WO2023097251A1 - Chimères ciblant le facteur de transcription et contenant des oligonucléotides - Google Patents

Chimères ciblant le facteur de transcription et contenant des oligonucléotides Download PDF

Info

Publication number
WO2023097251A1
WO2023097251A1 PCT/US2022/080390 US2022080390W WO2023097251A1 WO 2023097251 A1 WO2023097251 A1 WO 2023097251A1 US 2022080390 W US2022080390 W US 2022080390W WO 2023097251 A1 WO2023097251 A1 WO 2023097251A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
occurrence
group
independently
ona
Prior art date
Application number
PCT/US2022/080390
Other languages
English (en)
Inventor
Craig Crews
Kusal SAMARASINGHE
Ling Chu
Original Assignee
Yale University
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 Yale University filed Critical Yale University
Publication of WO2023097251A1 publication Critical patent/WO2023097251A1/fr

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/55Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound the modifying agent being also a pharmacologically or therapeutically active agent, i.e. the entire conjugate being a codrug, i.e. a dimer, oligomer or polymer of pharmacologically or therapeutically active compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/549Sugars, nucleosides, nucleotides or nucleic acids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/46Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
    • C07K14/47Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/1025Acyltransferases (2.3)
    • C12N9/104Aminoacyltransferases (2.3.2)

Definitions

  • PROTACs are small molecule-based heterobifunctional molecules that recruit an E3 ligase complex to a protein of interest. By doing so, PROTACs induce the ubiquitination and degradation of the protein of interest via the proteasome. Although PROTACs have the potential to induce the degradation of numerous proteins, the identification of small molecule recruiting ligands for several classes of proteins is still challenging and time-consuming. Therefore, the development of alternative, proximity- inducing strategies would help to target disease-causing proteins, such as transcription factors.
  • TFs Transcription factors
  • Many diseases such as neurological disorders, autoimmunity, developmental syndromes, and many cancers result from abnormalities in the TF-controlled gene-regulatory circuitry within the diseased cell.
  • the TF c-Myc the most frequently amplified oncogene, has been extensively studied and established as a direct mediator of tumorigenesis in numerous cancers.
  • indirect approaches such as bromodomain protein inhibitors and PROTACs have been explored to control c-Myc levels, direct inhibition or degradation of c-Myc has been an as yet unrealized goal.
  • T-box transcription factor In contrast to c-Myc, T-box transcription factor (“brachyury”) is expressed only in a restricted set of cancer types and minimally expressed in normal cells.
  • Brachyury is a master developmental TF that plays a pivotal role during early embryonic development in vertebrates. Brachyury expression is limited only to embryonic developmental stages and, in general, expression levels are highly downregulated in adult tissues. In addition to its regulatory functions during development, brachyury expression in remnant notochord cells in adults has been shown to be a key oncogenic driver in the rare bone cancer, chordoma. Chromosomal aberrations such as chromosome 6 gains and partial polysomy have been identified as potential molecular mechanisms in brachyury-dependent chordoma tumors.
  • TFs such as NF-kB, STAT3/5, the androgen receptor (AR) and the estrogen receptor (ER) are other known oncogenic drivers that also rewire transcriptional circuitry in various cancer types.
  • AR androgen receptor
  • ER estrogen receptor
  • ONA is an oligonucleotide having a 3'-end and a 5'-end and comprising one or more phosphodiester or phosphorothioate internucleotide linkages
  • LNK is a chemical linker covalently bonding the ONA and the UBL
  • UBL is an E3 ubiquitin ligase ligand.
  • a compound of formula II, or a salt, enantiomer, diastereomer, or tautomer thereof is provided: wherein: ONA is an oligonucleotide having a 3'-end and a 5'-end; LNK is a chemical linker covalently bonding the ONA and the C ⁇ C ⁇ H.
  • Compounds of formula I are useful in preventing, treating, and/or ameliorating cancer in subjects, including human subjects.
  • a method of preventing, treating, and/or ameliorating cancer in a subject is provided. The method includes administering to the subject a therapeutically effective amount of at least one compound of formula I, which is optionally formulated as a pharmaceutical composition further comprising at least one pharmaceutically acceptable carrier.
  • FIG.1 is a schematic representation of oligoTRAFTAC-mediated transcription factor (TF) and E3 ligase recruitment, and proximity-dependent TF ubiquitination, in accordance with various embodiments.
  • FIGs.2A-2E illustrate show activity and aspects of oligoTRAFTACs, according to various embodiments. OligoTRAFTAC induces c-Myc degradation. (FIG.2A) The oligonucleotide selected for the c-Myc oligoTRAFTAC engages c-Myc.
  • FIG.2C OligoTRAFTAC-induced c-Myc degradation occurred via the proteasomal pathway.
  • HEK293T cells were treated with c-Myc-targeting oligoTRAFTAC with and without the neddylation inhibitor, MLN-4924 (1 mM), and then analyzed for c-Myc levels.
  • FIG.2D HEK293 cells were preincubated with and without 10 mM VHL ligand followed by OT7 transfection and analyzed for c-Myc levels.
  • FIG.2E Chemical structure of a c-Myc- targeting oligoTRAFTAC (OT7), according to various embodiments.
  • FIGs.3A-3E show brachyury-GFP degradation by oligoTRAFTACs.
  • FIG.3A Brachyury-targeting oligonucleotide used in the oligoTRAFTAC design engaged with brachyury-GFP.
  • BRCH biotinylated oligonucleotide
  • SCRM scrambled oligonucleotide
  • FIG.3D Washout experiment after 12 h of OT3 transfection. Cells were incubated continuously for 24 h in transfection medium or OT3 was aspirated after 12 h of transfection and fresh medium added to cells. Washout cells incubated for another 12 h and 24 h prior to harvesting.
  • FIGs.4A-4D show oligoTRAFTACs induce brachyury-GFP degradation via the proteasomal pathway.
  • FIG.4B OT3 induced brachyury degradation is VHL- dependent.
  • HEK293T cells were preincubated with and without 10 mM of VHL ligand for 1.5 h prior to OT3 transfection. After 20 h of transfection, cells lysates were prepared and analyzed for brachyury degradation.
  • FIGs.5A-5F show endogenous brachyury degradation by oligoTRAFTACs constructed with phosphorothioate backbone.
  • FIG.5C UM-Chor1 cells were transfected with 60 nM of OT17 and harvested at subsequent different time points as indicated.
  • FIG.5F OT3- and OT17-induced brachyury ubiquitination.
  • FIGs.6A-6D show microinjection of brachyury-targeting oligoTRAFTAC into zebrafish embryos: demonstration of in vivo activity. (FIG.
  • FIG.6A Schematic representation of OT17 and OT20 microinjection into zebrafish embryos.
  • FIGs.7A-7C show the oligonucleotide synthesis and click reaction. Oligonucleotide synthesis and click reaction.
  • FIG.7A Oligonucleotides were custom synthesized with a terminal alkyne either at 3' or 5' end of the oligo. Oligonucleotide sequences for both 3' and 5' alkyne targeting c-Myc (left panel) and brachyury (right panel).
  • FIG.7B Chemical structure of azido-VHL ligand and click reaction conditions.
  • FIG.7C Chemical structures of 3' modified oligonucleotides (OT17- with a phosphorothioate backbone) after the click reaction with azido-VHL ligand.
  • FIGs.8A-8G show oligoTRAFTAC mediated c-Myc degradation.
  • FIG.8B Varying concentrations of OT7 were transfected into HEK293T cells and lysed after 20 h. Cell lysates were separated and transferred to a PVDF membrane followed by immunoblotting with antibodies against c-Myc and GAPDH.
  • FIG.8F Degradation of E-box binding transcription factor, TCF3.
  • OT7 transfected cells were analyzed for c-Myc and TCF3.
  • FIGs.9A-9B show EMSA and brachyury-GFP degradation data.
  • FIG.9A shows click reaction mixtures with or without VHL ligand were loaded on to a 1.2% agarose gel and separated over 1 h at constant 120 mV.
  • FIG.9B shows that OT1 through 4 were transfected into HEK293T cells overexpressing brachyury-GFP and lysed after 30 h.
  • FIGs.10A-10B show time course and washout experiments for brachyury targeting OTs.
  • FIG.10A shows OT2 through 4 were transfected at 75 nM into HEK293T cells overexpressing brachyury-GFP and lysed after 12 h, 24 h, and 36 h. Lysates were probed for brachyury and GAPDH.
  • FIG.10B shows OT3 was transfected into HEK293T cells and washed out after 6 h and 12 h. Cell lysates were probed with antibodies against brachyury and GAPDH.
  • FIGs.11A-11C show brachyury degradation by oligoTRAFTACs in HEK293T and UM-Chor1 cells.
  • FIG.11C Proteasome dependent brachyury degradation in UM-Chor1 cells.
  • DETAILED DESCRIPTION OF THE INVENTION Reference will now be made in detail to certain embodiments of the disclosed subject matter. While the disclosed subject matter will be described in conjunction with the enumerated claims, it will be understood that the exemplified subject matter is not intended to limit the claims to the disclosed subject matter.
  • values expressed in a range format should be interpreted in a flexible manner to include not only the numerical values explicitly recited as the limits of the range, but also to include all the individual numerical values or sub-ranges encompassed within that range as if each numerical value and sub-range is explicitly recited.
  • a range of "about 0.1% to about 5%” or "about 0.1% to 5%” should be interpreted to include not just about 0.1% to about 5%, but also the individual values (e.g., 1%, 2%, 3%, and 4%) and the sub-ranges (e.g., 0.1% to 0.5%, 1.1% to 2.2%, 3.3% to 4.4%) within the indicated range.
  • substantially refers to a majority of, or mostly, as in at least about 50%, 60%, 70%, 80%, 90%, 95%, 96%, 97%, 98%, 99%, 99.5%, 99.9%, 99.99%, or at least about 99.999% or more, or 100%.
  • substantially free of can mean having none or having a trivial amount of, such that the amount of material present does not affect the material properties of the composition including the material, such that the composition is about 0 wt% to about 5 wt% of the material, or about 0 wt% to about 1 wt%, or about 5 wt% or less, or less than, equal to, or greater than about 4.5 wt%, 4, 3.5, 3, 2.5, 2, 1.5, 1, 0.9, 0.8, 0.7, 0.6, 0.5, 0.4, 0.3, 0.2, 0.1, 0.01, or about 0.001 wt% or less.
  • substantially free of can mean having a trivial amount of, such that a composition is about 0 wt% to about 5 wt% of the material, or about 0 wt% to about 1 wt%, or about 5 wt% or less, or less than, equal to, or greater than about 4.5 wt%, 4, 3.5, 3, 2.5, 2, 1.5, 1, 0.9, 0.8, 0.7, 0.6, 0.5, 0.4, 0.3, 0.2, 0.1, 0.01, or about 0.001 wt% or less, or about 0 wt%.
  • organic group refers to any carbon-containing functional group.
  • Examples can include an oxygen-containing group such as an alkoxy group, aryloxy group, aralkyloxy group, oxo(carbonyl) group; a carboxyl group including a carboxylic acid, carboxylate, and a carboxylate ester; a sulfur-containing group such as an alkyl and aryl sulfide group; and other heteroatom-containing groups.
  • an oxygen-containing group such as an alkoxy group, aryloxy group, aralkyloxy group, oxo(carbonyl) group
  • a carboxyl group including a carboxylic acid, carboxylate, and a carboxylate ester such as an alkyl and aryl sulfide group
  • sulfur-containing group such as an alkyl and aryl sulfide group
  • Non-limiting examples of organic groups include OR, OOR, OC(O)N(R) 2 , CN, CF 3 , OCF 3 , R, C(O), methylenedioxy, ethylenedioxy, N(R) 2 , SR, SOR, SO 2 R, SO 2 N(R) 2 , SO 3 R, C(O)R, C(O)C(O)R, C(O)CH 2 C(O)R, C(S)R, C(O)OR, OC(O)R, C(O)N(R) 2 , OC(O)N(R) 2 , C(S)N(R) 2 , (CH 2 )0- 2 N(R)C(O)R, (CH 2 ) 0-2 N(R)N(R) 2 , N(R)N(R)C(O)R, N(R)N(R)C(O)OR, N(R)N(R)CON(R) 2 , N(R)SO 2 R,
  • substituted refers to the state in which one or more hydrogen atoms contained therein are replaced by one or more non-hydrogen atoms.
  • the substitution can be direct substitution, whereby the hydrogen atom is replaced by a functional group or substituent, or an indirect substitution, whereby an intervening linker group replaces the hydrogen atom, and the substituent or functional group is bonded to the intervening linker group.
  • direct substitution is: RR-H ⁇ RR-Cl, wherein RR is an organic moiety/fragment/molecule.
  • a non-limiting example of indirect substitution is: RR-H ⁇ RR- (LL) zz -Cl, wherein RR is an organic moiety/fragment/molecule, LL is an intervening linker group, and 'zz' is an integer from 0 to 100 inclusive. When zz is 0, LL is absent, and direct substitution results.
  • (LL) zz can be lin -ear, branched, cyclic, acyclic, and combinations thereof. In certain embodiments, two consecutive LL's cannot be both O.
  • a halogen e.g., F, Cl, Br, and I
  • an oxygen atom in groups such as hydroxy groups, alkoxy groups, aryloxy groups, aralkyloxy groups, oxo(carbonyl) groups, carboxyl groups including carboxylic acids, carboxylates, and carboxylate esters
  • a sulfur atom in groups such as thiol groups, alkyl and aryl sulfide groups, sulfoxide groups, sulfone groups, sulfonyl groups, and sulfonamide groups
  • a nitrogen atom in groups such as amines, hydroxyamines, nitriles, nitro groups, N-oxides, hydrazides, azides, and enamines; and other hetero
  • Non-limiting examples of substituents that can be bonded to a substituted carbon (or other) atom include F, Cl, Br, I, OR, OC(O)N(R) 2 , CN, NO, NO 2 , ONO 2 , azido, CF 3 , OCF 3 , R, O (oxo), S (thiono), C(O), S(O), methylenedioxy, ethylenedioxy, N(R) 2 , SR, SOR, SO 2 R, SO 2 N(R) 2 , SO 3 R, C(O)R, C(O)C(O)R, C(O)CH 2 C(O)R, C(S)R, C(O)OR, OC(O)R, C(O)N(R) 2 , OC(O)N(R) 2 , C(S)N(R) 2 , (CH 2 ) 0-2 N(R)C(O)R, (CH 2 )N(R)N(R) 2
  • alkyl refers to straight chain and branched alkyl groups and cycloalkyl groups having from 1 to 40 carbon atoms, 1 to about 20 carbon atoms, 1 to 12 carbons or, in some embodiments, from 1 to 8 carbon atoms.
  • straight chain alkyl groups include those with from 1 to 8 carbon atoms such as methyl, ethyl, n-propyl, n-butyl, n-pentyl, n-hexyl, n-heptyl, and n-octyl groups.
  • branched alkyl groups include, but are not limited to, isopropyl, iso-butyl, sec-butyl, t-butyl, neopentyl, isopentyl, and 2,2- dimethylpropyl groups.
  • alkyl encompasses n-alkyl, isoalkyl, and anteisoalkyl groups as well as other branched chain forms of alkyl.
  • Representative substituted alkyl groups can be substituted one or more times with any of the groups listed herein, for example, amino, hydroxy, cyano, carboxy, nitro, thio, alkoxy, and halogen groups.
  • alkenyl refers to straight and branched chain and cyclic alkyl groups as defined herein, except that at least one double bond exists between two carbon atoms.
  • alkenyl groups have from 2 to 40 carbon atoms, or 2 to about 20 carbon atoms, or 2 to 12 carbon atoms or, in some embodiments, from 2 to 8 carbon atoms.
  • alkynyl refers to straight and branched chain alkyl groups, except that at least one triple bond exists between two carbon atoms.
  • alkynyl groups have from 2 to 40 carbon atoms, 2 to about 20 carbon atoms, or from 2 to 12 carbons or, in some embodiments, from 2 to 8 carbon atoms. Examples include, but are not limited to – C ⁇ CH, -C ⁇ C(CH 3 ), -C ⁇ C(CH 2 CH 3 ), -CH 2 C ⁇ CH, -CH 2 C ⁇ C(CH 3 ), and -CH 2 C ⁇ C(CH 2 CH 3 ) among others.
  • acyl refers to a group containing a carbonyl moiety wherein the group is bonded via the carbonyl carbon atom.
  • the carbonyl carbon atom is bonded to a hydrogen forming a "formyl" group or is bonded to another carbon atom, which can be part of an alkyl, aryl, aralkyl cycloalkyl, cycloalkylalkyl, heterocyclyl, heterocyclylalkyl, heteroaryl, heteroarylalkyl group or the like.
  • An acyl group can include 0 to about 12, 0 to about 20, or 0 to about 40 additional carbon atoms bonded to the carbonyl group.
  • An acyl group can include double or triple bonds within the meaning herein.
  • An acryloyl group is an example of an acyl group.
  • An acyl group can also include heteroatoms within the meaning herein.
  • a nicotinoyl group (pyridyl-3-carbonyl) is an example of an acyl group within the meaning herein.
  • Other examples include acetyl, benzoyl, phenylacetyl, pyridylacetyl, cinnamoyl, and acryloyl groups and the like.
  • the group containing the carbon atom that is bonded to the carbonyl carbon atom contains a halogen
  • the group is termed a "haloacyl” group.
  • An example is a trifluoroacetyl group.
  • cycloalkyl refers to cyclic alkyl groups such as, but not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, and cyclooctyl groups.
  • the cycloalkyl group can have 3 to about 8-12 ring members, whereas in other embodiments the number of ring carbon atoms range from 3 to 4, 5, 6, or 7.
  • Cycloalkyl groups further include polycyclic cycloalkyl groups such as, but not limited to, norbornyl, adamantyl, bornyl, camphenyl, isocamphenyl, and carenyl groups, and fused rings such as, but not limited to, decalinyl, and the like. Cycloalkyl groups also include rings that are substituted with straight or branched chain alkyl groups as defined herein.
  • Representative substituted cycloalkyl groups can be mono-substituted or substituted more than once, such as, but not limited to, 2,2-, 2,3-, 2,4- 2,5- or 2,6-disubstituted cyclohexyl groups or mono-, di- or tri-substituted norbornyl or cycloheptyl groups, which can be substituted with, for example, amino, hydroxy, cyano, carboxy, nitro, thio, alkoxy, and halogen groups.
  • cycloalkenyl alone or in combination denotes a cyclic alkenyl group.
  • heterocycloalkyl refers to a cycloalkyl group as defined herein in which one or more carbon atoms in the ring are replaced by a heteroatom such as O, N, S, P, and the like, each of which may be substituted as described herein if an open valence is present, and each may be in any suitable stable oxidation state.
  • aryl refers to cyclic aromatic hydrocarbon groups that do not contain heteroatoms in the ring.
  • aryl groups include, but are not limited to, phenyl, azulenyl, heptalenyl, biphenyl, indacenyl, fluorenyl, phenanthrenyl, triphenylenyl, pyrenyl, naphthacenyl, chrysenyl, biphenylenyl, anthracenyl, and naphthyl groups.
  • aryl groups contain about 6 to about 14 carbons in the ring portions of the groups.
  • Aryl groups can be unsubstituted or substituted, as defined herein.
  • substituted aryl groups can be mono-substituted or substituted more than once, such as, but not limited to, a phenyl group substituted at any one or more of 2-, 3-, 4-, 5-, or 6-positions of the phenyl ring, or a naphthyl group substituted at any one or more of 2- to 8-positions thereof.
  • aralkyl refers to alkyl groups as defined herein in which a hydrogen or carbon bond of an alkyl group is replaced with a bond to an aryl group as defined herein.
  • aralkyl groups include benzyl and phenylethyl groups and fused (cycloalkylaryl)alkyl groups such as 4-ethyl-indanyl.
  • Aralkenyl groups are alkenyl groups as defined herein in which a hydrogen or carbon bond of an alkyl group is replaced with a bond to an aryl group as defined herein.
  • heterocyclyl refers to aromatic and non-aromatic ring compounds containing three or more ring members, of which one or more is a heteroatom such as, but not limited to, N, O, and S.
  • a heterocyclyl can be a cycloheteroalkyl, or a heteroaryl, or if polycyclic, any combination thereof.
  • heterocyclyl groups include 3 to about 20 ring members, whereas other such groups have 3 to about 15 ring members.
  • a heterocyclyl group designated as a C 2 -heterocyclyl can be a 5-ring with two carbon atoms and three heteroatoms, a 6-ring with two carbon atoms and four heteroatoms and so forth.
  • a C 4 -heterocyclyl can be a 5-ring with one heteroatom, a 6-ring with two heteroatoms, and so forth.
  • the number of carbon atoms plus the number of heteroatoms equals the total number of ring atoms.
  • a heterocyclyl ring can also include one or more double bonds.
  • a heteroaryl ring is an embodiment of a heterocyclyl group.
  • the phrase "heterocyclyl group" includes fused ring species including those that include fused aromatic and non-aromatic groups.
  • a dioxolanyl ring and a benzdioxolanyl ring system are both heterocyclyl groups within the meaning herein.
  • the phrase also includes polycyclic ring systems containing a heteroatom such as, but not limited to, quinuclidyl. Heterocyclyl groups can be unsubstituted, or can be substituted as discussed herein.
  • Heterocyclyl groups include, but are not limited to, pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, pyrrolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, thiazolyl, pyridinyl, thiophenyl, benzothiophenyl, benzofuranyl, dihydrobenzofuranyl, indolyl, dihydroindolyl, azaindolyl, indazolyl, benzimidazolyl, azabenzimidazolyl, benzoxazolyl, benzothiazolyl, benzothiadiazolyl, imidazopyridinyl, isoxazolopyridinyl, thianaphthalenyl, purinyl, xanthinyl, adeninyl, guaninyl, quinolinyl, isoquino
  • heterocyclyl groups can be mono-substituted or substituted more than once, such as, but not limited to, piperidinyl or quinolinyl groups, which are 2-, 3-, 4-, 5-, or 6- substituted, or disubstituted with groups such as those listed herein.
  • heteroaryl refers to aromatic ring compounds containing 5 or more ring members, of which, one or more is a heteroatom such as, but not limited to, N, O, and S; for instance, heteroaryl rings can have 5 to about 8-12 ring members.
  • a heteroaryl group is a variety of a heterocyclyl group that possesses an aromatic electronic structure.
  • a heteroaryl group designated as a C 2 -heteroaryl can be a 5-ring with two carbon atoms and three heteroatoms, a 6-ring with two carbon atoms and four heteroatoms and so forth.
  • a C 4 -heteroaryl can be a 5-ring with one heteroatom, a 6-ring with two heteroatoms, and so forth.
  • the number of carbon atoms plus the number of heteroatoms sums up to equal the total number of ring atoms.
  • a heterocyclyl ring designated C x-y can be any ring containing 'x' members up to 'y' members, including all intermediate integers between 'x' and 'y' and that contains one or more heteroatoms, as defined herein.
  • Heterocyclyl rings designated C x-y can also be polycyclic ring systems, such as bicyclic or tricyclic ring systems.
  • Heteroaryl groups include, but are not limited to, groups such as pyrrolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, thiazolyl, pyridinyl, thiophenyl, benzothiophenyl, benzofuranyl, indolyl, azaindolyl, indazolyl, benzimidazolyl, azabenzimidazolyl, benzoxazolyl, benzothiazolyl, benzothiadiazolyl, imidazopyridinyl, isoxazolopyridinyl, thianaphthalenyl, purinyl, xanthinyl, adeninyl,
  • Heteroaryl groups can be unsubstituted, or can be substituted with groups as is discussed herein. Representative substituted heteroaryl groups can be substituted one or more times with groups such as those listed herein. Additional examples of aryl and heteroaryl groups include but are not limited to phenyl, biphenyl, indenyl, naphthyl (1-naphthyl, 2-naphthyl), N-hydroxytetrazolyl, N- hydroxytriazolyl, N-hydroxyimidazolyl, anthracenyl (1-anthracenyl, 2-anthracenyl, 3- anthracenyl), thiophenyl (2-thienyl, 3-thienyl), furyl (2-furyl, 3-furyl) , indolyl, oxadiazolyl, isoxazolyl, quinazolinyl, fluorenyl, xanthenyl, isoindanyl, benzhydry
  • heterocyclylalkyl refers to alkyl groups as defined herein in which a hydrogen or carbon bond of an alkyl group as defined herein is replaced with a bond to a heterocyclyl group as defined herein.
  • Representative heterocyclyl alkyl groups include, but are not limited to, furan-2-yl methyl, furan-3-yl methyl, pyridine-3-yl methyl, tetrahydrofuran-2-yl ethyl, and indol-2-yl propyl.
  • heteroarylalkyl refers to alkyl groups as defined herein in which a hydrogen or carbon bond of an alkyl group is replaced with a bond to a heteroaryl group as defined herein.
  • alkoxy refers to an oxygen atom connected to an alkyl group, including a cycloalkyl group, as are defined herein. Examples of linear alkoxy groups include but are not limited to methoxy, ethoxy, propoxy, butoxy, pentyloxy, hexyloxy, and the like.
  • Examples of branched alkoxy include but are not limited to isopropoxy, sec-butoxy, tert-butoxy, isopentyloxy, isohexyloxy, and the like.
  • Examples of cyclic alkoxy include but are not limited to cyclopropyloxy, cyclobutyloxy, cyclopentyloxy, cyclohexyloxy, and the like.
  • An alkoxy group can include about 1 to about 12, about 1 to about 20, or about 1 to about 40 carbon atoms bonded to the oxygen atom, and can further include double or triple bonds, and can also include heteroatoms.
  • an allyloxy group or a methoxyethoxy group is also an alkoxy group within the meaning herein, as is a methylenedioxy group in a context where two adjacent atoms of a structure are substituted therewith.
  • amine refers to primary, secondary, and tertiary amines having, e.g., the formula N(group) 3 wherein each group can independently be H or non-H, such as alkyl, aryl, and the like.
  • Amines include but are not limited to R-NH 2 , for example, alkylamines, arylamines, alkylarylamines; R 2 NH wherein each R is independently selected, such as dialkylamines, diarylamines, aralkylamines, heterocyclylamines and the like; and R 3 N wherein each R is independently selected, such as trialkylamines, dialkylarylamines, alkyldiarylamines, triarylamines, and the like.
  • amine also includes ammonium ions as used herein.
  • amino group refers to a substituent of the form -NH 2 , - NHR, -NR 2 , -NR 3 + , wherein each R is independently selected, and protonated forms of each, except for -NR 3 + , which cannot be protonated. Accordingly, any compound substituted with an amino group can be viewed as an amine.
  • An “amino group” within the meaning herein can be a primary, secondary, tertiary, or quaternary amino group.
  • alkylamino includes a monoalkylamino, dialkylamino, and trialkylamino group.
  • halo means, unless otherwise stated, a fluorine, chlorine, bromine, or iodine atom.
  • haloalkyl includes mono-halo alkyl groups, poly- halo alkyl groups wherein all halo atoms can be the same or different, and per-halo alkyl groups, wherein all hydrogen atoms are replaced by halogen atoms, such as fluoro.
  • haloalkyl examples include trifluoromethyl, 1,1-dichloroethyl, 1,2-dichloroethyl, 1,3-dibromo-3,3- difluoropropyl, perfluorobutyl, and the like.
  • epoxy-functional or "epoxy-substituted” as used herein refers to a functional group in which an oxygen atom, the epoxy substituent, is directly attached to two adjacent carbon atoms of a carbon chain or ring system.
  • epoxy-substituted functional groups include, but are not limited to, 2,3-epoxypropyl, 3,4-epoxybutyl, 4,5- epoxypentyl, 2,3-epoxypropoxy, epoxypropoxypropyl, 2-glycidoxyethyl, 3-glycidoxypropyl, 4-glycidoxybutyl, 2-(glycidoxycarbonyl)propyl, 3-(3,4-epoxycylohexyl)propyl, 2-(3,4- epoxycyclohexyl)ethyl, 2-(2,3-epoxycylopentyl)ethyl, 2-(4-methyl-3,4- epoxycyclohexyl)propyl, 2-(3,4-epoxy-3-methylcylohexyl)-2-methylethyl, and 5,6- epoxyhexyl.
  • the term "monovalent” as used herein refers to a substituent connecting via a single bond to a substituted molecule. When a substituent is monovalent, such as, for example, F or Cl, it is bonded to the atom it is substituting by a single bond.
  • the term "hydrocarbon” or “hydrocarbyl” as used herein refers to a molecule or functional group that includes carbon and hydrogen atoms. The term can also refer to a molecule or functional group that normally includes both carbon and hydrogen atoms but wherein all the hydrogen atoms are substituted with other functional groups.
  • hydrocarbyl refers to a functional group derived from a straight chain, branched, or cyclic hydrocarbon, and can be alkyl, alkenyl, alkynyl, aryl, cycloalkyl, acyl, or any combination thereof. Hydrocarbyl groups can be shown as (Ca- C b )hydrocarbyl, wherein a and b are integers and mean having any of a to b number of carbon atoms.
  • (C1-C4)hydrocarbyl means the hydrocarbyl group can be methyl (C1), ethyl (C 2 ), propyl (C 3 ), or butyl (C 4 ), and (C 0 -C b )hydrocarbyl means in certain embodiments there is no hydrocarbyl group.
  • the term "C 6-10 -5-6 membered heterobiaryl” means a C 6-10 aryl moiety covalently bonded through a single bond to a 5- or 6-membered heteroaryl moiety.
  • the C 6-10 aryl moiety and the 5-6-membered heteroaryl moiety can be any of the suitable aryl and heteroaryl groups described herein.
  • Non-limiting examples of a C 6-10 -5-6 membered heterobiaryl include When the C 6-10- 5-6 membered heterobiaryl is listed as a substituent (e.g., as an "R" group), the C 6-10- 5-6 membered heterobiaryl is bonded to the rest of the molecule through the C 6-10 moiety.
  • the term "5-6 membered- C 6-10 heterobiaryl" is the same as a C 6-10 -5- 6 membered heterobiaryl, except that when the 5-6 membered- C 6-10 heterobiaryl is listed as a substituent (e.g., as an "R” group), the 5-6 membered- C 6-10 heterobiaryl is bonded to the rest of the molecule through the 5-6-membered heteroaryl moiety.
  • the term "C 6-10 - C 6-10 biaryl” means a C 6-10 aryl moiety covalently bonded through a single bond to another C 6-10 aryl moiety.
  • the C 6-10 aryl moiety can be any of the suitable aryl groups described herein.
  • Non-limiting example of a C 6-10 - C 6-10 biaryl include biphenyl and binaphthyl.
  • solvent refers to a liquid that can dissolve a solid, liquid, or gas.
  • Non-limiting examples of solvents are silicones, organic compounds, water, alcohols, ionic liquids, and supercritical fluids.
  • independently selected from refers to referenced groups being the same, different, or a mixture thereof, unless the context clearly indicates otherwise.
  • X 1 , X 2 , and X 3 are independently selected from noble gases” would include the scenario where, for example, X 1 , X 2 , and X 3 are all the same, where X 1 , X 2 , and X 3 are all different, where X 1 and X 2 are the same but X 3 is different, and other analogous permutations.
  • room temperature refers to a temperature of about 15 °C to 28 °C.
  • standard temperature and pressure refers to 20 °C and 101 kPa.
  • composition refers to a mixture of at least one compound described herein with a pharmaceutically acceptable carrier.
  • the pharmaceutical composition facilitates administration of the compound to a patient or subject. Multiple techniques of administering a compound exist in the art including, but not limited to, intravenous, oral, aerosol, parenteral, ophthalmic, pulmonary and topical administration.
  • a “disease” is a state of health of an animal wherein the animal cannot maintain homeostasis, and wherein if the disease is not ameliorated then the animal's health continues to deteriorate.
  • a disorder in an animal is a state of health in which the animal is able to maintain homeostasis, but in which the animal's state of health is less favorable than it would be in the absence of the disorder. Left untreated, a disorder does not necessarily cause a further decrease in the animal's state of health.
  • the terms "effective amount,” “pharmaceutically effective amount” and “therapeutically effective amount” refer to a nontoxic but sufficient amount of an agent to provide the desired biological result. That result may be reduction and/or alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system. An appropriate therapeutic amount in any individual case may be determined by one of ordinary skill in the art using routine experimentation.
  • the term “efficacy” refers to the maximal effect (Emax) achieved within an assay.
  • pharmaceutically acceptable refers to a material, such as a carrier or diluent, which does not abrogate the biological activity or properties of the compound, and is relatively non-toxic, i.e., the material may be administered to an individual without causing undesirable biological effects or interacting in a deleterious manner with any of the components of the composition in which it is contained.
  • pharmaceutically acceptable salt refers to a salt of the administered compounds prepared from pharmaceutically acceptable non-toxic acids or bases, including inorganic acids or bases, organic acids or bases, solvates, hydrates, or clathrates thereof.
  • Suitable pharmaceutically acceptable acid addition salts may be prepared from an inorganic acid or from an organic acid.
  • inorganic acids include hydrochloric, hydrobromic, hydriodic, nitric, carbonic, sulfuric (including sulfate and hydrogen sulfate), and phosphoric acids (including hydrogen phosphate and dihydrogen phosphate).
  • Appropriate organic acids may be selected from aliphatic, cycloaliphatic, aromatic, araliphatic, heterocyclic, carboxylic and sulfonic classes of organic acids, examples of which include formic, acetic, propionic, succinic, glycolic, gluconic, lactic, malic, tartaric, citric, ascorbic, glucuronic, maleic, malonic, saccharin, fumaric, pyruvic, aspartic, glutamic, benzoic, anthranilic, 4-hydroxybenzoic, phenylacetic, mandelic, embonic (pamoic), methanesulfonic, ethanesulfonic, benzenesulfonic, pantothenic, trifluoromethanesulfonic, 2- hydroxyethanesulfonic, p-toluenesulfonic, sulfanilic, cyclohexylaminosulfonic, stearic, alginic,
  • Suitable pharmaceutically acceptable base addition salts of compounds described herein include, for example, ammonium salts, metallic salts including alkali metal, alkaline earth metal and transition metal salts such as, for example, calcium, magnesium, potassium, sodium and zinc salts.
  • Pharmaceutically acceptable base addition salts also include organic salts made from basic amines such as, for example, N,N'-dibenzylethylene-diamine, chloroprocaine, choline, diethanolamine, ethylenediamine, meglumine (N-methylglucamine) and procaine. All of these salts may be prepared from the corresponding compound by reacting, for example, the appropriate acid or base with the compound.
  • the term "pharmaceutically acceptable carrier” or “pharmaceutically acceptable excipient” means a pharmaceutically acceptable material, composition or carrier, such as a liquid or solid filler, stabilizer, dispersing agent, suspending agent, diluent, excipient, thickening agent, solvent or encapsulating material, involved in carrying or transporting a compound described herein within or to the patient such that it may perform its intended function.
  • a pharmaceutically acceptable material, composition or carrier such as a liquid or solid filler, stabilizer, dispersing agent, suspending agent, diluent, excipient, thickening agent, solvent or encapsulating material, involved in carrying or transporting a compound described herein within or to the patient such that it may perform its intended function.
  • Such constructs are carried or transported from one organ, or portion of the body, to another organ, or portion of the body.
  • Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation, including the compound(s) described herein, and not injuri
  • materials that may serve as pharmaceutically acceptable carriers include: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol; polyols, such as glycerin, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffering agents, such as magnesium hydroxide and aluminum hydroxide; surface active agents; alginic acid; pyrogen-free water; isotonic saline
  • pharmaceutically acceptable carrier also includes any and all coatings, antibacterial and antifungal agents, and absorption delaying agents, and the like that are compatible with the activity of the compound(s) described herein, and are physiologically acceptable to the patient. Supplementary active compounds may also be incorporated into the compositions.
  • the "pharmaceutically acceptable carrier” may further include a pharmaceutically acceptable salt of the compound(s) described herein.
  • Other additional ingredients that may be included in the pharmaceutical compositions used with the methods or compounds described herein are known in the art and described, for example in Remington's Pharmaceutical Sciences (Genaro, Ed., Mack Publishing Co., 1985, Easton, PA), which is incorporated herein by reference.
  • patient refers to any animal, or cells thereof whether in vitro or in situ, amenable to the methods described herein.
  • the patient, subject or individual is a human.
  • potency refers to the dose needed to produce half the maximal response (ED 50 ).
  • a “therapeutic” treatment is a treatment administered to a subject who exhibits signs of pathology, for the purpose of diminishing or eliminating those signs.
  • treatment is defined as the application or administration of a therapeutic agent, i.e., a compound or compounds as described herein (alone or in combination with another pharmaceutical agent), to a patient, or application or administration of a therapeutic agent to an isolated tissue or cell line from a patient (e.g., for diagnosis or ex vivo applications), who has a condition contemplated herein or a symptom of a condition contemplated herein, with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve or affect a condition contemplated herein, or the symptoms of a condition contemplated herein.
  • Such treatments may be specifically tailored or modified, based on knowledge obtained from the field of pharmacogenomics.
  • homologous refers to the subunit sequence identity between two polymeric molecules, e.g., between two nucleic acid molecules, such as, two DNA molecules or two RNA molecules, or between two polypeptide molecules. When a subunit position in both of the two molecules is occupied by the same monomeric subunit; e.g., if a position in each of two DNA molecules is occupied by adenine, then they are homologous at that position.
  • the homology between two sequences is a direct function of the number of matching or homologous positions; e.g., if half (e.g., five positions in a polymer ten subunits in length) of the positions in two sequences are homologous, the two sequences are 50% homologous; if 90% of the positions (e.g., 9 of 10), are matched or homologous, the two sequences are 90% homologous.
  • the following abbreviations for the commonly occurring nucleic acid bases are used.
  • nucleic acids are polymers of nucleotides.
  • nucleic acids and polynucleotides as used herein are interchangeable.
  • nucleic acids are polynucleotides, which can be hydrolyzed into the monomeric “nucleotides.” The monomeric nucleotides can be hydrolyzed into nucleosides.
  • polynucleotides include, but are not limited to, all nucleic acid sequences which are obtained by any means available in the art, including, without limitation, recombinant means, i.e., the cloning of nucleic acid sequences from a recombinant library or a cell genome, using ordinary cloning technology and PCR, and the like, and by synthetic means.
  • recombinant means i.e., the cloning of nucleic acid sequences from a recombinant library or a cell genome, using ordinary cloning technology and PCR, and the like, and by synthetic means.
  • oligonucleotide refers to short polynucleotides, such as from two (2) to 50 nucleotides.
  • RNA sequence i.e., A, U, C, G
  • U replaces "T”
  • 3'-end refers to the terminal residue of a DNA or RNA strand that terminates at the hydroxyl group of the 3' carbon in a deoxyribose (DNA) or ribose (RNA).
  • the term "5'-end” refers to the terminal residue of a DNA or RNA strand that terminates at a hydroxyl group of the 5' carbon in a deoxyribose (DNA) or ribose (RNA).
  • a phosphate or phosphorothioate group can be attached to the hydroxyl group on the 5'-carbon.
  • the term "DNA” means deoxyribonucleic acid.
  • RNA means ribonucleic acid.
  • a compound of formula I or a salt, enantiomer, diastereomer, or tautomer thereof: wherein: ONA is an oligonucleotide having a 3'-end and a 5'-end and comprising one or more phosphodiester or phosphorothioate internucleotide linkages; LNK is a chemical linker covalently bonded to the ONA and the UBL; and UBL is an E3 ubiquitin ligase ligand.
  • ONA is an oligonucleotide having a 3'-end and a 5'-end and comprising one or more phosphodiester or phosphorothioate internucleotide linkages
  • LNK is a chemical linker covalently bonded to the ONA and the UBL
  • UBL is an E3 ubiquitin ligase ligand.
  • Oligonucleotide Suitable salts include any of the pharmaceutically acceptable salts described herein.
  • the ONA is bonded to the LNK at the 3'-end or the 5'-end.
  • the ONA is bonded to the LNK at the 3'-end.
  • the ONA is bonded to the LNK at the 5' end.
  • the ONA comprises deoxyribonucleotide(s) (DNA), ribonucleotide(s) (RNA), or any combinations thereof.
  • the ONA comprises deoxyribonucleotide(s) (DNA).
  • the ONA comprises a c-Myc-binding nucleotide sequence.
  • At least two nucleotides flank each end of the c-Myc-binding nucleotide sequence. In various embodiments, 2 to 20 nucleotides flank each end of the c- Myc-binding nucleotide sequence. The number of flanking nucleotides at each end of the c- Myc-binding nucleotide sequence can be the same or different, and the sequences can be the same, different, reverse sequences, complementary sequences, or reverse complementary sequences. In various embodiments, the c-Myc-binding nucleotide sequence is flanked on each side by 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 nucleotides.
  • the c-Myc-binding nucleotide sequence comprises a sequence that is at least 60, 65, 70, 75, 80, 85, 90, 95, 96, 97, 98, or 99% similar to 5'- CACGTGGTTGCCACGTG-3' (SEQ ID NO: 1). In various embodiments, the c-Myc-binding nucleotide sequence comprises the sequence 5'- CACGTGGTTGCCACGTG-3' (SEQ ID NO: 1)..
  • the c-Myc binding nucleotide sequence is 5'- TGGGAGCACGTGGTTGCCACGTGGTTGGG-3' (SEQ ID NO: 2) or 3'- GGGTTGGTGCACCGTTGGTGCACGAGGGT-5' (SEQ ID NO: 3).
  • the ONA comprises a brachyury-binding nucleotide sequence. In various embodiments, at least two nucleotides flank each end of the brachyury- binding nucleotide sequence. In various embodiments, 2 to 20 nucleotides flank each end of the brachyury-binding nucleotide sequence.
  • flanking nucleotides at each end of the brachyury-binding nucleotide sequence can be the same or different, and the sequences can be the same, different, reverse sequences, complementary sequences, or reverse complementary sequences.
  • the brachyury-binding nucleotide sequence is flanked on each side by 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 nucleotides.
  • the brachyury-binding nucleotide sequence comprises a sequence that is at least 60, 65, 70, 75, 80, 85, 90, 95, 96, 97, 98, or 99% similar to 5'- AATTTCACACCTAGGTGTGAAATT-3' (SEQ ID NO: 4). In various embodiments, the brachyury-binding nucleotide sequence comprises the sequence 5'- AATTTCACACCTAGGTGTGAAATT-3' (SEQ ID NO: 4).
  • the brachyury-binding nucleotide sequence is 5'-CTTTCCAATTTCACACCTAGGTGTGAAATTGGGGAC-3' (SEQ ID NO: 5) or 3'-CAGGGGTTAAAGTGTGGATCCACACTTTAACCTTTC-5' (SEQ ID NO: 6).
  • at least one of the phosphodiester groups in the internucleotide linkages comprising the ONA is replaced with a non-bridging phosphorothioate (PS) group, in which the sulfur atom in the PS group does not connect to a ribose or deoxyribose.
  • PS non-bridging phosphorothioate
  • phosphorothioate group has the structure: and can be present as a salt thereof. Suitable salts include any of the pharmaceutically acceptable salts described herein. In various embodiments, the salt is an sodium or potassium salt.
  • the terminal nucleotide in ONA at the 3'-end has the structure: wherein: Y is H or OH; and In various embodiments, the terminal nucleotide in ONA at the 5'-end has the structure: wherein: Y is H or OH; Z is O or S; and , , , , , The in the terminal nucleotide sugar, whether at the 3'-end or the 5'-end, designates the point of attachment of the rest of the nucleotides in ONA. The in NUC designates the point of attachment of the respective base to the ribose or deoxyribose.
  • A is phenylene or a C 5-18 heterocyclylene;
  • R is independently at each occurrence H, C 1-12 alkyl, C 2-12 alkenyl, C 2-12 alkynyl, C 6-10 aryl, C 5-10 heteroaryl, halogen, or combinations thereof;
  • R' is independently at each occurrence H, C 1-12
  • a given quantity of LL units can be used to form any suitable stable chemical structure, including branched, cyclic, and acyclic structures.
  • A is a heterocyclylene that contains one or more aliphatic carbons.
  • A is heteroarylene.
  • A is arylene.
  • suitable structures for –(LL) aa –A–(LL) bb — include:
  • suitable structures for –(LL) aa –A–(LL) bb — include:
  • A is a C5-18 heteroaryl, such as but not limited to a 1,2,3- triazolyl or a 1,2,4-triazolyl.
  • LNK-X1 any two X groups bonded to each other cannot be -O- such that (X) m does not contain a peroxide functionality.
  • X in LNK-X1 is independently at each occurrence -O- or -CH 2 -.
  • Contemplated herein are all combinations of m, X, and pp in LNK-X1.
  • LNK-X2 any two X groups bonded to each other cannot be -O- such that (X) m does not contain a peroxide functionality.
  • X in LNK-X2 is independently at each occurrence -O- or -CH 2 -.
  • Contemplated herein are all combinations of m, X, and pp in LNK-X2.
  • LNK has the structure: wherein pp is an integer from 1 to 10, X is CH 2 or O, k is an integer from 1 to 3; and wherein the atom marked with * is covalently bonded to the 3'-end or the 5'-end of the ONA.
  • X is CH 2 .
  • LNK-1 in LNK-1, X is O. In various embodiments, in LNK-1, k is 1. In various embodiments, in LNK-1, k is 2. In various embodiments, in LNK-1, k is 3. Contemplated herein are all combinations of k, X, and pp in LNK-1. In various embodiments, LNK has the structure: wherein pp is an integer from 1 to 10, X is CH 2 or O, k is an integer from 1 to 3; and wherein the atom marked with * is covalently bonded to the 3'-end or the 5'-end of the ONA. In various embodiments, in LNK-2, X is CH 2 . In various embodiments, in LNK-2, X is O.
  • k is 1. In various embodiments, in LNK-2, k is 2. In various embodiments, in LNK-2, k is 3. Contemplated herein are all combinations of k, X, and pp in LNK-2.
  • aa is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100.
  • bb is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100.
  • zz is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81, 82, 83, 84, 85, 86, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, or 100.
  • LNK-X1, LNK-X2, LNK-1, or LNK-2 When LNK-X1, LNK-X2, LNK-1, or LNK-2 is bonded the 3'-end, the O* atom connects directly to the ribose or deoxyribose 3' carbon.
  • LNK-X1, LNK-X2, LNK-1, or LNK-2 is bonded the 5'-end, the O* atom connects directly to the phosphorus atom in the 5'-phosphate in the ribose or deoxyribose.
  • pp in LNK-X1, LNK-X2, LNK-1, or LNK-2 is 1. In various embodiments, pp in LNK-X1, LNK-X2, LNK-1, or LNK-2 is 2.
  • pp in LNK-X1, LNK-X2, LNK-1, or LNK-2 is 3. In various embodiments, pp in LNK-X1, LNK-X2, LNK-1, or LNK-2 is 4. In various embodiments, pp in LNK-X1, LNK-X2, LNK-1, or LNK-2 is 5. In various embodiments, in LNK-X1, LNK- X2, LNK-1, or LNK-2 pp is 6. In various embodiments, in LNK-X1, LNK-X2, LNK-1, or LNK-2 pp is 7. In various embodiments, pp in LNK-X1, LNK-X2, LNK-1, or LNK-2 is 8.
  • pp in LNK-X1, LNK-X2, LNK-1, or LNK-2 is 9. In various embodiments, pp in LNK-X1, LNK-X2, LNK-1, or LNK-2 is 10. In various embodiments, pp in LNK-X1, LNK-X2, LNK-1, or LNK-2 is 2, 3, or 5.
  • LNK comprises a group represented by a general structure selected from the group consisting of: -NR(CH 2 ) n -(lower alkyl)-, -NR(CH 2 ) n -(lower alkoxyl)- , -NR(CH 2 ) n -(lower alkoxyl)-OCH 2 -, -NR(CH 2 ) n -(lower alkoxyl)-(lower alkyl)-OCH 2 -, - NR(CH 2 ) n -(cycloalkyl)-(lower alkyl)-OCH 2 -, -NR(CH 2 ) n -(heterocycloalkyl)-, - NR(CH 2 CH 2 O) n -(lower alkyl)-O-CH 2 -, -NR(CH 2 CH 2 O) n -(heterocycloalkyl)-O-CH 2 -, -NR(CH 2 CH
  • LNK comprises a group represented by a general structure selected from the group consisting of: -N(R)-(CH 2 ) m -O(CH 2 ) n -O(CH 2 ) o -O(CH 2 ) p -O(CH 2 ) q -O(CH 2 ) r -OCH 2 -, -O-(CH 2 ) m -O(CH 2 ) n -O(CH 2 )o-O(CH 2 ) p -O(CH 2 ) q -O(CH 2 )r-OCH 2 -, -O-(CH 2 ) m -O(CH 2 ) n -O(CH 2 ) o -O(CH 2 ) p -O(CH 2 ) q -O(CH 2 )r-OCH 2 -, -O-(CH 2 ) m -O(CH 2 ) n -O(CH 2 )
  • the linker (L) is selected from the group consisting of:
  • LNK is selected from the group consisting of:
  • LNK is selected from the group consisting of:
  • the UBL is a ligand of an E3 ubiquitin ligase selected from the group consisting of von Hippel-Lindau (VHL), cereblon (CRBN), RING-type zinc-finger protein 114 (RNF114), cellular inhibitor of apoptosis (cIAP), mouse double minute 2 homologue (MDM2), damage-specific DNA binding protein 1 (DDB1)-CUL4 associated factor 16 (DCAF16), and Kelch-like ECH-associated protein 1 (KEAP1).
  • VHL von Hippel-Lindau
  • CRBN cereblon
  • RRF114 RING-type zinc-finger protein 114
  • cIAP cellular inhibitor of apoptosis
  • MDM2 mouse double minute 2 homologue
  • DDB1-CUL4 associated factor 16 DCAF16
  • KEAP1 Kelch-like ECH-associated protein 1
  • UBL is a ligand of VHL or CRBN.
  • any suitable E3 ubiquitin ligase recruiting ligand can be incorporated into the compounds of formula I.
  • suitable E3 ubiquitin ligase ligands and their syntheses can be found in Bricelj, A. et al., Front. Chem., 05 July 2021, DOI: 10.3389/fchem.2021.707317.
  • the UBL is a ligand having the structure: wherein R 1 is H or CH 3 ; and R 2 is selected from the group consisting of In the CRBN ligands, the attachment point shown in structures such as can be through any suitable atom/fragment as defined in LL herein.
  • the CRBN ligands can be attached via any of the following connectivities:
  • the UBL has the structure:
  • the compound of formula I has the structure of:
  • pp is an integer from 1 to 10; and wherein the atom marked with * is covalently bonded to the 3'-end or the 5'-end of the ONA.
  • Modified Oligonucleotides a compound of formula II is provided: ONA–LNK–C ⁇ C-H (formula II), wherein: ONA is an oligonucleotide having a 3'-end and a 5'-end; LNK is a chemical linker connecting the ONA and the C ⁇ C-H. In various embodiments, LNK has any linker structure described elsewhere herein.
  • A is phenylene or a C5-18 heterocyclylene;
  • R is independently at each occurrence H, C 1-12 alkyl, C 2-12 alkenyl, C 2-12 alkynyl, C 6-10 aryl, C 5-10 heteroaryl, halogen, or combinations thereof;
  • R' is independently at each occurrence H,
  • Y in II-A is independently at each occurrence -O- or -CH 2 -; and wherein the atom marked with * is covalently bonded to the 3'-end or the 5'-end of the ONA.
  • d in II-A is 2. In various embodiments, d in II-A is 3. In various embodiments, d in II-A is 4. In various embodiments, d in II-A is 5. In various embodiments, d in II-A is 6. In various embodiments, d in II-A is 7. In various embodiments, d in II-A is 8. In various embodiments, d in II-A is 9. In various embodiments, d in II-A is 10. In various embodiments, d in II-A is 11.
  • d in II-A is 12. In various embodiments, d in II-A is 13. In various embodiments, d in II-A is 14. In various embodiments, d in II-A is 15. In various embodiments, the compound of formula II has the structure: wherein pp is an integer from 1 to 10; and wherein the atom marked with * is covalently bonded to the 3'-end or the 5'-end of the ONA. Degradation of c-Myc by oligoTRAFTACs Myc transcription factors are dysregulated in a range of cancers. Even though c-Myc has been targeted by indirect approaches, development of direct-targeting methods is hindered by the paucity of ligandable pockets and their highly disordered structure.
  • c- Myc binds to specific DNA sequences with a conserved E-box sequence (CACGTG)
  • CACGTG conserved E-box sequence
  • OT oligoTRAFTAC
  • a Myc binding consensus sequence (5' CACGTGGTTGCCACGTG 3') was taken from one of its target gene promoters. Additionally, a flanking sequence was included at both 3' and 5' end of the c-Myc- targeting oligonucleotide sequence to facilitate successful double strand formation of the recognition sequence while providing a flexibility for oligoTRAFTACs.
  • a c-Myc-targeting oligonucleotide sequence was synthesized with an orthogonal alkyne handle on either side as a reactive moiety to append an azide-containing VHL ligand (FIG.7A). Copper-catalyzed alkyne-azide cycloaddition (CuAAC) click reaction was performed to synthesize two c-Myc targeting oligoTRAFTACs (OT7: VHL ligand at the 5' end and OT10: VHL ligand at the 3' end of the oligonucleotide) (FIG.7B, 7C). After 18 h at room temperature, the crude mixture was purified and analyzed by HPLC to verify reaction completion (FIG.8A).
  • CuAAC Copper-catalyzed alkyne-azide cycloaddition
  • the reaction was also monitored by electrophoretic mobility shift assay (EMSA) (FIG.8B).
  • ESA electrophoretic mobility shift assay
  • the single stranded oligoTRAFTACs were then purified by HPLC, dried and reconstituted in water.
  • an annealing reaction was performed in the presence of the reverse complementary oligonucleotide by heating to 95 0 C and slowly cooling to room temperature over a 1.5 h period.
  • a biotinylated version of the same c-Myc-targeting oligonucleotide was also synthesized to generate a double stranded oligonucleotide via the same annealing reaction conditions.
  • OT7 and OT10 induces c-Myc degradation via the proteasomal pathway.
  • the cell viability in response to the transfection of OT7 and OT12 was assessed.
  • the induction of OT7-mediated c-Myc degradation inhibited cell proliferation compared to scrambled control-transfected cells (FIG.8E).
  • OligoTRAFTAC-mediated brachyury degradation T-box transcription factor (brachyury) is a DNA-interacting TF that regulates gene expression during early embryonic development in vertebrates.
  • Brachyury is crucial for early mesoderm formation and plays a key role in notochord development where, as a homodimer, it recognizes and binds to a consensus palindromic DNA sequence.
  • a brachyury-binding DNA sequence AATTTCACACCTAGGTGTGAAATT
  • Oligonucleotides were synthesized with flanking bases and terminal alkyne moieties at either end (3' and 5') of the oligonucleotide (FIG.7A, right panel).
  • oligoTRAFTACs To generate brachyury-targeting oligoTRAFTACs, two azido-VHL ligands were synthesized with a long (5 PEG units) and a short (2 PEG units) linker (FIG.7C, FIG.9A). To test whether the oligonucleotide used in oligoTRAFTAC design could recruit brachyury, a streptavidin pull-down experiment was performed using a biotin-oligonucleotide.
  • brachyury-oligoTRAFTACs were transfected into brachyury-GFP-expressing HEK293T cells for 24 h, after which cells were collected and lysed, followed by western blotting. OligoTRAFTACs with 5' VHL-ligand and a longer linker (OT1) did not induce significant degradation of brachyury-GFP (FIG.9B), while its counterpart the shorter linker (OT2) successfully induced target degradation.
  • oligoTRAFTACs with 3' VHL-ligand both showed comparable, better degradation profiles relative to OT1 and OT2 (FIG.9B, last two panels).
  • OT3 and OT4 were transfected into HEK293T cells and lysed after 20 h.
  • the time course experiment suggested that OT3 can induce significant brachyury-GFP degradation at 12 h that is maintained up to 36 h post-transfection (FIG.3C and FIG.10A).
  • brachyury degradation was prominent even after the cells incubated in fresh medium for longer time (36 h), which is consistent with oligoTRAFTAC OT7 targeting c-Myc-targeting (FIG.3D, last panel).
  • brachyury levels at 36 h had increased relative to the 24 h period, indicating a progressive loss of brachyury degradability by OT3. This could be partially explained by the reduced stability of oligoTRAFTACs due to the oligonucleotide sensitivity towards intracellular nucleases, increased deubiquitinase (DUB) activity, or increased c-Myc resynthesis.
  • DAB deubiquitinase
  • oligoTRAFTAC-mediated brachyury recruitment and degradation is oligonucleotide-dependent
  • OT5 and OT6 scrambled- oligoTRAFTACs
  • VHL competition rescued oligoTRAFTAC-mediated brachyury degradation (FIG.4B), confirming that observed brachyury degradation is a result of VHL E3 ligase recruitment by OT3.
  • OT3-mediated brachyury degradation was evaluated in the presence of a neddylation inhibitor to further confirm that the intended mechanism is via the proteasomal pathway.
  • the neddylation inhibitor, MLN-4924 was pre-incubated with cells at 1 mM concentration. After 1.5 h, OT3 was transfected into the cell for 20 h following which the cell lysates were analyzed as indicated in FIG.4C.
  • OT3 Consistent with the brachyury-GFP degradation in HEK293T cells, 60 nM of OT3 induces ⁇ 70% brachyury degradation in UM- Chor1 cells 24 h post-transfection (FIG.11B). Although OT3 induced comparable levels of brachyury degradation in both cell lines, phosphodiester linkages within OT3 are susceptible to cleavage by both extra- and intracellular nucleases. Therefore, oligoTRAFTACs with a phosphorothioate (PS) backbone were synthesized. The addition of extra non-bridging sulfur atoms in the inter-nucleotide phosphate group has been shown to have both increased stability against nucleases and improved cell permeability.
  • PS phosphorothioate
  • brachyury-targeting oligoTRAFTAC OT17
  • UM-Chor1 chordoma cells
  • FIG.5A A similar degradation pattern was noticed in another chordoma cell line, JHC-7 (FIG.5B), although OT17 was slightly less potent in them, requiring 30 nM to induce brachyury knockdown comparable to UM-Chor1 cells.
  • oligoTRAFTAC-mediated brachyury ubiquitination resulting from induced proximity to VHL HA-ubiquitin was transfected into HEK293 cells overexpressing brachyury-GFP. After cells pre-incubated with proteasome inhibitor epoxomicin (500 nM), OT3 and OT17 were transfected and incubated for 12 hours.
  • brachyury overexpression in adult tissue is one of the key factors that leads to tumorigenesis in chordoma
  • brachyury is widely known for its essential biological activity in vertebrate notochord formation at early stages of embryonic development.
  • tail deformation in OT3-injected zebrafish embryos was examined relative to mock-injected embryos (FIG.6A).
  • OT3 could induce brachyury degradation in cells, OT3 did not induce tail deformation in zebrafish, possibly due to the sensitivity of its phosphodiester backbone to nucleases.
  • Described herein are coupled TF-binding short DNA sequences from target gene promoters with VHL ligands to create bifunctional molecules for the targeted degradation of those same TFs. Since the binding sequences already have been identified for many TFs, and since synthetic routes for oligonucleotide synthesis are well established and economical, these oligonucleotide sequences can be rapidly employed in oligoTRAFTAC design to use as a versatile tool for both basic discovery biology and therapy development.
  • the TRAFTACs described herein were synthesized by directly attaching a VHL ligand to c-Myc or brachyury-binding oligonucleotide sequences via click chemistry.
  • OligoTRAFTACs targeting c-Myc TF displayed a robust degradation. Addition of VHL ligand to either side of the oligonucleotide did not significantly alter its ability to induce c- Myc degradation. This can be partially attributed to the flexibility of oligoTRAFTACs provided by the extra flanking nucleotides between the VHL ligand and TF-recruiting oligonucleotide. Other oligoTRAFTACs used in this study induced the degradation of ectopically expressed brachyury-GFP and endogenous brachyury via the proteasomal pathway.
  • oligoTRAFTACs are not as kinetically efficient as conventional PROTACs
  • the transient binding nature of oligoTRAFTACs to both TF and VHL proteins suggests the probability of a similar catalytic degradation mechanism.
  • the compositions and methods described herein represent the first evidence of the degradability of untagged, endogenous brachyury in multiple chordoma cell lines.
  • OT3 containing a phosphodiester backbone
  • first generation TRAFTACs where oligonucleotides are partly protected by the ribonuclear complex, induced defective tails in zebrafish. Therefore, a potential reason for the failure of OT3 to induce the intended phenotype within the embryos may be their high sensitivity and exposure of oligoTRAFTACs to embryonic nucleases present at the early stages of development. Therefore, phosphodiester oligoTRAFTACs might not endure in vivo at the intracellular concentrations needed to achieve a significant brachyury degradation and the intended phenotype.
  • oligoTRAFTACs are programmable heterobifunctional molecules comprised of a TF-binding oligonucleotide sequence and a VHL binding ligand, which induce TF degradation in cells and displayed robust in vivo activity. Due to the simple and modular nature of their structure, oligoTRAFTACs can be rapidly designed for many non-ligandable DNA-binding TFs both for use as a chemical biology tool as well as a potential therapeutic strategy.
  • TFs such as TCF3
  • TCF3 also recognizes a similar E-box consensus sequence as c-Myc
  • the evaluation of TCF3 degradation indicated a sequence-dependent off-target activity of OT7 (FIG.8F).
  • NF-kB p65 protein levels in response to brachyury-targeting OT17.
  • p65 NF-kB
  • oligoTRAFTACs are programmable heterobifunctional molecules comprised of a TF-binding oligonucleotide sequence and a VHL binding ligand, which induce TF degradation in cells and displayed robust in vivo activity. Due to the simple and modular nature of their structure, oligoTRAFTACs can be rapidly designed for many non-ligandable DNA-binding TFs both for use as a chemical biology tool as well as a potential therapeutic strategy.
  • the compounds described herein can possess one or more stereocenters, and each stereocenter can exist independently in either the (R) or (S) configuration. In certain embodiments, compounds described herein are present in optically active or racemic forms.
  • the compounds described herein encompass racemic, optically-active, regioisomeric and stereoisomeric forms, or combinations thereof that possess the therapeutically useful properties described herein.
  • Preparation of optically active forms is achieved in any suitable manner, including by way of non-limiting example, by resolution of the racemic form with recrystallization techniques, synthesis from optically-active starting materials, chiral synthesis, or chromatographic separation using a chiral stationary phase.
  • a mixture of one or more isomer is utilized as the therapeutic compound described herein.
  • compounds described herein contain one or more chiral centers.
  • These compounds are prepared by any means, including stereoselective synthesis, enantioselective synthesis and/or separation of a mixture of enantiomers and/ or diastereomers. Resolution of compounds and isomers thereof is achieved by any means including, by way of non-limiting example, chemical processes, enzymatic processes, fractional crystallization, distillation, and chromatography.
  • the methods and formulations described herein include the use of N-oxides (if appropriate), crystalline forms (also known as polymorphs), solvates, amorphous phases, and/or pharmaceutically acceptable salts of compounds having the structure of any compound(s) described herein, as well as metabolites and active metabolites of these compounds having the same type of activity.
  • Solvates include water, ether (e.g., tetrahydrofuran, methyl tert-butyl ether) or alcohol (e.g., ethanol) solvates, acetates and the like.
  • the compounds described herein exist in solvated forms with pharmaceutically acceptable solvents such as water, and ethanol.
  • the compounds described herein exist in unsolvated form.
  • the compound(s) described herein can exist as tautomers. All tautomers are included within the scope of the compounds presented herein.
  • compounds described herein are prepared as prodrugs.
  • a "prodrug" refers to an agent that is converted into the parent drug in vivo.
  • a prodrug upon in vivo administration, is chemically converted to the biologically, pharmaceutically or therapeutically active form of the compound.
  • a prodrug is enzymatically metabolized by one or more steps or processes to the biologically, pharmaceutically or therapeutically active form of the compound.
  • sites on, for example, the aromatic ring portion of compound(s) described herein are susceptible to various metabolic reactions. Incorporation of appropriate substituents on the aromatic ring structures may reduce, minimize or eliminate this metabolic pathway.
  • the appropriate substituent to decrease or eliminate the susceptibility of the aromatic ring to metabolic reactions is, by way of example only, a deuterium, a halogen, or an alkyl group.
  • Compounds described herein also include isotopically-labeled compounds wherein one or more atoms is replaced by an atom having the same atomic number, but an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
  • isotopes suitable for inclusion in the compounds described herein include and are not limited to 2 H, 3 H, 11 C, 13 C, 14 C, 36 Cl, 18 F, 123 I, 125 I, 13 N, 15 N, 15 O, 17 O, 18 O, 32 P, and 35 S.
  • isotopically-labeled compounds are useful in drug and/or substrate tissue distribution studies.
  • substitution with heavier isotopes such as deuterium affords greater metabolic stability (for example, increased in vivo half-life or reduced dosage requirements).
  • substitution with positron emitting isotopes, such as 11 C, 18 F, 15 O and 13 N is useful in Positron Emission Topography (PET) studies for examining substrate receptor occupancy.
  • Isotopically-labeled compounds are prepared by any suitable method or by processes using an appropriate isotopically-labeled reagent in place of the non-labeled reagent otherwise employed.
  • the compounds described herein are labeled by other means, including, but not limited to, the use of chromophores or fluorescent moieties, bioluminescent labels, or chemiluminescent labels.
  • the compounds described herein, and other related compounds having different substituents are synthesized using techniques and materials described herein and as described, for example, in Fieser & Fieser's Reagents for Organic Synthesis, Volumes 1-17 (John Wiley and Sons, 1991); Rodd's Chemistry of Carbon Compounds, Volumes 1-5 and Supplementals (Elsevier Science Publishers, 1989); Organic Reactions, Volumes 1-40 (John Wiley and Sons, 1991), Larock's Comprehensive Organic Transformations (VCH Publishers Inc., 1989), March, Advanced Organic Chemistry 4 th Ed., (Wiley 1992); Carey & Sundberg, Advanced Organic Chemistry 4th Ed., Vols.
  • Protecting groups are used to block some or all of the reactive moieties and prevent such groups from participating in chemical reactions until the protective group is removed.
  • each protective group is removable by a different means.
  • Protective groups that are cleaved under totally disparate reaction conditions fulfill the requirement of differential removal.
  • protective groups are removed by acid, base, reducing conditions (such as, for example, hydrogenolysis), and/or oxidative conditions.
  • Groups such as trityl, dimethoxytrityl, acetal and t-butyldimethylsilyl are acid labile and are used to protect carboxy and hydroxy reactive moieties in the presence of amino groups protected with Cbz groups, which are removable by hydrogenolysis, and Fmoc groups, which are base labile.
  • Carboxylic acid and hydroxy reactive moieties are blocked with base labile groups such as, but not limited to, methyl, ethyl, and acetyl, in the presence of amines that are blocked with acid labile groups, such as t-butyl carbamate, or with carbamates that are both acid and base stable but hydrolytically removable.
  • carboxylic acid and hydroxy reactive moieties are blocked with hydrolytically removable protective groups such as the benzyl group, while amine groups capable of hydrogen bonding with acids are blocked with base labile groups such as Fmoc.
  • Carboxylic acid reactive moieties are protected by conversion to simple ester compounds as exemplified herein, which include conversion to alkyl esters, or are blocked with oxidatively-removable protective groups such as 2,4-dimethoxybenzyl, while co- existing amino groups are blocked with fluoride labile silyl carbamates. Allyl blocking groups are useful in the presence of acid- and base- protecting groups since the former are stable and are subsequently removed by metal or pi-acid catalysts.
  • an allyl-blocked carboxylic acid is deprotected with a palladium-catalyzed reaction in the presence of acid labile t-butyl carbamate or base-labile acetate amine protecting groups.
  • Another form of protecting group is a resin to which a compound or intermediate is attached. As long as the residue is attached to the resin, that functional group is blocked and does not react. Once released from the resin, the functional group is available to react.
  • blocking/protecting groups may be selected from: .
  • compositions containing the compound(s) described herein include a pharmaceutical composition comprising at least one compound as described herein and at least one pharmaceutically acceptable carrier.
  • the composition is formulated for an administration route such as oral or parenteral, for example, transdermal, transmucosal (e.g., sublingual, lingual, (trans)buccal, (trans)urethral, vaginal (e.g., trans- and perivaginally), (intra)nasal and (trans)rectal, intravesical, intrapulmonary, intraduodenal, intragastrical, intrathecal, subcutaneous, intramuscular, intradermal, intra-arterial, intravenous, intrabronchial, inhalation, and topical administration.
  • Methods of Treatment, Amelioration, and/or Prevention The disclosure includes a method of treating, ameliorating, and/or preventing cancer using the compounds of formula I.
  • Non-limiting examples of cancer include squamous-cell carcinoma, basal cell carcinoma, adenocarcinoma, hepatocellular carcinomas, renal cell carcinoma, cancer of the bladder, bowel, breast, cervix, colon, esophagus, head, kidney, liver, lung, neck, ovary, pancreas, prostate, and stomach; leukemias; benign and malignant lymphomas, particularly Burkitt's lymphoma and Non-Hodgkin's lymphoma; benign and malignant melanomas; myeloproliferative diseases; multiple myeloma, sarcomas, including Ewing's sarcoma, hemangiosarcoma, Kaposi's sarcoma, liposarcoma, myosarcomas, peripheral neuroepithelioma, synovial sarcoma, gliomas, astrocytomas, oligodendrogliomas, ependymomas, gli
  • the methods described herein include administering to the subject a therapeutically effective amount of at least one compound described herein, which is optionally formulated in a pharmaceutical composition.
  • a therapeutically effective amount of at least one compound described herein present in a pharmaceutical composition is the only therapeutically active compound in a pharmaceutical composition.
  • the method further comprises administering to the subject an additional therapeutic agent that treats cancer.
  • administering the compound(s) described herein to the subject allows for administering a lower dose of the additional therapeutic agent as compared to the dose of the additional therapeutic agent alone that is required to achieve similar results in treating a cancer in the subject.
  • the compound(s) described herein enhance(s) the activity of the additional therapeutic compound, thereby allowing for a lower dose of the additional therapeutic compound to provide the same effect.
  • the compound(s) described herein and the therapeutic agent are co-administered to the subject.
  • the compound(s) described herein and the therapeutic agent are coformulated and co-administered to the subject.
  • the subject is a mammal. In other embodiments, the mammal is a human.
  • Combination Therapies The compounds useful within the methods described herein can be used in combination with one or more additional therapeutic agents useful for treating, ameliorating, and/or preventing cancer.
  • additional therapeutic agents may comprise compounds that are commercially available or synthetically accessible to those skilled in the art. These additional therapeutic agents are known to treat, ameliorate, prevent, and/or reduce the symptoms, of cancer.
  • the compounds described herein can be used in combination with radiation therapy.
  • the combination of administration of the compounds described herein and application of radiation therapy is more effective in treating, ameliorating, or preventing cancer than application of radiation therapy by itself.
  • the combination of administration of the compounds described herein and application of radiation therapy allows for use of lower amount of radiation therapy in treating the subject.
  • a synergistic effect is observed when a compound as described herein is administered with one or more additional therapeutic agents or compounds.
  • a synergistic effect may be calculated, for example, using suitable methods such as, for example, the Sigmoid-Emax equation (Holford & Scheiner, 1981, Clin. Pharmacokinet. 6:429-453), the equation of Loewe additivity (Loewe & Muischnek, 1926, Arch. Exp. Pathol Pharmacol.114:313-326) and the median-effect equation (Chou & Talalay, 1984, Adv. Enzyme Regul.22:27-55).
  • Sigmoid-Emax equation Holford & Scheiner, 1981, Clin. Pharmacokinet. 6:429-453
  • Loewe additivity Loewe & Muischnek, 1926, Arch. Exp. Pathol Pharmacol.114:313-326
  • the median-effect equation Chou & Talalay, 1984, Adv. Enzyme Regul.22:27-55.
  • the corresponding graphs associated with the equations referred to above are the concentration-effect curve, isobologram curve and combination index curve, respectively.
  • Administration/Dosage/Formulations The regimen of administration may affect what constitutes an effective amount.
  • the therapeutic formulations may be administered to the subject either prior to or after the onset of cancer. Further, several divided dosages, as well as staggered dosages may be administered daily or sequentially, or the dose may be continuously infused, or may be a bolus injection. Further, the dosages of the therapeutic formulations may be proportionally increased or decreased as indicated by the exigencies of the therapeutic or prophylactic situation.
  • compositions described herein to a patient may be carried out using known procedures, at dosages and for periods of time effective to treat cacner in the patient.
  • An effective amount of the therapeutic compound necessary to achieve a therapeutic effect may vary according to factors such as the state of the disease or disorder in the patient; the age, sex, and weight of the patient; and the ability of the therapeutic compound to treat cancer in the patient. Dosage regimens may be adjusted to provide the optimum therapeutic response. For example, several divided doses may be administered daily or the dose may be proportionally reduced as indicated by the exigencies of the therapeutic situation.
  • a non-limiting example of an effective dose range for a therapeutic compound described herein is from about 1 and 5,000 mg/kg of body weight/per day.
  • One of ordinary skill in the art would be able to study the relevant factors and make the determination regarding the effective amount of the therapeutic compound without undue experimentation.
  • Actual dosage levels of the active ingredients in the pharmaceutical compositions described herein may be varied so as to obtain an amount of the active ingredient that is effective to achieve the desired therapeutic response for a particular patient, composition, and mode of administration, without being toxic to the patient.
  • the selected dosage level depends upon a variety of factors including the activity of the particular compound employed, the time of administration, the rate of excretion of the compound, the duration of the treatment, other drugs, compounds or materials used in combination with the compound, the age, sex, weight, condition, general health and prior medical history of the patient being treated, and like factors well, known in the medical arts.
  • a medical doctor e.g., physician or veterinarian, having ordinary skill in the art may readily determine and prescribe the effective amount of the pharmaceutical composition required.
  • the physician or veterinarian could start doses of the compounds described herein employed in the pharmaceutical composition at levels lower than that required in order to achieve the desired therapeutic effect and gradually increase the dosage until the desired effect is achieved.
  • Dosage unit form refers to physically discrete units suited as unitary dosages for the patients to be treated; each unit containing a predetermined quantity of therapeutic compound calculated to produce the desired therapeutic effect in association with the required pharmaceutical vehicle.
  • the dosage unit forms of the compound(s) described herein are dictated by and directly dependent on (a) the unique characteristics of the therapeutic compound and the particular therapeutic effect to be achieved, and (b) the limitations inherent in the art of compounding/formulating such a therapeutic compound.
  • the compositions described herein are formulated using one or more pharmaceutically acceptable excipients or carriers.
  • the pharmaceutical compositions described herein comprise a therapeutically effective amount of a compound described herein and a pharmaceutically acceptable carrier.
  • the carrier may be a solvent or dispersion medium containing, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils.
  • the proper fluidity may be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants.
  • compositions described herein are administered to the patient in dosages that range from one to five times per day or more.
  • compositions described herein are administered to the patient in range of dosages that include, but are not limited to, once every day, every two, days, every three days to once a week, and once every two weeks. It is readily apparent to one skilled in the art that the frequency of administration of the various combination compositions described herein varies from individual to individual depending on many factors including, but not limited to, age, disease or disorder to be treated, gender, overall health, and other factors. Thus, administration of the compounds and compositions described herein should not be construed to be limited to any particular dosage regime and the precise dosage and composition to be administered to any patient is determined by the attending physician taking all other factors about the patient into account.
  • the compound(s) described herein for administration may be in the range of from about 1 ⁇ g to about 10,000 mg, about 20 ⁇ g to about 9,500 mg, about 40 ⁇ g to about 9,000 mg, about 75 ⁇ g to about 8,500 mg, about 150 ⁇ g to about 7,500 mg, about 200 ⁇ g to about 7,000 mg, about 350 ⁇ g to about 6,000 mg, about 500 ⁇ g to about 5,000 mg, about 750 ⁇ g to about 4,000 mg, about 1 mg to about 3,000 mg, about 10 mg to about 2,500 mg, about 20 mg to about 2,000 mg, about 25 mg to about 1,500 mg, about 30 mg to about 1,000 mg, about 40 mg to about 900 mg, about 50 mg to about 800 mg, about 60 mg to about 750 mg, about 70 mg to about 600 mg, about 80 mg to about 500 mg, and any and all whole or partial increments therebetween.
  • the dose of a compound described herein is from about 1 mg and about 2,500 mg. In some embodiments, a dose of a compound described herein used in compositions described herein is less than about 10,000 mg, or less than about 8,000 mg, or less than about 6,000 mg, or less than about 5,000 mg, or less than about 3,000 mg, or less than about 2,000 mg, or less than about 1,000 mg, or less than about 500 mg, or less than about 200 mg, or less than about 50 mg.
  • a dose of a second compound as described herein is less than about 1,000 mg, or less than about 800 mg, or less than about 600 mg, or less than about 500 mg, or less than about 400 mg, or less than about 300 mg, or less than about 200 mg, or less than about 100 mg, or less than about 50 mg, or less than about 40 mg, or less than about 30 mg, or less than about 25 mg, or less than about 20 mg, or less than about 15 mg, or less than about 10 mg, or less than about 5 mg, or less than about 2 mg, or less than about 1 mg, or less than about 0.5 mg, and any and all whole or partial increments thereof.
  • a composition as described herein is a packaged pharmaceutical composition
  • Formulations may be employed in admixtures with conventional excipients, i.e., pharmaceutically acceptable organic or inorganic carrier substances suitable for oral, parenteral, nasal, intravenous, subcutaneous, enteral, or any other suitable mode of administration, known to the art.
  • the pharmaceutical preparations may be sterilized and if desired mixed with auxiliary agents, e.g., lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure buffers, coloring, flavoring and/or aromatic substances and the like. They may also be combined where desired with other active agents, e.g., other analgesic agents.
  • auxiliary agents e.g., lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure buffers, coloring, flavoring and/or aromatic substances and the like.
  • auxiliary agents e.g., lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure buffers, coloring, flavoring and/or aromatic substances and the like.
  • other active agents e.g., other analgesic agents.
  • the compounds for use in the compositions described herein can be formulated for administration by any suitable route, such as for oral or parenteral, for example, transdermal, transmucosal (e.g., sublingual, lingual, (trans)buccal, (trans)urethral, vaginal (e.g., trans- and perivaginally), (intra)nasal and (trans)rectal), intravesical, intrapulmonary, intraduodenal, intragastrical, intrathecal, subcutaneous, intramuscular, intradermal, intra-arterial, intravenous, intrabronchial, inhalation, and topical administration.
  • transdermal e.g., sublingual, lingual, (trans)buccal, (trans)urethral
  • vaginal e.g., trans- and perivaginally
  • intravesical, intrapulmonary, intraduodenal, intragastrical intrathecal
  • compositions and dosage forms include, for example, tablets, capsules, caplets, pills, gel caps, troches, dispersions, suspensions, solutions, syrups, granules, beads, transdermal patches, gels, powders, pellets, magmas, lozenges, creams, pastes, plasters, lotions, discs, suppositories, liquid sprays for nasal or oral administration, dry powder or aerosolized formulations for inhalation, compositions and formulations for intravesical administration and the like. It should be understood that the formulations and compositions described herein are not limited to the particular formulations and compositions that are described herein.
  • compositions intended for oral use may be prepared according to any method known in the art and such compositions may contain one or more agents selected from the group consisting of inert, non-toxic pharmaceutically excipients that are suitable for the manufacture of tablets.
  • excipients include, for example an inert diluent such as lactose; granulating and disintegrating agents such as cornstarch; binding agents such as starch; and lubricating agents such as magnesium stearate.
  • the tablets may be uncoated or they may be coated by known techniques for elegance or to delay the release of the active ingredients.
  • Formulations for oral use may also be presented as hard gelatin capsules wherein the active ingredient is mixed with an inert diluent.
  • the compound(s) described herein can be in the form of tablets or capsules prepared by conventional means with pharmaceutically acceptable excipients such as binding agents (e.g., polyvinylpyrrolidone, hydroxypropylcellulose or hydroxypropyl methylcellulose); fillers (e.g., cornstarch, lactose, microcrystalline cellulose or calcium phosphate); lubricants (e.g., magnesium stearate, talc, or silica); disintegrates (e.g., sodium starch glycollate); or wetting agents (e.g., sodium lauryl sulphate).
  • binding agents e.g., polyvinylpyrrolidone, hydroxypropylcellulose or hydroxypropyl methylcellulose
  • fillers e.g., cornstarch, lactose, microcrystalline cellulose or calcium phosphate
  • the tablets may be coated using suitable methods and coating materials such as OPADRYTM film coating systems available from Colorcon, West Point, Pa. (e.g., OPADRYTM OY Type, OYC Type, Organic Enteric OY-P Type, Aqueous Enteric OY-A Type, OY-PM Type and OPADRYTM White, 32K18400).
  • OPADRYTM film coating systems available from Colorcon, West Point, Pa. (e.g., OPADRYTM OY Type, OYC Type, Organic Enteric OY-P Type, Aqueous Enteric OY-A Type, OY-PM Type and OPADRYTM White, 32K18400).
  • Liquid preparation for oral administration may be in the form of solutions, syrups or suspensions.
  • the liquid preparations may be prepared by conventional means with pharmaceutically acceptable additives such as suspending agents (e.g., sorbitol syrup, methyl cellulose or hydrogenated edible fats); emulsifying agent (e.g., lecithin or acacia); non-aqueous vehicles (e.g., almond oil, oily esters or ethyl alcohol); and preservatives (e.g., methyl or propyl p-hydroxy benzoates or sorbic acid).
  • suspending agents e.g., sorbitol syrup, methyl cellulose or hydrogenated edible fats
  • emulsifying agent e.g., lecithin or acacia
  • non-aqueous vehicles e.g., almond oil, oily esters or ethyl alcohol
  • preservatives e.g., methyl or propyl p-hydroxy benzoates or sorbic acid.
  • Compositions as described herein can be prepared, packaged, or sold in a
  • Compressed tablets may be prepared by compressing, in a suitable device, the active ingredient in a free-flowing form such as a powder or granular preparation, optionally mixed with one or more of a binder, a lubricant, an excipient, a surface active agent, and a dispersing agent. Molded tablets may be made by molding, in a suitable device, a mixture of the active ingredient, a pharmaceutically acceptable carrier, and at least sufficient liquid to moisten the mixture.
  • Pharmaceutically acceptable excipients used in the manufacture of tablets include, but are not limited to, inert diluents, granulating and disintegrating agents, dispersing agents, surface-active agents, disintegrating agents, binding agents, and lubricating agents.
  • Suitable dispersing agents include, but are not limited to, potato starch, sodium starch glycollate, poloxamer 407, or poloxamer 188.
  • One or more dispersing agents can each be individually present in the composition in an amount of about 0.01% w/w to about 90% w/w relative to weight of the dosage form.
  • One or more dispersing agents can each be individually present in the composition in an amount of at least, greater than, or less than about 0.01%, 0.05%, 0.1%, 0.5%, 1%, 2%, 3%, 4%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, or 90% w/w relative to weight of the dosage form.
  • Surface-active agents include cationic, anionic, or non-ionic surfactants, or combinations thereof.
  • Suitable surfactants include, but are not limited to, behentrimonium chloride, benzalkonium chloride, benzethonium chloride, benzododecinium bromide, carbethopendecinium bromide, cetalkonium chloride, cetrimonium bromide, cetrimonium chloride, cetylpyridine chloride, didecyldimethylammonium chloride, dimethyldioctadecylammonium bromide, dimethyldioctadecylammonium chloride, domiphen bromide, lauryl methyl gluceth-10 hydroxypropyl dimonium chloride, tetramethylammonium hydroxide, thonzonium bromide, stearalkonium chloride, octenidine dihydrochloride, olaflur, N-oleyl-1,3-propanediamine, 2-acrylamido-2-methylpropane sulfonic acid, alkylbenzen
  • One or more surfactants can each be individually present in the composition in an amount of about 0.01% w/w to about 90% w/w relative to weight of the dosage form.
  • One or more surfactants can each be individually present in the composition in an amount of at least, greater than, or less than about 0.01%, 0.05%, 0.1%, 0.5%, 1%, 2%, 3%, 4%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, or 90% w/w relative to weight of the dosage form.
  • Suitable diluents include, but are not limited to, calcium carbonate, magnesium carbonate, magnesium oxide, sodium carbonate, lactose, microcrystalline cellulose, calcium phosphate, calcium hydrogen phosphate, and sodium phosphate, Cellactose ® 80 (75 % ⁇ - lactose monohydrate and 25 % cellulose powder), mannitol, pre-gelatinized starch, starch, sucrose, sodium chloride, talc, anhydrous lactose, and granulated lactose.
  • One or more diluents can each be individually present in the composition in an amount of about 0.01% w/w to about 90% w/w relative to weight of the dosage form.
  • One or more diluents can each be individually present in the composition in an amount of at least, greater than, or less than about 0.01%, 0.05%, 0.1%, 0.5%, 1%, 2%, 3%, 4%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, or 90% w/w relative to weight of the dosage form.
  • Suitable granulating and disintegrating agents include, but are not limited to, sucrose, copovidone, corn starch, microcrystalline cellulose, methyl cellulose, sodium starch glycollate, pregelatinized starch, povidone, sodium carboxy methyl cellulose, sodium alginate, citric acid, croscarmellose sodium, cellulose, carboxymethylcellulose calcium, colloidal silicone dioxide, crosspovidone and alginic acid.
  • One or more granulating or disintegrating agents can each be individually present in the composition in an amount of about 0.01% w/w to about 90% w/w relative to weight of the dosage form.
  • One or more granulating or disintegrating agents can each be individually present in the composition in an amount of at least, greater than, or less than about 0.01%, 0.05%, 0.1%, 0.5%, 1%, 2%, 3%, 4%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, or 90% w/w relative to weight of the dosage form.
  • Suitable binding agents include, but are not limited to, gelatin, acacia, pre-gelatinized maize starch, polyvinylpyrrolidone, anhydrous lactose, lactose monohydrate, hydroxypropyl methylcellulose, methylcellulose, povidone, polyacrylamides, sucrose, dextrose, maltose, gelatin, polyethylene glycol.
  • One or more binding agents can each be individually present in the composition in an amount of about 0.01% w/w to about 90% w/w relative to weight of the dosage form.
  • One or more binding agents can each be individually present in the composition in an amount of at least, greater than, or less than about 0.01%, 0.05%, 0.1%, 0.5%, 1%, 2%, 3%, 4%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, or 90% w/w relative to weight of the dosage form.
  • Suitable lubricating agents include, but are not limited to, magnesium stearate, calcium stearate, hydrogenated castor oil, glyceryl monostearate, glyceryl behenate, mineral oil, polyethylene glycol, poloxamer 407, poloxamer 188, sodium laureth sulfate, sodium benzoate, stearic acid, sodium stearyl fumarate, silica, and talc.
  • One or more lubricating agents can each be individually present in the composition in an amount of about 0.01% w/w to about 90% w/w relative to weight of the dosage form.
  • One or more lubricating agents can each be individually present in the composition in an amount of at least, greater than, or less than about 0.01%, 0.05%, 0.1%, 0.5%, 1%, 2%, 3%, 4%, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, or 90% w/w relative to weight of the dosage form.
  • Tablets can be non-coated or they may be coated using known methods to achieve delayed disintegration in the gastrointestinal tract of a subject, thereby providing sustained release and absorption of the active ingredient.
  • a material such as glyceryl monostearate or glyceryl distearate may be used to coat tablets.
  • tablets may be coated using methods described in U.S. Patent Nos.4,256,108; 4,160,452; and 4,265,874 to form osmotically controlled release tablets.
  • Tablets may further comprise a sweetening agent, a flavoring agent, a coloring agent, a preservative, or some combination of these in order to provide for pharmaceutically elegant and palatable preparation. Tablets can also be enterically coated such that the coating begins to dissolve at a certain pH, such as at about pH 5.0 to about pH 7.5, thereby releasing a compound as described herein.
  • the coating can contain, for example, EUDRAGIT ® L, S, FS, and/or E polymers with acidic or alkaline groups to allow release of a compound as described herein in a particular location, including in any desired section(s) of the intestine.
  • the coating can also contain, for example, EUDRAGIT ® RL and/or RS polymers with cationic or neutral groups to allow for time controlled release of a compound as described herein by pH-independent swelling.
  • Parenteral Administration For parenteral administration, the compounds as described herein may be formulated for injection or infusion, for example, intravenous, intramuscular or subcutaneous injection or infusion, or for administration in a bolus dose and/or continuous infusion.
  • Suspensions, solutions or emulsions in an oily or aqueous vehicle, optionally containing other formulatory agents such as suspending, stabilizing and/or dispersing agents may be used.
  • Sterile injectable forms of the compositions described herein may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may 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.
  • the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • Sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or di-glycerides.
  • 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.
  • These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, such as such as lauryl, stearyl, or oleyl alcohols, or similar alcohol.
  • Additional Administration Forms Additional dosage forms suitable for use with the compound(s) and compositions described herein include dosage forms as described in U.S.
  • Controlled Release Formulations and Drug Delivery Systems can be, but are not limited to, short-term, rapid-offset, as well as controlled, for example, sustained release, delayed release and pulsatile release formulations.
  • sustained release is used in its conventional sense to refer to a drug formulation that provides for gradual release of a drug over an extended period of time, and that may, although not necessarily, result in substantially constant blood levels of a drug over an extended time period.
  • the period of time may be as long as a month or more and should be a release which is longer that the same amount of agent administered in bolus form.
  • the compounds may be formulated with a suitable polymer or hydrophobic material which provides sustained release properties to the compounds.
  • the compounds for use with the method(s) described herein may be administered in the form of microparticles, for example, by injection or in the form of wafers or discs by implantation.
  • the dosage forms to be used can be provided as slow or controlled- release of one or more active ingredients therein using, for example, hydropropylmethyl cellulose, other polymer matrices, gels, permeable membranes, osmotic systems, multilayer coatings, microparticles, liposomes, or microspheres or a combination thereof to provide the desired release profile in varying proportions.
  • Suitable controlled-release formulations known to those of ordinary skill in the art, including those described herein can be readily selected for use with the pharmaceutical compositions described herein.
  • single unit dosage forms suitable for oral administration such as tablets, capsules, gelcaps, and caplets, that are adapted for controlled-release are encompassed by the compositions and dosage forms described herein.
  • controlled-release pharmaceutical products have a common goal of improving drug therapy over that achieved by their non-controlled counterparts.
  • the use of an optimally designed controlled-release preparation in medical treatment is characterized by a minimum of drug substance being employed to cure or control the condition in a minimum amount of time.
  • Advantages of controlled-release formulations include extended activity of the drug, reduced dosage frequency, and increased patient compliance.
  • controlled-release formulations can be used to affect the time of onset of action or other characteristics, such as blood level of the drug, and thus can affect the occurrence of side effects.
  • Most controlled-release formulations are designed to initially release an amount of drug that promptly produces the desired therapeutic effect, and gradually and continually release of other amounts of drug to maintain this level of therapeutic effect over an extended period of time.
  • Controlled-release of an active ingredient can be stimulated by various inducers, for example pH, temperature, enzymes, water, or other physiological conditions or compounds.
  • the term "controlled-release component" is defined herein as a compound or compounds, including, but not limited to, polymers, polymer matrices, gels, permeable membranes, liposomes, or microspheres or a combination thereof that facilitates the controlled-release of the active ingredient.
  • the compound(s) described herein are administered to a patient, alone or in combination with another pharmaceutical agent, using a sustained release formulation.
  • the compound(s) described herein are administered to a patient, alone or in combination with another pharmaceutical agent, using a sustained release formulation.
  • delayed release is used herein in its conventional sense to refer to a drug formulation that provides for an initial release of the drug after some delay following drug administration and that mat, although not necessarily, includes a delay of from about 10 minutes up to about 12 hours.
  • pulsatile release is used herein in its conventional sense to refer to a drug formulation that provides release of the drug in such a way as to produce pulsed plasma profiles of the drug after drug administration.
  • immediate release is used in its conventional sense to refer to a drug formulation that provides for release of the drug immediately after drug administration.
  • short-term refers to any period of time up to and including about 8 hours, about 7 hours, about 6 hours, about 5 hours, about 4 hours, about 3 hours, about 2 hours, about 1 hour, about 40 minutes, about 20 minutes, or about 10 minutes and any or all whole or partial increments thereof after drug administration after drug administration.
  • rapid-offset refers to any period of time up to and including about 8 hours, about 7 hours, about 6 hours, about 5 hours, about 4 hours, about 3 hours, about 2 hours, about 1 hour, about 40 minutes, about 20 minutes, or about 10 minutes, and any and all whole or partial increments thereof after drug administration.
  • the therapeutically effective amount or dose of a compound described herein depends on the age, sex and weight of the patient, the current medical condition of the patient and the progression of cancer in the patient being treated. The skilled artisan is able to determine appropriate dosages depending on these and other factors.
  • a suitable dose of a compound described herein can be in the range of from about 0.01 mg to about 5,000 mg per day, such as from about 0.1 mg to about 1,000 mg, for example, from about 1 mg to about 500 mg, such as about 5 mg to about 250 mg per day.
  • the dose may be administered in a single dosage or in multiple dosages, for example from 1 to 4 or more times per day. When multiple dosages are used, the amount of each dosage may be the same or different.
  • a dose of 1 mg per day may be administered as two 0.5 mg doses, with about a 12-hour interval between doses. It is understood that the amount of compound dosed per day may be administered, in non-limiting examples, every day, every other day, every 2 days, every 3 days, every 4 days, or every 5 days. For example, with every other day administration, a 5 mg per day dose may be initiated on Monday with a first subsequent 5 mg per day dose administered on Wednesday, a second subsequent 5 mg per day dose administered on Friday, and so on.
  • the administration of the compound(s) described herein is optionally given continuously; alternatively, the dose of drug being administered is temporarily reduced or temporarily suspended for a certain length of time (i.e., a "drug holiday").
  • the length of the drug holiday optionally varies between 2 days and 1 year, including by way of example only, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 10 days, 12 days, 15 days, 20 days, 28 days, 35 days, 50 days, 70 days, 100 days, 120 days, 150 days, 180 days, 200 days, 250 days, 280 days, 300 days, 320 days, 350 days, or 365 days.
  • the dose reduction during a drug holiday includes from 10%-100%, including, by way of example only, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, or 100%.
  • a maintenance dose is administered if necessary.
  • the dosage or the frequency of administration, or both is reduced to a level at which the improved disease is retained.
  • patients require intermittent treatment on a long-term basis upon any recurrence of symptoms and/or infection.
  • the compounds described herein can be formulated in unit dosage form.
  • unit dosage form refers to physically discrete units suitable as unitary dosage for patients undergoing treatment, with each unit containing a predetermined quantity of active material calculated to produce the desired therapeutic effect, optionally in association with a suitable pharmaceutical carrier.
  • the unit dosage form may be for a single daily dose or one of multiple daily doses (e.g., about 1 to 4 or more times per day). When multiple daily doses are used, the unit dosage form may be the same or different for each dose. Toxicity and therapeutic efficacy of such therapeutic regimens are optionally determined in cell cultures or experimental animals, including, but not limited to, the determination of the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population).
  • the dose ratio between the toxic and therapeutic effects is the therapeutic index, which is expressed as the ratio between LD50 and ED 50 .
  • the data obtained from cell culture assays and animal studies are optionally used in formulating a range of dosage for use in human.
  • the dosage of such compounds lies preferably within a range of circulating concentrations that include the ED 50 with minimal toxicity.
  • the dosage optionally varies within this range depending upon the dosage form employed and the route of administration utilized. Examples Various embodiments of the present application can be better understood by reference to the following Examples which are offered by way of illustration. The scope of the present application is not limited to the Examples given herein.
  • DMEM fetal bovine serum
  • IMDM fetal bovine serum
  • RPMI fetal bovine serum
  • DMEM/F-12 fetal bovine serum
  • Primary antibodies for GAPDH (2118S), Vinculin (13901S), brachyury (81694S), HA-tag (3724S), protein A magnetic beads (73778S) and streptavidin magnetic beads (5947S) were purchased from Cell Signaling Technologies.
  • Primary antibody for c-Myc (sc-40) was purchased from Santa Cruz. Secondary rabbit (NA934) and mouse (NA931) antibodies were purchased from GE Health Care.
  • RNAiMAX (13778-150) transfecting reagent was purchased from Thermo Fisher Scientific.
  • MLN4924 (S7109) was purchased from Selleckchem. Copper (II) sulfate pentahydrate (209198) and L-Ascorbic acid (A4403) were purchased from Millipore Sigma, and THPTA purchased from Lumiprobe (H4050). All the oligonucleotide modifiers were purchased from Glen Research, 3' Alkyne (20-2992-41), 5' Alkyne (10-1992-90) and biotin (10-5950-90). All the oligonucleotides were custom synthesized by Yale Keck oligo synthesis facility.
  • DMEM Dulbecco's Modified Eagles Medium
  • FBS heat inactivated fetal bovine serum
  • streptomycin 5 ⁇ g/mL
  • penicillin 95 U/mL penicillin 95 U/mL
  • OligoTRAFTAC transfection One day prior to oligoTRAFTACs transfection, cells (HEK293T: 0.7X10 6 /well, HeLa: 0.2 X10 6 /well, UM-Chor1: 0.2X10 6 /well and JHC7: 0.4X10 6 /well) were propagated into 6-well plates containing appropriate complete growth medium. Prior to transfection, complete medium was replaced with 1.75 mL of transfection medium (2%FBS, no Penstrep). Chimeric oligoTRAFTAC transfection was performed using RNAi-Max reagent according to the protocols provided by the manufacture.
  • oligoTRAFTACs concentrations of oligoTRAFTACs were calculated according to this volume (2 mL). Briefly, for 50 nM concentration, 4 ⁇ L from a 25 ⁇ M oligo TRAFTAC stock was added to a tube containing 125 ⁇ L of OPTIM-MEM and 12.5 ⁇ L of RNAi-Max reagent was added to a separate tube containing 125 ⁇ L of OPTIM-MEM (added ⁇ 4 ⁇ g of oligoTRAFTAC to 2 mL cell culture medium).
  • oligoTRAFTAC containing OPTI-MEM was then slowly added to the second tube with RNAi-MAX.
  • the solution in the tube was mixed well by pipetting up and down several times. After incubating for 10 minutes at room temperature, 250 ⁇ L of oligoTRAFTAC:RNAi-MAX complex was added drop wise onto cells containing the transfection medium. Transfection medium was mixed well before transferring the 6-well plate into the incubator. After appropriate time, cells were either harvested or transfection medium containing oligoTRAFTAC:RNAi-MAX complex was replaced with fresh medium and incubated for desired time point prior to harvesting.
  • MLN-4924 and VHL ligand competition assay were pre-incubated in the transfection medium (1.75 mL) for 1 h prior to transfection of oligoTRAFTACs.
  • Cell lysates were prepared by incubating cells in RIPA lysis buffer (25 mM Tris pH 7.6, 150 mM NaCl, 1% NP40, 1% deoxycholate, 0.1% SDS, 1X protease inhibitor cocktail from Roche and 1 mM of PMSF) on ice for 30 minutes and cell lysate was clarified by centrifugation at high speed (15000 rpm) for 20 minutes. Clear supernatant was collected for further experiments.
  • RIPA lysis buffer 25 mM Tris pH 7.6, 150 mM NaCl, 1% NP40, 1% deoxycholate, 0.1% SDS, 1X protease inhibitor cocktail from Roche and 1 mM of PMSF
  • the program was set with a flow rate of 5 mL/min for 150 minutes, and a gradient of ACN increasing from 0-80%.
  • Annealing Reaction FPLC purified single stranded oligo conjugated to VHL (oligo-VHL) ligand and its reverse complement oligo were dissolved in ultra-pure water.
  • Single stranded oligo-VHL and single stranded reverse complement oligos were mixed 1:1 molar ratio (final concentrations of TRAFTACs were set to 25 ⁇ M) in 1X annealing buffer (10 mM Tris, pH 7.5, 50 mM NaCl and 1 mM EDTA) and incubated for 10 minutes in a water bath at 95 degrees Celsius.
  • Double stranded oligoTRAFTACs were mixed by gently vortexing and aliquoted and stored at -20 degrees Celsius.
  • Reverse complementary sequences OT3/17'- 5'CCCAATTTCACACCTAGGTGTGAAATTGGA3', OT7/10- 5' AACCACGTGGCAACCACGTGCTC 3'.
  • Western Blotting Protein concentration in all the cell lysates were measured by BCA protein assay kit and equal amounts from each lysate were mixed with 4X loading dye and boiled for 5 minutes followed by 2 minutes centrifugation prior to loading into SDS-PAGE gel.
  • EMSA Electrophoretic Mobility Shift Assay
  • transfected cells were split into three 10 cm cell culture dishes and incubated for 24 h prior to transfection of oligoTRAFTACs.
  • Epoxomicin (1 ⁇ M) was preincubated with cells for 1 h and oligoTRAFTACs (mock, OT3 and OT17) were then transfected using RNAi-MAX in 5 mL/dish of transfection medium.
  • RNAi-MAX 5 mL/dish of transfection medium.
  • cells were harvested and lysed using immunoprecipitation buffer (25 mM Tris pH 7.4, 150 mM NaCl, 0.4% NP40, 5% glycerol, 1X protease inhibitor cocktail from Roche and 1 mM of PMSF).
  • lysate from each sample was incubated with brachyury antibody at 4 o C for 4 h.
  • Protein A agarose beads (30 ⁇ L) were added to antibody, lysates mixture and rock at 4 o C for ⁇ 18 h. Beads were washed with 1X TBS for three times with 5-minute incubation during each wash. Immunoprecipitated proteins were eluted by boiling agarose beads in 2X loading buffer (containing 10% ß-ME) for 8 minutes and centrifuged at high speed for 5 minutes prior to the loading into SDS-PAGE gel followed by western blot analysis. Cell viability assay Cells were split and subjected to oligoTRAFTAC transfection as described herein.
  • CellTiter-Glo reagent was prepared according to the manufactures recommendation and mixed with cell culture medium with 1:1 ratio. CellTiter-Glo reagent (100 ⁇ L) was added to each well and incubated for 10 minutes before taking the reading. Zebrafish Husbandry and Microinjection Tüpfel-longfin zebrafish were raised according to standard protocols approved by the Institutional Animal Care and Use Committee. Experiments were performed before sex determination in zebrafish 1 . Embryos were injected at the one cell stage with 180 picoliters of a 25 ⁇ M oligoTRAFTAC solution.
  • OTs 1-20 can have a linker LNK of formula LNK-X1, LNK- X2, LNK-1, or LNK-2.
  • the linker PEG number (n) which represents the number of ethylene glycol (-CH 2 CH 2 O-) units, is the same as variable pp in formulas LNK-X1, LNK-X2, LNK- 1, and LNK-2. Table 2. Oligonucleotide sequence, modification and modification site of the oligonucleotides used for biotin pull-down experiments.
  • Embodiment 1 provides a compound of formula I, or a salt, enantiomer, diastereomer, or tautomer thereof: wherein: ONA is an oligonucleotide having a 3'-end and a 5'-end and comprising one or more phosphodiester or phosphorothioate internucleotide linkages; LNK is a chemical linker covalently bonding the ONA and the UBL; and UBL is an E3 ubiquitin ligase ligand.
  • Embodiment 2 provides the compound of embodiment 1, wherein the 3'-end or the 5'- end of the ONA is covalently bonded to the LNK.
  • Embodiment 3 provides the compound of any one of embodiments 1-2, wherein at least one internucleotide linkage comprises a phosphorothioate.
  • Embodiment 4 provides the compound of any one of embodiments 1-3, wherein all internucleotide linkages are independently phosphorothioates.
  • Embodiment 5 provides the compound of any one of embodiments 1-4, wherein the ONA comprises at least one deoxyribonucleotide and/or ribonucleotide.
  • Embodiment 6 provides the compound of any one of embodiments 1-5, wherein the ONA comprises a c-Myc-binding nucleotide sequence.
  • Embodiment 7 provides the compound of any one of embodiments 1-6, wherein at least two nucleotides flank each end of the c-Myc-binding nucleotide sequence.
  • Embodiment 8 provides the compound of any one of embodiments 1-7, wherein the c- Myc-binding nucleotide sequence comprises 5'-CACGTGGTTGCCACGTG-3'.
  • Embodiment 9 provides the compound of any one of embodiments 1-8, wherein the ONA comprises a brachyury-binding nucleotide sequence.
  • Embodiment 10 provides the compound of any one of embodiments 1-9, wherein at least two nucleotides flank each end of the brachyury-binding nucleotide sequence.
  • Embodiment 11 provides the compound of any one of embodiments 1-10, wherein the brachyury-binding nucleotide sequence comprises 5'- AATTTCACACCTAGGTGTGAAATT-3'.
  • Embodiment 15 provides the compound of any one of embodiments 1-14, wherein A is phenylene or a C 5-18 heteroarylene.
  • Embodiment 16 provides the compound of any one of embodiments 1-15, wherein A Embodiment 17 provides the compound of any one of embodiments 1-16, wherein LNK is –(LL) aa –A–(LL) bb –.
  • Embodiment 20 provides the compound of any one of embodiments 1-19, wherein LNK-X1 has the structure wherein X is CH 2 or O; and k is 1, 2, or 3.
  • Embodiment 22 provides the compound of any one of embodiments 1-21, wherein LNK-X2 has the structure wherein X is CH 2 or O; and k is 1, 2, or 3.
  • Embodiment 23 provides the compound of any one of embodiments 1-22, wherein LNK has the structure: , wherein pp is 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; and wherein the atom marked with * is covalently bonded to the 3'-end or the 5'-end of the ONA.
  • Embodiment 24 provides the compound of any one of embodiments 1-23, wherein pp is 2, 3, or 5.
  • Embodiment 25 provides the compound of any one of embodiments 1-24, wherein pp is 2, 3, or 5.
  • Embodiment 26 provides the compound of any one of embodiments 1-25, wherein the UBL is a ligand of an E3 ubiquitin ligase selected from the group consisting of von Hippel- Lindau (VHL), cereblon (CRBN), RING-type zinc-finger protein 114 (RNF114), cellular inhibitor of apoptosis (cIAP), mouse double minute 2 homologue (MDM2), damage-specific DNA binding protein 1 (DDB1)-CUL4 associated factor 16 (DCAF16), and Kelch-like ECH- associated protein 1 (KEAP1).
  • Embodiment 27 provides the compound of any one of embodiments 1-26, wherein the UBL is a ligand of VHL or CRBN.
  • Embodiment 28 provides the compound of any one of embodiments 1-27, wherein the UBL is a ligand having the structure:
  • Embodiment 29 provides the compound of any one of embodiments 1-28, wherein the UBL has the structure: .
  • Embodiment 30 provides the compound of any one of embodiments 1-29, wherein the compound is selected from the group consisting of: , , wherein pp is 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; and wherein the atom marked with * is covalently bonded to the 3'-end or the 5'-end of the ONA.
  • Embodiment 31 provides the compound of any one of embodiments 1-30, wherein the ONA has a sequence selected from the group consisting of: 5'-TGGGAGCACGTGGTTGCCACGTGGTTGGG-3', 3'-GGGTTGGTGCACCGTTGGTGCACGAGGGT-5', 5'-CTTTCCAATTTCACACCTAGGTGTGAAATTGGGGAC-3', and 3'-CAGGGGTTAAAGTGTGGATCCACACTTTAACCTTTC-5'.
  • Embodiment 33 provides the compound of embodiment 32, wherein the terminal nucleotide at the 3'-end of ONA has the structure: , wherein: zz is an integer from 1 to 100; Y is H or OH; and Embodiment 34 provides the compound of any one of embodiments 32-33, wherein the terminal nucleotide at the 5'-end of ONA has the structure: , wherein: zz is an integer from 1 to 100; Y is H or OH; Z is O or S; and Embodiment 35 provides the compound of any one of embodiments 32-34, wherein the ONA comprises at least one deoxyribonucleotide or ribonucleotide, or a combination thereof.
  • Embodiment 36 provides the compound of any one of embodiments 32-35, wherein the ONA comprises a c-Myc-binding nucleotide sequence.
  • Embodiment 37 provides the compound of any one of embodiments 32-36, wherein at least two nucleotides flank each end of the c-Myc-binding nucleotide sequence.
  • Embodiment 38 provides the compound of any one of embodiments 32-37, wherein the c-Myc-binding nucleotide sequence comprises 5'- CACGTGGTTGCCACGTG-3'.
  • Embodiment 39 provides the compound of any one of embodiments 32-38, wherein the ONA comprises a brachyury-binding nucleotide sequence.
  • Embodiment 40 provides the compound of any one of embodiments 32-39, wherein at least two nucleotides flank each end of the brachyury-binding nucleotide sequence.
  • Embodiment 41 provides the compound of any one of embodiments 32-40, wherein the brachyury-binding nucleotide sequence comprises 5'- AATTTCACACCTAGGTGTGAAATT-3'.
  • Embodiment 42 provides the compound of any one of embodiments 32-41, having the structure: wherein pp is 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10; and wherein the atom marked with * is covalently bonded to the 3'-end or the 5'-end of the ONA.
  • Embodiment 43 provides the compound of any one of embodiments 32-42, wherein the ONA has a sequence selected from the group consisting of: 5'-TGGGAGCACGTGGTTGCCACGTGGTTGGG-3', 3'-GGGTTGGTGCACCGTTGGTGCACGAGGGT-5', 5'-CTTTCCAATTTCACACCTAGGTGTGAAATTGGGGAC-3', and 3'-CAGGGGTTAAAGTGTGGATCCACACTTTAACCTTTC-5'.
  • Embodiment 44 provides a pharmaceutical composition comprising the compound of any one of embodiments 1-31, and a pharmaceutically acceptable carrier, additive, and/or excipient.
  • Embodiment 45 provides a method of preventing, treating, and/or ameliorating cancer in a subject, the method comprising: administering to the subject a therapeutically effective amount of at least one compound of any one of embodiments 1-31, which is optionally formulated as a pharmaceutical composition further comprising at least one pharmaceutically acceptable carrier.
  • Embodiment 46 provides the method of embodiment 45, wherein the cancer is at least one selected from the group consisting of squamous-cell carcinoma, basal cell carcinoma, adenocarcinoma, hepatocellular carcinomas, renal cell carcinoma, cancer of the bladder, bowel, breast, cervix, colon, esophagus, head, kidney, liver, lung, neck, ovary, pancreas, prostate, and stomach; leukemia; benign or malignant lymphoma; benign or malignant melanoma; myeloproliferative disease; multiple myeloma, sarcoma, glioma, astrocytoma, oligodendroglioma, ependymoma, gliobastoma, neuroblastoma, ganglioneuroma, ganglioglioma, medulloblastoma, pineal cell tumor, meningioma, meningeal sarcoma, neurofibrom
  • Embodiment 47 provides the method of any one of embodiments 45-46, wherein: the benign or malignant lymphoma comprises Burkitt's lymphoma and/or Non-Hodgkin's lymphoma; and the sarcoma comprises Ewing's sarcoma, hemangiosarcoma, Kaposi's sarcoma, liposarcoma, myosarcoma, peripheral neuroepithelioma, and/or synovial sarcoma.
  • the benign or malignant lymphoma comprises Burkitt's lymphoma and/or Non-Hodgkin's lymphoma
  • the sarcoma comprises Ewing's sarcoma, hemangiosarcoma, Kaposi's sarcoma, liposarcoma, myosarcoma, peripheral neuroepithelioma, and/or synovial sarcoma.
  • Embodiment 48 provides the method of any one of embodiments 45-47, wherein the compound and/or composition is administered by a route selected from the group consisting of oral, transdermal, transmucosal, (intra)nasal, (trans)rectal, intravesical, intrapulmonary, intraduodenal, intragastrical, intrathecal, subcutaneous, intramuscular, intradermal, intra- arterial, intravenous, intrabronchial, inhalation, and topical.
  • Embodiment 49 provides the method of any one of embodiments 45-48, wherein the subject is a mammal.
  • Embodiment 50 provides the method of any one of embodiments 45-49, wherein the subject is human.

Abstract

L'invention concerne des composés contenant un oligonucléotide de liaison au TF et un ligand de recrutement de ligase E3 ("oligoTRAFTAC"). Ces oligoTRAFTAC induisent la dégradation de facteurs de transcription (TF) oncogènes, c-Myc et Brachyury. Ces composés constituent une plate-forme généralisable pour des TF difficiles à cibler et peuvent être dégradés par l'intermédiaire de la voie protéasomique, ce qui est utile dans le traitement d'une grande variété de cancers.
PCT/US2022/080390 2021-11-23 2022-11-23 Chimères ciblant le facteur de transcription et contenant des oligonucléotides WO2023097251A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202163282298P 2021-11-23 2021-11-23
US63/282,298 2021-11-23

Publications (1)

Publication Number Publication Date
WO2023097251A1 true WO2023097251A1 (fr) 2023-06-01

Family

ID=86540368

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2022/080390 WO2023097251A1 (fr) 2021-11-23 2022-11-23 Chimères ciblant le facteur de transcription et contenant des oligonucléotides

Country Status (1)

Country Link
WO (1) WO2023097251A1 (fr)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040146910A1 (en) * 2002-11-20 2004-07-29 Affymetrix, Inc. Methods of genetic analysis of rat
US20160376585A1 (en) * 2013-07-11 2016-12-29 Alnylam Pharmaceuticals, Inc. Oligonucleotide-ligand conjugates and process for their preparation
US20200390894A1 (en) * 2019-06-12 2020-12-17 Northwestern University Proteolysis-targeting chimeric molecules (protacs) that induce degradation of c-myc protein
WO2022183006A1 (fr) * 2021-02-25 2022-09-01 Mayo Foundation For Medical Education And Research Composés pour la dégradation de protéine programmable et procédés d'utilisation pour le traitement d'une maladie
WO2022251614A1 (fr) * 2021-05-28 2022-12-01 Beth Israel Deaconess Medical Center, Inc. Protac avec des fractions ciblant un facteur de transcription

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20040146910A1 (en) * 2002-11-20 2004-07-29 Affymetrix, Inc. Methods of genetic analysis of rat
US20160376585A1 (en) * 2013-07-11 2016-12-29 Alnylam Pharmaceuticals, Inc. Oligonucleotide-ligand conjugates and process for their preparation
US20200390894A1 (en) * 2019-06-12 2020-12-17 Northwestern University Proteolysis-targeting chimeric molecules (protacs) that induce degradation of c-myc protein
WO2022183006A1 (fr) * 2021-02-25 2022-09-01 Mayo Foundation For Medical Education And Research Composés pour la dégradation de protéine programmable et procédés d'utilisation pour le traitement d'une maladie
WO2022251614A1 (fr) * 2021-05-28 2022-12-01 Beth Israel Deaconess Medical Center, Inc. Protac avec des fractions ciblant un facteur de transcription

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
LIU JING, CHEN HE, KANISKAN H. ÜMIT, XIE LING, CHEN XIAN, JIN JIAN, WEI WENYI: "TF-PROTACs Enable Targeted Degradation of Transcription Factors", JOURNAL OF THE AMERICAN CHEMICAL SOCIETY, AMERICAN CHEMICAL SOCIETY, vol. 143, no. 23, 16 June 2021 (2021-06-16), pages 8902 - 8910, XP093070826, ISSN: 0002-7863, DOI: 10.1021/jacs.1c03852 *

Similar Documents

Publication Publication Date Title
US11603532B2 (en) Oligonucleotide compositions and methods of use thereof
KR20190027373A (ko) 유전적 장애 치료를 위한, 특징이 개선된, 비사이클릭 스캐폴드 모이어티를 포함하는 프리-mrna 스플라이스 스위칭 또는 조정 올리고뉴클레오티드
US20080119426A1 (en) Compositions and methods for the treatment of muscle wasting
EP2093292A2 (fr) Inhibition spécifique d'isoformes d'histone deacetylase
US20220204976A1 (en) RNAi Agents for Inhibiting Expression of HIF-2 alpha (EPAS1), Compositions Thereof, and Methods of Use
AU2006345724B2 (en) Compositions and methods for the treatment of muscle wasting
KR101344084B1 (ko) 암세포 특이적 세포증식 억제제
CN110248956B (zh) HIF-1α反义寡核苷酸
AU2020292419A1 (en) Acetyl-CoA synthetase 2 (ACSS2) inhibitors and methods using same
WO2008005019A1 (fr) Compositions et procédés pour le traitement de l'atrophie musculaire
US20140128450A1 (en) Cancer Therapy
WO2023097251A1 (fr) Chimères ciblant le facteur de transcription et contenant des oligonucléotides
US20220267308A1 (en) Modulators of Excitatory Amino Acid Transporters and Methods Using Same
US20230399303A1 (en) Macrophage Migration Inhibitory Factor Inhibitors, and Methods of Making and Using Same
US20240092759A1 (en) Non-Covalent Inhibitors of the Main Protease of SARS-CoV-2 and Methods of Use
US20230271994A1 (en) Molecular degraders of extracellular proteins
KR102329524B1 (ko) 변형된 rt-let7을 유효성분으로 포함하는 간암의 예방 또는 치료용 조성물
KR101497930B1 (ko) Hs2st1 유전자 또는 상기 유전자가 암호화하는 단백질에 대한 억제제를 포함하는 세포 노화 유도용 조성물 및 이를 이용한 세포 노화 유도 방법
US20210317157A1 (en) Substituted Alloxazine Nucleosides and Nucleotides, and Methods of Making Same
WO2024026410A2 (fr) Inhibiteurs de récepteur tyrosine kinase neurotrophique (ntrk) et méthodes associées
WO2017159739A1 (fr) Agent anticancéreux
WO2020257753A1 (fr) Inhibiteurs de la phosphatidylsérine décarboxylase
JP2009225672A (ja) FGFR2に対するsiRNAによるスキルス胃がん細胞の増殖阻害
JP2009131162A (ja) 卵巣明細胞腺癌におけるアネキシン4の発現を増強する遺伝子領域

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: 22899536

Country of ref document: EP

Kind code of ref document: A1