WO2024049994A1 - Traitement de la polypose adénomateuse familiale à l'aide d'un macrolide à 13 chaînons - Google Patents

Traitement de la polypose adénomateuse familiale à l'aide d'un macrolide à 13 chaînons Download PDF

Info

Publication number
WO2024049994A1
WO2024049994A1 PCT/US2023/031687 US2023031687W WO2024049994A1 WO 2024049994 A1 WO2024049994 A1 WO 2024049994A1 US 2023031687 W US2023031687 W US 2023031687W WO 2024049994 A1 WO2024049994 A1 WO 2024049994A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
apc
acid
optionally substituted
group
Prior art date
Application number
PCT/US2023/031687
Other languages
English (en)
Inventor
Vasudeo BADARINARAYANA
Yoshitaka Ichikawa
Roger B. Clark
Shuhao Shi
Vijay Modur
Original Assignee
Zikani 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 Zikani Therapeutics, Inc. filed Critical Zikani Therapeutics, Inc.
Publication of WO2024049994A1 publication Critical patent/WO2024049994A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system

Definitions

  • Familial adenomatous polyposis is characterized by the development of numerous (100 – 1,000) non-malignant adenomas in the colon and rectum starting in adolescence that eventually progress to colorectal cancer in mid-life if left untreated. This is commonly associated with clinical symptoms such as rectal bleeding and resultant anemia in ⁇ 40% of patients [1].
  • the primary driver of the disease is deficient adenomatous polyposis coli (APC) protein activity resulting from germline mutations in the gene.
  • the mutations are present in 80% of FAP patients and inherited in an autosomal dominant pattern [2-4]. Mutations can be insertions, deletions or nonsense which result in loss of function of the APC protein and are primarily located in the 5’ region of the 8.5-kb coding region [5, 6].
  • Premature stop codons in the APC gene are generally caused by nonsense (30%) or frameshift (68%) mutations resulting in the synthesis of a truncated, dysfunctional protein and is causative in the majority of FAP patients [2-5].
  • the APC gene encodes for a large 312 kDa protein composed of multiple functional domains that are involved with cell division, adhesion and cellular polarization during embryonic development [8, 9].
  • the general structure of the protein consists of 4 domains which permit the protein to interact and form active complexes with other proteins such as b-catenin, axis inhibition protein (AXIN) and microtubule plus-end binding protein (EB1) [8-10].
  • APC acts as a tumor suppressor gene and the protein negatively regulates the b-catenin/wnt-signaling pathway by mediating the degradation of b-catenin in the cytoplasm [11].
  • the central region of APC between codon 1284 and codon 1580 is referred to as the mutation cluster region (MCR) and approximately 60% of all APC mutations occur in this area, with a majority being nonsense or frameshift mutations generating a truncated protein [2, 12, 13].
  • the MCR region is involved with b-catenin ubiquitination and in the absence of this function, cytoplasmic levels of b-catenin increase [8, 9].
  • APC min multiple intestinal neoplasia mouse model of FAP was developed in C57BL/6 mice by exposure to the mutagen N-ethyl-N-nitrosourea and is a widely used model of the disease [18, 19].
  • Pathology is due to a nonsense mutation at codon 850 of the murine homolog of the APC gene resulting in a nonfunctional, truncated protein [18, 20]. This mutation is similar to the mutations in the APC gene carried in human FAP patients [21].
  • the APC min model utilizes mice that are heterozygous for the mutation (homozygous APC min mice are nonviable) having an average lifespan of 119 days with chronic anemia caused by intestinal polyps as the major causative factor of lethality [18].
  • Moribund APC min mice present with numerous ( ⁇ 30/GI tract) adenomas, mainly located in the small intestines [18-20].
  • Adenomas are detectable in APC min mice at 5 weeks of age and these early lesions appear as crypts crowded with cells of similar morphology observed in low grade colonic lesions found in FAP patients [19, 21]. Accumulation of nuclear b-catenin can be observed in adenomas in APC min mice by immuno-histochemistry [19]. Additionally, disruption of nuclear translocation of b-catenin leads to reduced expression of genes that are transcriptionally co-regulated by b-catenin such as c-myc resulting in decreased polyp formation in APC min mice [22].
  • Macrolides and other ribosome targeting antibiotics have been shown to induce read- through of premature stop codons in eukaryotic cells leading to translation of the full-length mRNA.
  • Investigations by Zilberg et al. [23] demonstrated that tylosin can suppress translation termination generated by nonsense mutations in the APC gene using colorectal cancer cell lines and in vivo colon carcinoma models.
  • Other studies have demonstrated that gentamycin and erythromycin treatment also suppressed nonsense mutations in the APC gene in human colon carcinoma cells, inhibited the in vivo growth of human colorectal xenografts possessing APC nonsense mutations; and reduced the number of intestinal polyps in a mouse model of FAP [23, 24].
  • Figure 1 summarizes the antiproliferative activity of a compound of the invention in human colon carcinoma cells.
  • Figure 2 summarizes the pharmacokinetic parameters of a compound of the invention after oral administration at 100 mg/kg bw in CD1 mice.
  • Figure 3 shows that treatment with a compound of the invention reduces nuclear translocation of beta-catenin colon carcinoma cells harboring a nonsense mutation of the APC.
  • Figure 4 shows decreased protein levels of c-myc in colon carcinoma cells treated with a compound of the invention.
  • Figure 4 shows that long-term survival and decreased anemic phenotype in APC min mice treated with a compound of the invention.
  • Figure 5 shows the reduced number of intestinal polyps and dyplasic lesions observed in APC min mice treated with a compound of the invention.
  • Figure 7 shows that epithelial cells in the small intestines from a compound of the invention have reduced nuclear levels off beta-catenin.
  • Detailed Description of the Invention Definitions [0016] Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.
  • Claims or descriptions that include “or” between one or more members of a group are considered satisfied if one, more than one, or all of the group members are present in, employed in, or otherwise relevant to a given product or process unless indicated to the contrary or otherwise evident from the context.
  • the invention includes embodiments in which exactly one member of the group is present in, employed in, or otherwise relevant to a given product or process.
  • the invention includes embodiments in which more than one, or all of the group members are present in, employed in, or otherwise relevant to a given product or process.
  • the term “about” as used herein means “approximately” and is used to modify a numerical value indicates a defined range around that value.
  • “about X” would generally indicate a value from 0.95X to 1.05X. Any reference to “about X” specifically indicates at least the values X, 0.95X, 0.96X, 0.97X, 0.98X, 0.99X, 1.01X, 1.02X, 1.03X, 1.04X, and 1.05X. Thus, “about X” is intended to teach and provide written description support for a claim limitation of, e.g., “0.98X.” When the quantity “X” only includes whole-integer values (e.g., “X carbons”), “about X” indicates from (X-1) to (X+1). In this case, “about X” as used herein specifically indicates at least the values X, X-1, and X+1.
  • the symbol “ ” refers to a group on a double-bond as occupying either position on the terminus of a double bond to which the symbol is attached; that is, the geometry, E- or Z-, of the double bond is ambiguous.
  • the “ ” symbol will be used at the end of the bond which was theoretically cleaved in order to separate the group from its parent structural Formula.
  • the “R” group may reside on either the 5-membered or the 6-membered ring of the fused or bridged ring system.
  • a group “R” is depicted as existing on a ring system containing saturated carbons, as for example in the following Formula where, in this example, “y” can be more than one, assuming each replaces a currently depicted, implied, or expressly defined hydrogen on the ring; then, unless otherwise defined, where the resulting structure is stable, two “R’s” may reside on the same carbon.
  • alkyl as used herein includes an aliphatic hydrocarbon chain that may be straight chain or branched. The chain may contain an indicated number of carbon atoms: For example, C 1 -C 10 indicates that the group may have from 1 to 10 (inclusive) carbon atoms in it. If not otherwise indicated, an alkyl group contains from 1 to about 20 carbon atoms. In some aspects, alkyl groups have 1 to about 10 carbon atoms.
  • alkyl groups (“lower alkyl”) have 1 to 8, 1 to 6, or 1 to 3 carbon atoms in the chain. Examples may include, but are not limited to, methyl, ethyl, propyl, isopropyl (iPr), 1-butyl, 2-butyl, isobutyl (iBu), tert-butyl, pentyl, 2-methylbutyl, 1,1-dimethylpropyl, hexyl, heptyl, octyl, nonyl, decyl, docecyl, cyclopentyl, or cyclohexyl. [0027] An alkyl group can be unsubstituted or optionally substituted.
  • one or more hydrogen atoms of the alkyl group may be replaced with a moiety independently selected from the group consisting of chloro, fluoro, hydroxy, alkoxy, amino, alkylamino, acylamino, thio, and alkylthio.
  • the alkyl group is unsubstituted or not optionally substituted.
  • aryl as used herein includes cyclic aromatic carbon ring systems containing from 6 to 18 carbons.
  • an aryl group examples include, but are not limited to, phenyl, naphthyl, anthracenyl, tetracenyl, biphenyl and phenanthrenyl.
  • An aryl group can be unsubstituted or optionally substituted. When optionally substituted, one or more hydrogen atoms of the aryl group (e.g., from 1 to 5, from 1 to 2, or 1) may be replaced with a moiety independently selected from the group consisting of alkyl, cyano, acyl, halo, haloalkyl, hydroxy, alkoxy, amino, alkylamino, acylamino, thio, and alkylthio.
  • the alkoxy group is unsubstituted or not optionally substituted.
  • arylalkyl or “aralkyl” as used herein includes an alkyl group as defined herein where at least one hydrogen substituent has been replaced with an aryl group as defined herein. Examples include, but are not limited to, benzyl, 1-phenylethyl, 4-methylbenzyl, and 1,1,- dimethyl-1-phenylmethyl.
  • An arylalkyl or aralkyl group can be unsubstituted or optionally substituted as per its component groups. For example, but without limitation, the aryl group of an arylalkyl group can be substituted, such as in 4-methylbenzyl.
  • the group is unsubstituted or not optionally substituted, especially if including a defined substituent, such as a hydroxyalkyl or alkylaminoalkoxy group.
  • a defined substituent such as a hydroxyalkyl or alkylaminoalkoxy group.
  • fluoroalkyl includes an alkyl group wherein the alkyl group includes one or more fluoro- substituents. Examples include, but are not limited to, trifluoromethyl.
  • “geminal” substitution includes two or more substituents that are directly attached to the same atom. An example is 3,3-dimethyl substitution on a cyclohexyl or spirocyclohexyl ring.
  • halo or “halogen” includes fluoro, chloro, bromo, and iodo.
  • heteroaryl or “heterocycloaryl” includes mono and bicyclic groups that are completely unsaturated or partically unsaturated of about 4 to about 14 ring atoms (e.g., 4 to 10 or 5 to 10 atoms) containing at least one heteroatom.
  • Heteroatom as used in the term heteroaryl refers to oxygen, sulfur and nitrogen. A nitrogen atom of a heteroaryl is optionally oxidized to the corresponding N-oxide.
  • Examples include, but are not limited to, pyrazinyl, furanyl, thienyl, pyridyl, pyrimidinyl, isoxazolyl, isothiazolyl, oxazolyl, thiazolyl, pyrazolyl, furazanyl, pyrrolyl, pyrazolyl, triazolyl, 1,2,4-thiadiazolyl, pyrazinyl, pyridazinyl, quinoxalinyl, phthalazinyl, imidazo[1,2-a]pyridine, imidazo[2,1-b]thiazolyl, benzofurazanyl, indolyl, azaindolyl, benzimidazolyl, benzothienyl, quinolinyl, imidazolyl, thienopyridyl, quinazolinyl, thienopyrimidyl, pyrrolopyridyl, imidazopyrid
  • heteroarylene or “heterocycloarylene” as used herein includes a heteroaryl group that is substituted at two points.
  • An heteroaryl group can be unsubstituted or optionally substituted.
  • one or more hydrogen atoms of the heteroaryl group e.g., from 1 to 5, from 1 to 2, or 1 may be replaced with a moiety independently selected from the group consisting of alkyl, cyano, acyl, halo, haloalkyl, hydroxy, oxo, alkoxy, amino, alkylamino, acylamino, thio, and alkylthio.
  • heteroaryl group is unsubstituted or not optionally substituted.
  • heteroaroyl as used herein includes a heteroaryl-C(O)- group wherein heteroaryl is as defined herein.
  • Heteroaroyl groups include, but are not limited to, thiophenoyl, nicotinoyl, pyrrol-2-ylcarbonyl, and pyridinoyl.
  • heterocycloalkyl may be used interchangeably herein, and as used herein includes a heterocyclyl-C(O)- group wherein heterocyclyl is as defined herein.
  • heterocyclyl (heterocyclo; heterocyclic; heterocycloalkyl) includes a non-aromatic saturated ring of about 3 to about 8 ring atoms (e.g., 5 to about 10 ring atoms, or 3 to about 6 ring atoms), in which one or more of the atoms in the ring system is an element or elements other than carbon, e.g., nitrogen, oxygen or sulfur.
  • a heterocyclyl group optionally comprises at least one sp 2 -hybridized atom (e.g., a ring incorporating an carbonyl, endocyclic olefin, or exocyclic olefin).
  • a nitrogen or sulfur atom of the heterocyclyl is optionally oxidized to the corresponding N-oxide, S-oxide or S,S-dioxide.
  • the monocyclic heterocycle means a three-, four-, five-, six-, seven-, or eight-membered ring containing at least one heteroatom independently selected from the group consisting of O, N, and S.
  • the three- or four-membered ring contains zero or one double bond, and one heteroatom selected from the group consisting of O, N, and S.
  • the five-membered ring contains zero or one double bond and one, two or three heteroatoms selected from the group consisting of O, N and S.
  • the six- membered ring contains zero, one or two double bonds and one, two, or three heteroatoms selected from the group consisting of O, N, and S.
  • the seven- and eight-membered rings contains zero, one, two, or three double bonds and one, two, or three heteroatoms selected from the group consisting of O, N, and S.
  • monocyclic heterocycles include, but are not limited to, azetidinyl, azepanyl, aziridinyl, diazepanyl, 1,3-dioxanyl, 1,3- dioxolanyl, 1,3-dithiolanyl, 1,3-dithianyl, imidazolinyl, imidazolidinyl, isothiazolinyl, isothiazolidinyl, isoxazolinyl, isoxazolidinyl, morpholinyl, oxadiazolinyl, oxadiazolidinyl, oxazolinyl, oxazolidinyl, piperazinyl, piperidinyl, pyranyl, pyrazolinyl, pyrazolidinyl, pyridazin- 3(2H)-onyl, pyridin-2(1H)-onyl, pyrrolinyl, pyr
  • heterocycloalkylene as used herein includes a heterocyclyl (heterocyclo; heterocyclic) group that is substituted at two points.
  • heterocyclyl also includes multicyclic rings such as a bicyclic heterocycle, or a tricyclic heterocycle which may be in a fused, bridged, or spiro orientation.
  • the bicyclic heterocycle is a monocyclic heterocycle fused to a phenyl group, or a monocyclic heterocycle fused to a monocyclic cycloalkyl, or a monocyclic heterocycle fused to a monocyclic cycloalkenyl, or a monocyclic heterocycle fused to a monocyclic heterocycle, or a bridged monocyclic heterocycle ring system in which two non-adjacent atoms of the ring are linked by an alkylene bridge of 1, 2, 3, or 4 carbon atoms, or an alkenylene bridge of two, three, or four carbon atoms.
  • bicyclic heterocycles include, but are not limited to, 3- azabicyclo[3.1.0]hexane, 3-azabicyclo[4.1.0]heptane, 3-azabicyclo[3.2.0]heptane, (3aR,6aS)- hexahydro-1H-2 ⁇ 2-cyclopenta[c]pyrrole, (3aR,7aS)-octahydro-2 ⁇ 2-isoindole.
  • Tricyclic heterocycles are exemplified by a bicyclic heterocycle fused to a phenyl group, or a bicyclic heterocycle fused to a monocyclic cycloalkyl, or a bicyclic heterocycle fused to a monocyclic cycloalkenyl, or a bicyclic heterocycle fused to a monocyclic heterocycle, or a bicyclic heterocycle in which two non-adjacent atoms of the bicyclic ring are linked by an alkylene bridge of 1, 2, 3, or 4 carbon atoms, or an alkenylene bridge of two, three, or four carbon atoms.
  • a heterocycyl group can be unsubstituted or optionally substituted.
  • one or more hydrogen atoms of the group may be replaced with a moiety independently selected from the group consisting of alkyl, halo, haloalkyl, oxo, acetyl, hydroxy, alkoxy, amino, alkylamino, acylamino, thio, and alkylthio.
  • a substituted heterocycyl group can incorporate an exo- or endocyclic alkene (e.g., cyclohex-2-en-1-yl).
  • the heterocycyl group is unsubstituted or not optionally substituted.
  • the monocyclic, bicyclic, and tricyclic heterocycles are connected to the parent molecular moiety through any carbon atom or any nitrogen atom contained within the rings, and can be unsubstituted or substituted.
  • the term “hydrophilic moiety” or “hydrophilic group” includes a moiety or a functional group that has a strong affinity to water. Examples may include, but are not limited to, a charged moiety, such as a cationic moiety or an anionic moiety, or a polar uncharged moiety, such as an alkoxy group or an amine group.
  • hydroxyalkyl includes an alkyl group where at least one hydrogen substituent has been replaced with an alcohol (-OH) group.
  • the hydroxyalkyl group has one alcohol group.
  • the hydroxyalkyl group has one or two alcohol groups, each on a different carbon atom.
  • the hydroxyalkyl group has 1, 2, 3, 4, 5, or 6 alcohol groups. Examples may include, but are not limited to, hydroxymethyl, 2-hydroxyethyl, and 1-hydroxyethyl.
  • the groups may be the same or different.
  • R a and R b are independently selected from the group consisting of alkyl, fluoro, amino, and hydroxyalkyl
  • a molecule with two R a groups and two R b groups could have all groups be an alkyl group (e.g., four different alkyl groups).
  • the first R a could be alkyl
  • the second R a could be fluoro
  • the first R b could be hydroxyalkyl
  • the second R b could be amino (or any other substituents taken from the group).
  • both R a and the first R b could be fluoro
  • the second R b could be alkyl (i.e., some pairs of substituent groups may be the same, while other pairs may be different).
  • amino protecting group is a protecting group that is suitable for preventing undesired reactions at an amino nitrogen.
  • Representative amino-protecting groups include, but are not limited to, formyl; acyl groups, for example alkanoyl groups, such as acetyl; alkoxycarbonyl groups, such as tert-butoxycarbonyl (Boc); arylmethoxycarbonyl groups, such as benzyloxycarbonyl (Cbz) and 9-fluorenylmethoxycarbonyl (Fmoc); arylmethyl groups, such as benzyl (Bn), trityl (Tr), and 1,1-di-(4'-methoxyphenyl)methyl; silyl groups, such as trimethylsilyl (TMS) and tert-butyldimethylsilyl (TBDMS); and the like.
  • “Hydroxyl protecting group” is a protecting group that is suitable for preventing undesired reactions at a hydroxyl oxygen.
  • Representative hydroxyl protecting groups include, but are not limited to, acyl groups such as acetyl; arylmethyl groups, such as benzyl (Bn), trityl (Tr), and 1,1-di-(4'-methoxyphenyl)methyl; silyl groups, such as trimethylsilyl (TMS) and tert- butyldimethylsilyl (TBDMS); ethers such as methoxymethyl (MOM), tetrahydropyranyl (THP), and benzyl (Bn); and the like.
  • acyl groups such as acetyl
  • arylmethyl groups such as benzyl (Bn), trityl (Tr), and 1,1-di-(4'-methoxyphenyl)methyl
  • silyl groups such as trimethylsilyl (TM
  • yield for each of the reactions described herein is expressed as a percentage of the theoretical yield.
  • Subject and “patient” are used interchangeably.
  • a “subject” or “patient” for the purposes of the present invention includes humans and other animals, particularly mammals, and other organisms. Thus the methods are applicable to both human therapy and veterinary applications. In a specific embodiment the patient is a mammal, and in a more specific embodiment the patient is human.
  • a “pharmaceutically acceptable salt” of a compound means a salt that is pharmaceutically acceptable and that possesses the desired pharmacological activity of the parent compound. It is understood that the pharmaceutically acceptable salts are non-toxic.
  • Examples of pharmaceutically acceptable acid addition salts include those formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like; as well as organic acids such as acetic acid, trifluoroacetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, oxalic acid, maleic acid, malonic acid, succinic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, 3-(4-hydroxybenzoyl)benzoic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1,2-ethanedisulfonic acid, 2-hydroxyethanesulfonic acid, benzenesulfonic acid, 4-chlorobenzenesulfonic acid, 2-naphthalenesulfonic acid,
  • Examples of a pharmaceutically acceptable base addition salts include those formed when an acidic proton present in the parent compound is replaced by a metal ion, such as sodium, potassium, lithium, ammonium, calcium, magnesium, iron, zinc, copper, manganese, aluminum salts and the like. Specific salts are the ammonium, potassium, sodium, calcium, and magnesium salts. Salts derived from pharmaceutically acceptable organic non-toxic bases include, but are not limited to, salts of primary, secondary, and tertiary amines, substituted amines including naturally occurring substituted amines, cyclic amines and basic ion exchange resins.
  • organic bases include isopropylamine, trimethylamine, diethylamine, triethylamine, tripropylamine, ethanolamine, 2-dimethylaminoethanol, 2-diethylaminoethanol, dicyclohexylamine, lysine, arginine, histidine, caffeine, procaine, hydrabamine, choline, betaine, ethylenediamine, glucosamine, methylglucamine, theobromine, purines, piperazine, piperidine, N-ethylpiperidine, tromethamine, N-methylglucamine, polyamine resins, and the like.
  • “Therapeutically effective amount” is an amount of a compound of the invention, that when administered to a patient, ameliorates a symptom of the disease.
  • the amount of a compound of the invention which constitutes a “therapeutically effective amount” will vary depending on the compound, the disease state and its severity, the age of the patient to be treated, and the like. The therapeutically effective amount can be determined routinely by one of ordinary skill in the art having regard to their knowledge and to this disclosure.
  • the phrase “genetic disease”, as used herein, means a genetic disorder, genetic disease, genetic condition or genetic syndrome.
  • Preventing” or “prevention” of a disease, disorder, or syndrome includes inhibiting the disease from occurring in a human, i.e. causing the clinical symptoms of the disease, disorder, or syndrome not to develop in an animal that may be exposed to or predisposed to the disease, disorder, or syndrome but does not yet experience or display symptoms of the disease, disorder, or syndrome.
  • Treating” or “treatment” of a disease, disorder, or syndrome includes (i) inhibiting the disease, disorder, or syndrome, i.e., arresting its development; and (ii) relieving the disease, disorder, or syndrome, i.e., causing regression of the disease, disorder, or syndrome.
  • FAP means an autosomal dominant inherited condition in which numerous adenomatous polyps form mainly in the epithelium of the large intestine. While these polyps start out benign, malignant transformation into colon cancer occurs when they are left untreated.
  • FAP attenuated FAP (originally called hereditary flat adenoma syndrome), and autosomal recessive FAP (originally called (or MUTYH-associated polyposis).
  • FAP and attenuated FAP are caused by APC gene defects on chromosome 5 while autosomal recessive FAP is caused by defects in the MUTYH gene on chromosome 1.
  • FAP itself is the most severe and most common; although for all three, the resulting colonic polyps and cancers are initially confined to the colon wall. Detection and removal before metastasis outside the colon can greatly reduce and in many cases eliminate the spread of cancer.
  • Embodiments [0061]
  • a method for treating FAP in a patient in need of such treatment comprising administering to the patient a compound of formula I I or a pharmaceutically acceptable salt thereof, wherein R9a is selected from the group consisting of H or optionally substituted C1-6 alkyl; and R10a is an optionally substituted heteroaryl.
  • R9a is selected from the group consisting of H or optionally substituted C1-6 alkyl; and R10a is an optionally substituted heteroaryl.
  • R 9a is H
  • R9a is optionally substituted C1-6 alkyl.
  • R9a is selected from optionally substituted methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, pentyl, isopentyl, neopentyl and hexyl.
  • R 9a is selected from methyl, ethyl, propyl, butyl, and isobutyl.
  • R10a is an optionally substituted heteroaryl selected from the , a d .
  • R 9a is selected from H, methyl, ethyl, propyl, butyl, and isobutyl and R 10a is a bicyclic heteroaryl selected from the group consisting of , .
  • R 9a is selected from the group consisting of H or optionally substituted C 1-6 alkyl; and is an optionally substituted 6-membered aromatic or heteroaromatic ring.
  • R 9a is H [0070] In another embodiment, R 9a is optionally substituted C 1-6 alkyl. [0071] In another embodiment, R9a is selected from optionally substituted methyl, ethyl, propyl, isopropyl, butyl, isobutyl, tert-butyl, pentyl, isopentyl, neopentyl and hexyl. [0072] In another embodiment, R 9a is selected from methyl, ethyl, propyl, butyl, and isobutyl. A [0073] In another embodiment, is an optionally substituted aromatic ring. A [0074] In another embodiment, is an optionally substituted .
  • Compounds of the invention include the compounds depicted in Table A or a pharmaceutically acceptable salt thereof. Processes for Preparing Compounds [0079] Compounds disclosed herein can be prepared as provided in WO 2020/106627, the entire contents of which is incorporated herein by reference, or as described herein. [0080] Compounds are prepared via two intermediates. The eastern half intermediate is a compound of formula P-1: , or salt thereof.
  • R 3 , R 4a , R 4b , R 5 , R 6a , R 6b , R 8a , and R 8b are as defined herein; and G 4 is of the formula: , , , , o ; each instance of R 15 is independently silyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl, or two R 15 groups are joined to form an optionally substituted heterocyclyl or heteroaryl ring; and each instance of R 16a is independently hydrogen, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl.
  • the uncyclized eastern half intermediate is a compound of formula P-2: or salt thereof, wherein: PG is a hydroxyl protecting group; R 4a , R 4b , R 5 , R 6a , R 6b , R 8a , and R 8b are as defined herein; G 4 is of formula: , , , , ; each instance of R 15 is independently silyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl, or two R 15 groups are joined to form an optionally substituted heterocyclyl or heteroaryl ring; and each instance of R 16a is independently hydrogen, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted ary
  • -OPG is –OBz.
  • a compound of Formula P-3 or salt thereof, wherein the variables are as defined herein.
  • Coupling and Macrolactonization [0084] In certain embodiments, compounds of the present disclosure are prepared by coupling a compound of Formula P1 (the eastern half) wherein R s is a sugar residue , wherein PG is a hydroxyl protecting group and “ ” indicates a point of attachment, and a compound of Formula P-4 (the western half) to provide an uncyclized compound precursor of Formula P-5 as depicted in the following Scheme.
  • Formula P-5 is cyclized to give, after deprotection of the sugar residue a compound of Formula I as depicted in the following scheme.
  • the compound precursor of Formula P-5 wherein R 9a is hydrogen is cyclized to provide a compound of Formula I, which can undergo reductive amination to provide a compound of Formula I wherein R 9a is other than H, as shown in the following Scheme.
  • Late-stage installment of the R 2b group can be achieved via treatment of a compound of Formula P-6 prepared as provide above with a base and a suitable electrophile group (e.g., halogenating agent or R2-LG, wherein LG is a leaving group) as depicted in the following Scheme. In this process, the sugar residue in P-6 is protected and R9a is H or alkyl.
  • a suitable electrophile group e.g., halogenating agent or R2-LG, wherein LG is a leaving group
  • LG is a leaving group
  • G 4 is of formula: , , , , or ; each instance of R 15 is independently silyl, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl, or two R 15 groups are joined to form an optionally substituted heterocyclyl or heteroaryl ring; and each instance of R 16a is independently hydrogen, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted carbocyclyl, optionally substituted heterocyclyl, optionally substituted aryl, or optionally substituted heteroaryl.
  • R s is the sugar moiety .
  • the sugar moiety is typically attached to the compound framework during synthesis of the eastern half, but may also be attached at other stages of the preparation.
  • the sugar moiety may be attached by a chemical or enzymatic glycosylation reaction between the hydroxyl group at the C5 position and a glycosyl donor.
  • the sugar moiety is attached to the compound framework as a thioglycoside.
  • substituents of the sugar moiety are modified after the glycosylation of the compound or compound precursor (e.g., eastern half).
  • the invention provides a method for treating FAP, attenuated FAP (originally called hereditary flat adenoma syndrome), and autosomal recessive FAP (originally called (or MUTYH-associated polyposis)., comprising administering to a subject in need of such treatment a therapeutically effective amount of a compound of Formula I.
  • the FAP is FAP.
  • the FAP is attenuated FAP.
  • the FAP is autosomal recessive FAP.
  • the invention provides a method for treating a genetic disease or genetic disorder caused by APC gene defects on chromosome 5 comprising administering to a subject in need of such treatment a therapeutically effective amount of a compound of Formula I.
  • the invention provides a method for treating a genetic disease or genetic disorder caused by a mutation of the APC gene on chromosome 5 comprising administering to a subject in need of such treatment a therapeutically effective amount of a compound of Formula I.
  • the APC mutation is at codon 1309.
  • the APC mutation is in codons 1250-1464.
  • the mutation is between 168 and 1580.
  • the mutation is between 5’ of codon 168 and 3’ of 1580. [0098] In these and other aspects, the mutation is in the 5’ part of the APC gene (codons 1-177) and the distal 3’ part of the gene. In a further embodiment, the mutation is an interstitial deletion of chromosome 5q22. In a further embodiment, the mutation is in the 5’end in region spanning exons 4 and 5. In a further embodiment, the mutation is in the 5’end in exon 9.
  • the invention provides a method for treating a genetic disease or genetic disorder caused by caused by a mutation of the MUTYH gene on chromosome 1 comprising administering to a subject in need of such treatment a therapeutically effective amount of a compound of Formula I.
  • the invention provides a method for treating a genetic disease or genetic disorder caused by caused by gene defects in the MUTYH gene on chromosome 1 comprising administering to a subject in need of such treatment a therapeutically effective amount of a compound of Formula I.
  • Pharmaceutical Compositions and Administration [0101] The present invention provides pharmaceutical compositions comprising a compound of the present invention and a pharmaceutically acceptable excipient.
  • the compound of the present invention is provided in an effective amount in the pharmaceutical composition.
  • the effective amount is a therapeutically effective amount.
  • the effective amount is a prophylactically effective amount.
  • Pharmaceutically acceptable excipients include any and all solvents, diluents, or other liquid vehicles, dispersions, suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired. General considerations in formulation and/or manufacture of pharmaceutical compositions agents can be found, for example, in Remington's Pharmaceutical Sciences, Sixteenth Edition, E. W.
  • compositions described herein can be prepared by any method known in the art of pharmacology. In general, such preparatory methods include the steps of bringing the compound of the present invention (the “active ingredient”) into association with a carrier and/or one or more other accessory ingredients, and then, if necessary and/or desirable, shaping and/or packaging the product into a desired single- or multi-dose unit.
  • Pharmaceutical compositions can be prepared, packaged, and/or sold in bulk, as a single unit dose, and/or as a plurality of single unit doses.
  • a “unit dose” is discrete amount of the pharmaceutical composition comprising a predetermined amount of the active ingredient.
  • the amount of the active ingredient is generally equal to the dosage of the active ingredient which would be administered to a subject and/or a convenient fraction of such a dosage such as, for example, one-half or one-third of such a dosage.
  • Relative amounts of the active ingredient, the pharmaceutically acceptable excipient, and/or any additional ingredients in a pharmaceutical composition of the invention will vary, depending upon the identity, size, and/or condition of the subject treated and further depending upon the route by which the composition is to be administered.
  • the composition may comprise between 0.1% and 100% (w/w) active ingredient.
  • compositions include inert diluents, dispersing and/or granulating agents, surface active agents and/or emulsifiers, disintegrating agents, binding agents, preservatives, buffering agents, lubricating agents, and/or oils. Excipients such as cocoa butter and suppository waxes, coloring agents, coating agents, sweetening, flavoring, and perfuming agents may also be present in the composition.
  • Exemplary diluents include calcium carbonate, sodium carbonate, calcium phosphate, dicalcium phosphate, calcium sulfate, calcium hydrogen phosphate, sodium phosphate lactose, sucrose, cellulose, microcrystalline cellulose, kaolin, mannitol, sorbitol, inositol, sodium chloride, dry starch, cornstarch, powdered sugar, and mixtures thereof.
  • Exemplary granulating and/or dispersing agents include potato starch, corn starch, tapioca starch, sodium starch glycolate, clays, alginic acid, guar gum, citrus pulp, agar, bentonite, cellulose and wood products, natural sponge, cation-exchange resins, calcium carbonate, silicates, sodium carbonate, cross-linked poly(vinyl-pyrrolidone) (crospovidone), sodium carboxymethyl starch (sodium starch glycolate), carboxymethyl cellulose, cross-linked sodium carboxymethyl cellulose (croscarmellose), methylcellulose, pregelatinized starch (starch 1500), microcrystalline starch, water insoluble starch, calcium carboxymethyl cellulose, magnesium aluminum silicate (Veegum), sodium lauryl sulfate, quaternary ammonium compounds, and mixtures thereof.
  • crospovidone cross-linked poly(vinyl-pyrrolidone)
  • sodium carboxymethyl starch sodium starch glycolate
  • Exemplary surface active agents and/or emulsifiers include natural emulsifiers (e.g. acacia, agar, alginic acid, sodium alginate, tragacanth, chondrux, cholesterol, xanthan, pectin, gelatin, egg yolk, casein, wool fat, cholesterol, wax, and lecithin), colloidal clays (e.g. bentonite [aluminum silicate] and Veegum [magnesium aluminum silicate]), long chain amino acid derivatives, high molecular weight alcohols (e.g.
  • stearyl alcohol cetyl alcohol, oleyl alcohol, triacetin monostearate, ethylene glycol distearate, glyceryl monostearate, and propylene glycol monostearate, polyvinyl alcohol), carbomers (e.g. carboxy polymethylene, polyacrylic acid, acrylic acid polymer, and carboxyvinyl polymer), carrageenan, cellulosic derivatives (e.g. carboxymethylcellulose sodium, powdered cellulose, hydroxymethyl cellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, methylcellulose), sorbitan fatty acid esters (e.g.
  • Cremophor polyoxyethylene ethers, (e.g. polyoxyethylene lauryl ether [Brij 30]), poly(vinyl-pyrrolidone), diethylene glycol monolaurate, triethanolamine oleate, sodium oleate, potassium oleate, ethyl oleate, oleic acid, ethyl laurate, sodium lauryl sulfate, Pluronic F 68, Poloxamer 188, cetrimonium bromide, cetylpyridinium chloride, benzalkonium chloride, docusate sodium, and/or mixtures thereof.
  • Exemplary binding agents include starch (e.g.
  • cornstarch and starch paste examples include gelatin, sugars (e.g. sucrose, glucose, dextrose, dextrin, molasses, lactose, lactitol, mannitol, etc.), natural and synthetic gums (e.g.
  • acacia sodium alginate, extract of Irish moss, panwar gum, ghatti gum, mucilage of isapol husks, carboxymethylcellulose, methylcellulose, ethylcellulose, hydroxyethylcellulose, hydroxypropyl cellulose, hydroxypropyl methylcellulose, microcrystalline cellulose, cellulose acetate, poly(vinyl-pyrrolidone), magnesium aluminum silicate (Veegum), and larch arabogalactan), alginates, polyethylene oxide, polyethylene glycol, inorganic calcium salts, silicic acid, polymethacrylates, waxes, water, alcohol, and/or mixtures thereof.
  • Exemplary preservatives include antioxidants, chelating agents, antimicrobial preservatives, antifungal preservatives, alcohol preservatives, acidic preservatives, and other preservatives.
  • Exemplary antioxidants include alpha tocopherol, ascorbic acid, acorbyl palmitate, butylated hydroxyanisole, butylated hydroxytoluene, monothioglycerol, potassium metabisulfite, propionic acid, propyl gallate, sodium ascorbate, sodium bisulfite, sodium metabisulfite, and sodium sulfite.
  • Exemplary chelating agents include ethylenediaminetetraacetic acid (EDTA) and salts and hydrates thereof (e.g., sodium edetate, disodium edetate, trisodium edetate, calcium disodium edetate, dipotassium edetate, and the like), citric acid and salts and hydrates thereof (e.g., citric acid monohydrate), fumaric acid and salts and hydrates thereof: malic acid and salts and hydrates thereof: phosphoric acid and salts and hydrates thereof: and tartaric acid and salts and hydrates thereof.
  • EDTA ethylenediaminetetraacetic acid
  • salts and hydrates thereof e.g., sodium edetate, disodium edetate, trisodium edetate, calcium disodium edetate, dipotassium edetate, and the like
  • citric acid and salts and hydrates thereof e.g., citric acid mono
  • antimicrobial preservatives include benzalkonium chloride, benzethonium chloride, benzyl alcohol, bronopol, cetrimide, cetylpyridinium chloride, chlorhexidine, chlorobutanol, chlorocresol, chloroxylenol, cresol, ethyl alcohol, glycerin, hexetidine, imidurea, phenol, phenoxyethanol, phenylethyl alcohol, phenylmercuric nitrate, propylene glycol, and thimerosal.
  • Exemplary antifungal preservatives include butyl paraben, methyl paraben, ethyl paraben, propyl paraben, benzoic acid, hydroxybenzoic acid, potassium benzoate, potassium sorbate, sodium benzoate, sodium propionate, and sorbic acid.
  • Exemplary alcohol preservatives include ethanol, polyethylene glycol, phenol, phenolic compounds, bisphenol, chlorobutanol, hydroxybenzoate, and phenylethyl alcohol.
  • Exemplary acidic preservatives include vitamin A, vitamin C, vitamin E, beta-carotene, citric acid, acetic acid, dehydroacetic acid, ascorbic acid, sorbic acid, and phytic acid.
  • Other preservatives include tocopherol, tocopherol acetate, deteroxime mesylate, cetrimide, butylated hydroxyanisol (BHA), butylated hydroxytoluened (BHT), ethylenediamine, sodium lauryl sulfate (SLS), sodium lauryl ether sulfate (SLES), sodium bisulfite, sodium metabisulfite, potassium sulfite, potassium metabisulfite, Glydant Plus, Phenonip, methylparaben, Germall 115, Germaben II, Neolone, Kathon, and Euxyl.
  • the preservative is an anti-oxidant. In other embodiments, the preservative is a chelating agent.
  • Exemplary buffering agents include citrate buffer solutions, acetate buffer solutions, phosphate buffer solutions, ammonium chloride, calcium carbonate, calcium chloride, calcium citrate, calcium glubionate, calcium gluceptate, calcium gluconate, D-gluconic acid, calcium glycerophosphate, calcium lactate, propanoic acid, calcium levulinate, pentanoic acid, dibasic calcium phosphate, phosphoric acid, tribasic calcium phosphate, calcium hydroxide phosphate, potassium acetate, potassium chloride, potassium gluconate, potassium mixtures, dibasic potassium phosphate, monobasic potassium phosphate, potassium phosphate mixtures, sodium acetate, sodium bicarbonate, sodium chloride, sodium citrate, sodium lactate, dibasic sodium phosphate, monobasic sodium phosphate, sodium phosphate mixtures, trometh
  • Exemplary lubricating agents include magnesium stearate, calcium stearate, stearic acid, silica, talc, malt, glyceryl behanate, hydrogenated vegetable oils, polyethylene glycol, sodium benzoate, sodium acetate, sodium chloride, leucine, magnesium lauryl sulfate, sodium lauryl sulfate, and mixtures thereof.
  • Exemplary natural oils include almond, apricot kernel, avocado, babassu, bergamot, black current seed, borage, cade, camomile, canola, caraway, carnauba, castor, cinnamon, cocoa butter, coconut, cod liver, coffee, corn, cotton seed, emu, eucalyptus, evening primrose, fish, flaxseed, geraniol, gourd, grape seed, hazel nut, hyssop, isopropyl myristate, jojoba, kukui nut, lavandin, lavender, lemon, litsea cubeba, macademia nut, mallow, mango seed, meadowfoam seed, mink, nutmeg, olive, orange, orange roughy, palm, palm kernel, peach kernel, peanut, poppy seed, pumpkin seed, rapeseed, rice bran, rosemary, safflower, sandalwood, sasquana, savoury, sea
  • Exemplary synthetic oils include, but are not limited to, butyl stearate, caprylic triglyceride, capric triglyceride, cyclomethicone, diethyl sebacate, dimethicone 360, isopropyl myristate, mineral oil, octyldodecanol, oleyl alcohol, silicone oil, and mixtures thereof.
  • Liquid dosage forms for oral and parenteral administration include pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage forms may comprise inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (e.g., cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate,
  • the oral compositions can include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
  • the conjugates of the invention are mixed with solubilizing agents such as Cremophor, alcohols, oils, modified oils, glycols, polysorbates, cyclodextrins, polymers, and mixtures thereof.
  • solubilizing agents such as Cremophor, alcohols, oils, modified oils, glycols, polysorbates, cyclodextrins, polymers, and mixtures thereof.
  • injectable preparations for example, sterile injectable aqueous or oleaginous suspensions can be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation can be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • a nontoxic parenterally acceptable diluent or solvent for example, as a solution in 1,3-butanediol.
  • acceptable vehicles and solvents that can be employed are water, Ringer's solution, U.S.P. and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil can be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid are used in the preparation of injectables.
  • the injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
  • a sterile injectable composition e.g., a sterile injectable aqueous or oleaginous suspension, can be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as Tween 80) and suspending agents.
  • the sterile injectable preparation can also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • a non-toxic parenterally acceptable diluent or solvent for example, as a solution in 1,3-butanediol.
  • acceptable vehicles and solvents that can be employed are mannitol, water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium (e.g., synthetic mono- or diglycerides).
  • Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically- acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • oil solutions or suspensions can also contain a long-chain alcohol diluent or dispersant, or carboxymethyl cellulose or similar dispersing agents.
  • Other commonly used surfactants such as Tweens or Spans or other similar emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms can also be used for the purposes of formulation.
  • Tweens or Spans or other similar emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms can also be used for the purposes of formulation.
  • compositions for rectal or vaginal administration are typically suppositories which can be prepared by mixing the conjugates of this invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active ingredient.
  • suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active ingredient.
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • the active ingredient is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite clay, and
  • the dosage form may comprise buffering agents.
  • Solid compositions of a similar type can be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally comprise opacifying agents and can be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner.
  • embedding compositions which can be used include polymeric substances and waxes. Solid compositions of a similar type can be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.
  • Pharmaceutical compositions, which can be used orally include push-fit capsules made of gelatin as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules may contain the active ingredients in admixture with filler such as lactose, binders such as starches, lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the compounds presented herein may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • suitable liquids such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers may be added. All formulations for oral administration should be in dosages suitable for the chosen route of administration.
  • Dragee cores are provided with suitable coatings. For this purpose, concentrated sugar solutions may be used which may optionally contain gum arabic, talc, polyvinyl pyrrolidone, carbopol gel, polyethylene glycol, titanium dioxide, lacquer solutions and suitable organic solvents or solvent mixtures. Dyestuffs or pigments may be added to the tablets or dragee coatings for identification or to characterize different combinations of active aminoglycoside compounds doses.
  • the active ingredient can be in micro-encapsulated form with one or more excipients as noted above.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art.
  • the active ingredient can be admixed with at least one inert diluent such as sucrose, lactose or starch.
  • Such dosage forms may comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose.
  • the dosage forms may comprise buffering agents. They may optionally comprise opacifying agents and can be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner
  • examples of embedding compositions which can be used include polymeric substances and waxes.
  • Dosage forms for topical and/or transdermal administration of a compound of this invention may include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants and/or patches.
  • the active ingredient is admixed under sterile conditions with a pharmaceutically acceptable carrier and/or any needed preservatives and/or buffers as can be required.
  • the present invention contemplates the use of transdermal patches, which often have the added advantage of providing controlled delivery of an active ingredient to the body.
  • dosage forms can be prepared, for example, by dissolving and/or dispensing the active ingredient in the proper medium.
  • the rate can be controlled by either providing a rate controlling membrane and/or by dispersing the active ingredient in a polymer matrix and/or gel.
  • Suitable devices for use in delivering intradermal pharmaceutical compositions described herein include short needle devices such as those described in U.S. Pat.
  • Intradermal compositions can be administered by devices which limit the effective penetration length of a needle into the skin, such as those described in PCT publication WO 99/34850 and functional equivalents thereof. Jet injection devices which deliver liquid vaccines to the dermis via a liquid jet injector and/or via a needle which pierces the stratum corneum and produces a jet which reaches the dermis are suitable. Jet injection devices are described, for example, in U.S. Pat. Nos.
  • a pharmaceutical composition of the invention can be prepared, packaged, and/or sold in a formulation suitable for pulmonary administration via the buccal cavity.
  • a formulation may comprise dry particles which comprise the active ingredient and which have a diameter in the range from about 0.5 to about 7 nanometers or from about 1 to about 6 nanometers.
  • compositions are conveniently in the form of dry powders for administration using a device comprising a dry powder reservoir to which a stream of propellant can be directed to disperse the powder and/or using a self-propelling solvent/powder dispensing container such as a device comprising the active ingredient dissolved and/or suspended in a low-boiling propellant in a sealed container.
  • a self-propelling solvent/powder dispensing container such as a device comprising the active ingredient dissolved and/or suspended in a low-boiling propellant in a sealed container.
  • Such powders comprise particles wherein at least 98% of the particles by weight have a diameter greater than 0.5 nanometers and at least 95% of the particles by number have a diameter less than 7 nanometers. Alternatively, at least 95% of the particles by weight have a diameter greater than 1 nanometer and at least 90% of the particles by number have a diameter less than 6 nanometers.
  • Dry powder compositions may include a solid fine powder diluent such as sugar and are conveniently provided in a unit dose form.
  • Low boiling propellants generally include liquid propellants having a boiling point of below 65 oF at atmospheric pressure. Generally, the propellant may constitute 50 to 99.9% (w/w) of the composition, and the active ingredient may constitute 0.1 to 20% (w/w) of the composition.
  • the propellant may further comprise additional ingredients such as a liquid non- ionic and/or solid anionic surfactant and/or a solid diluent (which may have a particle size of the same order as particles comprising the active ingredient).
  • compositions of the invention formulated for pulmonary delivery may provide the active ingredient in the form of droplets of a solution and/or suspension.
  • Such formulations can be prepared, packaged, and/or sold as aqueous and/or dilute alcoholic solutions and/or suspensions, optionally sterile, comprising the active ingredient, and may conveniently be administered using any nebulization and/or atomization device.
  • Such formulations may further comprise one or more additional ingredients including, but not limited to, a flavoring agent such as saccharin sodium, a volatile oil, a buffering agent, a surface-active agent, and/or a preservative such as methylhydroxybenzoate.
  • the droplets provided by this route of administration may have an average diameter in the range from about 0.1 to about 200 nanometers.
  • Formulations described herein as being useful for pulmonary delivery are useful for intranasal delivery of a pharmaceutical composition of the invention.
  • Another formulation suitable for intranasal administration is a coarse powder comprising the active ingredient and having an average particle from about 0.2 to 500 micrometers. Such a formulation is administered by rapid inhalation through the nasal passage from a container of the powder held close to the nares.
  • Formulations for nasal administration may, for example, comprise from about as little as 0.1% (w/w) and as much as 100% (w/w) of the active ingredient, and may comprise one or more of the additional ingredients described herein.
  • a pharmaceutical composition of the invention can be prepared, packaged, and/or sold in a formulation for buccal administration.
  • Such formulations may, for example, be in the form of tablets and/or lozenges made using conventional methods, and may contain, for example, 0.1 to 20% (w/w) active ingredient, the balance comprising an orally dissolvable and/or degradable composition and, optionally, one or more of the additional ingredients described herein.
  • formulations for buccal administration may comprise a powder and/or an aerosolized and/or atomized solution and/or suspension comprising the active ingredient.
  • Such powdered, aerosolized, and/or aerosolized formulations when dispersed, may have an average particle and/or droplet size in the range from about 0.1 to about 200 nanometers, and may further comprise one or more of the additional ingredients described herein.
  • pharmaceutical compositions are principally directed to pharmaceutical compositions which are suitable for administration to humans, it will be understood by the skilled artisan that such compositions are generally suitable for administration to animals of all sorts. Modification of pharmaceutical compositions suitable for administration to humans in order to render the compositions suitable for administration to various animals is well understood, and the ordinarily skilled veterinary pharmacologist can design and/or perform such modification with ordinary experimentation.
  • compositions of the present invention are typically formulated in dosage unit form for ease of administration and uniformity of dosage. It will be understood, however, that the total daily usage of the compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment.
  • the specific therapeutically effective dose level for any particular subject or organism will depend upon a variety of factors including the disease, disorder, or condition being treated and the severity of the disorder; the activity of the specific active ingredient employed; the specific composition employed; the age, body weight, general health, sex and diet of the subject; the time of administration, route of administration, and rate of excretion of the specific active ingredient employed; the duration of the treatment; drugs used in combination or coincidental with the specific active ingredient employed; and like factors well known in the medical arts.
  • the above-described compound or its pharmaceutical composition can be administered intravenously, intravitreally, intradermally, transdermally, intrathecally, intraarterially, intraperitoneally, intranasally, intravaginally, intrarectally, intraosseously, periprosthetically, topically, intramuscularly, subcutaneously, mucosally, intraosseosly, periprosthetically, in utero, orally, topically, locally, via inhalation (e.g., aerosol inhalation), by injection, by infusion, by continuous infusion, by localized perfusion bathing target cells directly, via a catheter, via a lavage, in cremes, in lipid compositions (e.g., liposomes), or by other method or any combination of the forgoing as would be known to one of ordinary skill in the art (see, for example, Remington's Pharmaceutical Sciences, 2003, incorporated herein by reference).
  • inhalation e.g., aerosol inhalation
  • the pharmaceutical composition and/or additional agent is formulated to be administered via an alimentary route.
  • Alimentary routes include all possible routes of administration in which the composition is in direct contact with the alimentary tract.
  • the pharmaceutical compositions disclosed herein may be administered orally, buccally, rectally, or sublingually.
  • compositions described herein may be administered via a parenteral route.
  • parenteral includes routes that bypass the alimentary tract.
  • the pharmaceutical compositions disclosed herein may be administered, for example but not limited to, intravenously, intradermally, intramuscularly, intraarterially, intrathecally, subcutaneous, or intraperitoneally [0143] According to some embodiments, the administration is effected orally.
  • the compounds presented herein can be formulated readily by combining the compounds with pharmaceutically acceptable carriers well known in the art.
  • Such carriers enable the compounds presented herein to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions, and the like, for oral ingestion by a patient.
  • Pharmacological preparations for oral use can be made using a solid excipient, optionally grinding the resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries if desired, to obtain tablets or dragee cores.
  • Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carbomethylcellulose; and/or physiologically acceptable polymers such as polyvinylpyrrolidone (PVP).
  • disintegrating agents may be added, such as cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • compositions may take the form of tablets or lozenges formulated in conventional manner.
  • the compounds presented herein are conveniently delivered in the form of an aerosol spray presentation (which typically includes powdered, liquefied and/or gaseous carriers) from a pressurized pack or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichloro- tetrafluoroethane or carbon dioxide.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichloro- tetrafluoroethane or carbon dioxide.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • Capsules and cartridges of, e.g., gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compounds presented herein and a suitable powder base such as, but not limited to, lactose or starch.
  • the compounds presented herein may be formulated in aqueous solutions, preferably in physiologically compatible buffers such as Hank's solution, Ringer's solution, or physiological saline buffer with or without organic solvents such as propylene glycol, polyethylene glycol.
  • Pharmaceutical compositions for topical administration may include the compositions formulated for a medicated application such as an ointment, paste, cream, or powder.
  • Ointments include all oleaginous, adsorption, emulsion, and water-soluble based compositions for topical application, while creams and lotions are those compositions that include an emulsion base only.
  • Topically administered medications may contain a penetration enhancer to facilitate adsorption of the active ingredients through the skin.
  • Suitable penetration enhancers include glycerin, alcohols, alkyl methyl sulfoxides, pyrrolidones and luarocapram.
  • Possible bases for compositions for topical application include polyethylene glycol, lanolin, cold cream and petrolatum as well as any other suitable absorption, emulsion or water-soluble ointment base.
  • Topical preparations may also include emulsifiers, gelling agents, and antimicrobial preservatives as necessary to preserve the composition and provide for a homogenous mixture.
  • Transdermal administration of the compositions may also comprise the use of a "patch.”
  • the patch may supply one or more compositions at a predetermined rate and in a continuous manner over a fixed period of time.
  • the compositions may be delivered by eye drops, intranasal sprays, inhalation, and/or other aerosol delivery vehicles. Methods for delivering compositions directly to the lungs via nasal aerosol sprays has been described in U.S. Pat. Nos.5,756,353 and 5,804,212 (each specifically incorporated herein by reference in their entirety).
  • compositions disclosed herein may be delivered via an aerosol.
  • aerosol refers to a colloidal system of finely divided solid or liquid particles dispersed in a liquefied or pressurized gas propellant.
  • the typical aerosol for inhalation consists of a suspension of active ingredients in liquid propellant or a mixture of liquid propellant and a suitable solvent.
  • Suitable propellants include hydrocarbons and hydrocarbon ethers.
  • Suitable containers will vary according to the pressure requirements of the propellant.
  • Administration of the aerosol will vary according to subject's age, weight and the severity and response of the symptoms.
  • penetrants are used in the formulation. Such penetrants are generally known in the art.
  • the compounds presented herein may be formulated for parenteral administration, e.g., by bolus injection or continuous infusion.
  • Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multidose containers with optionally, an added preservative.
  • the compositions may be suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • the compounds presented herein may be in powder form for constitution with a suitable vehicle, e.g., sterile, pyrogen-free water, before use.
  • the exact amount of a compound required to achieve an effective amount will vary from subject to subject, depending, for example, on species, age, and general condition of a subject, severity of the side effects or disorder, identity of the particular compound(s), mode of administration, and the like.
  • the desired dosage can be delivered three times a day, two times a day, once a day, every other day, every third day, every week, every two weeks, every three weeks, or every four weeks.
  • the desired dosage can be delivered using multiple administrations (e.g., two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, or more administrations).
  • an effective amount of a compound for administration one or more times a day to a 70 kg adult human may comprise about 0.0001 mg to about 3000 mg, about 0.0001 mg to about 2000 mg, about 0.0001 mg to about 1000 mg, about 0.001 mg to about 1000 mg, about 0.01 mg to about 1000 mg, about 0.1 mg to about 1000 mg, about 1 mg to about 1000 mg, about 1 mg to about 100 mg, about 10 mg to about 1000 mg, or about 100 mg to about 1000 mg, of a compound per unit dosage form.
  • the compounds of the invention may be administered orally or parenterally at dosage levels sufficient to deliver from about 0.001 mg/kg to about 100 mg/kg, from about 0.01 mg/kg to about 50 mg/kg, preferably from about 0.1 mg/kg to about 40 mg/kg, preferably from about 0.5 mg/kg to about 30 mg/kg, from about 0.01 mg/kg to about 10 mg/kg, from about 0.1 mg/kg to about 10 mg/kg, and more preferably from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect.
  • dose ranges as described herein provide guidance for the administration of provided pharmaceutical compositions to an adult.
  • the amount to be administered to, for example, a child or an adolescent can be determined by a medical practitioner or person skilled in the art and can be lower or the same as that administered to an adult.
  • a compound or composition, as described herein can be administered in combination with one or more additional therapeutically active agents.
  • the compounds or compositions can be administered in combination with additional therapeutically active agents that improve their bioavailability, reduce and/or modify their metabolism, inhibit their excretion, and/or modify their distribution within the body.
  • the therapy employed may achieve a desired effect for the same disorder, and/or it may achieve different effects.
  • the compound or composition can be administered concurrently with, prior to, or subsequent to, one or more additional therapeutically active agents.
  • each agent will be administered at a dose and/or on a time schedule determined for that agent.
  • the additional therapeutically active agent utilized in this combination can be administered together in a single composition or administered separately in different compositions.
  • the particular combination to employ in a regimen will take into account compatibility of the inventive compound with the additional therapeutically active agent and/or the desired therapeutic effect to be achieved.
  • additional therapeutically active agents utilized in combination be utilized at levels that do not exceed the levels at which they are utilized individually. In some embodiments, the levels utilized in combination will be lower than those utilized individually.
  • kits e.g., pharmaceutical packs.
  • the kits provided may comprise an inventive pharmaceutical composition or compound and a container (e.g., a vial, ampule, bottle, syringe, and/or dispenser package, or other suitable container).
  • a container e.g., a vial, ampule, bottle, syringe, and/or dispenser package, or other suitable container.
  • provided kits may optionally further include a second container comprising a pharmaceutical excipient for dilution or suspension of an inventive pharmaceutical composition or compound.
  • inventive pharmaceutical composition or compound provided in the container and the second container are combined to form one-unit dosage form.
  • Examples [0160] In order that the invention described herein may be more fully understood, the following examples are set forth. The synthetic and biological examples described in this application are offered to illustrate the compounds, pharmaceutical compositions, and methods provided herein and are not to be construed in any way as limiting their scope. Compounds were prepared as described in WO 2020/106627 and as described below. Intermediate Scheme 6 IS6-1 [0161] tert-butyl (R)-(1,5-dihydroxypentan-2-yl)carbamate (IS6-1).
  • IS6-2 [0163] tert-butyl (R)-4-(3-hydroxypropyl)-2,2-dimethyloxazolidine-3-carboxylate (IS6-2).
  • DMEM Dulbecco Modified Eagle Medium
  • FBS Fetal Bovine serum
  • Penicillin-Streptomycin Gibco
  • Western blots Cells were plated at a density of 4x10 6 cells/100 mm dish. After 48 hours, cells were treated with either DMSO or a compound of the invention as recited in Table A (50 mM).
  • Non-phosphorylated b-catenin (Ser33/37/Thr41) (1:1000, catalogue 8814, Cell Signaling Technology, Danvers, MA): c-myc (1:1000, catalogue ab32072, Abcam, Waltham, MA), MEK1/2 (1:1000, catalogue 8727, Cell Signaling Technology), Histone H3 (1:1000, catalogue 4499, Cell Signaling Technology) and a-tubulin (1:10,000, catalogue T6199, MilliporeSigma, Burlington, MA).
  • Animal studies. Pharmacokinetic studies were performed at WuXi AppTec Laboratory Testing Division (Cranbury, NJ) in male CD-1 mice sourced from Hilltop Lab Animals, Inc. (Scottdale, PA) 6 – 8 weeks of age.
  • mice were allowed free access to water and rodent chow pellets (LabDiet, Certified Rodent Diet #5002). Animals received a single oral dose of a compound of the invention as recited in Table A (100 mg/kg body weight) from a sterile stock solution of 10 mg/ml saline. Blood samples were collected in EDTA tubes from peripheral veins at 0.083, 0.250, 0.500, 1.00, 2.00, 4.00, 8.00 and 24.0 hour time points post-dosing and processed for plasma. After the last blood collection, mice were euthanized, backs were shaved and a 1 cm 2 skin sample was excised and weighed. A 2 cm section of jejunum was harvested, rinsed with saline and weighed.
  • Tissue samples and plasma was stored at -70 o C.
  • Thawed skin samples were finely minced, extracted with acetonitrile, centrifuged at 3000 x g and supernatants were collected for LC-MS/MS analysis.
  • Water was added to thawed intestinal tissue at a 6:1 ratio (ml water/mg tissue), homogenized and processed for LC-MS/MS analysis.
  • a SCIEX Triple Quad 6500+ LC-MS/MS system (SCIEX, Framingham, MA) was used to analyze plasma, jejunum homogenate and skin/acetonitrile samples.
  • proteins were precipitated using IS solution (100 ng/mL Labetalol & 100 ng/mL Tolbutamide in Acetonitrile) and centrifuged at 3900 x g. Supernatant was mixed with an equal volume of water and 2.00 – 4.00 mL was injected for LC-MS/MS analysis. Calibration curves in the range of 1.00 – 3000 ng/ml were generated in corresponding tissue preparations from na ⁇ ve CD-1 mice. Data analysis was performed using Phoenix WinNonlin software (Certara, Princeton, NJ).
  • mice of C57BL/6 strain were obtained from Jackson Laboratory (Bar Harbor, ME) and female APC min mice 10 weeks of age with body weight greater than 15 g were used. Mice were housed using individually ventilated caging system and allowed free access to sterile rodent chow (Teklad 2919) and water. Clinical observations and body weights were recorded daily and mice with body weight loss greater than 20% were removed from study. Study was conducted under compliance of the UK Animals Scientific Procedures Act 1986 and was performed at Crown Biosciences UK Ltd (Loughborough, UK). The compound was formulated in sterile saline to a working concentration of 5 mg/ml.
  • mice were treated daily with oral doses of the compound (50, 25 or 12.5 mg/kg body weight) or saline for 8 weeks. Drug was well tolerated with no adverse events noted. As animals became moribund or at the end of study, mice were euthanized; a full necropsy was performed; spleens weighed; and the complete gastro- intestinal tract was removed for analysis. Whole blood was collected in EDTA tubes and complete blood counts (CBC) were as performed on a Woodley InSight 5 Diff Retic Haematology Analyzer and compared to normal murine CBC ranges (Pinmoore Animal Laboratory Services Limited, Cheshire, UK). [0247] Polyp scoring, histo-pathology and immuno-histochemistry.
  • CBC complete blood counts
  • the excised intestinal tract (small and large bowel) was flushed with PBS and sliced longitudinally; 10 cm sections were visually inspected; and polyps were enumerated. The mid-sections of the small intestines were fixed in 10% formalin; fashioned into swiss rolls; processed into FFPE blocks; and 4 mm sections were used for analysis.
  • a Tissue-Tek Prisma Plus autostainer (Sakura Finetek USA, Torrance, CA) was used for hematoxylin and eosin (H&E) staining. Sections were scanned with Pannoramic Digital Slide Scanners at 40x magnification (3DHISTECH, Pannoramic SCAN, Budapest, Hungary).
  • IHC Immuno-histochemistry
  • Rabbit IgG mAb for b-catenin was obtained from Cell Signaling Technology. A polyclonal anti-rabbit IgG conjugated to horseradish peroxidase was used for detection (Leica Biosystems, Buffalo Grove, IL). Non-specific rabbit IgG (Abcam) was used for an isotype control. Sections were rinsed and counterstained with H&E. [0248] Statistical analysis. GraphPad Prism software version 9 (GraphPad Software, La Jolla, CA) was used for data analysis and the generation of graphs. Mean values are plotted with the standard deviation (SD) and standard error of the mean (SEM) indicated by bars. The unpaired student t-test was used to compare the means of the treatment groups to control group. In survival analysis, the log-rank test was used to determine significance between the control and treatment groups. p-values less than 0.05 were considered significant.
  • Tumor cells were cultured in the presence of titrated concentrations of a compound as disclosed herein and the drug concentration that inhibited 50% of cell growth as compared to vehicle treated cells (GI50) is shown in Table 1 ( Figure 1).
  • the compound had no effect on the growth of HCT116 cells at all the concentrations tested (upto100 mM) and the GI50s for SW1417 and SW403 cells were 34.8 and 39.8 mM, respectively.
  • Treatment of human colon cancer cells with a compound as disclosed herein results in decreased levels of nuclear b-catenin and c-myc.
  • Adenomatous polyposis coli is a large, complex protein (312 kDa) which is directly involved in ubiquitin degradation of phosphorylated b-catenin in the cytoplasm [34].
  • APC activity In the absence of APC activity, there is increased translocation of b-catenin to the nucleus. Therefore, the amount of nuclear b-catenin relative to that in the cytoplasm is used as an indicator of the level of functional APC, particularly since there is a lack of reliable, commercially available antibodies for the detection of APC [35, 36].
  • SW403 and SW1417 cell were treated with 50 mM of the compound for 48 hours and b-catenin levels were determined by western blot in the nuclear and cytoplasmic extracts.
  • colon carcinoma cells exhibit a significant reduction of nuclear b-catenin accumulation in response to compound exposure.
  • the nuclear level of b-catenin was reduced by 40% in SW1417 and SW403 treated cells as compared to DMSO treated cells. This indicates increased levels of functional APC resulting from the compound mediated read-through of APC nonsense mutations. Cytoplasmic levels of b-catenin in cells treated with DMSO or the compound was similar. [0251] Expression of the c-myc gene is transcriptionally co-regulated by b-catenin [8, 37].
  • the compound of the invention as recited in Table A (50 mM, 48 hours) treatment reduced c-myc protein levels by 25% in SW1417 cells and by 30% in SW403 cells relative to cells treated with DMSO.
  • Table A 50 mM, 48 hours
  • treatment leads to increased levels of active APC, promoting cytoplasmic degradation of b- catenin and a reduction of nuclear levels of b-catenin, resulting in decreased expression of c- myc.
  • Treatment with a compound as disclosed herein improves the survival of APC min mice and reduces anemic phenotype.
  • a single dose mouse study was conducted to determine the pharmacokinetics and exposure of a compound as disclosed herein in intestinal tissue after oral administration.
  • a well-established model of FAP is the APC min mouse in which the human APC homologue gene (min) contains a nonsense mutation generating a non-functional, truncated protein.
  • APC min mice develop numerous small intestinal adenomas as the primary phenotype and anemia as a secondary, lethal condition resulting in an average lifespan of 120 days [18-20]. The majority of adenomas are present in APC min mice at 5 - 8 weeks of age and the number of adenomas does not significantly increase thereafter [39].
  • APC min mice were treated daily with oral doses of a compound as disclosed herein(50 mg/kg body weight) or vehicle saline.
  • mice in the vehicle control group began to show severe clinical signs of disease progression and had to be terminated. Only 5 vehicle treated mice (50%) remained in the study at the endpoint. In contrast, all the mice treated with the compound remained on study for the full duration.
  • Similar efficacy results were obtained in a repeat study using daily oral doses of the compound at 50, 25 and 12.5 mg/kg body weight with 100% survival achieved at the lowest dose used in the 8-week study.
  • Anemia resulting from chronic blood loss from the numerous intestinal lesions in APC min mice is considered to be the major cause of animal death [18].
  • Compound treatment significantly improved the anemia associated with the APC min model as indicated by an approximately 35% reduction of splenomegaly, and increased levels of hemoglobin (approximately 45%), packed cell volume (approximately 55%) and red blood cell counts (approximately 60%) in the mice at all 3 dose levels tested (Fig.3B)( Figure 5bB.
  • Clinical observations, consistent with the APC min model include pale limb extremities and discolored feces, were reported in both groups and body weight gain was comparable in vehicle and the treatment groups through-out the course of the study.
  • Compound treatment reduces the number of dysplastic intestinal polyps and lesion area in APC min mice.
  • the APC protein consists of multiple functional domains. The regions between amino acids 1020 – 1169 and 1342 – 2075 are essential for b-catenin binding and degradation [8-10]. Nonsense mutations often result in the synthesis of a truncated APC protein lacking these domains which are unable to mediate b-catenin degradation. [0256] Aminoglycoside and macrolide induced read-through typically results in restoration of 2 - 10% of normal protein levels.
  • macrolide antibiotics target the proximal region of the nascent peptide exit tunnel on the large ribosomal subunit, interfering the peptidyl transferase center and slowing-down synthesis of the nascent polypeptide. This stalling allows for the insertion of a near-cognate tRNA amino acid at the premature termination codon site and continued translation [43, 44].
  • the traditional macrolide antibiotics that have demonstrated read-through activity are not suitable for chronic dosing that would be required to treat FAP patients. Prolonged use of these traditional macrolide antibiotics has the potential to result in cardiac and liver toxicity.
  • APC mutations in sporadic colorectal tumors A mutational "hotspot” and interdependence of the "two hits".
  • 18 Moser AR, Pitot HC, Dove WF. A dominant mutation that predisposes to multiple intestinal neoplasia in the mouse. Science 1990; 247: 322-324. 19 Washington K, Zemper AED. Apc-related models of intestinal neoplasia: a brief review for pathologists. Surgical and Experimental Pathology 2019; 2.
  • Moser AR, Luongo C Gould KA, McNeley MK, Shoemaker AR, Dove WF.
  • ApcMin a mouse model for intestinal and mammary tumorigenesis. Eur J Cancer 1995; 31A: 1061- 1064. 21 Heyer J, Yang K, Lipkin M, Edelmann W, Kucherlapati R. Mouse models for colorectal cancer. Oncogene 1999; 18: 5325-5333. 22 Zhang J, Cao H, Zhang B, Cao H, Xu X, Ruan H et al. Berberine potently attenuates intestinal polyps growth in ApcMin mice and familial adenomatous polyposis patients through inhibition of Wnt signalling. J Cell Mol Med 2013; 17: 1484-1493. 23 Zilberberg A, Lahav L, Rosin-Arbesfeld R.
  • the invention encompasses all variations, combinations, and permutations in which one or more limitations, elements, clauses, and descriptive terms from one or more of the listed claims is introduced into another claim.
  • any claim that is dependent on another claim can be modified to include one or more limitations found in any other claim that is dependent on the same base claim.
  • elements are presented as lists, e.g., in Markush group format, each subgroup of the elements is also disclosed, and any element(s) can be removed from the group.

Landscapes

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

Abstract

L'invention concerne un procédé de traitement de la PAF chez un patient ayant besoin d'un tel traitement, ledit procédé comprenant l'administration au patient d'un composé représenté par la formule I : ou un sel pharmaceutiquement acceptable de celui-ci, formule dans laquelle R9a est choisi dans le groupe constitué par H, alkyle en C1-6, alkylène-OH en C1-6, C1-6alkylène- O-C1-6alkyle, alkyle en C(=O)C 1-6 alkyle, alkylène-cyclolkyl en C1-6, C(=O)cycloalkyle, et C(=O)NH-aryle ; et R10a est un hétéroaryle.
PCT/US2023/031687 2022-09-01 2023-08-31 Traitement de la polypose adénomateuse familiale à l'aide d'un macrolide à 13 chaînons WO2024049994A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202263403130P 2022-09-01 2022-09-01
US63/403,130 2022-09-01

Publications (1)

Publication Number Publication Date
WO2024049994A1 true WO2024049994A1 (fr) 2024-03-07

Family

ID=88192299

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2023/031687 WO2024049994A1 (fr) 2022-09-01 2023-08-31 Traitement de la polypose adénomateuse familiale à l'aide d'un macrolide à 13 chaînons

Country Status (1)

Country Link
WO (1) WO2024049994A1 (fr)

Citations (34)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4270537A (en) 1979-11-19 1981-06-02 Romaine Richard A Automatic hypodermic syringe
US4596556A (en) 1985-03-25 1986-06-24 Bioject, Inc. Hypodermic injection apparatus
US4790824A (en) 1987-06-19 1988-12-13 Bioject, Inc. Non-invasive hypodermic injection device
US4886499A (en) 1986-12-18 1989-12-12 Hoffmann-La Roche Inc. Portable injection appliance
US4940460A (en) 1987-06-19 1990-07-10 Bioject, Inc. Patient-fillable and non-invasive hypodermic injection device assembly
US4941880A (en) 1987-06-19 1990-07-17 Bioject, Inc. Pre-filled ampule and non-invasive hypodermic injection device assembly
US5015235A (en) 1987-02-20 1991-05-14 National Carpet Equipment, Inc. Syringe needle combination
US5064413A (en) 1989-11-09 1991-11-12 Bioject, Inc. Needleless hypodermic injection device
US5141496A (en) 1988-11-03 1992-08-25 Tino Dalto Spring impelled syringe guide with skin penetration depth adjustment
US5190521A (en) 1990-08-22 1993-03-02 Tecnol Medical Products, Inc. Apparatus and method for raising a skin wheal and anesthetizing skin
US5312335A (en) 1989-11-09 1994-05-17 Bioject Inc. Needleless hypodermic injection device
US5328483A (en) 1992-02-27 1994-07-12 Jacoby Richard M Intradermal injection device with medication and needle guard
US5334144A (en) 1992-10-30 1994-08-02 Becton, Dickinson And Company Single use disposable needleless injector
US5339163A (en) 1988-03-16 1994-08-16 Canon Kabushiki Kaisha Automatic exposure control device using plural image plane detection areas
US5383851A (en) 1992-07-24 1995-01-24 Bioject Inc. Needleless hypodermic injection device
US5417662A (en) 1991-09-13 1995-05-23 Pharmacia Ab Injection needle arrangement
US5466220A (en) 1994-03-08 1995-11-14 Bioject, Inc. Drug vial mixing and transfer device
US5480381A (en) 1991-08-23 1996-01-02 Weston Medical Limited Needle-less injector
US5527288A (en) 1990-12-13 1996-06-18 Elan Medical Technologies Limited Intradermal drug delivery device and method for intradermal delivery of drugs
US5569189A (en) 1992-09-28 1996-10-29 Equidyne Systems, Inc. hypodermic jet injector
US5599302A (en) 1995-01-09 1997-02-04 Medi-Ject Corporation Medical injection system and method, gas spring thereof and launching device using gas spring
WO1997013537A1 (fr) 1995-10-10 1997-04-17 Visionary Medical Products Corporation Dispositif d'injection sans aiguille et a gaz comprime
US5649912A (en) 1994-03-07 1997-07-22 Bioject, Inc. Ampule filling device
WO1997037705A1 (fr) 1996-04-11 1997-10-16 Weston Medical Limited Distributeur a usage medical entraine par ressort
US5725871A (en) 1989-08-18 1998-03-10 Danbiosyst Uk Limited Drug delivery compositions comprising lysophosphoglycerolipid
US5756353A (en) 1991-12-17 1998-05-26 The Regents Of The University Of California Expression of cloned genes in the lung by aerosol-and liposome-based delivery
US5780045A (en) 1992-05-18 1998-07-14 Minnesota Mining And Manufacturing Company Transmucosal drug delivery device
US5804212A (en) 1989-11-04 1998-09-08 Danbiosyst Uk Limited Small particle compositions for intranasal drug delivery
US5893397A (en) 1996-01-12 1999-04-13 Bioject Inc. Medication vial/syringe liquid-transfer apparatus
WO1999034850A1 (fr) 1998-01-08 1999-07-15 Fiderm S.R.L. Dispositif de commande de la profondeur de penetration d'une aiguille conçu pour etre utilise avec une seringue d'injection
US5993412A (en) 1997-05-19 1999-11-30 Bioject, Inc. Injection apparatus
US20130090326A1 (en) 2009-10-16 2013-04-11 Rib-X Pharmaceuticals, Inc. Antimicrobial Compounds and Methods of Making and Using the Same
WO2020106627A1 (fr) 2018-11-19 2020-05-28 Zikani Therapeutics, Inc. Macrolides à 13 chaînons à substitution alkylène en c10 et leurs utilisations
WO2022251730A1 (fr) * 2021-05-28 2022-12-01 Zikani Therapeutics, Inc. Composés pour le traitement de maladies génétiques

Patent Citations (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4270537A (en) 1979-11-19 1981-06-02 Romaine Richard A Automatic hypodermic syringe
US4596556A (en) 1985-03-25 1986-06-24 Bioject, Inc. Hypodermic injection apparatus
US4886499A (en) 1986-12-18 1989-12-12 Hoffmann-La Roche Inc. Portable injection appliance
US5015235A (en) 1987-02-20 1991-05-14 National Carpet Equipment, Inc. Syringe needle combination
US4790824A (en) 1987-06-19 1988-12-13 Bioject, Inc. Non-invasive hypodermic injection device
US4940460A (en) 1987-06-19 1990-07-10 Bioject, Inc. Patient-fillable and non-invasive hypodermic injection device assembly
US4941880A (en) 1987-06-19 1990-07-17 Bioject, Inc. Pre-filled ampule and non-invasive hypodermic injection device assembly
US5339163A (en) 1988-03-16 1994-08-16 Canon Kabushiki Kaisha Automatic exposure control device using plural image plane detection areas
US5141496A (en) 1988-11-03 1992-08-25 Tino Dalto Spring impelled syringe guide with skin penetration depth adjustment
US5725871A (en) 1989-08-18 1998-03-10 Danbiosyst Uk Limited Drug delivery compositions comprising lysophosphoglycerolipid
US5804212A (en) 1989-11-04 1998-09-08 Danbiosyst Uk Limited Small particle compositions for intranasal drug delivery
US5503627A (en) 1989-11-09 1996-04-02 Bioject, Inc. Ampule for needleless injection
US5064413A (en) 1989-11-09 1991-11-12 Bioject, Inc. Needleless hypodermic injection device
US5312335A (en) 1989-11-09 1994-05-17 Bioject Inc. Needleless hypodermic injection device
US5190521A (en) 1990-08-22 1993-03-02 Tecnol Medical Products, Inc. Apparatus and method for raising a skin wheal and anesthetizing skin
US5527288A (en) 1990-12-13 1996-06-18 Elan Medical Technologies Limited Intradermal drug delivery device and method for intradermal delivery of drugs
US5480381A (en) 1991-08-23 1996-01-02 Weston Medical Limited Needle-less injector
US5417662A (en) 1991-09-13 1995-05-23 Pharmacia Ab Injection needle arrangement
US5756353A (en) 1991-12-17 1998-05-26 The Regents Of The University Of California Expression of cloned genes in the lung by aerosol-and liposome-based delivery
US5328483A (en) 1992-02-27 1994-07-12 Jacoby Richard M Intradermal injection device with medication and needle guard
US5780045A (en) 1992-05-18 1998-07-14 Minnesota Mining And Manufacturing Company Transmucosal drug delivery device
US5520639A (en) 1992-07-24 1996-05-28 Bioject, Inc. Needleless hypodermic injection methods and device
US5383851A (en) 1992-07-24 1995-01-24 Bioject Inc. Needleless hypodermic injection device
US5704911A (en) 1992-09-28 1998-01-06 Equidyne Systems, Inc. Needleless hypodermic jet injector
US5569189A (en) 1992-09-28 1996-10-29 Equidyne Systems, Inc. hypodermic jet injector
US5334144A (en) 1992-10-30 1994-08-02 Becton, Dickinson And Company Single use disposable needleless injector
US5649912A (en) 1994-03-07 1997-07-22 Bioject, Inc. Ampule filling device
US5466220A (en) 1994-03-08 1995-11-14 Bioject, Inc. Drug vial mixing and transfer device
US5599302A (en) 1995-01-09 1997-02-04 Medi-Ject Corporation Medical injection system and method, gas spring thereof and launching device using gas spring
WO1997013537A1 (fr) 1995-10-10 1997-04-17 Visionary Medical Products Corporation Dispositif d'injection sans aiguille et a gaz comprime
US5893397A (en) 1996-01-12 1999-04-13 Bioject Inc. Medication vial/syringe liquid-transfer apparatus
WO1997037705A1 (fr) 1996-04-11 1997-10-16 Weston Medical Limited Distributeur a usage medical entraine par ressort
US5993412A (en) 1997-05-19 1999-11-30 Bioject, Inc. Injection apparatus
WO1999034850A1 (fr) 1998-01-08 1999-07-15 Fiderm S.R.L. Dispositif de commande de la profondeur de penetration d'une aiguille conçu pour etre utilise avec une seringue d'injection
US20130090326A1 (en) 2009-10-16 2013-04-11 Rib-X Pharmaceuticals, Inc. Antimicrobial Compounds and Methods of Making and Using the Same
WO2020106627A1 (fr) 2018-11-19 2020-05-28 Zikani Therapeutics, Inc. Macrolides à 13 chaînons à substitution alkylène en c10 et leurs utilisations
WO2022251730A1 (fr) * 2021-05-28 2022-12-01 Zikani Therapeutics, Inc. Composés pour le traitement de maladies génétiques

Non-Patent Citations (51)

* Cited by examiner, † Cited by third party
Title
BEROUD CSOUSSI T: "APC gene: database of germline and somatic mutations in human tumors and cell lines", NUCLEIC ACIDS RES, vol. 24, 1996, pages 121 - 124
BIAN JDANNAPPEL MWAN CFIRESTEIN R: "Transcriptional Regulation of Wnt/beta-Catenin Pathway in Colorectal Cancer", CELLS, vol. 9, 2020
BIDOU LBUGAUD OBELAKHOV VBAASOV TNAMY O: "Characterization of new-generation aminoglycoside promoting premature termination codon readthrough in cancer cells", RNA BIOL, vol. 14, 2017, pages 378 - 388
BRINKER AD, WASSEL RT, LYNDLY J, SERRANO J, AVIGAN M, LEE WM: "Telithromycin-associated hepatotoxicity: Clinical spectrum and causality assessment of 42 cases", HEPATOLOGY, vol. 49, 2009, pages 250 - 257
CASPI MFIRSOW ARAJKUMAR RSKALKA NMOSHKOVITZ IMUNITZ A ET AL.: "A flow cytometry-based reporter assay identifies macrolide antibiotics as nonsense mutation read-through agents", J MOL MED (BERL, vol. 94, 2016, pages 469 - 482, XP035801357, DOI: 10.1007/s00109-015-1364-1
CASPI MICHAL ET AL: "A flow cytometry-based reporter assay identifies macrolide antibiotics as nonsense mutation read-through agents", JOURNAL OF MOLECULAR MEDICINE, SPRINGER BERLIN HEIDELBERG, BERLIN/HEIDELBERG, vol. 94, no. 4, 1 December 2015 (2015-12-01), pages 469 - 482, XP035801357, ISSN: 0946-2716, [retrieved on 20151201], DOI: 10.1007/S00109-015-1364-1 *
CHLIPALA EBENDZINSKI CMCHU KJOHNSON IIBROUS MCOPELAND K ET AL.: "Optical density-based image analysis method for the evaluation of hematoxylin and eosin staining precision", JHISTOTECHNOL, vol. 43, 2020, pages 29 - 37
DABROWSKI MACIEJ ET AL: "Advances in therapeutic use of a drug-stimulated translational readthrough of premature termination codons", vol. 24, no. 1, 1 December 2018 (2018-12-01), Washington , DC, XP055954670, ISSN: 1076-1551, Retrieved from the Internet <URL:https://molmed.biomedcentral.com/track/pdf/10.1186/s10020-018-0024-7.pdf> DOI: 10.1186/s10020-018-0024-7 *
DAVIES MLROBERTS GTSTUART NWAKEMAN JA: "Analysis of a panel of antibodies to APC reveals consistent activity towards an unidentified protein", BR J CANCER, vol. 97, 2007, pages 384 - 390, XP093024408, DOI: 10.1038/sj.bjc.6603873
DE QUEIROZ ROSSANESE LBDE LIMA MARSON FARIBEIRO JDCOY CSBERTUZZO CS: "APC germline mutations in families with familial adenomatous polyposis", ONCOL REP, vol. 30, 2013, pages 2081 - 2088
FEARNHEAD NSBRITTON MPBODMER WF: "The ABC of APC", HUM MOL GENET, vol. 10, 2001, pages 721 - 733, XP002250494, DOI: 10.1093/hmg/10.7.721
FLOQUET C, ROUSSET JP, BIDOU L.: "Readthrough of premature termination codons in the adenomatous polyposis coli gene restores its biological activity in human cancer cells", PLOS ONE, vol. 6, 2011, pages e24125, XP055417114, DOI: 10.1371/journal.pone.0024125
FREDERICKS EDEALTRY GROUX S: "beta-Catenin Regulation in Sporadic Colorectal Carcinogenesis: Not as Simple as APC", CAN JGASTROENTEROLHEPATOL, vol. 2018, 2018, pages 4379673
GALIATSATOS PFOULKES WD: "Familial adenomatous polyposis", AM J GASTROENTEROL, vol. 101, 2006, pages 385 - 398
GRAF MARTIN R. ET AL: "Novel read through agent: ZKN-0013 demonstrates efficacy in APCmin model of familial adenomatous polyposis", JOURNAL OF MOLECULAR MEDICINE, vol. 101, no. 4, 20 February 2023 (2023-02-20), Berlin/Heidelberg, pages 375 - 385, XP093098350, ISSN: 0946-2716, Retrieved from the Internet <URL:https://link.springer.com/article/10.1007/s00109-023-02291-x/fulltext.html> DOI: 10.1007/s00109-023-02291-x *
GUO DCAI YCHAI DLIANG BBAI NWANG R: "The cardiotoxicity of macrolides: a systematic review", PHARMAZIE, vol. 65, 2010, pages 631 - 640
HAJIMIRZAEI NKHALILI NPBOROUMAND BSAFARI FPOURHOSSEINI AJUDI-CHELAN R ET AL.: "Comparative Study of the Effect of Macrolide Antibiotics Erythromycin, Clarithromycin, and Azithromycin on the ERG1 Gene Expression in H9c2 Cardiomyoblast Cells", DRUG RES (STUTTG), vol. 70, 2020, pages 341 - 347
HALF EBERCOVICH DROZEN P: "Familial adenomatous polyposis", ORPHANET J RARE DIS, vol. 4, no. 22, 2009
HANKEY WFRANKEL WLGRODEN J: "Functions of the APC tumor suppressor protein dependent and independent of canonical WNT signaling: implications for therapeutic targeting", CANCER METASTASIS REV, vol. 37, 2018, pages 159 - 172, XP036419749, DOI: 10.1007/s10555-017-9725-6
HE TCSPARKS ABRAGO CHERMEKING HZAWEL LDA COSTA LT ET AL.: "Identification of c-MYC as a target of the APC pathway", SCIENCE, vol. 281, 1998, pages 1509 - 1512
HEYER JYANG KLIPKIN MEDELMANN W: "Kucherlapati R. Mouse models for colorectal cancer", ONCOGENE, vol. 18, 1999, pages 5325 - 5333
HORAI YAKATSUKA AMIZUKAWA MNIISHINA HNISHIKAWA SONO Y ET AL.: "Current status and prospects for quantitative analysis of digital image of pathological specimen using image processing software including artificial intelligence", TRANSLATIONAL AND, vol. 2, 2020, pages 72 - 79
HUI-LING ROSE LEE ET AL: "Pharmaceutical therapies to recode nonsense mutations in inherited diseases", PHARMACOLOGY & THERAPEUTICS, vol. 136, no. 2, 1 November 2012 (2012-11-01), pages 227 - 266, XP055068955, ISSN: 0163-7258, DOI: 10.1016/j.pharmthera.2012.07.007 *
KARIV RCASPI MFLISS-ISAKOV NSHORER YSHOR YROSNER G ET AL.: "Resorting the function of the colorectal cancer gatekeeper adenomatous polyposis coli", INT J CANCER, vol. 146, 2020, pages 1064 - 1074, XP071291817, DOI: 10.1002/ijc.32557
KEELING KMLANIER JDU MSALAS-MARCO JGAO LKAENJAK-ANGELETTI A ET AL.: "Leaky termination at premature stop codons antagonizes nonsense-mediated mRNA decay in S. cerevisiae", RNA, vol. 10, 2004, pages 691 - 703
KETTUNEN HLKETTUNEN ASRAUTONEN NE: "Intestinal immune responses in wild-type and Apcmin/+ mouse, a model for colon cancer", CANCER RES, vol. 63, 2003, pages 5136 - 5142
KIRST HASIDES GD: "New directions for macrolide antibiotics: pharmacokinetics and clinical efficacy", ANTIMICROB AGENTS CHEMOTHER, vol. 33, 1989, pages 1419 - 1422, XP001080742
KOLLIGS FTBOMMER GGOKE B: "Wnt/beta-catenin/tcf signaling: a critical pathway in gastrointestinal tumorigenesis", DIGESTION, vol. 66, 2002, pages 131 - 144
LECARPENTIER YSCHUSSLER OHEBERT JLVALLEE A: "Multiple Targets of the Canonical WNT/beta-Catenin Signaling in Cancers", FRONT ONCOL, vol. 9, 2019, pages 1248
LEE HLDOUGHERTY JP: "Pharmaceutical therapies to recode nonsense mutations in inherited diseases", PHARMACOL THER, vol. 136, 2012, pages 227 - 266, XP055068955, DOI: 10.1016/j.pharmthera.2012.07.007
LEOZ ML, CARBALLAL S, MOREIRA L, OCANA T, BALAGUER F.: " The genetic basis of familial adenomatous polyposis and its implications for clinical practice and risk management", APPL CLIN GENET, vol. 8, 2015, pages 95 - 107
LI XWANG MLIU GZHOU LWANG ZLI C: "Macrolides use and the risk of sudden cardiac death", EXPERT REV ANTI INFECT THER, vol. 14, 2016, pages 535 - 537
MCCAUGHAN KK, BROWN CM, DALPHIN ME, BERRY MJ, TATE WP.: "Translational termination efficiency in mammals is influenced by the base following the stop codon.", PROC NATL ACAD SET USA, vol. 92, 1995, pages 5431 - 5435
MORGAN RETRAUNER MVAN STADEN CJLEE PHRAMACHANDRAN BESCHENBERG M ET AL.: "Interference with bile salt export pump function is a susceptibility factor for human liver injury in drug development", TOXICOLSCI, vol. 118, 2010, pages 485 - 500
MOSER ARLUONGO CGOULD KAMCNELEY MKSHOEMAKER ARDOVE WF: "ApcMin: a mouse model for intestinal and mammary tumorigenesis", EUR J CANCER, vol. 31A, 1995, pages 1061 - 1064
MOSER ARPITOT HCDOVE WF: "A dominant mutation that predisposes to multiple intestinal neoplasia in the mouse", SCIENCE, vol. 247, 1990, pages 322 - 324, XP002973456, DOI: 10.1126/science.2296722
PAI SGCARNEIRO BAMOTA JMCOSTA RLEITE CABARROSO-SOUSA R ET AL.: "Wnt/beta-catenin pathway: modulating anticancer immune response", J HEMATOL ONCOL, vol. 10, 2017, pages 101, XP055702542, DOI: 10.1186/s13045-017-0471-6
POLAKIS P: "Mutations in the APC gene and their implications for protein structure and function", CURR OPIN GENET DEV, vol. 5, 1995, pages 66 - 71, XP004556369, DOI: 10.1016/S0959-437X(95)90055-1
RIPA RBISGAARD MLBULOW SNIELSEN FC: "De novo mutations in familial adenomatous polyposis (FAP", EUR J HUM GENET, vol. 10, 2002, pages 631 - 637
ROWAN AJLAMLUM HILYAS MWHEELER JSTRAUB JPAPADOPOULOU A ET AL.: "APC mutations in sporadic colorectal tumors: A mutational ''hotspot'' and interdependence of the ''two hits", PROC NATL ACAD SCI USA, vol. 97, 2000, pages 3352 - 3357
S. M. BERGE ET AL.: "Pharmaceutical Salts", J. PHARM. SCI., vol. 66, 1977, pages 1 - 19, XP002675560, DOI: 10.1002/jps.2600660104
SEGDITSAS S, TOMLINSON I.: "Colorectal cancer and genetic alterations in the Wnt pathway.", ONCOGENE, vol. 25, 2006, pages 7531 - 7537, XP037741719, DOI: 10.1038/sj.onc.1210059
SENDA TSHIMOMURA AIIZUKA-KOGO A: "Adenomatous polyposis coli (Apc) tumor suppressor gene as a multifunctional gene", ANAT SCI INT, vol. 80, 2005, pages 121 - 131
SOTHISELVAM SNEUNER SRIGGER LKLEPACKI DMICURA RVAZQUEZ-LASLOP N ET AL.: "Binding of Macrolide Antibiotics Leads to Ribosomal Selection against Specific Substrates Based on Their Charge and Size", CELL REP, vol. 16, 2016, pages 1789 - 1799
STAMOS JLWEIS WI: "The beta-catenin destruction complex", COLD SPRING HARB PERSPECT, vol. 5, 2013, pages a007898
TIAN XLIU ZNIU BZHANG JTAN TKLEE SR ET AL.: "E-cadherin/beta-catenin complex and the epithelial barrier", J BIOMED BIOTECHNOL, vol. 2011, 2011, pages 567305
VAZQUEZ-LASLOP NMANKIN AS: "How Macrolide Antibiotics Work", TRENDS BIOCHEM, vol. 43, 2018, pages 668 - 684, XP085447985, DOI: 10.1016/j.tibs.2018.06.011
WASHINGTON K: "Zemper AED. Apc-related models of intestinal neoplasia: a brief review for pathologists", SURGICAL AND EXPERIMENTAL PATHOLOGY, vol. 2, 2019
WOODHEAD JLYANG KOLDACH DMACLAUCHLIN CFERNANDES PWATKINS PB ET AL.: "Analyzing the Mechanisms Behind Macrolide Antibiotic-Induced Liver Injury Using Quantitative Systems Toxicology Modeling", PHARM RES, vol. 36, no. 48, 2019
ZHANG JCAO HZHANG BCAO HXU XRUAN H ET AL.: "Berberine potently attenuates intestinal polyps growth in ApcMin mice and familial adenomatous polyposis patients through inhibition of Wnt signalling", J CELL MOL MED, vol. 17, 2013, pages 1484 - 1493
ZILBERBERG ALAHAV LROSIN-ARBESFELD R: "Restoration of APC gene function in colorectal cancer cells by aminoglycoside- and macrolide-induced read-through of premature termination codons", GUT, vol. 59, 2010, pages 496 - 507, XP055417112, DOI: 10.1136/gut.2008.169805

Similar Documents

Publication Publication Date Title
US10577366B2 (en) Crystalline forms of a compound that inhibits bromodomain
KR100829875B1 (ko) 항암제로서의 병용 의약 조성물
AU2018208231B2 (en) Imidazopyrazine compound, preparation method therefor and use thereof
AU2007352397B2 (en) Viral inhibitors
WO2023020519A9 (fr) Dérivés de 1, 4-oxazépane et leurs utilisations
ES2694297T3 (es) Formas cristalinas de ácido {[1-ciano-5-(4-clorofenoxi)-4-hidroxi-isoquinolina-3-carbonilo]-amino}-acético
US20220048922A1 (en) Compounds targeting prmt5
CN113924146A (zh) 用于治疗疾病的新化合物及其药物组合物
US9884874B2 (en) Bromodomain-inhibiting compounds and pharmaceutical composition comprising same for preventing or treating a cancer
US7884099B2 (en) Quinolone carboxylic acid-substituted rifamycin derivatives
US20230055237A1 (en) Compounds and methods for the treatment of cystic fibrosis
CN117580839A (zh) 抗癌核激素受体靶向化合物
WO2020156189A1 (fr) Dérivé de camptothécine et promédicament soluble dans l&#39;eau à base de celui-ci, composition pharmaceutique le contenant, procédé de préparation et utilisation
WO2022002142A1 (fr) Composés de tétrahydroisoquinoline et leur utilisation
WO2022089389A1 (fr) Composé hétérocyclique, procédé de préparation s&#39;y rapportant, composition pharmaceutique associée et application associée
TW202043223A (zh) 做為腺苷受體拮抗劑之5-氮雜吲唑衍生物
WO2021155288A1 (fr) Thérapie antifibrotique dirigée sur une protéine d&#39;activation des fibroblastes (fap)
WO2024009191A1 (fr) Composés pyrido[4,3-d]pyrimidines
WO2024049994A1 (fr) Traitement de la polypose adénomateuse familiale à l&#39;aide d&#39;un macrolide à 13 chaînons
WO2021076124A1 (fr) Utilisation d&#39;inhibiteurs de jak1 pour le traitement du lupus érythémateux cutané et du lichen plan (l.p.)
CN114728938B (zh) 作为prmt5抑制剂的四氢异喹啉螺环化合物
WO2022078462A1 (fr) Nouveaux analogues de glutamine
CN108358953B (zh) 铜与咪唑并吡啶类化合物的配合物及其应用
EP3004099B1 (fr) Dérivés de tétrahydropyridine servant d&#39;inhibiteurs de fabi
WO2022135390A1 (fr) Inhibiteur de cétohexokinase et son utilisation

Legal Events

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

Ref document number: 23776761

Country of ref document: EP

Kind code of ref document: A1