2 is independently hydrogen, substituted or unsubsituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl; substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, substituted or xmsubstituted heteroaryl, alkoxy, aryloxy, heteroaryloxy, halogen or CF 3 .
  • Preferred compounds of the invention include, but are not limited to,
  • Potency and efficacy activities are ranked as either extremely high, very high or significant. The combination of these activities is used to determine the activity ranking.
  • Compounds which were found to have both extremely high potency and extremely high efficacy of protein synthesis are classified as "*****”
  • Compounds which were found to have extremely high potency of protein synthesis and very high efficacy were classified as "****”.
  • Compounds which were found to have very high potency of protein synthesis and extremely high efficacy were classified as "**** »
  • Compounds which were found to have both very high potency and very high efficacy of protein synthesis are classified as "***”.
  • Compounds which were found to have very high potency of protein synthesis and significant efficacy were classified as "**”.
  • Compounds which were found to have significant potency of protein synthesis and very high efficacy were classified as "**”.
  • compounds which were found to have significant potency of protein synthesis and very high efficacy were classified as "*”.
  • the present invention encompasses the in vitro or in vivo use of a compound of the invention, and the inco ⁇ oration of a compoxmd of the invention into pharmaceutical compositions and single unit dosage forms useful in the treatment and prevention of a variety of diseases and disorders.
  • Specific diseases and disorders include those ameliorated by the suppression of a nonsense mutation in messenger RNA.
  • compositions including dosage forms of the invention which comprise a compound of the invention or a pharmaceutically acceptable polymo ⁇ h,tician _ ,_ ti ⁇ ⁇ prodrug, salt, clathrate, solvate or hydrate thereof, can be used in the methods of the invention.
  • a first embodiment of the invention relates to a method of modulating premature translation termination and/or nonsense-mediated mRNA decay comprising contacting a cell exhibiting a nonsense mutation with an effective amount of a compound of the invention, or a pharmaceutically acceptable prodrug, metabolite, polymo ⁇ h, salt, solvate, hydrate, or clathrate thereof.
  • the invention relates to a method of inducing nonsense suppression comprising contacting a cell exhibiting a nonsense mutation with an effective amount of a compound of the invention, or a pharmaceutically acceptable prodrug, metabolite, polymo ⁇ h, salt, solvate, hydrate, or clathrate thereof.
  • BIOLOGICAL ASSAYS AND ANIMAL STUDIES Compounds that modulate premature translation termination and or nonsense- mediated mRNA decay can be identified by a number of techniques. For example, methods for screening compounds that modulate the post-transcriptional expression of any gene with a premature translation stop codon are described in International Patent Publication No. WO 01/44516 A2, inco ⁇ orated herein in its entirety by reference.
  • a mRNA with a premature termination codon is franslated in vitro and is used to screen a library of test compounds.
  • the mRNA with a premature termination codon is a reporter gene with a premature termination codon.
  • the first assay is a cell-based luciferase reporter assay and the second is a biochemical assay consisting of rabbit reticulocyte lysate and a nonsense-containing luciferase reporter mRNA.
  • a luciferase reporter construct containing a UGA premature termination codon was stably fransfected in 293T Human Embryonic Kidney cells.
  • mRNA containing a UGA premature termination codon was used as a reporter in an in vitro translation reaction using rabbit reticulocyte lysate supplemented with tRNA, hemin, creatine kinase, amino acids, KOAc, Mg(OAc)2, and creatine phosphate.
  • Translation of the mRNA was initiated within a virus ___ , _ >t _ derived leader sequence, which significantly reduced the cost of the assay because capped RNA was not required.
  • Synthetic mRNA was prepared in vitro using the T7 promoter and the MegaScript in vitro transcription kit (Ambion). hi both of the biochemical and cell- based assays, addition of gentamicin, a small molecule known to allow readthrough of premature termination codons, resulted in increased luciferase activity and was, therefore, used as an internal standard.
  • Animal model systems can also be used to demonstrate the safety and efficacy of compounds of formula I, II, HI, IV or V.
  • the compounds of formula I, II, III, IV or V can be tested for biological activity using animal models for a disease, condition, or syndrome of interest. These include animals engineered to contain the target RNA element coupled to a functional readout system, such as a transgenic mouse.
  • Examples of animal models for cystic fibrosis include, but are not limited to, cftr(-/-) mice (see, e.g., Freedman et al, 2001, Gasfroenterology 121(4):950-7), cftr(tmlHGU/tmlHGU) mice (see, e.g., Bernhard et al, 2001, Exp Lung Res 27(4):349- 66), CFTR-deficient mice with defective cAMP-mediated Cl(-) conductance (see, e.g., Stotland et al, 2000, Pediafr Pulmonol 30(5):413-24), and C57BL/6- Cffr(mlUNC)/Cftr(mlUNC) knockout mice (see, e.g., Stotland et al, 2000, Pediafr Pulmonol 30(5):413-24).
  • Examples of animal models for muscular dystrophy include, but are not limited to, mouse, hamster, cat, dog, and C. elegans.
  • Examples of mouse models for muscular dystrophy include, but are not limited to, the dy-/- mouse (see, e.g., Connolly et al, 2002, J Neuroimmunol 127(l-2):80-7), a muscular dystrophy with myositis (mdm) mouse mutation (see, e.g., Garvey et al, 2002, Genomics 79(2): 146-9), the mdx mouse (see, e.g., Nakamura et al, 2001, Neuromuscul Disord 11(3):251-9), the utrophin-dystrophin knockout (dko) mouse (see, e.g., Nakamura et al, 2001, Nexxromuscul Disord 11(3):251-9), the dy/dy mouse (see, e.
  • Examples of hamster models for muscular dystrophy include, but are not limited to, sarcoglycan-deficient hamsters (see, e.g., Nakamura et al, 2001, Am J Physiol Cell Physiol 281(2):C690-9) and the BIO 14.6 dystrophic hamster (see, e.g., Schlenker & Burbach, 1991, J Appl Physiol 71(5):1655-62).
  • An example of a feline model for muscular dystrophy includes, but is not limited to, the hypertrophic feline muscular dystrophy model (see, e.g., Gaschen & Burgunder, 2001, Acta Neuropathol (Berl) 101(6):591-600).
  • Canine models for muscular dystrophy include, but are not limited to, golden retriever muscular dystrophy (see, e.g., Fletcher et al, 2001, Neuromuscul Disord ll(3):239-43) and canine X-linked muscular dystrophy (see, e.g., Valentine et al, 1992, Am J Med Genet 42(3):352-6).
  • golden retriever muscular dystrophy see, e.g., Fletcher et al, 2001, Neuromuscul Disord ll(3):239-43
  • canine X-linked muscular dystrophy see, e.g., Valentine et al, 1992, Am J Med Genet 42(3):352-6).
  • Examples of C. elegans models for muscular dystrophy are described in Chamberlain & Benian, 2000, Cun Biol 10(21):R795-7 and Culette & Sattelle, 2000, Hum Mol Genet 9(6): 869-77.
  • mice lacking functional LDL receptor genes see, e.g., Aji et al, 1997, Circulation 95(2):430-7
  • Yoshida rats see, e.g., Fantappie et al, 1992, Life Sci 50(24):1913-24
  • the JCR:LA-cp rat see, e.g., Richardson et al, 1998, Atherosclerosis 138(l):135-46
  • swine see, e.g., Hasler-Rapacz et al, 1998, Am J Med Genet 76(5):379- 86
  • Watanabe heritable hyperlipidaemic rabbit see, e.g., Tsutsumi et al, 2000, Arzneiffenforschung 50(2):118-21; Harsch et al, 1998, Br J Pharmacol 124(2):227-82; and Tanaka et al,
  • an animal model for human cancer in general includes, but is not limited to, spontaneously occurring tumors of companion animals (see, e.g., Nail & MacEwen, 2000, Cancer Invest 18(8):781-92).
  • animal models for lung cancer include, but are not limited to, lung cancer animal models described by Zhang & Roth (1994, In Vivo 8(5):755-69) and a transgenic mouse model with disrupted p53 function (see, e.g., Morris et al, 1998, J La State Med Soc 150(4): 179-85).
  • An example of an animal model for breast cancer includes, but is not limited to, a transgenic mouse that overexpresses cyclin DI (see, e.g., Hosokawa et al, 2001, Transgenic Res 10(5):471-8).
  • An example of an animal model for colon cancer includes, but is not limited to, a TCRbeta and p53 double knockout mouse (see, e.g., Kado et al, 2001, Cancer Res 61(6):2395-8).
  • animal models for pancreatic cancer include, but are not limited to, a metastatic model of Panc02 murine pancreatic adenocarcinoma (see, e.g., Wang et al, 2001, Int J Pancreatol 29(l):37-46) and nu-nu mice generated in subcutaneous pancreatic tumours (see, e.g., Ghaneh et al, 2001, Gene Ther 8(3): 199-208).
  • animal models for non-Hodgkin's lymphoma include, but are not limited to, a severe combined immunodeficiency ("SCID") mouse (see, e.g.
  • SCID severe combined immunodeficiency
  • an animal model for esophageal cancer includes, but is not limited to, a mouse transgenic for the human papillomavirus type 16 E7 oncogene (see, e.g., Herber et al, 1996, J Nirol 70(3): 1873-81).
  • animal models for colorectal carcinomas include, but are not limited to, Ape mouse models (see, e.g., Fodde & Smits, 2001, Trends Mol Med 7(8):369-73 and Kuraguchi et al, 2000, Oncogene 19(50):5755-63).
  • An example of an animal model for neurofibromatosis includes, but is not limited to, mutant NF1 mice (see, e.g., Cichowski et al, 1996, Semin Cancer Biol 7(5):291- 8).
  • animal models for retinoblastoma include, but are not limited to, transgenic mice that expression the simian virus 40 T antigen in the retina (see, e.g., Howes et al, 1994, Invest Ophthahnol Vis Sci 35(2):342-51 and Windle et al, 1990, Nature 343(6259):665-9) and inbred rats (see, e.g., Nishida et al, 1981, Curr Eye Res l(l):53-5 and Kobayasbi et al, 1982, Acta Neuropathol (Berl) 57(2-3) :203-8).
  • Examples of animal models for Wilm's tumor include, but are not limited to, a WTl knockout mice (see, e.g., Scharnhorst et al, 1997, Cell Growth Differ 8(2):133-43), a rat subline with a high incidence of neuphroblastoma (see, e.g., Mesfin & Breech, 1996, Lab Anim Sci 46(3):321- 6), and a Wistar/Furth rat with Wilms' tumor (see, e.g., Mu ⁇ hy et al, 1987, Anticancer Res 7(4B):717-9).
  • Examples of animal models for retinitis pigmentosa include, but are not limited to, the Royal College of Surgeons ("RCS") rat (see, e.g., Nollrath et al, 2001, Proc ⁇ atl Acad Sci USA 98(22);12584-9 and Hanitzsch et al, 1998, Acta Anat (Basel) 162(2-3): 119-26), a rhodopsin knockout mouse (see, e.g., Jaissle et al, 2001, Invest Ophthahnol Vis Sci 42(2):506-13), and Wag/Rij rats (see, e.g., Lai et al, 1980, Am J Pathol 98(l):281-4).
  • RCS Royal College of Surgeons
  • animal models for cirrhosis include, but are not limited to, CC1 4 - exposed rats (see, e.g., Kloehn et al, 2001, Horm Mexab Res 33(7):394-401) and rodent models instigated by bacterial cell components or colitis (see, e.g., Nierling, 2001, Best Pract Res Clin Gastroenterol 15(4):591-610).
  • animal models for hemophilia include, but are not limited to, rodent models for hemophilia A (see, e.g. , Reipert et al, 2000, Thromb Haemost 84(5):826-32; Jarvis et al,.
  • Examples of animal models for von Willebrand disease include, but are not limited to, an inbred mouse strain RHIS/J (see, e.g., Nichols et al, 1994, 83(11):3225-31 and Sweeney et al, 1990, 76(11):2258-65), rats injected with botrocetin (see, e.g., Sanders et al, 1988, Lab Invest 59(4):443-52), and porcine models for von Willebrand disease (see, e.g., Nichols et al, 1995, Proc Natl Acad Sci USA 92(7):2455-9; Johnson & Bowie, 1992, J Lab Clin Med 120(4):553-8); and Brinkhous et al, 1991, Mayo Clin Proc 66(7):733-42).
  • animal models for b-thalassemia include, but are not limited to, murine models with mutations in globin genes (see, e.g., Lewis et al, 1998, Blood 91(6):2152-6; Raja et al, 1994, Br J Haematol 86(1): 156-62; Popp et al, 1985, 445:432-44; and Skow et al, 1983, Cell 34(3): 1043-52).
  • kidney stones examples include, but are not limited to, genetic hypercalciuric rats (see, e.g., Bushinsky et al, 1999, Kidney hit 55(l):234-43 and Bushinsky et al, 1995, Kidney hit 48(6): 1705-13), chemically treated rats (see, e.g., Grases et al, 1998, Scand J Urol Nephrol 32(4):261-5; Burgess et al, 1995, Urol Res 23(4):239- 42; Kumar et al, 1991, J Urol 146(5):1384-9; Okada et al, 1985, Hinyokika Kiyo
  • animal models for ataxia-telangiectasia include, but are not limited to, murine models of ataxia-telangiectasia (see, e.g., Barlow et al, 1999, Proc Natl Acad Sci USA 96(17):9915-9 and Inoue et al, 1986, Cancer Res 46(8):3979-82).
  • animal models for lysosomal storage diseases include, but are not limited to, mouse models for mucopolysaccharidosis type Nil (see, e.g., Brooks et al, 2002, Proc ⁇ atl Acad Sci U S A. 99(9):6216-21 ; Monroy et al, 2002, Bone 30(2):352-9; Nogler et al, 2001, Pediafr Dev Pathol. 4(5):421-33; Nogler et al, 2001, Pediafr Res. 49(3):342-8; and Wolfe et al., 2000, Mol Ther.
  • mice model for metachromatic leukodystrophy see, e.g., Matzner et al, 2002, Gene Ther. 9(l):53-63
  • a mouse model of Sandhoff disease see, e.g., Sango et al, 2002, ⁇ europathol Appl ⁇ eurobiol.
  • mice for mucopolysaccharidosis type III A (see, e.g., Bhattacharyya et al., 2001, Glycobiology 11(1):99-10 and Bhaumik et al., 1999, Glycobiology 9(12): 1389-96.), arylsulfatase A (ASA)-deficient mice (see, e.g., D'Hooge et al, 1999, Brain Res. 847(2):352-6 and D'Hooge et al, 1999, ⁇ eurosci Lett.
  • ASA arylsulfatase A
  • mice with an aspartylglucosaminuria mutation see, e.g., Jalanko et al, 1998, Hum Mol Genet. 7(2):265- 72
  • feline models of mucopolysaccharidosis type VI see, e.g-., Crawley et al, 1998, J Clin Invest. 101(1):109-19 and Norrdin et al., 1995, Bone 17(5):485-9
  • a feline model of Niemann-Pick disease type C see, e.g., March et al, 1997, Acta Neuropathol (Berl ).
  • TSC tuberous sclerosis
  • TSC1 mouse model of TSC 1
  • Tscl TSC1 homologue
  • TSC2 gene mutant(Eker) rat model see, e.g., Hino 2000, Nippon Rinsho 58(6):1255-61; Mizuguchi et al, 2000, J Neuropathol Exp Neurol.
  • the compounds of the invention can be obtained via standard, well-known synthetic methodology, see e.g. March, J. Advanced Organic Chemistry; Reactions Mechanisms, and Structure, 4th ed., 1992. Some convenient methods are illustrated in Schemes 1 and 2. Starting materials useful for preparing the compounds of the invention and intermediates therefor, are commercially available or can be prepared from commercially available materials using known synthetic methods and reagents.
  • a compound of structure Al is coupled to an alcohol (or thiol) reagent A2.
  • the group L represents a leaving group of some sort, and can include (but is not limited to) halide, alkylsulfonate, arylsulfonate, diazonium, etc.
  • Typical alkylation conditions for the coulpling of Al and A2 might include the presence of a basic reagent as a catalyst.
  • Basic reagents that are useful here include trialkylamines, sodium or potassium carbonate, sodium hydride, etc.
  • L can also include hydroxy, which can be activated in-situ using conditions such as those employed in the Mitsonobu reaction (typically, use of a trisubstituted phosphine reagent in conjunction with an azodicarboxylate compound).
  • An alternative method involves the coupling of aniline compoxmd 3 with the reagent 4.
  • L' may also be OH, and the reaction is then a peptide-type coupling which is familiar to those skilled in the art.
  • Such reactions are mediated by in-situ activation with reagents such as dialkylcarbodiimide, carbonyldiimidazole or isobutylchloroformate.
  • Modifications of the carboxyl group may be performed at any time in the synthesis which proves compatible with the substrate and chemistry.
  • R 6 is not H
  • the ester can be cleaved to the free carbox ylate using a number of standard methods. This includes (but is not limited to) sodium or potassium hydroxide, lithium iodide, boron tribromide, fluoride reagents (for certain silicon-bearing R 6 groups) and catalytic hydrogenation (for certain benzyl-type R 6 groups).
  • the compound may also be attached through the carboxylate group to a solid-phase resin, which facilitates the synthesis by allowing easy removal of impurities and byproducts from each reaction by simple filtration and washing with an appropriate solvent. Cleavage of the product from the resin may then be accomplished in various ways to afford the final
  • a compound of formula CI is prepared using methods described above.
  • the carbonyl bond may then be thiolated using an appropriate thiolating reagent, such as Lawesson's reagent or phosphorus pentasulfide, typically in inert solvents at elevated temperature.
  • an appropriate thiolating reagent such as Lawesson's reagent or phosphorus pentasulfide
  • the amide nifrogen atom of the unsubstituted compound C2 may be alkylated using an alkylating agent C3 (wherein L" is a leaving group such as halide or alkyl or arylsulfonate) basic reagent such as sodium hydride or potassium hexamethyldisilazane in polar aprotic solvents at temperatures ranging from sub-ambient to heated.
  • alkylating agent C3 wherein L" is a leaving group such as halide or alkyl or arylsulfonate
  • basic reagent such as sodium hydride or potassium hexamethyldisilazane in polar aprotic solvents at temperatures ranging from sub-ambient to heated.
  • Nitration conditions typically include concentrated nitric acid or sodium nitrite optionally in the presence of a , _ u . .,,,,,, , , deliberately, ] ⁇ sulfuric acid.
  • the nitro group of compound D2 may be reduced to an aniline, using conditions such as catalytic hydrogenation or the use of reducing agents such as tin (II) dichloride, iron powder or sodium hydrosulfite.
  • the aniline compound may then be functionahzed as desired (if Rl is not H) to afford compound A3.
  • Alkylation reactions may be used, employing appropriate alkylating reagents such as alkyl halides, etc. and bases.
  • Anilines may also be functionahzed by reductive alkylations, whereby an intermediate Schiff base compound (formed by condensation of the aniline with an aldehyde or ketone reagent) is reduced in situ (typically by catalytic hydrogenation or reagents such as sodium cyanoborohydride).
  • An alternative route involves the reaction of the lithium reagent D3 with an carboxylating reagent to give the compound A3.
  • the nitrogen atom in compound D3 must usually be protected with group P, which can include pivaloyl or tert- butoxycarbonyl.
  • group P can include pivaloyl or tert- butoxycarbonyl.
  • the 3-lithio reagent may be generated by direct deprotonation for some substrates using strong bases such as sec- or tert-butyllithium at temperatures typically ranging slightly below to at ambient.
  • the lithium reagent may also be generated by lithium- halogen exchange from the bromo or iodo analogue and an alkyllithium reagent, usually at reduced temperatures.
  • Carboxylating reagents that are useful here include solid carbon dioxide or an alkyl chloroformate. After the carboxylation, the protecting group P may be removed by methods specific to the group employed, well-known in the area of protecting group chemistry.
  • the invention encompasses methods of treating and preventing diseases or disorders ameliorated by the suppression of premature translation termination and/or nonsense- mediated mRNA decay in a patient which comprise administering to a patient in need of such treatment or prevention a therapeutically effective amount of a compoxmd of the invention, or a pharmaceutically acceptable prodrug, solvate, metabolite, polymo ⁇ h, salt, solvate, hydrate, or clathrate thereof.
  • the present invention encompasses the treatment or prevention of any disease which is associated with a gene exhibiting premature translation termination and/or nonsense-mediated mRNA decay.
  • the disease is due, in part, to the lack of expression of the gene resulting from a premature stop codon.
  • Specific examples of genes which may exhibit premature translation termination and/or nonsense-mediated mRNA decay and diseases associated with premature translation termination and/or nonsense-mediated mRNA decay are found in U.S. Patent Application No. 60/390,747, titled: Methods For Identifying Small Molecules That Modulate Premature Translation Termination And Nonsense Mediated mRNA Decay, filed June 21, 2002, which is inco ⁇ orated herein by reference in its entirety.
  • Diseases ameliorated by the suppression of premature translation termination and or nonsense-mediated mRNA decay include, but are not limited to: a genetic disease, cancer, an autoimmune disease, a blood disease, a collagen disease, diabetes, a neurodegenerative disease, a proliferative disease, a cardiovascular disease, a pulmonary disease, an inflammatory disease or central nervous system disease.
  • Specific genetic diseases within the scope of the methods of the invention include, but are not limited to, amyloidosis, hemophilia, Alzheimer's disease, Tay Sachs disease, atherosclerosis, giantism, dwarfism, hypothyroidism, hyperthyroidism, aging, obesity, Parkinson's disease, Niemann Pick's disease, cystic fibrosis, muscular dystrophy, heart disease, kidney stones, ataxia-telangiectasia, familial hypercholesterolemia, retinitis pigmentosa, lysosomal storage disease, tuberous sclerosis, Duchenne muscular dystrophy, and Marfan syndrome. Both solid tumor and other cancers are included within the methods of the invention.
  • the genetic disease is an autoimmune disease.
  • the autoimmune disease is rheumatoid arthritis or graft versus host disease.
  • the genetic disease is a blood disease.
  • the blood disease is hemophilia, Von Willebrand disease, ataxia- telangiectasia, b-thalassemia or kidney stones.
  • the genetic disease is a collagen disease.
  • the collagen disease is osteogenesis imperfecta or cirrhosis.
  • the genetic disease is diabetes. ⁇ ⁇ personally administrating .
  • the genetic disease is an inflammatory disease. In a preferred embodiment, the inflammatory disease is arthritis.
  • the genetic disease is a central nervous system disease.
  • the central nervous system disease is a neurodegenerative disease.
  • the central nervous system disease is multiple sclerosis, muscular dystrophy, Duchenne muscular dystrophy, Alzheimer's disease, Tay Sachs disease, late infantile neuronal ceroid lipofuscinosis (LINCL) or Parkinson's disease.
  • the genetic disease is cancer.
  • the cancer is of the head and neck, eye, skin, mouth, throat, esophagus, chest, bone, lung, colon, sigmoid, rectum, stomach, prostate, breast, ovaries, kidney, liver, pancreas, brain, intestine, heart or adrenals.
  • the cancer is associated with the p53 gene.
  • Nonsense mutations have been identified in the p53 gene and have been implicated in cancer.
  • Several nonsense mutations in the p53 gene have been identified (see, e.g., Masuda et al, 2000, Tokai J Exp Clin Med. 25(2):69-77; Oh et al, 2000, Mol Cells 10(3):275-80; Li et al, 2000, Lab Invest. 80(4):493-9; Yang et al, 1999, Zhonghua Zhong Liu Za Zhi 21(2):114-8; Finkelstein et al, 1998, Mol Diagn. 3(1):37-41; Kajiyama et al, 1998, Dis Esophagus.
  • diseases to be treated or prevented by administering to a patient in need thereof an effective amount of a compound of formula I include solid tumor, sarcoma, carcinomas, fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal
  • the invention encompasses a method of treating or preventing a disease ameliorated by modulation of premature translation termination and or nonsense-mediated mRNA decay, or ameliorating one or more symptoms associated therewith comprising contacting a cell with an effective amount of a compound of formula I, II, III, IV or V.
  • Cells encompassed by the present methods include animal cells, mammalian cells, bacterial cells, plant cells and virally infected cells.
  • the nonsense codon was present in the progenitor DNA.
  • the nonsense codon resulted from mutagenesis.
  • a compound of formula I, II, III, IV or V, or a pharmaceutically acceptable salt thereof is administered to a patient, preferably a mammal, more preferably a human, as a preventative measure against a disease associated with premature translation termination and or nonsense-mediated mRNA decay.
  • the patient is suffering from a disease associate with premature translation termination and/or nonsense-mediated mRNA decay.
  • the patient has undergone a screening process to determine the presence of a nonsense mutation comprising the steps of screening a subject, or cells extracted therefrom, by an acceptable nonsense mutation screening assay.
  • the DNA of the patient can be sequenced or subject to Southern Blot, polymerase chain reaction (PCR), use of the Short Tandem Repeat (STR), or polymo ⁇ hic length restriction fragments (RFLP) analysis to determine if a nonsense mutation is present in the DNA of the patient.
  • the patient is an unborn child who has undergone screening in utero for the presence of a nonsense mutation.
  • Administration of a compound of formula I, II, III, IV or V can occur either before or after birth.
  • the therapy is personalized in that the patient is screened for a nonsense mutation screening assay and treated by the administration of one or more compounds of the invention; particularly, the patient may be treated with a compoxmd particularly suited for the mutations in question; e.g., depending upon the disease type, cell type, and the gene in question.
  • a compoxmd particularly suited for the mutations in question; e.g., depending upon the disease type, cell type, and the gene in question.
  • the cells e.g., animal cells, mammalian cells, bacterial cells, plant cells and virally infected cells
  • a method such as that described above (i.e., the DNA of the cell can be sequenced or subjected to Southern Blot, polymerase chain reaction (PCR), use of the Short Tandem Repeat (STR), or polymo ⁇ hic length restriction fragments (RFLP) analysis to determine if a nonsense mutation is present in the DNA of the cell).
  • PCR polymerase chain reaction
  • STR Short Tandem Repeat
  • RFLP polymo ⁇ hic length restriction fragments
  • Specific methods of the invention further comprise the administration of an additional therapeutic agent (i.e. a therapeutic agent other than a compound of the invention).
  • an additional therapeutic agent i.e. a therapeutic agent other than a compound of the invention.
  • the compounds of the invention can be used in combination with at least one other therapeutic agent.
  • Therapeutic agents include, but are not limited to non-opioid analgesics; non-steroid anti-inflammatory agents; antiemetics; ⁇ -adrenergic blockers; anticonvulsants; antidepressants; Ca 2+ -channel blockers; anticancer agent and mixtures thereof.
  • the compounds of formula I, II, III, IV or V can be administered or formulated in combination with anticancer agents.
  • Suitable anticancer agents include, but are not limited to, alkylating agents; nitrogen mustards; folate antagonists; purine antagonists; pyrimidine antagonists; spindle poisons; topoisomerase inhibitors; apoptosis inducing agents; angiogenesis inhibitors; podophyllotoxins; nitrosoureas; cisplatin; carboplatin; interferon; asparginase; tamoxifen; leuprolide; flutamide; megestrol; mitomycin; bleomycin; doxorubicin; irinotecan and taxol.
  • the compounds of formula I, II, III, IV or V can be administered or formulated in combination with antibiotics
  • the antibiotic is a macrolide (e.g., tobramycin (Tobi®)), a cephalosporin (e.g., cephalexin (Keflex®), cephradine (Nelosef®), cefixroxime (Ceftin®), cefprozil (Cefzil®), cefaclor (Ceclor®), cefixime (Suprax®) or cefadroxil (Duricef®)), a clarithromycin (e.g., clarithromycin (Biaxin®)), an erythromycin (e.g., erythromycin (EMycin®)), a penicillin (e.g., penicillin N (N-Cillin K® or Pen Nee K®)) or a quinolone (e.g., ofloxacin (Floxin®), ciprof
  • the antibiotic is active against Pseudomonas aeruginosa.
  • the compounds of the invention and the other therapeutics agent can act additively or, more preferably, synergistically.
  • a composition comprising a compound of the invention is administered concurrently with the adminisfration of another therapeutic agent, which can be part of the same composition or in a different composition from that comprising the compounds of the invention.
  • a compound of the invention is administered prior to or subsequent to administration of another therapeutic agent.
  • a prophylactic or therapeutic dose of a particular active ingredient of the invention in the acute or chronic management of a disease or condition will vary, however, with the nature and severity of the disease or condition, and the route by which the active ingredient is administered.
  • the dose, and perhaps the dose frequency will also vary according to the age, body weight, and response of the individual patient. Suitable dosing regimens can be readily selected by those skilled in the art with due consideration of such factors.
  • the recommended daily dose range for the conditions described herein lie within the range of from about 0.1 mg to about 2000 mg per day, given as a single once- a-day dose, preferably as divided doses throughout a day.
  • the daily dose is administered in a single dose or in equally divided doses.
  • a daily dose range should be from about 5 mg to about 500 mg per day, more specifically, between about 10 mg and about 200 mg per day.
  • the therapy should be initiated at a lower dose, perhaps about 1 mg to about 25 mg, and increased if necessary up to about ⁇ _ _ ⁇ ⁇
  • therapeutically effective amount encompasses the above described dosage amounts and dose frequency schedules. Different therapeutically effective amounts may be applicable for different diseases and conditions, as will be readily known by those of ordinary skill in the art. Similarly, amounts sufficient to treat or prevent such diseases, but insufficient to cause, or sufficient to reduce, adverse effects associated with conventional therapies are also encompassed by the above described dosage amounts and dose frequency schedules. 4.5 PHARMACEUTICAL COMPOSITIONS
  • compositions and single unit dosage forms comprising a compound of the invention, or a pharmaceutically acceptable polymo ⁇ h, prodrug, salt, solvate, hydrate, or clathrate thereof, are also encompassed by the invention.
  • Individual dosage forms of the invention may be suitable for oral, mucosal (including sublingual, buccal, rectal, nasal, or vaginal), parenteral (including subcutaneous, intramuscular, bolus injection, mfraarterial, or intravenous), transdermal, or topical administration.
  • compositions and dosage forms of the invention comprise a compoxmd of the invention, or a pharmaceutically acceptable prodrug, polymo ⁇ h, salt, solvate, hydrate, or clathrate thereof.
  • Pharmaceutical compositions and dosage forms of the invention typically also comprise one or more pharmaceutically acceptable excipients.
  • a particular pharmaceutical composition encompassed by this embodiment comprises a compound of the invention, or a pharmaceutically acceptable polymo ⁇ h, prodrug, salt, solvate, hydrate, or clathrate thereof, and at least one additional therapeutic agent.
  • additional therapeutic agents include, but are not limited to: anti-cancer drugs and anti-inflammation therapies including, but not limited to, those listed above in Section 4.3.
  • Single unit dosage forms of the invention are suitable for oral, mucosal (e.g., nasal, sublingual, vaginal, buccal, or rectal), parenteral (e.g., subcutaneous, intravenous, bolus injection, intramuscular, or intraarterial), or transdermal adminisfration to a patient. ,...,, undertaken. ,, , context consider herein. ..,, ... modix.
  • parenteral e.g., subcutaneous, intravenous, bolus injection, intramuscular, or intraarterial
  • transdermal adminisfration e.g., transdermal adminisfration to a patient.
  • mucosal e.g., nasal, sublingual, vaginal, buccal, or rectal
  • parenteral e.g., subcutaneous, intravenous, bolus injection, intramuscular, or intraarterial
  • transdermal adminisfration e.g., transdermal adminisfration
  • dosage forms include, but are not limited to: tablets; caplets; capsules, such as soft elastic gelatin capsules; cachets; troches; lozenges; dispersions; suppositories; ointments; cataplasms (poultices); pastes; powders; dressings; creams; plasters; solutions; patches; aerosols (e.g., nasal sprays or inhalers); gels; liquid dosage forms suitable for oral or mucosal adminisfration to a patient, including suspensions (e.g., aqueous or non-aqueous liquid suspensions, oil-in- water emulsions, or a water-in-oil liquid emulsions), solutions, and elixirs; liquid dosage forms suitable for parenteral administration to a patient; and sterile solids (e.g., crystalline or amo ⁇ hous solids) that can be reconstituted to provide liquid dosage forms suitable for parenteral administration to a patient.
  • suspensions e.g.,
  • composition, shape, and type of dosage forms of the invention will typically vary depending on their use.
  • a dosage form used in the acute treatment of inflammation or a related disease may contain larger amounts of one or more of the active ingredients it comprises than a dosage form used in the chronic treatment of the same disease.
  • a parenteral dosage form may contain smaller amounts of one or more of the active ingredients it comprises than an oral dosage form used to treat the same disease or disorder.
  • Typical pharmaceutical compositions and dosage forms comprise one or more carriers, excipients or diluents.
  • Suitable excipients are well known to those skilled in the art of pharmacy, and non-limiting examples of suitable excipients are provided herein. Whether a particular excipient is suitable for inco ⁇ oration into a pharmaceutical composition or dosage form depends on a variety of factors well known in the art including, but not limited to, the way in which the dosage form will be administered to a patient.
  • oral dosage forms such as tablets may contain excipients not suited for use in parenteral dosage forms. The suitability of a particular excipient may also depend on the specific active ingredients in the dosage form.
  • This invention further encompasses anhydrous pharmaceutical compositions and dosage forms comprising active ingredients, since water can facilitate the degradation of some compounds.
  • water e.g., 5%
  • water is widely accepted in the pharmaceutical arts as a means of simulating long-term storage in order to determine characteristics such as shelf-life or the stability of formulations over time. See, e.g., Jens T. Carstensen, Drug Stability: Principles & Practice, 2d. Ed., Marcel Dekker, NY, NY, 1995, pp. 379-80.
  • water and heat accelerate the decomposition of some compounds.
  • the effect of water on a formulation can be of great significance since moisture and/or humidity are commonly encountered during manufacture, handling, packaging, storage, shipment, and use of formulations.
  • Anhydrous pharmaceutical compositions and dosage forms of the invention can be prepared using anhydrous or low moisture containing ingredients and low moisture or low humidity conditions.
  • Pharmaceutical compositions and dosage forms that comprise lactose and at least one active ingredient that comprises a primary or secondary amine are preferably anhydrous if substantial contact with moisture and/or humidity during manufacturing, packaging, and/or storage is expected.
  • An anhydrous pharmaceutical composition should be prepared and stored such that its anhydrous nature is maintained.
  • anhydrous compositions are preferably packaged using materials known to prevent exposure to water such that they can be included in suitable formulary kits. Examples of suitable packaging include, but are not limited to, hermetically sealed foils, plastics, unit dose containers (e.g., vials), blister packs, and strip packs.
  • compositions and dosage forms that comprise one or more compounds that reduce the rate by which an active ingredient will decompose.
  • compounds which are referred to herein as “stabilizers,” include, but are not limited to, antioxidants such as ascorbic acid, pH buffers, or salt buffers.
  • antioxidants such as ascorbic acid, pH buffers, or salt buffers.
  • the amounts and specific types of active ingredients in a dosage form may differ depending on factors such as, but not limited to, the route by which it is to be administered to patients.
  • typical dosage forms of the invention comprise a compound of the invention, or a pharmaceutically acceptable salt, solvate, clathrate, hydrate, polymoprh or prodrug thereof lie within the range of from about 0.1 mg to about 2000 mg per day, given as a single once-a-day dose in the morning but preferably as divided doses throughout the day taken with food. More specifically, the daily dose is administered twice daily in equally divided doses. Specifically, a daily dose range should be from about 5 mg to about 500 mg per day, more specifically, between about 10 mg and about 200 mg per day.
  • the therapy should be initiated at a lower dose, perhaps about 1 mg to about 25 mg, and increased if necessary up to about 200 mg to about 2000 mg per day as either a single dose or divided doses, depending on the patient's global response.
  • compositions of the invention that are suitable for oral admimstration can be presented as discrete dosage forms, such as, but are not limited to, tablets (e.g., chewable tablets), caplets, capsules, and liquids (e.g., flavored syrups).
  • dosage forms contain predetermined amounts of active ingredients, and may be prepared by methods of pharmacy well known to those skilled in the art. See generally, Remington 's Pharmaceutical Sciences, 18th ed., Mack Publishing, Easton PA (1990).
  • Typical oral dosage forms of the invention are prepared by combining the active ingredient(s) in an intimate admixture with at least one excipient according to conventional pharmaceutical compounding techniques.
  • Excipients can take a wide variety of forms depending on the form of preparation desired for admimstration.
  • excipients suitable for use in oral liquid or aerosol dosage forms include, but are not limited to, water, glycols, oils, alcohols, flavoring agents, preservatives, and coloring agents.
  • excipients suitable for use in solid oral dosage forms include, but are not limited to, starches, sugars, micro-crystalline cellulose, diluents, granulating agents, lubricants, binders, and disintegrating agents.
  • tablets and capsules represent the most advantageous oral dosage unit forms, in which case solid excipients are employed. If desired, tablets can be coated by standard aqueous or nonaqueous techniques. Such dosage forms can be prepared by any of the methods of pharmacy. In general, pharmaceutical compositions and dosage forms are prepared by uniformly and intimately admixing the active ingredients with liquid carriers, finely divided solid carriers, or both, and then shaping the product into the desired presentation if necessary.
  • a tablet can be prepared by compression or molding.
  • Compressed tablets can be prepared by compressing in a suitable machine the active ingredients in a free-flowing form such as powder or granules, optionally mixed with an excipient.
  • Molded tablets can be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • excipients that can be used in oral dosage forms of the invention include, but are not limited to, binders, fillers, disintegrants, and lubricants.
  • Binders suitable for use in pharmaceutical compositions and dosage forms include, but are not limited to, corn starch, potato starch, or other starches, gelatin, natural and synthetic gums such as acacia, sodium alginate, alginic acid, other alginates, powdered tragacanth, guar gum, cellulose and its derivatives (e.g., ethyl cellulose, cellulose acetate, carboxymethyl u _ _ .. dots .
  • fillers suitable for use in the pharmaceutical compositions and dosage forms disclosed herein include, but are not limited to, talc, calcium carbonate (e.g., granules or powder), microcrystalline cellulose, powdered cellulose, dextrates, kaolin, mannitol, silicic acid, sorbitol, starch, pre-gelatinized starch, and mixtures thereof.
  • the binder or filler in pharmaceutical compositions of the invention is typically present in from about 50 to about 99 weight percent of the pharmaceutical composition or dosage form.
  • Suitable forms of microcrystalline cellulose include, but are not limited to, the materials sold as AVICEL-PH-101, AVICEL-PH-103 AVICEL RC-581, AVICEL-PH-105 (available from FMC Co ⁇ oration, American Viscose Division, Avicel Sales, Marcus Hook, PA), and mixtures thereof.
  • An specific binder is a mixture of microcrystalline cellulose and sodium carboxymethyl cellulose sold as AVICEL RC-581.
  • Suitable anhydrous or low moisture excipients or additives include AVICEL-PH-103TM and Starch 1500 LM.
  • Disintegrants are used in the compositions of the invention to provide tablets that disintegrate when exposed to an aqueous environment. Tablets that contain too much disintegrant may disintegrate in storage, while those that contain too little may not disintegrate at a desired rate or under the desired conditions. Thus, a sufficient amount of disintegrant that is neither too much nor too little to detrimentally alter the release of the active ingredients should be used to form solid oral dosage forms of the invention.
  • the amount of disintegrant used varies based upon the type of formulation, and is readily discernible to those of ordinary skill in the art.
  • Typical pharmaceutical compositions comprise from about 0.5 to about 15 weight percent of disintegrant, specifically from about 1 to about 5 weight percent of disintegrant.
  • Disintegrants that can be used in pharmaceutical compositions and dosage forms of the invention include, but are not limited to, agar-agar, alginic acid, calcium carbonate, microcrystalline cellulose, croscarmellose sodium, crospovidone, polacrilin potassium, sodium starch glycolate, potato or tapioca starch, pre-gelatinized starch, other starches, clays, other algins, other celluloses, gums, and mixtures thereof.
  • Lubricants that can be used in pharmaceutical compositions and dosage forms of the invention include, but are not limited to, calcium stearate, magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc, hydrogenated vegetable oil (e.g., peanut oil, cottonseed oil, sunflower oil, sesame oil, olive oil, corn oil, and soybean oil), zinc stearate, ethyl oleate, ethyl laureate, agar, and mixtures thereof.
  • calcium stearate e.g., magnesium stearate, mineral oil, light mineral oil, glycerin, sorbitol, mannitol, polyethylene glycol, other glycols, stearic acid, sodium lauryl sulfate, talc
  • hydrogenated vegetable oil e.g., peanut oil, cottonseed oil
  • Additional lubricants include, for example, a syloid silica gel (AEROSIL 200, manufactured by W.R. Grace Co. of Baltimore, MD), a coagulated aerosol of synthetic silica (marketed by Degussa Co. of Piano, TX), CAB-O-SIL (a pyrogenic silicon dioxide product sold by Cabot Co. of Boston, MA), and mixtures thereof. If used at all, lubricants are typically used in an amount of less than about 1 weight percent of the pharmaceutical compositions or dosage forms into which they are inco ⁇ orated.
  • AEROSIL 200 a syloid silica gel
  • a coagulated aerosol of synthetic silica marketed by Degussa Co. of Piano, TX
  • CAB-O-SIL a pyrogenic silicon dioxide product sold by Cabot Co. of Boston, MA
  • lubricants are typically used in an amount of less than about 1 weight percent of the pharmaceutical compositions or dosage forms into which they are inco ⁇ orated.
  • Active ingredients of the invention can be administered by controlled release means or by delivery devices that are well known to those of ordinary skill in the art. Examples include, but are not limited to, those described in U.S. Patent Nos.: 3,845,770; 3,916,899; 3,536,809; 3,598,123; and 4,008,719, 5,674,533, 5,059,595, 5,591,767, 5,120,548, 5,073,543, 5,639,476, 5,354,556, and 5,733,566, each of which is inco ⁇ orated herein by reference.
  • Such dosage forms can be used to provide slow or controlled-release of one or more active ingredients using, for example, hydropropylmethyl cellulose, other polymer matrices, gels, permeable membranes, osmotic systems, multilayer coatings, microparticles, liposomes, microspheres, or a combination thereof to provide the desired release profile in varying proportions.
  • Suitable controlled-release formulations known to those of ordinary skill in the art, including those described herein, can be readily selected for use with the active ingredients of the invention.
  • the invention thus encompasses single unit dosage forms suitable for oral administration such as, but not limited to, tablets, capsules, gelcaps, and caplets that are adapted for controlled-release.
  • controlled-release pharmaceutical products have a common goal of improving drug therapy over that achieved by their non-controlled counte ⁇ arts.
  • the use of an optimally designed controlled-release preparation in medical treatment is characterized by a minimum of drug substance being employed to cure or control the condition in a minimum amount of time.
  • Advantages of confrolled-release formulations include extended activity of the drug, reduced dosage frequency, and increased patient compliance.
  • controlled-release formulations can be used to affect the time of onset of action or other characteristics, such as blood levels of the drug, and can thus affect the occurrence of side (e.g., adverse) effects.
  • Controlled-release formulations are designed to initially release an amount of drug (active ingredient) that promptly produces the desired therapeutic effect, and gradually . _ and continually release of other amounts of drug to maintain this level of therapeutic or prophylactic effect over an extended period of time. In order to maintain this constant level of drug in the body, the drug must be released from the dosage form at a rate that will replace the amount of drug being metabolized and excreted from the body. Controlled- release of an active ingredient can be stimulated by various conditions including, but not limited to, pH, temperature, enzymes, water, or other physiological conditions or compounds.
  • Parenteral dosage forms can be administered to patients by various routes including, but not limited to, subcutaneous, intravenous (including bolus injection), intramuscular, and intraarterial. Because their administration typically bypasses patients' natural defenses against contaminants, parenteral dosage forms are preferably sterile or capable of being sterilized prior to administration to a patient. Examples of parenteral dosage forms include, but are not limited to, solutions ready for injection, dry products ready to be dissolved or suspended in a pharmaceutically acceptable vehicle for injection, suspensions ready for injection, and emulsions.
  • Suitable vehicles that can be used to provide parenteral dosage forms of the invention are well known to those skilled in the art. Examples include, but are not limited to: Water for Injection USP; aqueous vehicles such as, but not limited to, Sodium Chloride Injection, Ringer's Injection, Dextrose Injection, Dextrose and Sodium Chloride Injection, and Lactated Ringer's Injection; water-miscible vehicles such as, but not limited to, ethyl alcohol, polyethylene glycol, and polypropylene glycol; and non-aqueous vehicles such as, but not limited to, corn oil, cottonseed oil, peanut oil, sesame oil, ethyl oleate, isopropyl myristate, and benzyl benzoate. Compounds that increase the solubility of one or more of the active ingredients disclosed herein can also be inco ⁇ orated into the parenteral dosage forms of the invention.
  • Transdermal and topical dosage forms of the invention include, but are not limited to, creams, lotions, ointments, gels, solutions, emulsions, suspensions, or other forms known to one of skill in the art. See, e.g., Remington 's Pharmaceutical Sciences, 18th eds.,
  • Transdermal dosage forms include "reservoir type” or “matrix type” patches, which can be applied to the skin and worn for a specific period of time to permit the penetration of a desired amount of active ingredients.
  • Suitable excipients e.g., carriers and diluents
  • other materials that can be used to provide transdermal and topical dosage forms encompassed by this invention are well known to those skilled in the pharmaceutical arts, and depend on the particular tissue to which a given pharmaceutical composition or dosage form will be applied.
  • typical excipients include, but are not limited to, water, acetone, ethanol, ethylene glycol, propylene glycol, butane- 1,3-diol, isopropyl myristate, isopropyl palmitate, mineral oil, and mixtures thereof to form lotions, tinctures, creams, emulsions, gels or ointments, which are non-toxic and pharmaceutically acceptable.
  • Moisturizers or humectants can also be added to pharmaceutical compositions and dosage forms if desired. Examples of such additional ingredients are well known in the art. See, e.g., Remington 's Pharmaceutical Sciences, 18th eds., Mack Publishing, Easton PA (1990).
  • penetration enhancers can be used to assist in delivering the active ingredients to the tissue.
  • Suitable penetration enhancers include, but are not limited to: acetone; various alcohols such as ethanol, oleyl, and tetrahydrofuryl; alkyl sulfoxides such as dimethyl sulfoxide; dimethyl acetamide; dimethyl formamide; polyethylene glycol; pyrrolidones such as polyvinylpyrrolidone; Kollidon grades (Povidone, Polyvidone); urea; and various water-soluble or insoluble sugar esters such as Tween 80 (polysorbate 80) and Span 60 (sorbitan monostearate).
  • the pH of a pharmaceutical composition or dosage form, or of the tissue to which the pharmaceutical composition or dosage form is applied may also be adjusted to improve delivery of one or more active ingredients.
  • the polarity of a solvent carrier, its ionic strength, or tonicity can be adjusted to improve delivery.
  • Compounds such as stearates can also be added to pharmaceutical compositions or dosage forms to advantageously alter the hydrophilicity or lipophilicity of one or more active ingredients so as to improve delivery.
  • stearates can serve as a lipid vehicle for the formulation, as an emulsifying agent or surfactant, and as a delivery-enhancing or penetration-enhancing agent.
  • Different salts, hydrates or solvates of the active ingredients can be used to further adjust the properties of the resulting composition.
  • Mucosal dosage forms of the invention include, but are not limited to, ophthalmic solutions, sprays and aerosols, or other forms known to one of skill in the art. See, e.g., Remington 's Pharmaceutical Sciences, 18th eds., Mack Publishing, Easton PA (1990); and Introduction to Pharmaceutical Dosage Forms, 4th ed., Lea & Febiger, Philadelphia (1985). Dosage forms suitable for treating mucosal tissues within the oral cavity can be formulated as mouthwashes or as oral gels.
  • the aerosol comprises a carrier. In another embodiment, the aerosol is carrier free.
  • a compound of formula V can also be administered directly to the lung by inhalation (see e.g., Tong et al., PCT Application, WO 97/39745; Clark et al, PCT Application, WO 99/47196, which are herein inco ⁇ orated by reference).
  • inhalation a compound of formula V can be conveniently delivered to the lung by a number of different devices.
  • a Metered Dose Inhaler which utilizes canisters that contain a suitable low boiling propellant, e.g., dichlorodifluoromethane, trichloroftuoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas can be used to deliver a compound of formula I directly to the lung.
  • MDI devices are available from a number of suppliers such as 3M Co ⁇ oration, Aventis, Boehringer Ingleheim, Forest Laboratories, Glaxo-Wellcome, Schering Plough and Nectura.
  • a Dry Powder Inhaler (DPI) device can be used to administer a compound of formula I to the lung (See, e.g., Raleigh et al., Proc. Amer. Assoc. Cancer Research Annual Meeting, 1999, 40, 397, which is herein inco ⁇ orated by reference).
  • DPI devices typically use a mechanism such as a burst of gas to create a cloud of dry powder inside a container, which can then be inhaled by the patient.
  • DPI devices are also well known in the art and can be purchased from a number of vendors which include, for example, Fisons, Glaxo-Wellcome, Inhale Therapeutic Systems, ML Laboratories, Qdose and Nectura.
  • MDDPI multiple dose DPI
  • a popular variation is the multiple dose DPI (“MDDPI”) system, which allows for the delivery of more than one therapeutic dose.
  • MDDPI devices are available from companies such as AstraZeneca, Glaxo Wellcome, IN AX, Schering Plough, SkyePharma and Nectura.
  • capsules and cartridges of gelatin for use in an inhaler or insufflator can be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch for these systems.
  • liquid spray device Another type of device that can be used to deliver a compound of formula V to the lung is a liquid spray device supplied, for example, by Aradigm Co ⁇ oration.
  • Liquid spray systems use extremely small nozzle holes to aerosolize liquid drug formulations that can then be directly inhaled into the lung.
  • a nebulizer device is used to deliver a compound of formula V to the lung.
  • Nebulizers create aerosols from liquid drug formulations by using, for example, ultrasonic energy to form fine particles that can be readily inhaled (See e.g., ⁇ > ⁇ ⁇ _ _ ⁇ _
  • nebulizers include devices supplied by Sheffield/Systemic Pulmonary Delivery Ltd. (See, Armer et al., U.S. Pat. No. 5,954,047; van der Linden et al., U.S. Pat. No. 5,950,619; van der Linden et al., U.S. Pat. No. 5,970,974, which are herein inco ⁇ orated by reference), Aventis and Batelle Pulmonary Therapeutics.
  • EHD aerosol device uses electrical energy to aerosolize liquid drug solutions or suspensions (see e.g., Noakes et al., U.S. Pat. No. 4,765,539; Coffee, U.S.
  • the electrochemical properties of the compound of formula I formulation may be important parameters to optimize when delivering this drug to the lung with an EHD aerosol device and such optimization is routinely performed by one of skill in the art.
  • EHD aerosol devices may more efficiently delivery drugs to the lung than existing pulmonary delivery technologies.
  • Other methods of infra-pulmonary delivery of a compound of formula V will be known to the skilled artisan and are within the scope of the invention.
  • Liquid drug formulations suitable for use with nebulizers and liquid spray devices and EHD aerosol devices will typically include a compound of formula I with a pharmaceutically acceptable carrier.
  • the pharmaceutically acceptable carrier is a liquid such as alcohol, water, polyethylene glycol or a perfluorocarbon.
  • another material maybe added to alter the aerosol properties of the solution or suspension of a compound of formula V.
  • this material is liquid such as an alcohol, glycol, polyglycol or a fatty acid.
  • Other methods of formulating liquid drug solutions or suspension suitable for use in aerosol devices are known to those of skill in the art (See, e.g., Biesalski, U.S. Pat. Nos.
  • a compound of formula I can also be formulated in rectal or vaginal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.
  • ___ ⁇ ⁇ r ⁇ ⁇ _ ____ _ h addition to the formulations described previously, a compound of formula V can also be formulated as a depot preparation.
  • Such long acting fonnulations can be administered by implantation (for example subcutaneously or intramuscularly) or by intramuscular injection.
  • the compounds can be formulated with suitable polymeric or hydrophobic materials (for example, as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • Liposomes and emulsions are well known examples of delivery vehicles that can be used to deliver a compound of formula V.
  • Certain organic solvents such as dimethylsulfoxide can also be employed, although usually at the cost of greater toxicity.
  • a compound of formula I can also be delivered in a controlled release system.
  • a pump can be used (Sefton, CRC Crit. Ref Biomed Eng., 1987, 14, 201; Buchwald et al., Surgery, 1980, 88, 507; Saudek et al., N. Engl. J Med, 1989, 321, 574).
  • polymeric materials can be used (see Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Fla. (1974); Controlled Drug Bioavailability, Drug Product Design and Performance, Smolen and Ball (eds.), Wiley, New York (1984); Ranger and Peppas, J Macromol. Sci. Rev. Macromol. Chem., 1983, 23, 61; see also Levy et al., Science 1985, 228, 190; During et al., Ann. Neural., 1989,25,351; Howard et al, 1989, J. Neurosurg. 71, 105).
  • a confrolled-release system can be placed in proximity of the target of the compounds of the invention, e.g., the lung, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115 (1984)).
  • Other confrolled-release system can be used (see e.g. Langer, Science, 1990, 249, 1527).
  • Suitable excipients (e.g., carriers and diluents) and other materials that can be used to provide mucosal dosage forms encompassed by this invention are well known to those skilled in the pharmaceutical arts, and depend on the particular site or method which a given pharmaceutical composition or dosage form will be administered.
  • excipients include, but are not limited to, water, ethanol, ethylene glycol, propylene glycol, butane- 1,3 -diol, isopropyl myristate, isopropyl palmitate, mineral oil, and mixtures thereof, which are non-toxic and pharmaceutically acceptable.
  • additional ingredients are well known in the art. See, e.g., Remington 's Pharmaceutical Sciences, 18th eds., Mack Publishing, Easton PA (1990).
  • the pH of a pharmaceutical composition or dosage form, or of the tissue to which the pharmaceutical composition or dosage form is applied can also be adjusted to improve delivery of one or more active ingredients.
  • the polarity of a solvent carrier, its ionic strength, or tonicity can be adjusted to improve delivery.
  • Compounds such as stearates can also be added to pharmaceutical compositions or dosage fonns to advantageously alter the hydrophilicity or lipophilicity of one or more active ingredients so as to improve delivery.
  • stearates can serve as a lipid vehicle for the fonnulation, as an emulsifying agent or surfactant, and as a delivery-enhancing or penetration-enhancing agent.
  • Different salts, hydrates or solvates of the active ingredients can be used to further adjust the properties of the resulting composition.
  • the mixture was allowed to stir for 1 h, then was treated with methyl iodide (0.60 g), and the resulting mixture stirred overnight. It was diluted with ethyl acetate (50 mL), and this was washed with three 50 mL portions of water and then brine. The aqueous phases were back-extracted in sequence with ethyl acetate, and the organic extracts were combined, dried over anhydrous magnesium sulfate, filtered, and evaporated.
  • Ribosomes prepared from HeLa cells were incubated with the small molecules (at a concentration of 100 ⁇ M), followed by treatment with chemical modifying agents (dimethyl sulfate [DMS] and kethoxal [KE]).
  • DMS dimethyl sulfate
  • KE kethoxal
  • rR ⁇ A was phenol- chlorofonn extracted, ethanol precipitated, analyzed in primer extension reactions using end-labeled oligonucleotides hybridizing to different regions of the three rR ⁇ As and resolved on 6% polyacrylamide gels.
  • the probes used for primer extension cover the entire 18S (7 oligonucleotide primers), 28S (24 oligonucleotide primers), and 5S (one primer) rR ⁇ As.
  • Controls in these experiments include DMSO (a control for changes in rR ⁇ A accessibility induced by DMSO), paromomycin (a marker for 18S rR ⁇ A binding), and anisomycin (a marker for 28S rR ⁇ A binding).
  • a bronchial epithelial cell line harboring a nonsense codon at amino acid 1282 (W1282X) was treated with 3-[2-(4-Isopropyl-3-methyl-phenoxy)- acetylamino] -benzoic acid, sodium salt (20 ⁇ M) and CFTR function was monitored as a cAMP-activated chloride channel using the SPQ assay (Yang et al., 1993, Hum Mol Genet. 2(8):1253-1261 and Howard et al., 1996, Nat Med. 2(4):467-469).
  • mice The mutation in the mdx mouse that premature termination of the 427 kDa dystrophin polypeptide has been shown to be a C to T transition at position 3185 in exon 23 (Sicinski et al., 1989, Science. 244(4912):1578-1580).
  • Mouse primary skeletal muscle cultures derived from 1-day old mdx mice were prepared as described previously (Barton- Davis et al., 1999, J Clin Invest. 104(4):375-381). Cells were cultured for 10 days in the presence of 3-[2-(4-Isopropyl-3-methyl-phenoxy)-acetylamino]-benzoic acid, sodium salt (20 ⁇ M).
  • mice were freated for 14 days, after which animals were anesthetized with ketamine and exsanguinated.
  • the tibialis anterior (TA) muscle of the experimental animals was then excised, frozen, and used for immunofluorescence analysis of dystrophin inco ⁇ oration into striated muscle.
  • the presence of dystrophin in TA muscles was detected by immunostaining, as described previously (Barton-Davis et al., 1999, J Clin Invest. 104(4):375-381).
  • Table 3 illustrates a batch formulation and single dosage formulation for a 200 mg 3-[2-(3'-methyl-biphen-4-yloxy)-acetylamino]-benzoic acid single dose unit, i.e., about 40 percent by weight.
  • the pregelatinized corn starch (SPRESS B-820) and 3-[2-(3'-methyl-biphen-4- yloxy)-acetylamino] -benzoic acid components are passed through a 710 ⁇ m screen and then are loaded into a Diffusion Mixer with a baffle insert and blended for 15 minutes.
  • the magnesium stearate is passed through a 210 ⁇ m screen and is added to the Diffusion Mixer.
  • the blend is then encapsulated in a size #0 capsule, 500 mg per capsule (8400 capsule batch size) using a Dosator type capsule filling machine.
  • Table 4 illustrates a batch formulation and a single dose unit formulation containing 100 mg of 3-[2-(4-cyclopentyl-phenoxy)-acetylamino]-benzoic acid.
  • microcrystalline cellulose, croscarmellose sodium, and 3-[2-(4-cyclopentyl- phenoxy)-acetylamino]-benzoic acid components are passed through a #30 mesh screen (about 430 ⁇ to about 655 ⁇ ).
  • the Pluronic F-68® (manufactured by JRH Biosciences, Inc. of Lenexa, KS) surfactant is passed through a #20 mesh screen (about 457 ⁇ to about 1041 ⁇ ).
  • the Pluronic F-68® surfactant and 0.5 kgs of croscarmellose sodium are loaded into a 16 qt. twin shell tumble blender and are mixed for about 5 minutes.
  • the mix is then transfened to a 3 cubic foot twin shell tumble blender where the microcrystalline cellulose is added and blended for about 5 minutes.
  • the thalidomide is added and blended for an additional 25 minutes.
  • This pre-blend is passed through a roller compactor with a hammer mill attached at the discharge of the roller compactor and moved back to the tumble blender.
  • croscarmellose sodium and magnesium stearate is added to the tumble blender and blended for about 3 minutes.
  • the final mixture is compressed on a rotary tablet press with 250 mg per tablet (200,000 tablet batch size). ⁇ ._ _ _
  • a concentrate is prepared by combining 3-[2-(4-trifluoromethoxy-phenoxy)- acetylaminoj-benzoic acid, and a 12.6 kg portion of the trichloromonofluoromethane in a sealed stainless steel vessel equipped with a high shear mixer. Mixing is carried out for about 20 minutes.
  • the bulk suspension is then prepared in the sealed vessel by combining the concentrate with the balance of the propellants in a bulk product tank that is temperature controlled to 21° to 27 °C. and pressure controlled to 2.8 to 4.0 BAR. 17 ml aerosol containers which have a metered valve which is designed to provide 100 inhalations of the composition of the invention. Each container is provided with the following:

Landscapes

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

Abstract

L'invention concerne de nouveaux composés d'acide acétyl amino benzoïque, des procédés d'utilisation d'un dérivé d'acide acétyl amino benzoïque et des compositions pharmaceutiques comprenant ce dernier. Il s'agit notamment de procédés de traitement ou de prévention d'une maladie qui sont améliorés par la modulation de la terminaison de translation prématurée ou la dégradation de l'ARNm médiée par les non-sens, ou qui améliorent au moins un symptôme qui y est associé.
EP03766013A 2002-07-24 2003-07-23 Composes d'acide acetylamino benzoique et leur utilisation pour la suppression de non-sens et le traitement de maladie Withdrawn EP1525185A1 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US39826702P 2002-07-24 2002-07-24
US398267P 2002-07-24
PCT/US2003/023183 WO2004009533A1 (fr) 2002-07-24 2003-07-23 Composes d'acide acetylamino benzoique et leur utilisation pour la suppression de non-sens et le traitement de maladie

Publications (1)

Publication Number Publication Date
EP1525185A1 true EP1525185A1 (fr) 2005-04-27

Family

ID=30771207

Family Applications (1)

Application Number Title Priority Date Filing Date
EP03766013A Withdrawn EP1525185A1 (fr) 2002-07-24 2003-07-23 Composes d'acide acetylamino benzoique et leur utilisation pour la suppression de non-sens et le traitement de maladie

Country Status (4)

Country Link
EP (1) EP1525185A1 (fr)
AU (1) AU2003254157A1 (fr)
CA (1) CA2493457A1 (fr)
WO (1) WO2004009533A1 (fr)

Families Citing this family (26)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7247741B2 (en) * 2005-01-21 2007-07-24 Ptc Therapeutics, Inc. Acetylamino benzoic acid compounds and their use for nonsense suppression and the treatment of disease
US20060025480A1 (en) 2004-08-02 2006-02-02 Boehringer Ingelheim International Gmbh Benzoic acid derivatives as non nucleoside reverse transcriptase inhibitors
BRPI0515015A (pt) 2004-08-11 2008-07-01 Kyorin Seiyaku Kk derivado cìclico de ácido aminobenzóico; medicamento; agonista ppar(alpha); agonista duplo ppar(alpha), y; agonista duplo ppar(alpha), (delta); modulador ppar; agente lipìdeo; agente profilático ou terapêutico compreendendo pelo menos um dos derivados cìclicos de ácido aminobenzóico ou sal do mesmo farmacêuticamente aceitável
CA2582885A1 (fr) * 2004-10-13 2006-04-27 Ptc Therapeutics, Inc. Composes pour la suppression de mutations non-sens et procedes d'utilisation associes
US20090304641A1 (en) 2005-10-17 2009-12-10 Children's Hospital Boston Methods and Compositions for Regulating Gene Expression
ATE518535T1 (de) * 2005-12-30 2011-08-15 Arbor Vita Corp Kleinmolekulare hemmer von pdz-wechselwirkungen
US8633160B2 (en) 2005-12-30 2014-01-21 Nono Inc. Small molecule inhibitors of PDZ interactions
WO2007117438A2 (fr) 2006-03-30 2007-10-18 Ptc Therapeutics, Inc. Méthodes de production d'une protéine fonctionnelle à partir d'un adn présentant une mutation non-sens et traitement des troubles associés
KR100787131B1 (ko) 2006-07-04 2007-12-21 한국생명공학연구원 Hif―1 활성을 저해하는 화합물, 이의 제조방법 및이를 유효성분으로 함유하는 약학적 조성물
FR2912745A1 (fr) * 2007-02-19 2008-08-22 Centre Nat Rech Scient Nouveaux composes derives d'indole et compositions pharmaceutiques les contenant
JP2010526038A (ja) * 2007-05-03 2010-07-29 ノイロサーチ アクティーゼルスカブ イオンチャネル調節剤として有用なβ−ケト−アミド誘導体
EP2062578A1 (fr) * 2007-11-12 2009-05-27 Institut National De La Sante Et De La Recherche Medicale (Inserm) Utilisation de composés chimiques pour traiter le SIDA
WO2009065131A1 (fr) * 2007-11-16 2009-05-22 Rigel Pharmaceuticals, Inc. Composés de carboxamide, de sulfonamide et d'amine pour des troubles métaboliques
CN102464618B (zh) * 2010-11-03 2014-07-23 中国中化股份有限公司 吡唑酰胺类化合物及其应用
JP6045828B2 (ja) * 2012-07-11 2016-12-14 千葉県 抗癌剤
JP2015530378A (ja) * 2012-08-29 2015-10-15 メルク パテント ゲゼルシャフト ミット ベシュレンクテル ハフツングMerck Patent Gesellschaft mit beschraenkter Haftung 変形性関節症の処置のためのddr2インヒビター
MA38837B1 (fr) 2013-07-18 2018-10-31 Novartis Ag Inhibiteurs de l'autotaxine contenant un noyau à cycle benzyle-amide cyclique hétéroaromatique
CN103554043B (zh) * 2013-09-03 2015-04-01 浙江医药高等专科学校 苯基取代的三唑酰胺类化合物及用途
CN103467399B (zh) * 2013-09-03 2015-01-07 浙江医药高等专科学校 三唑酰胺类化合物、其制备方法和其用途
US10800742B2 (en) 2015-04-24 2020-10-13 The Johns Hopkins University Small molecule compounds targeting PBX1 transcriptional complex
CN106478447B (zh) * 2015-09-01 2018-10-30 上海医药工业研究院 羧酸衍生物及其作为fxr拮抗剂的应用
WO2017049409A1 (fr) * 2015-09-25 2017-03-30 The Centre For Drug Research And Development Compositions permettant de favoriser la translecture de codons de terminaison prématurée, et leurs procédés d'utilisation
JP7304697B2 (ja) 2015-12-23 2023-07-07 ムーンショット ファーマ エルエルシー 未成熟終止コドンのリードスルーを促進することにより免疫反応を誘発するための方法
WO2018237326A1 (fr) 2017-06-22 2018-12-27 1AlMOONSHOT PHARMA LLC Méthodes de traitement du cancer au moyen de compositions comprenant de l'amlexanox et des immunomodulateurs
WO2020012339A1 (fr) * 2018-07-09 2020-01-16 Glaxosmithkline Intellectual Property Development Limited Composés chimiques
CN114716343B (zh) * 2020-12-22 2023-03-21 清华大学 新型nampt酶激动剂及其制备与用途

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS51149240A (en) * 1975-06-17 1976-12-22 Mitsubishi Yuka Yakuhin Kk Process for preparing new 3-substituted aminophenylacetic acid derivat ives
JPH07138221A (ja) * 1993-11-11 1995-05-30 Fujisawa Pharmaceut Co Ltd アミジノ化合物
DE19624155A1 (de) * 1996-06-18 1998-01-08 Hoechst Ag Substituierte Benzoesäurederivate, Verfahren zu ihrer Herstellung und die Anwendung der Verbindungen zur Behandlung von Krankheiten
EP0973513A4 (fr) * 1996-10-23 2003-03-19 Zymogenetics Inc Compositions et methodes pour traiter les etats associes a un deficit osseux
US6458538B1 (en) * 1999-12-14 2002-10-01 Ptc Therapeutics, Inc. Methods of assaying for compounds that inhibit premature translation termination and nonsense-mediated RNA decay
WO2002018321A2 (fr) * 2000-08-29 2002-03-07 Abbott Laboratories Inhibiteurs de proteine tyrosine phosphatase d'acide amino(oxo)acetique

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2004009533A1 *

Also Published As

Publication number Publication date
WO2004009533A1 (fr) 2004-01-29
AU2003254157A1 (en) 2004-02-09
CA2493457A1 (fr) 2004-01-29
AU2003254157A8 (en) 2004-02-09

Similar Documents

Publication Publication Date Title
US9861617B2 (en) Compositions of 1,2,4-oxadiazole benzoic acid compounds and methods for their use
US7763656B2 (en) Use of Acetylamino benzoic acid compounds for nonsense suppression and the treatment of disease
US8334313B2 (en) Ureido substituted benzoic acid compounds and their use for nonsense suppression and the treatment of disease
EP1525185A1 (fr) Composes d'acide acetylamino benzoique et leur utilisation pour la suppression de non-sens et le traitement de maladie

Legal Events

Date Code Title Description
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

17P Request for examination filed

Effective date: 20050217

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LI LU MC NL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL LT LV MK

DAX Request for extension of the european patent (deleted)
RIN1 Information on inventor provided before grant (corrected)

Inventor name: BECKMANN, HOLGER

Inventor name: RUBENSTEIN, STEVEN, MARC

Inventor name: ALMSTEAD, NEIL, G.

Inventor name: TAKASUGI, JAMES, JAN

Inventor name: WELCH. ELLEN, M.

Inventor name: WILDE, RICHARD, G.

17Q First examination report despatched

Effective date: 20091203

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

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20120519