WO2022020247A1 - Small molecule correctors of mammalian slc6a8 function - Google Patents

Small molecule correctors of mammalian slc6a8 function Download PDF

Info

Publication number
WO2022020247A1
WO2022020247A1 PCT/US2021/042208 US2021042208W WO2022020247A1 WO 2022020247 A1 WO2022020247 A1 WO 2022020247A1 US 2021042208 W US2021042208 W US 2021042208W WO 2022020247 A1 WO2022020247 A1 WO 2022020247A1
Authority
WO
WIPO (PCT)
Prior art keywords
alkyl
haloalkyl
heteroalkyl
heteroaryl
aryl
Prior art date
Application number
PCT/US2021/042208
Other languages
French (fr)
Inventor
Giovanni MUNCIPINTO
Dean G. Brown
Jaclyn L. HENDERSON
Original Assignee
Jnana Therapeutics, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Jnana Therapeutics, Inc. filed Critical Jnana Therapeutics, Inc.
Publication of WO2022020247A1 publication Critical patent/WO2022020247A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D213/00Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members
    • C07D213/02Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members
    • C07D213/04Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D213/60Heterocyclic compounds containing six-membered rings, not condensed with other rings, with one nitrogen atom as the only ring hetero atom and three or more double bonds between ring members or between ring members and non-ring members having three double bonds between ring members or between ring members and non-ring members having no bond between the ring nitrogen atom and a non-ring member or having only hydrogen or carbon atoms directly attached to the ring nitrogen atom with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D213/78Carbon atoms having three bonds to hetero atoms, with at the most one bond to halogen, e.g. ester or nitrile radicals
    • C07D213/81Amides; Imides
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings
    • C07D401/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D417/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00
    • C07D417/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and sulfur atoms as the only ring hetero atoms, not provided for by group C07D415/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/08Bridged systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/02Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D498/08Bridged systems

Definitions

  • Creatine transporter deficiency has been reported to be the most common cerebral creatine deficiency syndrome (CCDS). Creatine transporter deficiency is an X-linked disorder caused by mutations in the SLC6A8 gene.
  • the SLC6A8 gene located on the short arm of the sex chromosome, provides instructions for making a protein that transports the compound creatine into cells. Creatine is needed for the body to store and use energy properly.
  • People with CTD have intellectual disability, which can range from mild to severe, and delayed speech development. Some affected individuals develop behavioral disorders such as attention deficit hyperactivity disorder or autistic behaviors that affect communication and social interaction. They may also experience seizures. Children with CTD may experience slow growth and exhibit delayed development of motor skills such as sitting and walking. CTD is difficult to treat because the actual transporter responsible for transporting creatine to the brain and muscles is defective. There is no current standard of care.
  • One aspect of the invention provides compounds, compositions, and methods useful for treating or preventing a disease or disorder associated with a SLC6A8 mutation.
  • a and A' are each independently selected from C-R' and N;
  • A" is selected from C-R" and N; provided that at least one of A, A', and A" is N; or A and A' are each independently selected from C-R and N, and A" is C-R", wherein R" taken together with R and the ring atoms to which they are bonded form a fused five-membered heteroaryl ring, such that formula I is a substituted indole or azaindole;
  • X is selected from Y is selected from n is 0, 1, 2, or 3;
  • m is 1;
  • R1, R2, R3, R4, R5 and R6 are independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, aryl, heteroaryl, -C(O)OR 11 , -C(O)NR 12 R 13 , and -C(O)R 14 ;
  • R 7, R 8 , R 9 , and R 10 are independently selected from -H,
  • Ri5 and Rie are independently selected from -H, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -C(0)Rn, -C(0)0Rn, and - C(0)NRi2Ri3; or Ris and Rie taken together with the nitrogen atom to which they are bonded form an un substituted or substituted C4-Cs cycloheteroalkyl; and provided that if A is N, A' and A" are each is selected from phenyl and 2-methoxy-5-fluorophenyl, then R is not
  • Another aspect of the invention relates to methods of treating or preventing a disease or disorder associated with a SLC6A8 mutation, treating an inflammatory disease, increasing transport of guanidinoacetic acid or a salt thereof across the blood-brain barrier, improving the function of a cellular creatine transporter, or decreasing the accumulation or the concentration of guanidinoacetic acid or a salt thereof in a cell, in a subject in need thereof comprising administering to the subject an effective amount of a compound of Formula (I): wherein:
  • a and A' are each independently selected from C-R and N;
  • A" is selected from C-R" and N; provided that at least one of A, A', and A" is N; or A and A' are each independently selected from C-R and N, and A" is C-R", wherein R" taken together with R and the ring atoms to which they are bonded form a fused five-membered heteroaryl ring, such that formula I is a substituted indole or azaindole;
  • X is selected from Y is selected from
  • « is 0, 1, 2, or 3; m is 1;
  • Ri, R2, R3, R4, RS and Re are independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, aryl, heteroaryl, -C(0)0Rn, -C(0)NRi2Ri3, and -C(0)Ri4;
  • Rv, Rs, R9, and Rio are independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, aryl, heteroaryl, -C(0)0Rn, -C(0)NRi2Ri3, and -C(0)Ri4;
  • Z is selected from -H, halogen, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O- cycloheteroalkyl, -O-halocycloalkyl, -NH-alkyl, -NH-heteroalkyl, -NH-haloalkyl, -NH- cycloalkyl, -NH-cycloheteroalkyl, -NH-halocycloalkyl, aryl, -O-aryl, -NH-aryl, heteroaryl, - O-heteroaryl, -NH-heteroaryl, -C(0)0Rn, -C(0)NRi2Ri3, -NR12R13, and -
  • R is selected from alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O-cycloheteroalkyl, - O-halocycloalkyl, aryl, -O-aryl, heteroaryl, -O-heteroaryl -OSO2R11, and -NRisRie;
  • R is selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O- cycloalkyl, -O-cycloheteroalkyl, -O-halocycloalkyl, -O-aryl, -O-heteroaryl, -OSO2R11, and
  • R" is selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O- cycloalkyl, -O-cycloheteroalkyl, -O-halocycloalkyl, -O-aryl, -O-heteroaryl, -OSO2R11, and
  • each occurrence of R11, R12 and R13 is independently selected from H, alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl; each occurrence of Ri4is independently selected from alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl;
  • Ri5 and Rie are independently selected from -H, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -C(0)Rn, -C(0)0Rn, and - C(0)NRI2RI3; or R15 and Rie taken together with the nitrogen atom to which they are bonded form an un substituted or substituted C4-C8 cycloheteroalkyl.
  • the subject is a mammal. In certain embodiments, the mammal is a human.
  • the invention provides several additional advantages.
  • the prophylactic and therapeutic methods described herein are also effective for treating creatine transporter deficiency and associated symptoms.
  • the therapeutic method is effective in treating motor dysfunction, intellectual disability, language delay, speech delay, seizures, behaviors associated with autism and attention deficit hyperactivity disorder, fatigue, muscular hypotonia, low weight gain, and gastrointestinal and cardiac disorders.
  • the therapeutic method is effective in treating inflammatory diseases.
  • the inflammatory disease is acute.
  • the inflammatory disease is chronic.
  • the inflammatory disease is selected from inflammatory bowel diseases (for example, ulcerative colitis or Crohn's disease), multiple sclerosis, psoriasis, arthritis, rheumatoid arthritis, osteoarthritis, juvenile arthritis, psoriatic arthritis, reactive arthritis, ankylosing spondylitis, cryopyrin associated periodic syndromes, Muckle-Wells syndrome, familial cold auto-inflammatory syndrome, neonatal- onset multisystem inflammatory disease, TNF receptor associated periodic syndrome, acute and chronic pancreatitis, atherosclerosis, gout, ankylosing spondylitis, fibrotic disorders (for example, hepatic fibrosis or idiopathic pulmonary fibrosis), nephropathy, sarcoidosis, scleroderma, anaphylaxis, diabetes (for example, chronic ulcerative colitis or Crohn's disease
  • host reaction for example, graft vs. host disease
  • allograft rejections for example, acute allograft rejection or chronic allograft rejection
  • early transplantation rejection for example, acute allograft rejection
  • reperfusion injury pain (for example, acute pain, chronic pain, neuropathic pain, or fibromyalgia), chronic infections, meningitis, encephalitis, myocarditis, gingivitis, post surgical trauma, tissue injury, traumatic brain injury, enterocolitis, sinusitis, uveitis, ocular inflammation, optic neuritis, gastric ulcers, esophagitis, peritonitis, periodontitis, dermatomyositis, gastritis, myositis, polymyalgia, pneumonia and bronchitis.
  • FIG. 1 is a table summarizing trafficking and correction data for exemplary compounds of the invention.
  • Trafficking Emax A: 1 ⁇ Emax ⁇ 2; B: 2 ⁇ Emax ⁇ 5; C: 5 ⁇ E max ⁇ 15.
  • Trafficking ECso A: 0.1 ⁇ ⁇ ECso ⁇ 20 ⁇ ; B: 20 ⁇ ⁇ ECso ⁇ 35 pM; C: 35 pM ⁇ ECso ⁇ 50 pM; D: 50 pM ⁇ ECso ⁇ 100 pM.
  • Correction E A: E max — 1; B: 1 ⁇ Emax ⁇ 2; C:
  • an element means one element or more than one element.
  • a reference to “A and/or B”, when used in conjunction with open-ended language such as “comprising” can refer, in one embodiment, to A only (optionally including elements other than B); in another embodiment, to B only (optionally including elements other than A); in yet another embodiment, to both A and B (optionally including other elements); etc.
  • the phrase “at least one,” in reference to a list of one or more elements, should be understood to mean at least one element selected from any one or more of the elements in the list of elements, but not necessarily including at least one of each and every element specifically listed within the list of elements and not excluding any combinations of elements in the list of elements.
  • This definition also allows that elements may optionally be present other than the elements specifically identified within the list of elements to which the phrase “at least one” refers, whether related or unrelated to those elements specifically identified.
  • “at least one of A and B” can refer, in one embodiment, to at least one, optionally including more than one, A, with no B present (and optionally including elements other than B); in another embodiment, to at least one, optionally including more than one, B, with no A present (and optionally including elements other than A); in yet another embodiment, to at least one, optionally including more than one, A, and at least one, optionally including more than one,
  • compositions of the present invention may exist in particular geometric or stereoisomeric forms.
  • polymers of the present invention may also be optically active.
  • the present invention contemplates all such compounds, including cis- and trans-isomers, R- and //-enantiomers, diastereomers, (D)-isomers, (L)- i somers, the racemic mixtures thereof, and other mixtures thereof, as falling within the scope of the invention.
  • Additional asymmetric carbon atoms may be present in a substituent such as an alkyl group. All such isomers, as well as mixtures thereof, are intended to be included in this invention.
  • “Geometric isomer” means isomers that differ in the orientation of substituent atoms in relationship to a carbon-carbon double bond, to a cycloalkyl ring, or to a bridged bicyclic system. Atoms (other than H) on each side of a carbon- carbon double bond may be in an E (substituents are on opposite sides of the carbon- carbon double bond) or Z (substituents are oriented on the same side) configuration. "R,” “S,” “S*,” “R*,” “E,” “Z,” “cis,” and “trans,” indicate configurations relative to the core molecule.
  • Certain of the disclosed compounds may exist in “atropisomeric” forms or as “atropisomers.”
  • Atropi somers are stereoisomers resulting from hindered rotation about single bonds where the steric strain barrier to rotation is high enough to allow for the isolation of the conformers.
  • the compounds of the invention may be prepared as individual isomers by either isomer-specific synthesis or resolved from a mixture of isomers.
  • Conventional resolution techniques include forming the salt of a free base of each isomer of an isomeric pair using an optically active acid (followed by fractional crystallization and regeneration of the free base), forming the salt of the acid form of each isomer of an isomeric pair using an optically active amine (followed by fractional crystallization and regeneration of the free acid), forming an ester or amide of each of the isomers of an isomeric pair using an optically pure acid, amine or alcohol (followed by chromatographic separation and removal of the chiral auxiliary), or resolving an isomeric mixture of either a starting material or a final product using various well known chromatographic methods.
  • a particular enantiomer of compound of the present invention may be prepared by asymmetric synthesis, or by derivation with a chiral auxiliary, where the resulting diastereomeric mixture is separated and the auxiliary group cleaved to provide the pure desired enantiomers.
  • the molecule contains a basic functional group, such as amino, or an acidic functional group, such as carboxyl, diastereomeric salts are formed with an appropriate optically-active acid or base, followed by resolution of the diastereomers thus formed by fractional crystallization or chromatographic means well known in the art, and subsequent recovery of the pure enantiomers.
  • Percent purity by mole fraction is the ratio of the moles of the enantiomer (or diastereomer) or over the moles of the enantiomer (or diastereomer) plus the moles of its optical isomer.
  • the stereochemistry of a disclosed compound is named or depicted by structure, the named or depicted stereoisomer is at least about 60%, about 70%, about 80%, about 90%, about 99% or about 99.9% by mole fraction pure relative to the other stereoisomers.
  • the depicted or named enantiomer is at least about 60%, about 70%, about 80%, about 90%, about 99% or about 99.9% by mole fraction pure.
  • the depicted or named diastereomer is at least about 60%, about 70%, about 80%, about 90%, about 99% or about 99.9% by mole fraction pure.
  • prodrug as used herein encompasses compounds that, under physiological conditions, are converted into therapeutically active agents.
  • a common method for making a prodrug is to include selected moieties that are hydrolyzed under physiological conditions to reveal the desired molecule.
  • the prodrug is converted by an enzymatic activity of the host animal.
  • phrases “pharmaceutically acceptable excipient” or “pharmaceutically acceptable carrier” as used herein means a pharmaceutically acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject chemical 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, not injurious to the patient, and substantially non-pyrogenic.
  • materials which can serve as pharmaceutically acceptable carriers include: (1) sugars, such as lactose, glucose, and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose, and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil, and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol, and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum
  • compositions of the present invention are non-pyrogenic, i.e., do not induce significant temperature elevations when administered to a patient.
  • pharmaceutically acceptable salts refers to the relatively non-toxic, inorganic and organic acid addition salts of the compound(s). These salts can be prepared in situ during the final isolation and purification of the compound(s), or by separately reacting a purified compound(s) in its free base form with a suitable organic or inorganic acid, and isolating the salt thus formed.
  • Representative salts include the hydrobromide, hydrochloride, sulfate, bisulfate, phosphate, nitrate, acetate, valerate, oleate, palmitate, stearate, laurate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, naphthylate, mesylate, glucoheptonate, lactobionate, and laurylsulphonate salts, and the like.
  • sulfate bisulfate
  • phosphate nitrate
  • acetate valerate
  • oleate palmitate
  • stearate laurate
  • benzoate lactate
  • phosphate tosylate
  • citrate maleate
  • fumarate succinate
  • tartrate naphthylate
  • mesylate glucoheptonate
  • lactobionate lactobionate
  • laurylsulphonate salts and the like.
  • the compounds useful in the methods of the present invention may contain one or more acidic functional groups and, thus, are capable of forming pharmaceutically acceptable salts with pharmaceutically acceptable bases.
  • pharmaceutically acceptable salts refers to the relatively non-toxic inorganic and organic base addition salts of a compound(s). These salts can likewise be prepared in situ during the final isolation and purification of the compound(s), or by separately reacting the purified compound(s) in its free acid form with a suitable base, such as the hydroxide, carbonate, or bicarbonate of a pharmaceutically acceptable metal cation, with ammonia, or with a pharmaceutically acceptable organic primary, secondary, or tertiary amine.
  • Representative alkali or alkaline earth salts include the lithium, sodium, potassium, calcium, magnesium, and aluminum salts, and the like.
  • Representative organic amines useful for the formation of base addition salts include ethylamine, diethylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine, and the like (see, for example, Berge et al., supra).
  • pharmaceutically acceptable cocrystals refers to solid coformers that do not form formal ionic interactions with the small molecule.
  • a “therapeutically effective amount” (or “effective amount”) of a compound with respect to use in treatment refers to an amount of the compound in a preparation which, when administered as part of a desired dosage regimen (to a mammal, preferably a human) alleviates a symptom, ameliorates a condition, or slows the onset of disease conditions according to clinically acceptable standards for the disorder or condition to be treated or the
  • a desired dosage regimen to a mammal, preferably a human
  • alleviates a symptom ameliorates a condition, or slows the onset of disease conditions according to clinically acceptable standards for the disorder or condition to be treated or the
  • proliferative or therapeutic treatment is art-recognized and includes administration to the host of one or more of the subject compositions.
  • the treatment is prophylactic, (i.e., it protects the host against developing the unwanted condition), whereas if it is administered after manifestation of the unwanted condition, the treatment is therapeutic, (i.e., it is intended to diminish, ameliorate, or stabilize the existing unwanted condition or side effects thereof).
  • patient or “subject” refers to a mammal in need of a particular treatment.
  • a patient is a primate, canine, feline, or equine. In certain embodiments, a patient is a human.
  • An aliphatic chain comprises the classes of alkyl, alkenyl and alkynyl defined below.
  • a straight aliphatic chain is limited to unbranched carbon chain moieties.
  • the term “aliphatic group” refers to a straight chain, branched-chain, or cyclic aliphatic hydrocarbon group and includes saturated and unsaturated aliphatic groups, such as an alkyl group, an alkenyl group, or an alkynyl group.
  • Alkyl refers to a fully saturated cyclic or acyclic, branched or unbranched carbon chain moiety having the number of carbon atoms specified, or up to 30 carbon atoms if no specification is made.
  • alkyl of 1 to 8 carbon atoms refers to moieties such as methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, and octyl, and those moieties which are positional isomers of these moieties.
  • Alkyl of 10 to 30 carbon atoms includes decyl, undecyl, dodecyl, tri decyl, tetradecyl, pentadecyl, hexadecyl, heptadecyl, octadecyl, nonadecyl, eicosyl, heneicosyl, docosyl, tri cosy 1 and tetracosyl.
  • a straight chain or branched chain alkyl has 30 or fewer carbon atoms in its backbone (e.g., C1-C30 for straight chains, C3-C30 for branched chains), and more preferably 20 or fewer.
  • Alkyl goups may be substituted or unsubstituted.
  • heteroalkyl refers to an alkyl moiety as hereinbefore defined which contain one or more oxygen, sulfur, nitrogen, phosphorus, or silicon atoms in place of carbon atoms.
  • haloalkyl refers to an alkyl group as hereinbefore defined substituted with at least one halogen.
  • hydroxy alkyl refers to an alkyl group as hereinbefore defined substituted with at least one hydroxyl.
  • alkylene refers to an alkyl group having the specified number of carbons, for example from 2 to 12 carbon atoms, that contains two points of attachment to the rest of the compound on its longest carbon chain.
  • alkylene groups include methylene -(CH 2 )-, ethylene -(CH 2 CH 2 )-, n-propylene - (CH 2 CH 2 CH 2 )-, isopropylene -(CH 2 CH(CH 3 ))-, and the like.
  • Alkylene groups can be cyclic or acyclic, branched or unbranched carbon chain moiety, and may be optionally substituted with one or more substituents.
  • Cycloalkyl means mono- or bicyclic or bridged or spirocyclic, or polycyclic saturated carbocyclic rings, each having from 3 to 12 carbon atoms. Preferred cycloalkyls have from 3-10 carbon atoms in their ring structure, and more preferably have 3-6 carbons in the ring structure. Cycloalkyl groups may be substituted or unsubstituted. As used herein, the term “halocycloalkyl” refers to an cycloalkyl group as hereinbefore defined substituted with at least one halogen.
  • Cycloheteroalkyl refers to an cycloalkyl moiety as hereinbefore defined which contain one or more oxygen, sulfur, nitrogen, phosphorus, or silicon atoms in place of carbon atoms.
  • Preferred cycloheteroalkyls have from 4-8 carbon atoms and heteroatoms in their ring structure, and more preferably have 4-6 carbons and heteroatoms in the ring structure. Cycloheteroalkyl groups may be substituted or unsubstituted.
  • lower alkyl means an alkyl group, as defined above, but having from one to ten carbons, more preferably from one to six carbon atoms in its backbone structure such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, and tert-butyl.
  • lower alkenyl and “lower alkynyl” have similar chain lengths.
  • preferred alkyl groups are lower alkyls.
  • a substituent designated herein as alkyl is a lower alkyl.
  • Alkenyl refers to any cyclic or acyclic, branched or unbranched unsaturated carbon chain moiety having the number of carbon atoms specified, or up to 26 carbon atoms if no limitation on the number of carbon atoms is specified; and having one or more double bonds in the moiety.
  • Alkenyl of 6 to 26 carbon atoms is exemplified by hexenyl, heptenyl, octenyl, nonenyl, decenyl, undecenyl, dodenyl, tridecenyl, tetradecenyl, pentadecenyl, hexadecenyl, heptadecenyl, octadecenyl, nonadecenyl, eicosenyl, heneicosoenyl, docosenyl, tricosenyl, and tetracosenyl, in their various isomeric forms, where the unsaturated bond(s) can be located anywhere in the moiety and can have either the (Z) or the (E) configuration about the double bond(s).
  • Alkynyl refers to hydrocarbyl moieties of the scope of alkenyl, but having one or more triple bonds in the moiety.
  • aryl as used herein includes 3- to 12-membered substituted or unsubstituted single-ring aromatic groups in which each atom of the ring is carbon (i.e., carbocyclic aryl) or where one or more atoms are heteroatoms (i.e., heteroaryl).
  • aryl groups include 5- to 12-membered rings, more preferably 6- to 10-membered rings
  • aryl also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is aromatic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls.
  • Carboycyclic aryl groups include benzene, naphthalene, phenanthrene, phenol, aniline, and the like.
  • Heteroaryl groups include substituted or unsubstituted aromatic 3- to 12-membered ring structures, more preferably 5- to 12- membered rings, more preferably 5- to 10-membered rings, whose ring structures include one to four heteroatoms.
  • Heteroaryl groups include, for example, pyrrole, furan, thiophene, imidazole, oxazole, thiazole, triazole, pyrazole, pyridine, pyrazine, pyridazine and pyrimidine, and the like.
  • Aryl and heteroaryl can be monocyclic, bicyclic, or polycyclic.
  • halo means halogen and includes, for example, and without being limited thereto, fluoro, chloro, bromo, iodo and the like, in both radioactive and non-radioactive forms. In a preferred embodiment, halo is selected from the group consisting of fluoro, chloro and bromo.
  • heterocyclyl or “heterocyclic group” refer to 3- to 12-membered ring structures, more preferably 5- to 12-membered rings, more preferably 5- to 10-membered rings, whose ring structures include one to four heteroatoms. Heterocycles can be monocyclic, bicyclic, spirocyclic, or polycyclic.
  • Heterocyclyl groups include, for example, thiophene, thianthrene, furan, pyran, isobenzofuran, chromene, xanthene, phenoxathiin, pyrrole, imidazole, pyrazole, isothiazole, isoxazole, pyridine, pyrazine, pyrimidine, pyridazine, indolizine, isoindole, indole, indazole, purine, quinolizine, isoquinoline, quinoline, phthalazine, naphthyridine, quinoxaline, quinazoline, cinnoline, pteridine, carbazole, carboline, phenanthridine, acridine, pyrimidine, phenanthroline, phenazine, phenarsazine, phenothiazine, furazan, phenoxazine, pyrrolidine, o
  • the heterocyclic ring can be substituted at one or more positions with such substituents as described above, as for example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, amino, nitro, sulfhydryl, imino, amido, phosphate, phosphonate, phosphinate, carbonyl, carboxyl, silyl, sulfamoyl, sulfinyl, ether, alkylthio, sulfonyl, ketone, aldehyde, ester, a heterocyclyl, an aromatic or heteroaromatic moiety, -CF3, -CN, and the like.
  • substituents as described above, as for example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, amino, nitro, sulfhydryl, imino, amid
  • substituted refers to moieties having substituents replacing a hydrogen on one or more carbons of the backbone. It will be understood that “substitution” or “substituted with” includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., which does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc. As used herein, the term “substituted” is contemplated to include all permissible substituents of organic compounds.
  • the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and non-aromatic substituents of organic compounds.
  • the permissible substituents can be one or more and the same or different for appropriate organic compounds.
  • the heteroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms.
  • Substituents can include any substituents described herein, for example, a halogen, a hydroxyl, a carbonyl (such as a carboxyl, an alkoxy carbonyl, a formyl, or an acyl), a thiocarbonyl (such as a thioester, a thioacetate, or a thioformate), an alkoxy, a phosphoryl, a phosphate, a phosphonate, a phosphinate, an amino, an ami do, an amidine, an imine, a cyano, a nitro, an azido, a sulfhydryl, an alkylthio, a sulfate, a sulfonate, a sulfamoyl, a sulfonamido, a sulfonyl, a heterocyclyl, an aralkyl, or an aromatic or heteroaromatic mo
  • the substituents on substituted alkyls are selected from Ci-6 alkyl, C3-6 cycloalkyl, halogen, carbonyl, cyano, or hydroxyl. In more preferred embodiments, the substituents on substituted alkyls are selected from fluoro, carbonyl, cyano, or hydroxyl. It will be understood by those skilled in the art that substituents can themselves be substituted, if appropriate. Unless specifically stated as “unsubstituted,” references to chemical moieties herein are understood to include substituted variants. For example, reference to an “aryl” group or moiety implicitly includes both substituted and unsubstituted variants.
  • each expression e.g., alkyl, m, n, etc., when it occurs more than once in any structure, is intended to be independent of its definition elsewhere in the same structure.
  • small molecules refers to small organic or inorganic molecules of molecular weight below about 3,000 Daltons.
  • small molecules useful for the invention have a molecular weight of less than 3,000 Daltons (Da).
  • the small molecules can be, e.g., from at least about 100 Da to about 3,000 Da (e.g., between about 100 to about 3,000 Da, about 100 to about 2500 Da, about 100 to about 2,000 Da, about 100 to about 1,750 Da, about 100 to about 1,500 Da, about 100 to about 1,250 Da, about 100 to about 1,000 Da, about 100 to about 750 Da, about 100 to about 500 Da, about 200 to about 1500, about 500 to about 1000, about 300 to about 1000 Da, or about 100 to about 250 Da).
  • a “small molecule” refers to an organic, inorganic, or organometallic compound typically having a molecular weight of less than about 1000. In some embodiments, a small molecule is an organic compound, with a size on the order of 1 nm. In some embodiments, small molecule drugs of the invention encompass oligopeptides and other biomolecules having a molecular weight of less than about 1000.
  • an “effective amount” is an amount sufficient to effect beneficial or desired results.
  • a therapeutic amount is one that achieves the desired therapeutic effect. This amount can be the same or different from a prophylactically effective amount, which is an amount necessary to prevent onset of disease or disease symptoms.
  • An effective amount can be administered in one or more administrations, applications or dosages.
  • a therapeutically effective amount of a composition depends on the composition selected. The compositions can be administered from one or more times per day to one or more times per week; including once every other day. The skilled artisan will appreciate that certain factors may influence the dosage and timing required to effectively treat a subject, including but not limited to the severity of the disease or disorder, previous treatments, the general health and/or age of the subject, and other diseases present.
  • treatment of a subject with a therapeutically effective amount of the compositions described herein can include a single treatment or a series of treatments.
  • “decrease,” “reduce,” “reduced”, “reduction”, “decrease,” and “inhibit” are all used herein generally to mean a decrease by a statistically significant amount relative to a reference.
  • “reduce,” “reduction” or “decrease” or “inhibit” typically means a decrease by at least 10% as compared to a reference level and can include, for example, a decrease by at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, at least about 99%, up to and including, for example, the complete absence of the given entity or parameter ascompared to the reference level, or any decrease between 10-99% as compared to the absence of a given treatment.
  • the terms “increased”, “increase” or “enhance” or “activate” are all used herein to generally mean an increase by a statically significant amount; for the avoidance of any doubt, the terms “increased”, “increase” or “enhance” or “activate” means an increase of at least 10% as compared to a reference level, for example an increase of at least about 20%, or at least about 30%, or at least about 40%, or at least about 50%, or at least about 60%, or at least about 70%, or at least about 80%, or at least about 90% or up to and including a 100% increase or any increase between 10-100% as compared to a reference level, or at least about a 2-fold, or at least about a 3-fold, or at least about a 4-fold, or at least about a 5-fold or at least about a 10-fold increase, or any increase between 2-fold and 10-fold or greater as compared to a reference level.
  • the term “modulate” includes up-regulation and down-regulation, e.g., enhancing or inhibiting a response.
  • a “radiopharmaceutical agent,” as defined herein, refers to a pharmaceutical agent which contains at least one radiation-emitting radioisotope. Radiopharmaceutical agents are routinely used in nuclear medicine for the diagnosis and/or therapy of various diseases.
  • the radiolabelled pharmaceutical agent for example, a radiolabelled antibody, contains a radioisotope (RI) which serves as the radiation source.
  • RI radioisotope
  • the term “radioisotope” includes metallic and non-metallic radioisotopes. The radioisotope is chosen based on the medical application of the radiolabeled pharmaceutical agents.
  • the radioisotope is a metallic radioisotope
  • a chelator is typically employed to bind the metallic radioisotope to the rest of the molecule.
  • the radioisotope is a non-metallic radioisotope
  • the non-metallic radioisotope is typically linked directly, or via a linker, to the rest of the molecule.
  • the chemical elements are identified in accordance with the Periodic Table of the Elements, CAS version, Handbook of Chemistry and Physics, 67th Ed., 1986-87, inside cover.
  • a and A' are each independently selected from C-R' and N; A'' is selected from C-R'' and N; provided that at least one of A, A', and A'' is N; or A and A' are each independently selected from C-R' and N, and A'' is C-R'', wherein R'' taken together with R and the ring atoms to which they are bonded form a fused five-membered heteroaryl ring, such that formula I is a substituted indole or azaindole; n is 0, 1, 2, or 3; m is 1; R 1 , R 2 , R 3 , R 4 , R 5 and R 6 are independently selected from -H, halogen, -CN, -CF 3 , alkyl, heteroalkyl, haloalkyl, aryl, heteroaryl, -C(O)OR11, -C
  • Y is selected from In some embodiments, R1, R2, R3, R4, R5, R6, R7 and R8 are -H. In some embodiments, one of R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 and R 8 is not -H. In some embodiments, two of R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 and R 8 are not -H. In some embodiments, three of R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , R 7 and R8 are not -H. In some embodiments, Y is . In some embodiments, R 7 and R 8 are -H.
  • R9 and R10 are independently selected from -H and alkyl. In some embodiments, R7 and R8 are -H; and wherein R9 and R10 are independently selected from -H and alkyl. In some embodiments, R 7 , R 8 , R 9 , and R 10 are each -H.
  • Z is aryl or heteroaryl. In some embodiments, Z is selected from unsubstituted or substituted phenyl, pyridine, pyrimidine, pyrazole, and triazole. In some embodiments, Z is selected from
  • Ri7, R18, Rig, R20, and R21 are independently selected from -H, halogen, CN, CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, -O- heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O-cycloheteroalkyl, -O-halocycloalkyl, -NH-alkyl, -NH-heteroalkyl, -NH-haloalkyl, -NH-cycloalkyl, -NH-cycloheteroalkyl, -NH- halocycloalkyl, aryl, and heteroaryl; and Rmis haloalkyl or alkyl.
  • Z is Ri7 and Rix are independently selected from -H, halogen, CN, CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, -O-heteroalkyl, -O- haloalkyl, -O-cycloalkyl, -O-cycloheteroalkyl, -O-halocycloalkyl, -NH-alkyl, -NH- heteroalkyl, -NH-haloalkyl, -NH-cycloalkyl, -NH-cycloheteroalkyl, -NH-halocycloalkyl, aryl, and heteroaryl, or R17 taken together with Rix and the ring atoms to which they are bonded form a fused ring, such that Z is a benzimidazole, dihydrobenzo
  • the compound having the structure selected from In some embodiments, the compound having the structure selected from In some embodiments, R is selected from aryl, heteroaryl, and -NR 15 R 16 ; and R 15 and R 16 taken together with the nitrogen atom to which they are bonded form an unsubstituted or substituted C4-C8 cycloheteroalkyl. In some embodiments, R is selected from aryl and heteroaryl. In some embodiments, R is phenyl.
  • R is -NR15R16; and R15 and R16 taken together with the nitrogen atom to which they are bonded form an unsubstituted or substituted piperidine, piperazine, pyrrolidine, morpholine, or azetidine.
  • R 22 is selected from alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -SO 2 R 23 , -C(O)OR 23 , -C(O)NR 24 R 25 , and -C(O)R 26 ;
  • Ra, Rb, Rc, Rd, Rg, Rh, Ri, and Rj are independently selected from -H, halogen, -CN, - CF3, alkyl, heteroalkyl, haloalkyl, hydroxyalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -SO 2 R 23 , -C(O)OR 23 , -C(O)NR 24 R 25 , and -C(O)R 26 ; or R a taken together with Rb and the carbon atom to
  • Ra, Rb, Ri and Rj are independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, - C(O)OR 23 , -C(O)NR 24 R 25 , and -C(O)R 26 ; or R a taken together with R b and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, ⁇ -lactam, or ⁇ - lactam; Rc, Rd, Re, Rf, Rg and Rh are independently selected from -H, halogen, -CN, -CF3, - OH, -NH2, alkyl, heteroalkyl, haloalkyl, hydroxyalkyl, cycloalkyl, cyclohetero
  • Ra, Rb, Rc, Rd, Rg, Rh Ri and Rj are independently selected from -H, halogen, -CN, CF 3 , - alkyl, heteroalkyl, haloalkyl, hydroxyalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -SO 2 R 23 , -C(O)OR 23 , -C(O)NR 24 R 25 , -OSO 2 (N 26 ) 2 , - SO2(N26)2, –C(O)R26 , and alkyl-NHSO2-R26; or Ra taken together with Rb and the carbon atom to which they are bonded form an unsubstituted or substituted C 3 -C 6 cycloalkyl, ⁇ - lactam, or ⁇ -lactam; or R c taken together with R d and the carbon atom to which they are bonded form
  • R is Ra, Rb, Re and Rf are independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, hydroxy alkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -SO2R23, -C(0)0R23, and -C(0)NR24R25; or R a taken together with Rb and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, ⁇ -lactam, or ⁇ -lactam;
  • Rc and Rd are independently selected from -H, halogen, -CN, -CF3, -OH, -NH2, alkyl, heteroalkyl, haloalkyl, hydroxy alkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, - O-heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O-cycloheteroalkyl, -O-halocycloalkyl, -NH- alkyl, -NH-heteroalkyl, -NH-haloalkyl, -NH-cycloalkyl, -NH-cycloheteroalkyl, -NH- halocycloalkyl, aryl, heteroaryl- SO2R23, -OSO2R23, -C(0)0R23, -NR24C(0)R25, -NR24SO2
  • R is selected from each occurrence of R22 is selected from alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -SO2R19, -C(0)Ri9, -C(0)0Ri9, and - C(0)NRI 9 R2O; each occurrence of Ra, Rb, Rc, Rd, Rg, Rh Ri and Rj is independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, hydroxy alkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -SO2R23, -C(0)0R23, -C(0)NR24R25, and -C(0)R26; or Ra taken together with Rb and the carbon atom to which they are bonded form an
  • each occurrence of Ra, Rb, Ri and Rj is independently selected from -H, halogen, -CN, -CF 3 , alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -C(O)OR 23 , -C(O)NR 24 R 25 , and -C(O)R 26 ; or R a taken together with R b and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, ⁇ -lactam, or ⁇ -lactam; each occurrence of Rc, Rd, Re, Rf, Rg and Rh is independently selected from -H, halogen, -CN, -CF 3 , -OH, -NH 2 , alkyl, heteroalkyl, haloalkyl, hydroxyalkyl, cycloal
  • R is selected from each occurrence of Ra, Rb, Rc, Rd, Rg, Rh Ri and Rj is independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, hydroxyalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -SO2R23, -C(0)0R23, -C(0)NR24R25, -0S02(N26)2, - S02(N26)2, -C(0)R26, and alkyl-NHS02-R26; or R a taken together with Rb and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, ⁇ - lactam, or ⁇ -lactam; each occurrence of R23, R24 and R25 is independently selected from -H, alkyl, heteroal
  • R is selected from , and each occurrence of Ra, Rb, Rg and Rh is independently selected from -H, halogen, - CN, -CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, hydroxyalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -SO2R23, -C(0)0R23, -C(0)NR24R25, -S02(N26)2, and - C(0)R26; or Ra taken together with Rb and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, ⁇ -lactam, or ⁇ -lactam; each occurrence of Rc, R d , Re and Rr is independently selected from -H, halogen, -CN, -CF3, -OH, -NH2, alkyl, heteroalkyl, halo
  • each occurrence Ra and Rb is independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, hydroxyalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -SO2R23, -C(O)OR23, and -C(O)NR24R25; or Ra taken together with Rb and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, ⁇ -lactam, or ⁇ -lactam; each occurrence R c and R d is independently selected from -H, halogen, -CN, -CF 3 , - OH, -NH 2 , alkyl, heteroalkyl, haloalkyl, hydroxyalkyl, cycloalkyl, cycloheteroalkyl,
  • each occurrence of R 26 is independently selected from -H, alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl.
  • the compound is selected from the following table:
  • R taken together with R'' and the ring atoms to which they are bonded form a fused five-membered heteroaryl ring, such that formula I is a substituted indole or azaindole.
  • the compound having the structure: wherein each occurrence of R27 is selected from alkyl, heteroalkyl, haloalkyl, hydroxyalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, and heteroaryl.
  • the compound is selected from the following table:
  • the compounds are atropisomers. Additionally, unless otherwise stated, structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms.
  • the (C1-C4)alkyl or the -O-(C1-C4)alkyl can be suitably deuterated (e.g., -CD3, -OCD3).
  • Any compound of the invention can also be radiolabed for the preparation of a radiopharmaceutical agent.
  • Another aspect of the invention relates to methods of treating or preventing a disease or disorder associated with a SLC6A8 mutation, treating an inflammatory disease, increasing transport of guanidinoacetic acid or a salt thereof across the blood-brain barrier, improving the function of a cellular creatine transporter, or decreasing the accumulation or the concentration of guanidinoacetic acid or a salt thereof in a cell, in a subject in need thereof comprising administering to the subject an effective amount of a compound of Formula (I): wherein: A and A' are each independently selected from C-R' and N; A'' is selected from C-R'' and N; provided that at least one of A, A', and A'' is N; or A and A' are each independently selected from C-R' and N, and A'' is C-R'', wherein R'' taken together with R and the ring atoms to which they are bonded form a fused five-membered heteroaryl ring, such that formula I is a substitute
  • « is 0, 1, 2, or 3; m is 1;
  • Ri, R2, R3, R4, RS and Re are independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, aryl, heteroaryl, -C(0)0Rn, -C(0)NRi2Ri3, and -C(0)Ri4;
  • Rv, Rs, R9, and Rio are independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, aryl, heteroaryl, -C(0)0Rn, -C(0)NRi2Ri3, and -C(0)Ri4;
  • Z is selected from -H, halogen, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O- cycloheteroalkyl, -O-halocycloalkyl, -NH-alkyl, -NH-heteroalkyl, -NH-haloalkyl, -NH- cycloalkyl, -NH-cycloheteroalkyl, -NH-halocycloalkyl, aryl, -O-aryl, -NH-aryl, heteroaryl, - O-heteroaryl, -NH-heteroaryl, -C(0)0Rn, -C(0)NRi2Ri3, -NR12R13, and -
  • R is selected from alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O-cycloheteroalkyl, - O-halocycloalkyl, aryl, -O-aryl, heteroaryl, -O-heteroaryl -OSO2R11, and -NRisRie;
  • R is selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O- cycloalkyl, -O-cycloheteroalkyl, -O-halocycloalkyl, -O-aryl, -O-heteroaryl, -OSO2R11, and -
  • R" is selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O- cycloalkyl, -O-cycloheteroalkyl, -O-halocycloalkyl, -O-aryl, -O-heteroaryl, -OSO2R11, and -
  • each occurrence of R11, R12 and R13 is independently selected from H, alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl; each occurrence of Ri4is independently selected from alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl;
  • Ri5 and Rie are independently selected from -H, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -C(0)Rn, -C(0)0Rn, and - C(0)NRI2RI3; or R15 and Rie taken together with the nitrogen atom to which they are bonded form an un substituted or substituted C4-C8 cycloheteroalkyl.
  • the above compounds wherein if A is N, A' and A'' are each C- H, X is , , and Z is selected from phenyl and 2-methoxy-5- fluorophenyl, then R is not .
  • Another aspect of the invention relates to methods of treating or preventing a disease or disorder associated with a SLC6A8 mutation, treating an inflammatory disease, increasing transport of guanidinoacetic acid or a salt thereof across the blood-brain barrier, improving the function of a cellular creatine transporter, or decreasing the accumulation or the concentration of guanidinoacetic acid or a salt thereof in a cell, in a subject in need thereof comprising administering to the subject an effective amount of any of the compounds disclosed herein.
  • Another aspect of the invention relates to methods of treating or preventing a disease or disorder associated with a SLC6A8 mutation, treating an inflammatory disease, increasing transport of guanidinoacetic acid or a salt thereof across the blood-brain barrier, improving the function of a cellular creatine transporter, or decreasing the accumulation or the concentration of guanidinoacetic acid or a salt thereof in a cell, in a subject in need thereof comprising administering to the subject an effective amount of any of the pharmaceutical compositions disclosed herein.
  • Creatine transporter deficiency (CTD) CTD is an inborn error of creatine metabolism in which creatine is not properly transported to the brain and muscles due to defective creatine transporters.
  • CTD is an X- linked disorder caused by mutations in the SLC6A8 gene.
  • the SLC6A8 gene is located on the short arm of the sex chromosome, Xq28.
  • Hemizygous males with CTD express speech and behavior abnormalities, intellectual disabilities, development delay, seizures, and autistic behavior.
  • Heterozygous females with CTD generally express fewer, less severe symptoms.
  • CCD is one of three different types of cerebral creatine deficiency (CCD).
  • the other two types of CCD are guanidinoacetate methyltransferase (GAMT) deficiency and L- arginine:glycine amidinotransferase (AGAT) deficiency.
  • GAMT guanidinoacetate methyltransferase
  • AGAT L- arginine:glycine amidinotransferase
  • CTD Clinical presentation of CTD is similar to that of GAMT and AGAT deficiency.
  • CTD was first identified in 2001 with the presence of a hemizygous nonsense mutation in the SLC6A8 gene in a male patient.
  • CTD is difficult to treat because the actual transporter responsible for transporting creatine to the brain and muscles is defective.
  • Studies in which oral creatine monohydrate supplements were given to patients with CTD found that patients did not respond to treatment.
  • patients that had GAMT or AGAT deficiency were given oral creatine monohydrate supplements found that patient’s clinical symptoms improved.
  • Patients with CTD are unresponsive to oral creatine monohydrate supplements because regardless of the amount of creatine they ingest, the creatine transporter is still defective, and therefore creatine is incapable of being transported across the BBB.
  • the invention provides methods of treating or preventing a disease or disorder associated with a SLC6A8 mutation in a subjet in need thereof, comprising administering to the subject an effective amount of a compound disclosed herein.
  • the disease or disorder is creatine transporter deficiency. In some embodiments, the disease or disorder is motor dysfunction. In some embodiments, the disease or disorder is intellectual disability. In some embodiments, the disease or disorder is language delay or speech delay. In some embodiments, the disease or disorder is hypotonia.
  • the disease or disorder is seizures. In some embodiments, the disease or disorder is behaviours associated with autism and attention deficit hyperactivity disorder.
  • the disease or disorder is fatigue. In some embodiments, the disease or disorder is muscular hypotonia. In some embodiments, the disease or disorder is low weight gain. In some embodiments, the disease or disorder is gastrointestinal disorders. In some embodiments, the disease or disorder is cardiac disorders.
  • the invention provides a method of improving the function of a cellular creatine transporter in a subject in need thereof, comprising administering to the subject an effective amount of a compound disclosed herein.
  • the invention provides a method of increasing cellular trafficking of a cellular creatine transporter in a subject in need thereof, comprising administering to the subject an effective amount of a compound disclosed herein.
  • the invention provides a method of correcting a defect in a cellular creatine transporter function in a subject in need thereof, comprising administering to the subject an effective amount of a compound disclosed herein.
  • the method wherein the cellular creatine transporter is SLC6A8. In some embodiments, the method wherein cellular creatine transporter is a mutant creatine transporter. In some embodiments, the method wherein mutant creatine transporter is mutant SLC6A8. In some embodiments, the method wherein the cellular concentration of creatine is increased.
  • the invention provides a method of decreasing accumulation or the concentration of guanidinoacetic acid or a salt thereof in a cell of a subject in need thereof, comprising administering to the subject an effective amount of a compound dislosed herein.
  • the invention provides a method of decreasing accumulation or the concentration of guanidinoacetic acid or a salt thereof in a cell of a subject in need thereof, comprising administering to the subject an effective amount of a compound dislosed herein that increases transport of guanidinoacetic acid or a salt thereof by a mutant creatine transporter.
  • the method wherein compound decreases the intracellular concentration of guanidinoacetic acid or a salt thereof. In some embodiments, the method wherein compound decreases the intracellular accumulation of guanidinoacetic acid or a salt thereof.
  • the invention provides a method of increasing transport of guanidinoacetic acid or a salt thereof across the blood-brain barrier in the subject in need thereof, comprising administering to the subject aneffective amount of a compound dislosed herein.
  • the invention provides a method of treating an inflammatory disease in a subject in need thereof, comprising administering to the subject aneffective amount of a compound dislosed herein.
  • the method wherein the inflammatory disease is acute. In some embodiments, the method wherein the inflammatory disease is chronic.
  • the method wherein the inflammatory disease is selected from inflammatory bowel diseases (for example, ulcerative colitis or Crohn's disease), multiple sclerosis, psoriasis, arthritis, rheumatoid arthritis, osteoarthritis, juvenile arthritis, psoriatic arthritis, reactive arthritis, ankylosing spondylitis, cryopyrin associated periodic syndromes, Muckle- Wells syndrome, familial cold auto-inflammatory syndrome, neonatal-onset multisystem inflammatory disease, TNF receptor associated periodic syndrome, acute and chronic pancreatitis, atherosclerosis, gout, ankylosing spondylitis, fibrotic disorders (for example, hepatic fibrosis or idiopathic pulmonary fibrosis), nephropathy, sarcoidosis, scleroderma, anaphylaxis, diabetes (for example, diabetes mellitus type 1 or diabetes mellitus type 2), diabetic retinopathy, Still's disease, vasculitis, s
  • host reaction for example, graft vs. host disease
  • allograft rejections for example, acute allograft rejection or chronic allograft rejection
  • early transplantation rejection for example, acute allograft rejection
  • reperfusion injury pain (for example, acute pain, chronic pain, neuropathic pain, or fibromyalgia), chronic infections, meningitis, encephalitis, myocarditis, gingivitis, post- surgical trauma, tissue injury, traumatic brain injury, enterocolitis, sinusitis, uveitis, ocular inflammation, optic neuritis, gastric ulcers, esophagitis, peritonitis, periodontitis, dermatomyositis, gastritis, myositis, polymyalgia, pneumonia, and bronchitis.
  • the cell is a brain cell. In some embodiments of any of the disclosed methods, wherein the subject is a mammal. In some embodiments of any of the disclosed methods, the mammal is a male. In some embodiments of any of the disclosed methods, the mammal is a female. In some embodiments of any of the disclosed methods, the mammal is a primate, equine, bovine, ovine, feline, or canine. In some embodiments of any of the disclosed methods, the mammal is a human. Pharmaceutical Compositions, Routes of Administration, and Dosing In certain embodiments, the invention is directed to a pharmaceutical composition, comprising a compound of the invention and a pharmaceutically acceptable carrier.
  • the pharmaceutical composition comprises a plurality of compounds of the invention and a pharmaceutically acceptable carrier.
  • a pharmaceutical composition of the invention further comprises at least one additional pharmaceutically active agent other than a compound of the invention.
  • the at least one additional pharmaceutically active agent can be an agent useful in the treatment of ischemia-reperfusion injury.
  • Pharmaceutical compositions of the invention can be prepared by combining one or more compounds of the invention with a pharmaceutically acceptable carrier and, optionally, one or more additional pharmaceutically active agents.
  • an “effective amount” refers to any amount that is sufficient to achieve a desired biological effect.
  • an effective prophylactic or therapeutic treatment regimen can be planned which does not cause substantial unwanted toxicity and yet is effective to treat the particular subject.
  • the effective amount for any particular application can vary depending on such factors as the disease or condition being treated, the particular compound of the invention being administered, the size of the subject, or the severity of the disease or condition.
  • One of ordinary skill in the art can empirically determine the effective amount of a particular compound of the invention and/or other therapeutic agent without necessitating undue experimentation.
  • a maximum dose may be used, that is, the highest safe dose according to some medical judgment.
  • intravenous administration of a compound may typically be from 0.1 mg/kg/day to 20 mg/kg/day. In one embodiment, intravenous administration of a compound may typically be from 0.1 mg/kg/day to 2 mg/kg/day. In one embodiment, intravenous administration of a compound may typically be from 0.5 mg/kg/day to 5 mg/kg/day. In one embodiment, intravenous administration of a compound may typically be from 1 mg/kg/day to 20 mg/kg/ day. In one embodiment, intravenous administration of a compound may typically be from 1 mg/kg/day to 10 mg/kg/day.
  • daily oral doses of a compound will be, for human subjects, from about 0.01 milligrams/kg per day to 1000 milligrams/kg per day. It is expected that oral doses in the range of 0.5 to 50 milligrams/kg, in one or more administrations per day, will yield therapeutic results. Dosage may be adjusted appropriately to achieve desired drug levels, local or systemic, depending upon the mode of administration. For example, it is expected that intravenous administration would be from one order to several orders of magnitude lower dose per day. In the event that the response in a subject is insufficient at such doses, even higher doses (or effective higher doses by a different, more localized delivery route) may be employed to the extent that patient tolerance permits. Multiple doses per day are contemplated to achieve appropriate systemic levels of the compound.
  • the therapeutically effective amount can be initially determined from animal models.
  • a therapeutically effective dose can also be determined from human data for compounds which have been tested in humans and for compounds which are known to exhibit similar pharmacological activities, such as other related active agents. Higher doses may be required for parenteral administration.
  • the applied dose can be adjusted based on the relative bioavailability and potency of the administered compound. Adjusting the dose to achieve maximal efficacy based on the methods described above and other methods as are well-known in the art is well within the capabilities of the ordinarily skilled artisan.
  • compositions of the invention can be administered in pharmaceutically acceptable solutions, which may routinely contain pharmaceutically acceptable concentrations of salt, buffering agents, preservatives, compatible carriers, adjuvants, and optionally other therapeutic ingredients.
  • an effective amount of the compound can be administered to a subject by any mode that delivers the compound to the desired surface.
  • Administering a pharmaceutical composition may be accomplished by any means known to the skilled artisan. Routes of administration include but are not limited to intravenous, intramuscular, intraperitoneal, intravesical (urinary bladder), oral, subcutaneous, direct injection (for example, into a tumor or abscess), mucosal (e.g., topical to eye), inhalation, and topical.
  • a compound of the invention can be formulated as a lyophilized preparation, as a lyophilized preparation of liposome-intercalated or -encapsulated active compound, as a lipid complex in aqueous suspension, or as a salt complex.
  • Lyophilized formulations are generally reconstituted in suitable aqueous solution, e.g., in sterile water or saline, shortly prior to administration.
  • the compounds can be formulated readily by combining the active compound(s) with pharmaceutically acceptable carriers well known in the art.
  • Such carriers enable the compounds of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a subject to be treated.
  • Pharmaceutical preparations for oral use can be obtained as solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores.
  • Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP).
  • disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate.
  • the oral formulations may also be formulated in saline or buffers, e.g., EDTA for neutralizing internal acid conditions or may be administered without any carriers.
  • oral dosage forms of the above component or components may be chemically modified so that oral delivery of the derivative is efficacious.
  • the chemical modification contemplated is the attachment of at least one moiety to the component molecule itself, where said moiety permits (a) inhibition of acid hydrolysis; and (b) uptake into the blood stream from the stomach or intestine.
  • the increase in overall stability of the component or components and increase in circulation time in the body examples include: polyethylene glycol, copolymers of ethylene glycol and propylene glycol, carboxymethyl cellulose, dextran, polyvinyl alcohol, polyvinyl pyrrolidone and polyproline.
  • the location of release may be the stomach, the small intestine (the duodenum, the jejunum, or the ileum), or the large intestine.
  • the stomach the small intestine (the duodenum, the jejunum, or the ileum), or the large intestine.
  • One skilled in the art has available formulations which will not dissolve in the stomach, yet will release the material in the duodenum or elsewhere in the intestine.
  • the release will avoid the deleterious effects of the stomach environment, either by protection of the compound of the invention (or derivative) or by release of the biologically active material beyond the stomach environment, such as in the intestine.
  • a coating impermeable to at least pH 5.0 is essential.
  • cellulose acetate trimellitate hydroxypropylmethylcellulose phthalate
  • HPMCP 50 HPMCP 55
  • PVAP polyvinyl acetate phthalate
  • CAP cellulose acetate phthalate
  • shellac shellac
  • These coatings may be used as mixed films.
  • a coating or mixture of coatings can also be used on tablets, which are not intended for protection against the stomach. This can include sugar coatings, or coatings which make the tablet easier to swallow.
  • Capsules may consist of a hard shell (such as gelatin) for delivery of dry therapeutic (e.g., powder); for liquid forms, a soft gelatin shell may be used.
  • the shell material of cachets could be thick starch or other edible paper.
  • moist massing techniques can be used.
  • the therapeutic can be included in the formulation as fine multi-particulates in the form of granules or pellets of particle size about 1 mm.
  • the formulation of the material for capsule administration could also be as a powder, lightly compressed plugs or even as tablets.
  • the therapeutic could be prepared by compression. Colorants and flavoring agents may all be included.
  • the compound of the invention may be formulated (such as by liposome or microsphere encapsulation) and then further contained within an edible product, such as a refrigerated beverage containing colorants and flavoring agents.
  • an edible product such as a refrigerated beverage containing colorants and flavoring agents.
  • One may dilute or increase the volume of the therapeutic with an inert material.
  • These diluents could include carbohydrates, especially mannitol, ⁇ -lactose, anhydrous lactose, cellulose, sucrose, modified dextrans and starch.
  • Certain inorganic salts may be also be used as fillers including calcium triphosphate, magnesium carbonate and sodium chloride.
  • Some commercially available diluents are Fast-Flo, Emdex, STA-Rx 1500, Emcompress and Avicell.
  • Disintegrants may be included in the formulation of the therapeutic into a solid dosage form.
  • Materials used as disintegrates include but are not limited to starch, including the commercial disintegrant based on starch, Explotab. Sodium starch glycolate, Amberlite, sodium carboxymethylcellulose, ultramylopectin, sodium alginate, gelatin, orange peel, acid carboxymethyl cellulose, natural sponge and bentonite may all be used.
  • Another form of the disintegrants are the insoluble cationic exchange resins.
  • Powdered gums may be used as disintegrants and as binders and these can include powdered gums such as agar, Karaya or tragacanth. Alginic acid and its sodium salt are also useful as disintegrants.
  • Binders may be used to hold the therapeutic agent together to form a hard tablet and include materials from natural products such as acacia, tragacanth, starch and gelatin. Others include methyl cellulose (MC), ethyl cellulose (EC) and carboxymethyl cellulose (CMC). Polyvinyl pyrrolidone (PVP) and hydroxypropylmethyl cellulose (HPMC) could both be used in alcoholic solutions to granulate the therapeutic.
  • MC methyl cellulose
  • EC ethyl cellulose
  • CMC carboxymethyl cellulose
  • PVP polyvinyl pyrrolidone
  • HPMC hydroxypropylmethyl cellulose
  • Lubricants may be used as a layer between the therapeutic and the die wall, and these can include but are not limited to; stearic acid including its magnesium and calcium salts, polytetrafluoroethylene (PTFE), liquid paraffin, vegetable oils and waxes. Soluble lubricants may also be used such as sodium lauryl sulfate, magnesium lauryl sulfate, polyethylene glycol of various molecular weights, Carbowax 4000 and 6000.
  • the glidants may include starch, talc, pyrogenic silica and hydrated silicoaluminate.
  • Surfactants may include anionic detergents such as sodium lauryl sulfate, dioctyl sodium sulfosuccinate and dioctyl sodium sulfonate.
  • anionic detergents such as sodium lauryl sulfate, dioctyl sodium sulfosuccinate and dioctyl sodium sulfonate.
  • Cationic detergents which can be used and can include benzalkonium chloride and benzethonium chloride.
  • Non-ionic detergents that could be included in the formulation as surfactants include lauromacrogol 400, polyoxyl 40 stearate, polyoxyethylene hydrogenated castor oil 10, 50 and 60, glycerol monostearate, polysorbate 40, 60, 65 and 80, sucrose fatty acid ester, methyl cellulose and carboxy methyl cellulose. These surfactants could be present in the formulation of the compound of the invention or derivative either alone or as a mixture in different ratios.
  • compositions which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol.
  • the push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers.
  • the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols.
  • stabilizers may be added.
  • Microspheres formulated for oral administration may also be used. Such microspheres have been well defined in the art.
  • compositions for oral administration should be in dosages suitable for such administration.
  • buccal administration the compositions may take the form of tablets or lozenges formulated in conventional manner.
  • topical administration the compound may be formulated as solutions, gels, ointments, creams, suspensions, etc. as are well-known in the art.
  • Systemic formulations include those designed for administration by injection, e.g., subcutaneous, intravenous, intramuscular, intrathecal or intraperitoneal injection, as well as those designed for transdermal, transmucosal oral or pulmonary administration.
  • compounds for use according to the present invention may be conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • a suitable propellant e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas.
  • the dosage unit may be determined by providing a valve to deliver a metered amount.
  • Capsules and cartridges of e.g., gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch.
  • the compound is delivered to the lungs of a mammal while inhaling and traverses across the lung epithelial lining to the blood stream.
  • Other reports of inhaled molecules include Adjei et al., Pharm Res 7:565-569 (1990); Adjei et al., Int J Pharmaceutics 63:135-144 (1990) (leuprolide acetate); Braquet et al., J Cardiovasc Pharmacol 13(suppl.
  • No.5,284,656 granulocyte colony stimulating factor; incorporated by reference.
  • a method and composition for pulmonary delivery of drugs for systemic effect is described in U.S. Pat. No.5,451,569 (incorporated by reference), issued Sep.19, 1995 to Wong et al.
  • Contemplated for use in the practice of this invention are a wide range of mechanical devices designed for pulmonary delivery of therapeutic products, including but not limited to nebulizers, metered dose inhalers, and powder inhalers, all of which are familiar to those skilled in the art.
  • Some specific examples of commercially available devices suitable for the practice of this invention are the Ultravent nebulizer, manufactured by Mallinckrodt, Inc., St.
  • each formulation is specific to the type of device employed and may involve the use of an appropriate propellant material, in addition to the usual diluents, adjuvants and/or carriers useful in therapy. Also, the use of liposomes, microcapsules or microspheres, inclusion complexes, or other types of carriers is contemplated.
  • Chemically modified compound of the invention may also be prepared in different formulations depending on the type of chemical modification or the type of device employed.
  • Formulations suitable for use with a nebulizer will typically comprise a compound of the invention (or derivative) dissolved in water at a concentration of about 0.1 to 25 mg of biologically active compound of the invention per mL of solution.
  • the formulation may also include a buffer and a simple sugar (e.g., for inhibitor stabilization and regulation of osmotic pressure).
  • the nebulizer formulation may also contain a surfactant, to reduce or prevent surface induced aggregation of the compound of the invention caused by atomization of the solution in forming the aerosol.
  • Formulations for use with a metered-dose inhaler device will generally comprise a finely divided powder containing the compound of the invention (or derivative) suspended in a propellant with the aid of a surfactant.
  • the propellant may be any conventional material employed for this purpose, such as a chlorofluorocarbon, a hydrochlorofluorocarbon, a hydrofluorocarbon, or a hydrocarbon, including trichlorofluoromethane, dichlorodifluoromethane, dichlorotetrafluoroethanol, and 1,1,1,2-tetrafluoroethane, or combinations thereof.
  • Suitable surfactants include sorbitan trioleate and soya lecithin. Oleic acid may also be useful as a surfactant.
  • Formulations for dispensing from a powder inhaler device will comprise a finely divided dry powder containing compound of the invention (or derivative) and may also include a bulking agent, such as lactose, sorbitol, sucrose, or mannitol in amounts which facilitate dispersal of the powder from the device, e.g., 50 to 90% by weight of the formulation.
  • the compound of the invention (or derivative) should advantageously be prepared in particulate form with an average particle size of less than 10 micrometers ( ⁇ m), most preferably 0.5 to 5 ⁇ m, for most effective delivery to the deep lung. Nasal delivery of a pharmaceutical composition of the present invention is also contemplated.
  • Nasal delivery allows the passage of a pharmaceutical composition of the present invention to the blood stream directly after administering the therapeutic product to the nose, without the necessity for deposition of the product in the lung.
  • Formulations for nasal delivery include those with dextran or cyclodextran.
  • a useful device is a small, hard bottle to which a metered dose sprayer is attached.
  • the metered dose is delivered by drawing the pharmaceutical composition of the present invention solution into a chamber of defined volume, which chamber has an aperture dimensioned to aerosolize and aerosol formulation by forming a spray when a liquid in the chamber is compressed.
  • the chamber is compressed to administer the pharmaceutical composition of the present invention.
  • the chamber is a piston arrangement.
  • Such devices are commercially available.
  • a plastic squeeze bottle with an aperture or opening dimensioned to aerosolize an aerosol formulation by forming a spray when squeezed is used.
  • the opening is usually found in the top of the bottle, and the top is generally tapered to partially fit in the nasal passages for efficient administration of the aerosol formulation.
  • the nasal inhaler will provide a metered amount of the aerosol formulation, for administration of a measured dose of the drug.
  • the compounds, when it is desirable to deliver them systemically, may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative.
  • compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents.
  • Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes.
  • Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethylcellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
  • the active compounds may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
  • a suitable vehicle e.g., sterile pyrogen-free water
  • the compounds may also be formulated in rectal or vaginal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.
  • a compound may also be formulated as a depot preparation.
  • Such long acting formulations may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
  • compositions also may comprise suitable solid or gel phase carriers or excipients.
  • suitable solid or gel phase carriers or excipients include but are not limited to calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols.
  • Suitable liquid or solid pharmaceutical preparation forms are, for example, aqueous or saline solutions for inhalation, microencapsulated, encochleated, coated onto microscopic gold particles, contained in liposomes, nebulized, aerosols, pellets for implantation into the skin, or dried onto a sharp object to be scratched into the skin.
  • the pharmaceutical compositions also include granules, powders, tablets, coated tablets, (micro)capsules, suppositories, syrups, emulsions, suspensions, creams, drops or preparations with protracted release of active compounds, in whose preparation excipients and additives and/or auxiliaries such as disintegrants, binders, coating agents, swelling agents, lubricants, flavorings, sweeteners or solubilizers are customarily used as described above.
  • the pharmaceutical compositions are suitable for use in a variety of drug delivery systems. For a brief review of methods for drug delivery, see Langer R, Science 249:1527-33 (1990).
  • the compound of the invention and optionally other therapeutics may be administered per se (neat) or in the form of a pharmaceutically acceptable salt or cocrystal.
  • a pharmaceutically acceptable salt or cocrystal When used in medicine the salts or cocrystals should be pharmaceutically acceptable, but non- pharmaceutically acceptable salts or cocrystals may conveniently be used to prepare pharmaceutically acceptable salts or cocrystals thereof.
  • Such salts include, but are not limited to, those prepared from the following acids: hydrochloric, hydrobromic, sulphuric, nitric, phosphoric, maleic, acetic, salicylic, p -toluene sulphonic, tartaric, citric, methane sulphonic, formic, malonic, succinic, naphthal ene-2- sulphoni c, and benzene sulphonic.
  • such salts can be prepared as alkaline metal or alkaline earth salts, such as sodium, potassium or calcium salts of the carboxylic acid group.
  • Suitable buffering agents include: acetic acid and a salt (1-2% w/v); citric acid and a salt (1-3% w/v); boric acid and a salt (0.5-2.5% w/v); and phosphoric acid and a salt (0.8-2% w/v).
  • Suitable preservatives include benzalkonium chloride (0.003-0.03% w/v); chlorobutanol (0.3-0.9% w/v); parabens (0.01-0.25% w/v) and thimerosal (0.004-0.02% w/v).
  • compositions of the invention contain an effective amount of a compound as described herein and optionally therapeutic agents included in a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable carrier means one or more compatible solid or liquid filler, diluents or encapsulating substances which are suitable for administration to a human or other vertebrate animal.
  • carrier denotes an organic or inorganic ingredient, natural or synthetic, with which the active ingredient is combined to facilitate the application.
  • the components of the pharmaceutical compositions also are capable of being commingled with the compounds of the present invention, and with each other, in a manner such that there is no interaction which would substantially impair the desired pharmaceutical efficiency.
  • the therapeutic agent(s), including specifically but not limited to a compound of the invention, may be provided in particles.
  • Particles as used herein means nanoparticles or microparticles (or in some instances larger particles) which can consist in whole or in part of the compound of the invention or the other therapeutic agent(s) as described herein.
  • the particles may contain the therapeutic agent(s) in a core surrounded by a coating, including, but not limited to, an enteric coating.
  • the therapeutic agent(s) also may be dispersed throughout the particles.
  • the therapeutic agent(s) also may be adsorbed into the particles.
  • the particles may be of any order release kinetics, including zero-order release, first-order release, second-order release, delayed release, sustained release, immediate release, and any combination thereof, etc.
  • the particle may include, in addition to the therapeutic agent(s), any of those materials routinely used in the art of pharmacy and medicine, including, but not limited to, erodible, nonerodible, biodegradable, or nonbiodegradable material or combinations thereof.
  • the particles may be microcapsules which contain the compound of the invention in a solution or in a semi-solid state. The particles may be of virtually any shape.
  • Both non-biodegradable and biodegradable polymeric materials can be used in the manufacture of particles for delivering the therapeutic agent(s).
  • Such polymers may be natural or synthetic polymers.
  • the polymer is selected based on the period of time over which release is desired.
  • Bioadhesive polymers of particular interest include bioerodible hydrogels described in Sawhney H S et al. (1993 ) Macromolecules 26:581-7, the teachings of which are incorporated herein.
  • polyhyaluronic acids casein, gelatin, glutin, polyanhydrides, polyacrylic acid, alginate, chitosan, poly(methyl methacrylates), poly(ethyl methacrylates), poly(butylmethacrylate), poly (isobutyl methacrylate), poly(hexylmethacrylate), poly(isodecyl methacrylate), poly(lauryl methacrylate), poly(phenyl methacrylate), poly(methyl acrylate), poly (isopropyl acrylate), poly (isobutyl acrylate), and poly(octadecyl acrylate).
  • controlled release is intended to refer to any drug-containing formulation in which the manner and profile of drug release from the formulation are controlled. This refers to immediate as well as non-immediate release formulations, with non-immediate release formulations including but not limited to sustained release and delayed release formulations.
  • sustained release also referred to as “extended 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 preferably, although not necessarily, results in substantially constant blood levels of a drug over an extended time period.
  • delayed release is used in its conventional sense to refer to a drug formulation in which there is a time delay between administration of the formulation and the release of the drug there from. “Delayed release” may or may not involve gradual release of drug over an extended period of time, and thus may or may not be “sustained release.”
  • Long-term sustained release implant may be particularly suitable for treatment of chronic conditions.
  • Long-term release as used herein, means that the implant is constructed and arranged to deliver therapeutic levels of the active ingredient for at least 7 days, and preferably 30-60 days.
  • Long-term sustained release implants are well-known to those of ordinary skill in the art and include some of the release systems described above.
  • Example 1 PathHunter MEM-EA Pharmacotrafficking Assay for SLC6A8 CTD mutants
  • Cell Lines Preparation of Cells U-2 OS MEM-EA cells were purchased from Eurofins (catalog #93-1101C3). From these parental cells, stable cell lines expressing SLC6A8 CTD mutants were made using standard cell culture protocols, involving transfections of plasmids followed by antibiotic selection. These plasmids encoded CTD mutant SLC6A8 proteins with a C-terminal ProLink2 tag.
  • U-2 OS MEM-EA cells and derived stable cell lines were grown in RPMI medium 1640 (Thermo Fisher Scientific, catalog #A10491-01) supplemented with 10% Fetal Bovine Serum (FBS), 200 ug/mL hygromycin B (Thermo Fisher Scientific, catalog #10687010), 100 mg/mL streptomycin, and 100 U/mL penicillin. Cells were grown at 37°C in a humidified CO 2 incubator. Assay U-2 OS MEM-EA cells stably expressing SLC6A8 CTD mutants were plated into white-walled 96-well plates (Corning, catalog #3903) at a density of 20,000 cells per well.
  • the parental U-2 OS MEM-EA cells were also plated. After 24 hrs, compounds were dispensed directly into the plated cells using the Tecan D300e Digital Dispenser. After an additional 24 hrs, the media with compound was again removed and white covers (Thermo Fisher Scientific, catalog #236272) were placed on the bottoms of the 96-well plates. Luminescence indicative of SLC6A8 CTD mutant cell surface localization was measured according to the manufacturer’s protocol, using the PathHunter Detection kit (Eurofins catalog #93-0001L) and an EnVision plate reader (PerkinElmer, 2104 multilabel reader). Data were analyzed in Excel.
  • Example 2 Corrector Assay for SLC6A8 CTD mutant Cell Lines Preparation of Cells
  • a number of SLC6A8 CTD mutant cell lines were made in U-2 OS MEM-EA cells, 293T cells, HeLa cells, and CHO cells. All cells lines were generated as described above for U-2 OS MEM-EA cells, namely stable cell lines expressing SLC6A8 CTD mutants were made using standard cell culture protocols involving transfections of plasmids followed by antibiotic selection.
  • Assay Stable cell lines expressing CTD mutants were plated into 96-well plates (Corning, catalog #3595) at a density of 40,000 cells per well. After 24 hrs, compounds were dispensed directly into the plated cells using a Tecan D300e Digital Dispenser. After an additional 24 hrs, the media with compound was removed. Cells were then incubated with a solution of 100 uM D3-creatine (SIGMA, 616249-1G) in media (without FBS). This solution was incubated with the cells for a 30 min incubation at 37°C. After the incubation, the media was removed, and the cells were washed once with 180 uL of phosphate buffered solution (PBS).
  • PBS phosphate buffered solution
  • Step 1 Synthesis of compound 2 PBr 3 (0.8 eq.) was slowly added at 0 o C over 30 minutes to a solution of compound 1 (1 eq.) in anhydrous DCM (0.3 M), and the resulting mixture was stirred at room temperature for 2 hours. The mixture was then poured into a saturated NaHCO3 solution and washed twice with DCM. The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was then purified via flash column chromatography (eluted with PE/EtOAc) to give pure compound 2.
  • Step 2 Synthesis of compound 3 1M TBAF in THF (1.5 eq.) was added dropwise at 0 o C over 30 minutes to a mixture of compound 2 (1 eq.) and TMSCN (1.5 eq.) in acetonitrile (0.3M). The resulting mixture was stirred at room temperature for 1-4 hours and monitored by TLC. The mixture was then diluted with water and extracted with EtOAc (x3). The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified via flash column chromatography (eluted with PE/EtOAc) to give pure compound 3.
  • Step 3 Synthesis of compound 4 EtMgBr (2.2 eq., 3 M in Et2O) was added dropwise at -78 o C under N2 atmosphere to a solution of Ti(O-iPr) 4 (1.2 eq.) in anhydrous THF (0.15M). After stirring at -78 o C for 1 hour, a solution of compound 3(1 eq) in anhydrous THF (15M) was added to the above mixture and the resulting mixture was slowly warmed to room temperature (2-4 hours). Boron trifluoride diethyl etherate (1.5-2 eq.) was then added, and the mixture was stirred at room temperature for 1hour. After completion, the mixture was quenched with 1 Maq.
  • Step 4 Synthesis of compound 6 EDCI (1.2 eq.), HOBt (1.2 eq.) and NMM (3 eq.) were added to a mixture of compound 4 (1 eq.) and compound 5 (1.1 eq.) in DMF (0.2M), and the resulting mixture was stirred at room temperature for 16 hours. The mixture was then diluted with saturated aq. NH 4 Cl solution and extracted twice with EtOAc.
  • Step 5 Synthesis of compound 7
  • the appropriate amine (2 eq.) was added to a mixture of compound 6 (1 eq.) and DIEA (3 eq.) in NMP (0.2M), and the resulting mixture was stirred in a sealed tube at 160 o C for 6-10 hours. The mixture was then cooled down, diluted with water, and extracted twice with EtOAc. The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated under vacuum.
  • the residue was purified via prep-HPLC (Gemini 5 ⁇ m C18 250*21.2mm, H 2 O/MeCN (5-95 %)/0.1% NH 3 . H 2 O) to give pure compound 7.
  • Step 2 Synthesis of M1-int-3 TBAF (12.7 mL, 1 M in THF) was added dropwise at 0 o C over 30 minutes to a mixture of M1- int-2 (1.82 g, 8.46 mmol) and TMSCN (1.27 g, 12.7 mmol) in acetonitrile (30 mL), andthe resulting mixture was stirred at room temperature for 3 hours.
  • Step 3 Synthesis of M1-int-4 EtMgBr (3.7 mL, 3 M in Et 2 O) was added dropwise at -78 o C under N 2 atmosphere to a solution of Ti(O- i Pr) 4 (1.72 g, 6.04 mmol) in anhydrous THF (40 mL). The reaction mixture was then stirred at -78 o C for 1 hour before a solution of M1-int-3(810 mg, 5.03 mmol) in anhydrous THF (3 mL) was added. The resulting mixture was then slowly warmed to room temperature (3 hours). Boron trifluoride diethyl etherate (1.07 g, 7.55 mmol) was then added and the mixture was stirred at room temperature for 1 hour.
  • Step 4 Synthesis of M1-int-6 EDCI (152 mg, 0.8 mmol), HOBt (108 mg, 0.8 mmol) and NMM (202 mg, 2 mmol) were added to a mixture of M1-int-4 (210 mg, 0.66 mmol) and M1-int-5(114 mg, 0.73 mmol) in DMF (4 mL), and the resulting mixture was stirred at room temperature for 16 hours. The mixture was diluted with saturated aq. NH4Cl (40 mL) and extracted twice with EtOAc (40 mL). The combined organic layers werewashed with brine, dried over anhydrous Na 2 SO 4 , filtered, and concentrated under vacuum.
  • M1-int-6 EDCI 152 mg, 0.8 mmol
  • HOBt 108 mg, 0.8 mmol
  • NMM 202 mg, 2 mmol
  • Step 5 Synthesis of M1 Cis-2,6-dimethylmorpholine (51 mg, 0.44 mmol) was added to a mixture of M1-int-6 (72 mg, 0.22 mmol) and DIEA (86 mg, 0.66 mmol) in NMP (3 mL), and the resulting mixture was stirred in a sealed tube at 160 o C for 8 hours.
  • EtMgBr (2.2-5.8 eq., 3 M in EtiO) was added dropwise at -78°C under N2 atmosphere to a solution of Ti(0-/Pr)4 (1.2-3 eq.) in anhydrous THE (0.25M).
  • the reaction mixture was stirred at -78°C for 1 hour before a solution of compound 6 (1 eq.) in anhydrous THF (0.5M) was added.
  • the resulting mixture was slowly warmed to room temperature (2-4 hours), and BF3- Et2O (1.5 ⁇ 2 eq.) was added.
  • the reaction mixture was then stirred at room temperature for 1 hour. After completion, the mixture was quenched with 1 M aq. HCl and extracted twice with MTBE.
  • Step 5 Synthesis of compound 8 EDCI (1.2 eq), HOBt (1.2 eq.) and NMM (3 eq) were added to a mixture of compound 7 (1 eq.) and compound 2 (1.1 eq.) in DMF (0.1M), and the resulting mixture was stirred at room temperature for 16 hours. The mixture was then diluted with saturated aq. NH4Cl and extracted twice with EtOAc.
  • EtMgBr (4.5 mL, 3 M in EtiO) was added dropwise at -78°C under N2 atmosphere to a solution of Ti(0-/Pr)4 (0.79 mL, 7.25 mmol) in anhydrous THF (30 mL).
  • the reaction mixture was stirred at -78°C for 1 hour before a solution of M2-int-5 (430 mg, 2.9 mmol) in anhydrous THF (5 mL) was added.
  • the resulting mixture was slowly warmed to room temperature over 3 hours, then BF3-Et20 (618 mg, 4.35 mmol) was added, and the mixture was stirred at room temperature for 1 hour. After completion, the mixture was quenched with 1 M aq.
  • Step 4 Synthesis of M2 EDCI (53 mg, 0.27 mmol), HOBt (37 mg, 0.27 mmol) and NMM (51 mg, 0.5 mmol) were added to a mixture of M2-int-6 (60 mg, 0.17 mmol) and M2-int-2 (44 mg, 0.19 mmol) in DMF (2 mL), andthe resulting mixture was stirred at room temperature for 16 hours. The mixture was then diluted with saturated aq. NH 4 Cl (30 mL) and extracted twice with EtOAc (30 mL). The combined organic layers were dried over anhydrous Na2SO4, filtered, and concentrated under vacuum.
  • Step 1 Synthesis of compound 2 n-BuLi (1.2 eq., 1.6 M in hexane) was added dropwise at -78°C under N2 atmosphere to a solution of compound 1 (1 eq.) in anhydrous THF (0.1M). After stirring at-78°C for 1 hour, BF3 Et20 (2 eq.) and the corresponding epoxide (2 eq.) were added to the above mixture. The resulting mixture was then stirred at -78°C for an additional hour, quenched with iced saturated aq. NHrCl, and extracted twice with EtOAc. The combined organic layers were washed with brine, dried over anhydrous Na2SOr, filtered, and concentrated under vacuum. The residue was purified by column chromatography on silica gel (eluted with PE/EtOAc) to give pure compound 2.
  • Step 3 Synthesis of compound 4 NaN 3 (2 eq.) was added to a solution of compound 3 (1 eq.) in DMF (0.2M), and the resulting mixture was stirred at 60°C for 8 hours. The mixture was then diluted with H2O and extracted twice with EtOAc. The combined organic layers were washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated under vacuum to give compound 4, which was used for next reaction without any further purification.
  • Step 4 Synthesis of compound 5 Pd/C (10%, w/w) was added to a solution of compound 4 (1 eq.) in EtOAc(0.05M), and the resulting mixture was stirred under 25 psi H2 atmosphere for 16 hours at room temperature.
  • Step 5 Synthesis of compound 7 EDCI (1.2 eq.), HOBt (1.3 eq.) and NMM (3 eq.) were added under N2 atmosphere to a mixture of compound 5 (1 eq.) and compound 6 (1.1 eq.) in DMF (0.1M), and the resulting mixture was stirred at room temperature for 8 hours. The mixture was then diluted with saturated aq. NH 4 Cl and extracted twice with EtOAc. The combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified via prep-HPLC (Gemini 5 ⁇ m C18 250*21.2mm, H 2 O/MeCN (5-95 %)/0.1% NH 3 . H 2 O) to give pure compound 7.
  • Step 2 Synthesis of M3-int-3 MsCl (528 mg, 4.6 mmol) was added at 0°C under N 2 atmosphere to a mixture of M3-int-2 (520 mg, 3.06 mmol) and DIEA (1.2 g, 9.18 mmol) in anhydrous DCM (20 mL), and the resulting mixture was allowed to warm to room temperature over 1 hour. The mixture was then quenched with saturated aq. NaHCO 3 and extracted twice with DCM. The combined organic layers were washed with brine, dried over anhydrous Na 2 SO 4 , filtered, and concentrated under vacuum to give crude M3-int-3 (660 mg, 87.0 % yield) as a yellow oil without any further purification.
  • Step 3 Synthesis of M3-int-4 NaN3 (278 mg, 4.26 mmol) was added to a solution of M3-int-3 (660 mg, 2.66 mmol) in DMF (15 mL) and the resulting mixture was stirred at 60°C for 6 hours The reaction mixture was then cooled down, diluted with H2O, and extracted twice with EtOAc. The combined organic layers were washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated under vacuum to give M3-int-4 (220 mg, 42.4 % yield) as a yellow oil without any further purification.
  • Step 5 Synthesis of M3 EDCI (122 mg, 0.64 mmol), HOBt (87 mg, 0.64 mmol) and NMM (160 mg, 1.6 mmol) were added under N2 atmosphere to a mixture of M3-int-5 (90 mg, 0.53 mmol) and M3-int-6 (138 mg, 0.59 mmol) in DMF (4 mL), and the resulting mixture was stirred at room temperature for 8 hours. The mixture was then diluted with saturated aq. NH4Cl (20 mL) and extracted twice with EtOAc (20 mL).
  • Step 2 Synthesis of compound 5
  • the appropriate Grignard reagent (2 eq.) was added dropwise at 0 o C under N2 atmosphere to a mixture of compound 3 (1 eq.) and BF 3 . Et 2 O (2eq.) in anhydrous THF.
  • the resulting mixture was stirred at 0 o C for 30 minutes and then warmed to room temperature over 2 hours.
  • the mixture was then poured into iced aq. NH4Cl and extracted twice with EtOAc.
  • the combined organic layers were washed with brine, dried over Na 2 SO 4 , filtered and concentrated under vacuum.
  • the residue was purified by column chromatography on silica gel (eluted with petroleum ether/EtOAc) to give compound 5.
  • Step 3 Synthesis of compound 6 Pd/C (15%, w/w) was added to a solution of compound 5 (1 eq.) in MeOH, and the resulting mixture was stirred under 25 psi H2 atmosphere at room temperature for 16 hours. The mixture was then filtered, and the filtrate was concentrated under vacuum to give crude compound 6, which was used for next step without any further purification.
  • Step 4 Synthesis of compound 8 EDCI (2 eq.), HOBt (2 eq.) and NMM (3 eq.) were added to a mixture of compound 6 (1 eq.) and compound 7 (1.1 eq.) in DMF, and the resulting mixture was stirred at room temperature for 16 hours. The mixture was then diluted with saturated aq.
  • Step 2 Synthesis of N,3-dibenzyloxetan-3-amine (M4-int-3) Benzyl magnesium bromide (3.7 mL, 1 M in THF) was added dropwise at 0o C under N2 atmosphere to a solution of M4-int-2 (300 mg, 1.86 mmol) and BF3 . Et2O (529 mg, 3.72 mmol) in THF (20 mL). The resulting mixture was then stirred at 0 o C for 30 minutes and warmed to room temperature over 2 hours. The mixture was then poured into iced aq. NH4Cl (30 mL) and extracted twice with EtOAc (30 mL).
  • Step 3 Synthesis of 3-benzyloxetan-3-amine (M4-int-4) Pd/C (30 mg) was added to a solution of N,3-dibenzyloxetan-3-amine (200 mg, 0.47 mmol) in MeOH (10 mL), and the resulting mixture was stirred under 25 psi H 2 atmosphere at room temperature for 16 hours. The mixture was then filtered and the filtrate was concentrated under vacuum to give crude M4-int-4 (50 mg, 65.3 % yield) as a yellow oil, which was used for next step without any further purification. LC/MS (ESI) m/z: 164 (M+H) + .
  • Step 4 Synthesis of N-(3-benzyloxetan-3-yl)-4-((2S,6R)-2,6-dimethylmorpholino) picolinamide (M4)
  • EDCI 76 mg, 0.49 mmol
  • HOBt 66 mg, 0.49 mmol
  • NMM 74 mg, 0.75mmol
  • 3-benzyloxetan-3-amine 40 mg, 0.25 mmol
  • M4-int-5 69 mg, 0.29 mmol
  • the mixture was then diluted with saturated aq. NH4Cl (30 mL) and extracted twice with EtOAc (30 mL).
  • Step 2 Synthesis of 1-(5-fluoro-2-methoxybenzyl)cyclopropan-1-amine (3)
  • Ethylmagnesium bromide (16 mL, 3M in ether) was slowly added at -60 o C under N 2 atmosphere to a stirred mixture of 2-(5-fluoro-2-methoxyphenyl)acetonitrile (4 g, 24 mmol) and Ti(Oi-Pr)4 (7.8 g, 27 mmol) in anhydrous THF (40 mL).
  • BF 3 -Et 2 O (6.82 g, 48 mmol) was added, and the resulting mixture was stirred for additional 2 hours.
  • Step 3 Synthesis of compound 4 EDCI (1.2 eq), HOBt (1.2 eq) and NMM (3.0 eq) were added under N 2 atmosphere to a mixture of 1-(5-fluoro-2-methoxybenzyl)cyclopropan-1-amine (1.0 eq) and the appropriate acid (1.1 eq.) in DMF, and the reaction was stirred at room temperature overnight. The reaction was then diluted with saturated aq. NH4Cl and extracted twice with EtOAc. The combined organic layers were dried over anhydrous Na 2 SO 4 , filtered and concentrated under vacuum.
  • Step 3 Synthesis of M5 EDCI (92 mg, 0.48 mmol), HOBt (65 mg, 0.48 mmol) and NMM (122 mg, 1.2 mmol) were added to a mixture of M5-int-4 (78 mg, 0.44 mmol) and M5-int-3 (78 mg, 0.4 mmol) in DMF (8 mL), and the resulting mixture was stirred at room temperature for 16 hours. The mixture was then diluted with saturated aq. NH4Cl (30 mL) and extracted twice with EtOAc (30 mL). The combined organic layers were dried over Na 2 SO 4 , filtered and concentrated under vacuum.
  • Method A K 2 CO 3 (2.5 eq.) and Pd(PPh 3 ) 4 (0.08 eq.) were added to a mixture of compound 1 (1 eq.) and the appropriate boronic acid (1.3 eq.) in 1,4-dioxane/H2O (9:1, V/V), and the resulting mixture was stirred at 90 o C under N 2 atmosphere for 16 hours. The mixture was then cooled to room temperature and diluted with EtOAc. The mixture was filtered and the filtrate was washed with saturated aq. NaHCO3 and brine. The organic layer was then dried over anhydrous Na2SO4, filtered and concentrated under vacuum.
  • Step 2 Synthesis of compound 4 AlMe3 (1 eq., 2M in hexane) was slowly added at 0 oC under N2 atmosphere to a mixture of compound 2 (1 eq.) and compound 3 (1.5 eq.) in anhydrous toluene, and the resulting mixture was stirred at 80 o C under N2 atmosphere for 16 hours. The mixture was then cooled to room temperature, quenched with saturated aq. NaHCO3 and extracted twice with EtOAc. The combined organic layers were dried over anhydrous Na 2 SO 4 , filtered and concentrated under vacuum.
  • Step 2 Synthesis of M6 AlMe3 (1.9 mL, 2M in hexane) was slowly added at 0 o C under N2 atmosphere to a mixture of M6-int-2 (780 mg, 3.66 mmol) and M6-int-3 (1.07 g, 5.5 mmol) in anhydrous toluene (40 mL), and the resulting mixture was stirred at 80 o C under N 2 atmosphere for 16 hours. The mixture was then cooled to room temperature, quenched with saturated aq. NaHCO3 (40 mL) and extracted twice with EtOAc (30 mL). The combined organic layerswere dried over anhydrous Na 2 SO 4 , filtered and concentrated under vacuum.
  • Step 2 Synthesis of compound 4 Hydroxylamine hydrochloride (2 eq.) and TEA (3 eq.) were added at 0 o C to a mixture of compound 3 (1 eq.) in EtOH, and the resulting mixture was stirred at 75 o C for 18 hours. The mixture was then diluted with water and extracted with EtOAc. The organic layer was washed with brine, dried over Na2SO4, filtered and concentrated under vacuumto give crude compound 4 without any further purification.
  • Step 3 Synthesis of compound 6 CDI (1.5 eq.) was added to a mixture of compound 4 (1.0 eq.) and compound 5 (1.0 eq.) in pyridine, and the resulting mixture was stirred at 100 o C for 3 hours.
  • the reaction mixture was cooled to room temperature, diluted with ethyl acetate and washed with water.
  • the organic layer was dried over anhydrous Na2SO4, filtered and concentrated under vacuum.
  • the residue was purified via prep-HPLC (Gemini 5 ⁇ m C18250*21.2mm, H 2 O/MeCN (5-95 %)/0.1% NH3 . H2O) to give compound 6.
  • Step 2 Synthesis of N-hydroxy-l-[(2-methoxyphenyl)methyl]cyclopropane-l- carboximidamide (M7-int-02)
  • Hydroxylamine hydrochloride (1.78g, 25.64 mmol) and TEA (5.345 mL, 38.45 mmol) were added at 0°C to a mixture of 1 -[(2-methoxyphenyl)methyl]cyclopropane- 1 -carbonitrile (2.4 g, 12.82 mmol) in EtOH (60 mL), and the resulting mixture was stirred at 75°C for 18 hours. The mixture was then diluted with water (50 mL) and extracted with EtOAc (50 mL).
  • CDI (111 mg, 0.681 mmol) was added to a mixture of M7-int-02 (100 mg, 0.454 mmol) and M7-int-03 (94.53 mg, 0.454 mmol) in pyridine (4 mL), and the resulting mixture was stirred at 100°C for 3 hours. After cooling, the mixture was diluted with EtOAc (20 mL) and washed with water (20 mL). The organic layer was then washed with brine, dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified via prep-HPLC (Gemini 5 ⁇ m C18250*21.2mm, H 2 O/MeCN (5-95 %)/0.1% NH 3 .
  • Step 2 Synthesis of compound 4 NaI (0.2 eq.) and NaHCO 3 (2 eq.) were added to a mixture of compound 2 and compound 3 (1 eq.) in a mixture of THF/H2O (15:1, V/V), and the resulting mixture was stirred at 80 oC for 2 hours. The reaction was then cooled down to room temperature, diluted with EtOAc, and washed with H 2 O and brine. The organic layer was dried over anhydrous Na 2 SO 4 , filtered and concentrated under vacuum. The residue was purified via flash column chromatography (eluted with DCM/MeOH) to give compound 4.
  • Step 3 Synthesis of compound 6
  • Compound 5 2.0 eq.
  • DIEA (3.0 eq.) were added to a mixture of compound 4 (1.0 eq.) in NMP, and the resulting mixture was stirred in a sealed tube at 160 oC for 7 hours.
  • the reaction mixture was then cooled down to room temperature, diluted with water and extracted with EtOAc.
  • the organic layer was dried over anhydrous Na2SO4, filtered and concentrated under vacuum.
  • the residue was purified via prep-HPLC (Gemini 5 ⁇ m C18250*21.2mm, H 2 O/MeCN (5-95 %)/0.1% NH 3 . H 2 O) to give compound 6.
  • Step 2 Synthesis of 2-[4-(1-benzylcyclopropyl)-1H-imidazol-2-yl]-4-chloropyridine (M8- int-3) NaI (43 mg, 0.383 mmol) and NaHCO3 (322 mg, 3.833 mmol) were added to a mixture of 1- (1-benzylcyclopropyl)-2-chloroethan-1-one (400 mg, 1.917 mmol) and 4-chloropyridine-2- carboximidamide (298 mg, 1.917 mmol) in a mixture of THF (15 mL) and H 2 O (1 mL). The resulting mixture was stirred at 80 o C for 2 hours.
  • Step 2 Synthesis of M8 Cis-2,6-dimethylmorpholine (75 mg, 0.648 mmol) and DIEA (127 mg, 0.972 mmol) were added to a mixture of M8-int-3 (100 mg, 0.324mmol) in NMP (5 mL), and the resulting mixture was stirred in a sealed tube at 160 o C for 7 hours. The reaction mixture was cooled to room temperature, diluted with water (30 mL) and extracted twice with EtOAc (30 mL). The combined organic layers were dried over anhydrous Na 2 SO 4 , filtered and concentrated under vacuum.
  • Step 1 Synthesis of compound 3
  • the appropriate amine 2 (2 eq.) was added to a mixture of compound 1 (1 eq.) and DIEA (3 eq.) in n-BuOH, and the resulting mixture was stirred in a sealed tube at 120 oC for 16 hours. After cooling, the mixture was concentrated under vacuum, and the residue was purified by column chromatography on silica gel (eluted with DCM/MeOH) to give compound 3.
  • Step 2 Synthesis of compound 5 EDCI (1.2 eq.), HOBt (1.2 eq.) and NMM (3 eq.) were added to a mixture of the appropriate compound 4 (1 eq.) and compound 3 (1.1 eq.) in DMF, and the resulting mixture was stirred at room temperature for 16 hours. The mixture was then diluted with saturated aq. NH4Cl and extracted twice with EtOAc. The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified via prep-HPLC (Gemini 5 ⁇ m C18250*21.2mm, H 2 O/MeCN (5-95 %)/0.1% NH 3 . H 2 O) to give compound 5.
  • EDCI 1.2 eq.
  • HOBt 1.2 eq.
  • NMM 3 eq.
  • Step 2 Synthesis of M9 EDCI (230 mg, 1.2 mmol), HOBt (162 mg, 1.2 mmol) and NMM (303 mg, 3 mmol) were added to a mixture of M9-int-3 (133 mg, 1 mmol) and M9-int-2 (260 mg, 1.1 mmol) in DMF (3 mL), and the resulting mixture was stirred at room temperature for 16 hours. The mixture was then diluted with saturated aq. NH 4 Cl (20 mL) and extracted twice with EtOAc (20 mL). The combined organic layers were dried over anhydrous Na 2 SO 4 , filtered and concentrated under vacuum.
  • Step 1 Synthesis of compound 3 LDA (1.2 eq.) was added at -78 o C under N2 atmosphere to a solution of compound 2 (1 eq.) in anhydrous THF, and the mixture was stirred at -78 oC for 1 hour. A solution of compound 1 (1 eq.) in THF was then added dropwise over 30 minutes and the resulting mixture was allowed to warm to room temperature over 3 hours under N2 atmosphere. The mixture was then quenched with saturated aq. NH4Cl and extracted twice with EtOAc.
  • Step 3 Synthesis of compound 5 DPPA (1.5 eq.) was added to a mixture of compound 4 (1 eq.) and Et3N (3 eq.) in t-BuOH, and the resulting mixture was stirred at 90 oC under N 2 atmosphere for 16 hours. After cooling, the mixture was diluted with DCM and washed with water. The organic layer was dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified via flash column chromatography on silica gel (PE/EtOAc) to give compound 5, which was used for next step without any further purification.
  • PE/EtOAc silica gel
  • Step 4 Synthesis of compound 6
  • Compound 5 (1 eq.) was added into a solution of TFA in DCM (1:3, V/V) at 0 o C and the resulting mixture was stirred at room temperature under N2 atmosphere for 3 hours. The mixture was then concentrated under vacuum to give crude compound 6, which was used for next step without any further purification.
  • Step 5 Synthesis of compound 8 EDCI (1.2 eq.), HOBt (1.2 eq.) and NMM (3 eq.) were added under N 2 atmosphere to a mixture of compound 6 (1 eq.) and compound 7 (1.1 eq.) in DMF, and the resulting mixture was stirred at room temperature for 8 hours. The mixture was then diluted with saturated aq.
  • Step 3 Synthesis of M10-int-4 DPPA (1.15 g, 4.17 mmol) was added to a mixture of M10-int-3 (620 mg, 2.61 mmol) and Et 3 N (790 mg, 7.82 mmol) in t-BuOH (40 mL), and the resulting mixture was stirred under N 2 atmosphere at 90 oC for 16 hours.
  • the reaction mixture was cooled down, diluted with DCM (60 mL), and washed with water (60 mL). The organic layer was then dried over anhydrous Na 2 SO 4, filtered and concentrated under vacuum.
  • Step 4 Synthesis of M10-int-5 M10-int-4 (210 mg, 0.68 mmol) was added at 0 oC into a solution of TFA (2 mL) in DCM (6 mL), and the resulting mixture was stirred under N 2 atmosphere at room temperature for 3 hours. The mixture was then concentrated under vacuum to give M10-int-5 (110 mg, 77.4 % yield) as a yellow oil without any further purification.
  • Step 5 Synthesis of M10 EDCI (122 mg, 0.64 mmol), HOBt (87 mg, 0.64 mmol) and NMM (160 mg, 1.6 mmol) were added under N2 atmosphere to a mixture of M10-int-5 (110 mg, 0.53 mmol) and M10-int-6 (139 mg, 0.59 mmol) in DMF (6 mL), and the resulting mixture was stirred at room temperature for 8 hours. The mixture was then diluted with saturated aq. NH4Cl (30 mL) and extracted twice with EtOAc (30 mL).
  • Step 2 Synthesis of compound 4 Ethylmagnesium bromide (2.2 eq.) was slowly added at -60 o C under N2 atmosphere to a stirred mixture of compound 3 (1 eq.) and Ti(O-iPr)4 (1.4 eq.) in anhydrous THF. The reaction mixture was then warmed to 0 oC over 3 hours and BF 3 -Et 2 O (1.2 eq.) was added. The resulting mixture was stirred for 2 hours and then poured into 1N aq. NaOH and extracted twice with EtOAc (20 mL). The combined organic layers were washed with brine, dried over anhydrous Na 2 SO 4 , filtered and concentrated under vacuum.
  • Step 3 Synthesis of compound 5 Pd/C (15%, w/w) was added to a solution of compound 4 (1 eq.) in MeOH, and the resulting mixture was stirred under 25 psi H2 atmosphere at room temperature for 16 hours. The vacuum to give crude compound 5, which was used for next step without any further purification.
  • Step 4 Synthesis of compound 7 EDCI (1.2 eq.), HOBt (1.2 eq.) and NMM (3 eq.) were added to a mixture of compound 5 (1 eq.) and compound 6 (1.1 eq.) in DMF, and the resulting mixture was stirred at room temperature for 16 hours.
  • Step 1 Synthesis of compound 3 PPh 3 (1.3 eq.) and DIAD (2 eq.) were added at 0 oC under N 2 atmosphere to a mixture of compound 1 (1 eq.) and compound 2 (1.1 eq.) in anhydrous THF, and the resulting mixture was stirred at room temperature for 16 hours.
  • Step 2 Synthesis of compound 4 Compound 3 (1 eq.) was added into a mixture of TFA/DCM (1:4, V/V) at 0 oC under N2 atmosphere, and the resulting mixture was stirred at room temperature for 2 hours. The mixture was then concentrated under vacuum and the residue was diluted with DCM and washed with saturated aq. NaHCO3.
  • Step 3 Synthesis of compound 6 EDCI (1.2 eq.), HOBt (1.2 eq.) and NMM (3 eq.) were added to a mixture of compound 4 (1 eq.) and compound 5 (1.1 eq.) in DMF, and the resulting mixture was stirred at room temperature for 16 hours. The mixture was then diluted with saturated aq. NaHCO3 and extracted twice with EtOAc. The combined organic layers were dried over Na2SO4, filtered and concentrated under vacuum.
  • Step 1 Synthesis of compound 2 A solution of compound 1 (1 eq.) in DCM was added dropwise at 0 oC under N 2 atmosphere into a solution of HCl/1,4-dioxane (4 M), and the resulting mixture was stirred at room temperature for 5 hours. The mixture was then concentrated under vacuum to give crude compound 2, which was used for next step without any further purification.
  • Step 2 Synthesis of compound 4 EDCI (1.2 eq.), HOBt (1.2 eq.) and NMM (3 eq.) were added to a mixture of compound 2 (1 eq.) and compound 3 (1.1 eq.) in DMF, and the resulting mixture was stirred at room temperature for 16 hours. The mixture was then diluted with saturated aq. NH4Cl and extracted twice with EtOAc. The combined organic layers were dried over anhydrous Na 2 SO 4 , filtered and concentrated under vacuum. The residue was purified by flash column chromatography (eluted with PE/EtOAc) to give compound 4.
  • Step 3 Synthesis of compound 5 Anhydrous DMSO (1.5 eq.) in dry DCM was added dropwise at -65°C under N 2 atmosphere to a solution of oxalyl chloride (1.5 eq.) in dry DCM. After stirring for 30 minutes, a solution of compound 4 (1 eq.) in dry DCM was added dropwise, and the reaction mixture was stirred for 2 hours before Et3N (3 eq.) was added dropwise. The mixture was then slowly warmed to room temperature until completion. Water was added and the mixture was extracted twice with DCM. The combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered and concentrated under vacuum.
  • Step 4 Synthesis of compound 7 4A MS and compound 6 (1 eq.) were added to a mixture of compound 5 (1 eq.) in DCE, and the mixture was stirred at room temperature for 2 hours before NaBH(OAc) 3 (2 eq.) was added. The resulting mixture was stirred at room temperature for 16 hours. The mixture was then diluted with DCM and washed with saturated aq. NaHCO3. The organic layer was dried over anhydrous Na 2 SO 4 , filtered and concentrated under vacuum.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Disclosed are compounds, compositions, and methods useful for treating or preventing a disease or disorder associated with mutation in a protein.

Description

SMALL MOLECULE CORRECTORS OF MAMMALIAN SLC6A8 FUNCTION
RELATED APPLICATION
This application claims benefit of priority to U S. Provisional Patent Application No. 63/054,000, filed July 20, 2020.
BACKGROUND
Creatine transporter deficiency (CTD) has been reported to be the most common cerebral creatine deficiency syndrome (CCDS). Creatine transporter deficiency is an X-linked disorder caused by mutations in the SLC6A8 gene. The SLC6A8 gene, located on the short arm of the sex chromosome, provides instructions for making a protein that transports the compound creatine into cells. Creatine is needed for the body to store and use energy properly. People with CTD have intellectual disability, which can range from mild to severe, and delayed speech development. Some affected individuals develop behavioral disorders such as attention deficit hyperactivity disorder or autistic behaviors that affect communication and social interaction. They may also experience seizures. Children with CTD may experience slow growth and exhibit delayed development of motor skills such as sitting and walking. CTD is difficult to treat because the actual transporter responsible for transporting creatine to the brain and muscles is defective. There is no current standard of care.
SUMMARY
One aspect of the invention provides compounds, compositions, and methods useful for treating or preventing a disease or disorder associated with a SLC6A8 mutation.
Accordingly, provided herein is a compound having the structure of Formula (I):
Figure imgf000002_0001
wherein:
A and A' are each independently selected from C-R' and N;
A" is selected from C-R" and N; provided that at least one of A, A', and A" is N; or A and A' are each independently selected from C-R and N, and A" is C-R", wherein R" taken together with R and the ring atoms to which they are bonded form a fused five-membered heteroaryl ring, such that formula I is a substituted indole or azaindole; X is selected from
Figure imgf000003_0002
Y is selected from
Figure imgf000003_0001
n is 0, 1, 2, or 3; m is 1; R1, R2, R3, R4, R5 and R6 are independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, aryl, heteroaryl, -C(O)OR11, -C(O)NR12R13, and -C(O)R14; R7, R8, R9, and R10 are independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, aryl, heteroaryl, -C(O)OR11, -C(O)NR12R13, and -C(O)R14; Z is selected from -H, halogen, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O- cycloheteroalkyl, -O-halocycloalkyl, -NH-alkyl, -NH-heteroalkyl, -NH-haloalkyl, -NH- cycloalkyl, -NH-cycloheteroalkyl, -NH-halocycloalkyl, aryl, -O-aryl, -NH-aryl, heteroaryl, - O-heteroaryl, -NH-heteroaryl, -C(O)OR11, -C(O)NR12R13, -NR12R13, and -C(O)R14; R is selected from alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O-cycloheteroalkyl, - O-halocycloalkyl, aryl, -O-aryl, heteroaryl, -O-heteroaryl -OSO2R11, and -NR15R16; R' is selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O- cycloalkyl, -O-cycloheteroalkyl, -O-halocycloalkyl, -O-aryl, -O-heteroaryl, -OSO2R11, and – NR12R13; R'' is selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O- cycloalkyl, -O-cycloheteroalkyl, -O-halocycloalkyl, -O-aryl, -O-heteroaryl, -OSO2R11, and – NR12R13; each occurrence of R11, R12 and R13 is independently selected from H, alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl; each occurrence of Rids independently selected from alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl;
Ri5 and Rie are independently selected from -H, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -C(0)Rn, -C(0)0Rn, and - C(0)NRi2Ri3; or Ris and Rie taken together with the nitrogen atom to which they are bonded form an un substituted or substituted C4-Cs cycloheteroalkyl; and provided that if A is N, A' and A" are each
Figure imgf000004_0004
is selected from phenyl and 2-methoxy-5-fluorophenyl, then R is not
Figure imgf000004_0003
Figure imgf000004_0005
Another aspect of the invention relates to methods of treating or preventing a disease or disorder associated with a SLC6A8 mutation, treating an inflammatory disease, increasing transport of guanidinoacetic acid or a salt thereof across the blood-brain barrier, improving the function of a cellular creatine transporter, or decreasing the accumulation or the concentration of guanidinoacetic acid or a salt thereof in a cell, in a subject in need thereof comprising administering to the subject an effective amount of a compound of Formula (I):
Figure imgf000004_0002
wherein:
A and A' are each independently selected from C-R and N;
A" is selected from C-R" and N; provided that at least one of A, A', and A" is N; or A and A' are each independently selected from C-R and N, and A" is C-R", wherein R" taken together with R and the ring atoms to which they are bonded form a fused five-membered heteroaryl ring, such that formula I is a substituted indole or azaindole;
X is selected from
Figure imgf000004_0001
Y is selected from
Figure imgf000005_0001
« is 0, 1, 2, or 3; m is 1;
Ri, R2, R3, R4, RS and Re are independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, aryl, heteroaryl, -C(0)0Rn, -C(0)NRi2Ri3, and -C(0)Ri4;
Rv, Rs, R9, and Rio are independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, aryl, heteroaryl, -C(0)0Rn, -C(0)NRi2Ri3, and -C(0)Ri4;
Z is selected from -H, halogen, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O- cycloheteroalkyl, -O-halocycloalkyl, -NH-alkyl, -NH-heteroalkyl, -NH-haloalkyl, -NH- cycloalkyl, -NH-cycloheteroalkyl, -NH-halocycloalkyl, aryl, -O-aryl, -NH-aryl, heteroaryl, - O-heteroaryl, -NH-heteroaryl, -C(0)0Rn, -C(0)NRi2Ri3, -NR12R13, and -C(0)Ri4;
R is selected from alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O-cycloheteroalkyl, - O-halocycloalkyl, aryl, -O-aryl, heteroaryl, -O-heteroaryl -OSO2R11, and -NRisRie;
R is selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O- cycloalkyl, -O-cycloheteroalkyl, -O-halocycloalkyl, -O-aryl, -O-heteroaryl, -OSO2R11, and
-NR12R13;
R" is selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O- cycloalkyl, -O-cycloheteroalkyl, -O-halocycloalkyl, -O-aryl, -O-heteroaryl, -OSO2R11, and
-NR12R13; each occurrence of R11, R12 and R13 is independently selected from H, alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl; each occurrence of Ri4is independently selected from alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl;
Ri5 and Rie are independently selected from -H, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -C(0)Rn, -C(0)0Rn, and - C(0)NRI2RI3; or R15 and Rie taken together with the nitrogen atom to which they are bonded form an un substituted or substituted C4-C8 cycloheteroalkyl. In certain embodiments, the subject is a mammal. In certain embodiments, the mammal is a human.
The invention provides several additional advantages. The prophylactic and therapeutic methods described herein are also effective for treating creatine transporter deficiency and associated symptoms. In some embodiments, the therapeutic method is effective in treating motor dysfunction, intellectual disability, language delay, speech delay, seizures, behaviors associated with autism and attention deficit hyperactivity disorder, fatigue, muscular hypotonia, low weight gain, and gastrointestinal and cardiac disorders.
In some embodiments, the therapeutic method is effective in treating inflammatory diseases. In some embodiments, the inflammatory disease is acute. In some embodiments, the inflammatory disease is chronic. In some embodiments, the inflammatory disease is selected from inflammatory bowel diseases (for example, ulcerative colitis or Crohn's disease), multiple sclerosis, psoriasis, arthritis, rheumatoid arthritis, osteoarthritis, juvenile arthritis, psoriatic arthritis, reactive arthritis, ankylosing spondylitis, cryopyrin associated periodic syndromes, Muckle-Wells syndrome, familial cold auto-inflammatory syndrome, neonatal- onset multisystem inflammatory disease, TNF receptor associated periodic syndrome, acute and chronic pancreatitis, atherosclerosis, gout, ankylosing spondylitis, fibrotic disorders (for example, hepatic fibrosis or idiopathic pulmonary fibrosis), nephropathy, sarcoidosis, scleroderma, anaphylaxis, diabetes (for example, diabetes mellitus type 1 or diabetes mellitus type 2), diabetic retinopathy, Still's disease, vasculitis, sarcoidosis, pulmonary inflammation, acute respiratory distress syndrome, wet and dry age-related macular degeneration, autoimmune hemolytic syndromes, autoimmune and inflammatory hepatitis, autoimmune neuropathy, autoimmune ovarian failure, autoimmune orchitis, autoimmune thrombocytopenia, silicone implant associated autoimmune disease, Sjogren's syndrome, familial Mediterranean fever, systemic lupus erythematosus, vasculitis syndromes (for example, temporal, Takayasu's and giant cell arteritis, Beliefs disease or Wegener's granulomatosis), vitiligo, secondary hematologic manifestation of autoimmune diseases (for example, anemias), drug-induced autoimmunity, Hashimoto's thyroiditis, hypophysitis, idiopathic thrombocytic pupura, metal-induced autoimmunity, myasthenia gravis, pemphigus, autoimmune deafness (for example, Meniere's disease), Goodpasture's syndrome, Graves' disease, HW -related autoimmune syndromes, Gullain-Barre disease, Addison's disease, antiphospholipid syndrome, asthma, atopic dermatitis, Celiac disease, Cushing's syndrome, dermatomyositis, idiopathic adrenal adrenal atrophy, idiopathic thrombocytopenia, Kawasaki syndrome, Lambert-Eaton Syndrome, pernicious anemia, pollinosis, polyarteritis nodosa, primary biliary cirrhosis, primary sclerosing cholangitis, Raynaud's, Reiter's Syndrome, relapsing polychondritis, Schmidt's syndrome, thyrotoxidosis, sepsis, septic shock, endotoxic shock, exotoxin-induced toxic shock, gram negative sepsis, toxic shock syndrome, glomerulonephritis, peritonitis, interstitial cystitis, hyperoxia-induced inflammations, chronic obstructive pulmonary disease (COPD), vasculitis, graft vs. host reaction (for example, graft vs. host disease), allograft rejections (for example, acute allograft rejection or chronic allograft rejection), early transplantation rejection (for example, acute allograft rejection), reperfusion injury, pain (for example, acute pain, chronic pain, neuropathic pain, or fibromyalgia), chronic infections, meningitis, encephalitis, myocarditis, gingivitis, post surgical trauma, tissue injury, traumatic brain injury, enterocolitis, sinusitis, uveitis, ocular inflammation, optic neuritis, gastric ulcers, esophagitis, peritonitis, periodontitis, dermatomyositis, gastritis, myositis, polymyalgia, pneumonia and bronchitis.
ETnless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the present invention, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.
Other features, objects, and advantages of the invention will be apparent from the detailed description, and from the claims.
BRIEF DESCRIPTION OF THE DRAWINGS FIG. 1: is a table summarizing trafficking and correction data for exemplary compounds of the invention. Trafficking Emax: A: 1 < Emax < 2; B: 2 < Emax < 5; C: 5 < E max < 15. Trafficking ECso: A: 0.1 μΜ < ECso < 20 μΜ; B: 20 μΜ < ECso < 35 pM; C: 35 pM < ECso< 50 pM; D: 50 pM < ECso< 100 pM. Correction E, : A: E max 1; B: 1 < Emax < 2; C:
2 < Emax < 5; D: 5 < E max < 15. Correction ECso: A: 0.1 pM < ECso < 20 pM; B: 20 pM < ECso < 35 pM; C: 35 pM < ECso< 50 pM; D: 50 pM < ECso< 100 pM.
DETAILED DESCRIPTION
Definitions
For convenience, before further description of the present invention, certain terms employed in the specification, examples and appended claims are collected here. These definitions should be read in light of the remainder of the disclosure and understood as by a person of skill in the art. Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by a person of ordinary skill in the art.
In order for the present invention to be more readily understood, certain terms and phrases are defined below and throughout the specification.
The articles “a” and “an” are used herein to refer to one or to more than one (i.e., to at least one) of the grammatical object of the article. By way of example, “an element” means one element or more than one element.
The phrase “and/or,” as used herein in the specification and in the claims, should be understood to mean “either or both” of the elements so conjoined, i.e., elements that are conjunctively present in some cases and disjunctively present in other cases. Multiple elements listed with “and/or” should be construed in the same fashion, i.e., “one or more” of the elements so conjoined. Other elements may optionally be present other than the elements specifically identified by the “and/or” clause, whether related or unrelated to those elements specifically identified. Thus, as a non-limiting example, a reference to “A and/or B”, when used in conjunction with open-ended language such as “comprising” can refer, in one embodiment, to A only (optionally including elements other than B); in another embodiment, to B only (optionally including elements other than A); in yet another embodiment, to both A and B (optionally including other elements); etc.
As used herein in the specification and in the claims, “or” should be understood to have the same meaning as “and/or” as defined above. For example, when separating items in a list, “or” or “and/or” shall be interpreted as being inclusive, i.e., the inclusion of at least one, but also including more than one, of a number or list of elements, and, optionally, additional unlisted items. Only terms clearly indicated to the contrary, such as “only one of’ or “exactly one of,” or, when used in the claims, “consisting of,” will refer to the inclusion of exactly one element of a number or list of elements. In general, the term “or” as used herein shall only be interpreted as indicating exclusive alternatives (i.e., “one or the other but not both”) when preceded by terms of exclusivity, such as “either, ')'> a one of,” “only one of,” or “exactly one of.” “Consisting essentially of,” when used in the claims, shall have its ordinary meaning as used in the field of patent law.
As used herein in the specification and in the claims, the phrase “at least one,” in reference to a list of one or more elements, should be understood to mean at least one element selected from any one or more of the elements in the list of elements, but not necessarily including at least one of each and every element specifically listed within the list of elements and not excluding any combinations of elements in the list of elements. This definition also allows that elements may optionally be present other than the elements specifically identified within the list of elements to which the phrase “at least one” refers, whether related or unrelated to those elements specifically identified. Thus, as a non-limiting example, “at least one of A and B” (or, equivalently, “at least one of A or B,” or, equivalently “at least one of A and/or B”) can refer, in one embodiment, to at least one, optionally including more than one, A, with no B present (and optionally including elements other than B); in another embodiment, to at least one, optionally including more than one, B, with no A present (and optionally including elements other than A); in yet another embodiment, to at least one, optionally including more than one, A, and at least one, optionally including more than one,
B (and optionally including other elements); etc.
It should also be understood that, unless clearly indicated to the contrary, in any methods claimed herein that include more than one step or act, the order of the steps or acts of the method is not necessarily limited to the order in which the steps or acts of the method are recited.
In the claims, as well as in the specification above, all transitional phrases such as “comprising,” “including,” “carrying,” “having,” “containing,” “involving,” “holding,” “composed of,” and the like are to be understood to be open-ended, i.e., to mean including but not limited to. Only the transitional phrases “consisting of’ and “consisting essentially of’ shall be closed or semi-closed transitional phrases, respectively, as set forth in the United States Patent Office Manual of Patent Examining Procedures, Section 2111.03.
Certain compounds contained in compositions of the present invention may exist in particular geometric or stereoisomeric forms. In addition, polymers of the present invention may also be optically active. The present invention contemplates all such compounds, including cis- and trans-isomers, R- and //-enantiomers, diastereomers, (D)-isomers, (L)- i somers, the racemic mixtures thereof, and other mixtures thereof, as falling within the scope of the invention. Additional asymmetric carbon atoms may be present in a substituent such as an alkyl group. All such isomers, as well as mixtures thereof, are intended to be included in this invention.
“Geometric isomer" means isomers that differ in the orientation of substituent atoms in relationship to a carbon-carbon double bond, to a cycloalkyl ring, or to a bridged bicyclic system. Atoms (other than H) on each side of a carbon- carbon double bond may be in an E (substituents are on opposite sides of the carbon- carbon double bond) or Z (substituents are oriented on the same side) configuration. "R," "S," "S*," "R*," "E," "Z," "cis," and "trans," indicate configurations relative to the core molecule. Certain of the disclosed compounds may exist in “atropisomeric” forms or as “atropisomers.” Atropi somers are stereoisomers resulting from hindered rotation about single bonds where the steric strain barrier to rotation is high enough to allow for the isolation of the conformers. The compounds of the invention may be prepared as individual isomers by either isomer-specific synthesis or resolved from a mixture of isomers. Conventional resolution techniques include forming the salt of a free base of each isomer of an isomeric pair using an optically active acid (followed by fractional crystallization and regeneration of the free base), forming the salt of the acid form of each isomer of an isomeric pair using an optically active amine (followed by fractional crystallization and regeneration of the free acid), forming an ester or amide of each of the isomers of an isomeric pair using an optically pure acid, amine or alcohol (followed by chromatographic separation and removal of the chiral auxiliary), or resolving an isomeric mixture of either a starting material or a final product using various well known chromatographic methods.
If, for instance, a particular enantiomer of compound of the present invention is desired, it may be prepared by asymmetric synthesis, or by derivation with a chiral auxiliary, where the resulting diastereomeric mixture is separated and the auxiliary group cleaved to provide the pure desired enantiomers. Alternatively, where the molecule contains a basic functional group, such as amino, or an acidic functional group, such as carboxyl, diastereomeric salts are formed with an appropriate optically-active acid or base, followed by resolution of the diastereomers thus formed by fractional crystallization or chromatographic means well known in the art, and subsequent recovery of the pure enantiomers.
Percent purity by mole fraction is the ratio of the moles of the enantiomer (or diastereomer) or over the moles of the enantiomer (or diastereomer) plus the moles of its optical isomer. When the stereochemistry of a disclosed compound is named or depicted by structure, the named or depicted stereoisomer is at least about 60%, about 70%, about 80%, about 90%, about 99% or about 99.9% by mole fraction pure relative to the other stereoisomers. When a single enantiomer is named or depicted by structure, the depicted or named enantiomer is at least about 60%, about 70%, about 80%, about 90%, about 99% or about 99.9% by mole fraction pure. When a single diastereomer is named or depicted by structure, the depicted or named diastereomer is at least about 60%, about 70%, about 80%, about 90%, about 99% or about 99.9% by mole fraction pure.
When a disclosed compound is named or depicted by structure without indicating the stereochemistry, and the compound has at least one chiral center, it is to be understood that the name or structure encompasses either enantiomer of the compound free from the corresponding optical isomer, a racemic mixture of the compound or mixtures enriched in one enantiomer relative to its corresponding optical isomer. When a disclosed compound is named or depicted by structure without indicating the stereochemistry and has two or more chiral centers, it is to be understood that the name or structure encompasses a diastereomer free of other diastereomers, a number of diastereomers free from other diastereomeric pairs, mixtures of diastereomers, mixtures of diastereomeric pairs, mixtures of diastereomers in which one diastereomer is enriched relative to the other diastereomer(s) or mixtures of diastereomers in which one or more diastereomer is enriched relative to the other diastereomers. The invention embraces all of these forms. Structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, compounds produced by the replacement of a hydrogen with deuterium or tritium, or of a carbon with a 13C- or 14C- enriched carbon are within the scope of this invention. The term “prodrug” as used herein encompasses compounds that, under physiological conditions, are converted into therapeutically active agents. A common method for making a prodrug is to include selected moieties that are hydrolyzed under physiological conditions to reveal the desired molecule. In other embodiments, the prodrug is converted by an enzymatic activity of the host animal. The phrase “pharmaceutically acceptable excipient” or “pharmaceutically acceptable carrier” as used herein means a pharmaceutically acceptable material, composition or vehicle, such as a liquid or solid filler, diluent, excipient, solvent or encapsulating material, involved in carrying or transporting the subject chemical 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, not injurious to the patient, and substantially non-pyrogenic. Some examples of materials which can serve as pharmaceutically acceptable carriers include: (1) sugars, such as lactose, glucose, and sucrose; (2) starches, such as corn starch and potato starch; (3) cellulose, and its derivatives, such as sodium carboxymethyl cellulose, ethyl cellulose, and cellulose acetate; (4) powdered tragacanth; (5) malt; (6) gelatin; (7) talc; (8) excipients, such as cocoa butter and suppository waxes; (9) oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil, and soybean oil; (10) glycols, such as propylene glycol; (11) polyols, such as glycerin, sorbitol, mannitol, and polyethylene glycol; (12) esters, such as ethyl oleate and ethyl laurate; (13) agar; (14) buffering agents, such as magnesium hydroxide and aluminum hydroxide; ethyl alcohol; (20) phosphate buffer solutions; and (21) other non-toxic compatible substances employed in pharmaceutical formulations. In certain embodiments, pharmaceutical compositions of the present invention are non-pyrogenic, i.e., do not induce significant temperature elevations when administered to a patient. The term “pharmaceutically acceptable salts” refers to the relatively non-toxic, inorganic and organic acid addition salts of the compound(s). These salts can be prepared in situ during the final isolation and purification of the compound(s), or by separately reacting a purified compound(s) in its free base form with a suitable organic or inorganic acid, and isolating the salt thus formed. Representative salts include the hydrobromide, hydrochloride, sulfate, bisulfate, phosphate, nitrate, acetate, valerate, oleate, palmitate, stearate, laurate, benzoate, lactate, phosphate, tosylate, citrate, maleate, fumarate, succinate, tartrate, naphthylate, mesylate, glucoheptonate, lactobionate, and laurylsulphonate salts, and the like. (See, for example, Berge et al. (1977) “Pharmaceutical Salts”, J. Pharm. Sci.66:1-19.) In other cases, the compounds useful in the methods of the present invention may contain one or more acidic functional groups and, thus, are capable of forming pharmaceutically acceptable salts with pharmaceutically acceptable bases. The term “pharmaceutically acceptable salts” in these instances refers to the relatively non-toxic inorganic and organic base addition salts of a compound(s). These salts can likewise be prepared in situ during the final isolation and purification of the compound(s), or by separately reacting the purified compound(s) in its free acid form with a suitable base, such as the hydroxide, carbonate, or bicarbonate of a pharmaceutically acceptable metal cation, with ammonia, or with a pharmaceutically acceptable organic primary, secondary, or tertiary amine. Representative alkali or alkaline earth salts include the lithium, sodium, potassium, calcium, magnesium, and aluminum salts, and the like. Representative organic amines useful for the formation of base addition salts include ethylamine, diethylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine, and the like (see, for example, Berge et al., supra). The term “pharmaceutically acceptable cocrystals” refers to solid coformers that do not form formal ionic interactions with the small molecule. A “therapeutically effective amount” (or “effective amount”) of a compound with respect to use in treatment, refers to an amount of the compound in a preparation which, when administered as part of a desired dosage regimen (to a mammal, preferably a human) alleviates a symptom, ameliorates a condition, or slows the onset of disease conditions according to clinically acceptable standards for the disorder or condition to be treated or the The term “prophylactic or therapeutic” treatment is art-recognized and includes administration to the host of one or more of the subject compositions. If it is administered prior to clinical manifestation of the unwanted condition (e.g., disease or other unwanted state of the host animal) then the treatment is prophylactic, (i.e., it protects the host against developing the unwanted condition), whereas if it is administered after manifestation of the unwanted condition, the treatment is therapeutic, (i.e., it is intended to diminish, ameliorate, or stabilize the existing unwanted condition or side effects thereof).
The term “patient” or “subject” refers to a mammal in need of a particular treatment.
In certain embodiments, a patient is a primate, canine, feline, or equine. In certain embodiments, a patient is a human.
An aliphatic chain comprises the classes of alkyl, alkenyl and alkynyl defined below. A straight aliphatic chain is limited to unbranched carbon chain moieties. As used herein, the term “aliphatic group” refers to a straight chain, branched-chain, or cyclic aliphatic hydrocarbon group and includes saturated and unsaturated aliphatic groups, such as an alkyl group, an alkenyl group, or an alkynyl group.
“Alkyl” refers to a fully saturated cyclic or acyclic, branched or unbranched carbon chain moiety having the number of carbon atoms specified, or up to 30 carbon atoms if no specification is made. For example, alkyl of 1 to 8 carbon atoms refers to moieties such as methyl, ethyl, propyl, butyl, pentyl, hexyl, heptyl, and octyl, and those moieties which are positional isomers of these moieties. Alkyl of 10 to 30 carbon atoms includes decyl, undecyl, dodecyl, tri decyl, tetradecyl, pentadecyl, hexadecyl, heptadecyl, octadecyl, nonadecyl, eicosyl, heneicosyl, docosyl, tri cosy 1 and tetracosyl. In certain embodiments, a straight chain or branched chain alkyl has 30 or fewer carbon atoms in its backbone (e.g., C1-C30 for straight chains, C3-C30 for branched chains), and more preferably 20 or fewer. Alkyl goups may be substituted or unsubstituted.
As used herein, the term “heteroalkyl” refers to an alkyl moiety as hereinbefore defined which contain one or more oxygen, sulfur, nitrogen, phosphorus, or silicon atoms in place of carbon atoms.
As used herein, the term “haloalkyl” refers to an alkyl group as hereinbefore defined substituted with at least one halogen.
As used herein, the term “hydroxy alkyl” refers to an alkyl group as hereinbefore defined substituted with at least one hydroxyl.
As used herein, the term “alkylene” refers to an alkyl group having the specified number of carbons, for example from 2 to 12 carbon atoms, that contains two points of attachment to the rest of the compound on its longest carbon chain. Non-limiting examples of alkylene groups include methylene -(CH2)-, ethylene -(CH2CH2)-, n-propylene - (CH2CH2CH2)-, isopropylene -(CH2CH(CH3))-, and the like. Alkylene groups can be cyclic or acyclic, branched or unbranched carbon chain moiety, and may be optionally substituted with one or more substituents. "Cycloalkyl" means mono- or bicyclic or bridged or spirocyclic, or polycyclic saturated carbocyclic rings, each having from 3 to 12 carbon atoms. Preferred cycloalkyls have from 3-10 carbon atoms in their ring structure, and more preferably have 3-6 carbons in the ring structure. Cycloalkyl groups may be substituted or unsubstituted. As used herein, the term “halocycloalkyl” refers to an cycloalkyl group as hereinbefore defined substituted with at least one halogen. "Cycloheteroalkyl" refers to an cycloalkyl moiety as hereinbefore defined which contain one or more oxygen, sulfur, nitrogen, phosphorus, or silicon atoms in place of carbon atoms. Preferred cycloheteroalkyls have from 4-8 carbon atoms and heteroatoms in their ring structure, and more preferably have 4-6 carbons and heteroatoms in the ring structure. Cycloheteroalkyl groups may be substituted or unsubstituted. Unless the number of carbons is otherwise specified, “lower alkyl,” as used herein, means an alkyl group, as defined above, but having from one to ten carbons, more preferably from one to six carbon atoms in its backbone structure such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, and tert-butyl. Likewise, “lower alkenyl” and “lower alkynyl” have similar chain lengths. Throughout the application, preferred alkyl groups are lower alkyls. In certain embodiments, a substituent designated herein as alkyl is a lower alkyl. “Alkenyl” refers to any cyclic or acyclic, branched or unbranched unsaturated carbon chain moiety having the number of carbon atoms specified, or up to 26 carbon atoms if no limitation on the number of carbon atoms is specified; and having one or more double bonds in the moiety. Alkenyl of 6 to 26 carbon atoms is exemplified by hexenyl, heptenyl, octenyl, nonenyl, decenyl, undecenyl, dodenyl, tridecenyl, tetradecenyl, pentadecenyl, hexadecenyl, heptadecenyl, octadecenyl, nonadecenyl, eicosenyl, heneicosoenyl, docosenyl, tricosenyl, and tetracosenyl, in their various isomeric forms, where the unsaturated bond(s) can be located anywhere in the moiety and can have either the (Z) or the (E) configuration about the double bond(s). “Alkynyl” refers to hydrocarbyl moieties of the scope of alkenyl, but having one or more triple bonds in the moiety. The term “aryl” as used herein includes 3- to 12-membered substituted or unsubstituted single-ring aromatic groups in which each atom of the ring is carbon (i.e., carbocyclic aryl) or where one or more atoms are heteroatoms (i.e., heteroaryl). Preferably, aryl groups include 5- to 12-membered rings, more preferably 6- to 10-membered rings The term “aryl” also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings wherein at least one of the rings is aromatic, e.g., the other cyclic rings can be cycloalkyls, cycloalkenyls, cycloalkynyls, aryls, heteroaryls, and/or heterocyclyls. Carboycyclic aryl groups include benzene, naphthalene, phenanthrene, phenol, aniline, and the like. Heteroaryl groups include substituted or unsubstituted aromatic 3- to 12-membered ring structures, more preferably 5- to 12- membered rings, more preferably 5- to 10-membered rings, whose ring structures include one to four heteroatoms. Heteroaryl groups include, for example, pyrrole, furan, thiophene, imidazole, oxazole, thiazole, triazole, pyrazole, pyridine, pyrazine, pyridazine and pyrimidine, and the like. Aryl and heteroaryl can be monocyclic, bicyclic, or polycyclic. The term “halo”, “halide”, or “halogen” as used herein means halogen and includes, for example, and without being limited thereto, fluoro, chloro, bromo, iodo and the like, in both radioactive and non-radioactive forms. In a preferred embodiment, halo is selected from the group consisting of fluoro, chloro and bromo. The terms “heterocyclyl” or “heterocyclic group” refer to 3- to 12-membered ring structures, more preferably 5- to 12-membered rings, more preferably 5- to 10-membered rings, whose ring structures include one to four heteroatoms. Heterocycles can be monocyclic, bicyclic, spirocyclic, or polycyclic. Heterocyclyl groups include, for example, thiophene, thianthrene, furan, pyran, isobenzofuran, chromene, xanthene, phenoxathiin, pyrrole, imidazole, pyrazole, isothiazole, isoxazole, pyridine, pyrazine, pyrimidine, pyridazine, indolizine, isoindole, indole, indazole, purine, quinolizine, isoquinoline, quinoline, phthalazine, naphthyridine, quinoxaline, quinazoline, cinnoline, pteridine, carbazole, carboline, phenanthridine, acridine, pyrimidine, phenanthroline, phenazine, phenarsazine, phenothiazine, furazan, phenoxazine, pyrrolidine, oxolane, thiolane, oxazole, piperidine, piperazine, morpholine, lactones, lactams such as azetidinones and pyrrolidinones, sultams, sultones, and the like. The heterocyclic ring can be substituted at one or more positions with such substituents as described above, as for example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxyl, amino, nitro, sulfhydryl, imino, amido, phosphate, phosphonate, phosphinate, carbonyl, carboxyl, silyl, sulfamoyl, sulfinyl, ether, alkylthio, sulfonyl, ketone, aldehyde, ester, a heterocyclyl, an aromatic or heteroaromatic moiety, -CF3, -CN, and the like.
The term “substituted” refers to moieties having substituents replacing a hydrogen on one or more carbons of the backbone. It will be understood that “substitution” or “substituted with” includes the implicit proviso that such substitution is in accordance with permitted valence of the substituted atom and the substituent, and that the substitution results in a stable compound, e.g., which does not spontaneously undergo transformation such as by rearrangement, cyclization, elimination, etc. As used herein, the term “substituted” is contemplated to include all permissible substituents of organic compounds. In a broad aspect, the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and non-aromatic substituents of organic compounds. The permissible substituents can be one or more and the same or different for appropriate organic compounds. For purposes of this invention, the heteroatoms such as nitrogen may have hydrogen substituents and/or any permissible substituents of organic compounds described herein which satisfy the valences of the heteroatoms. Substituents can include any substituents described herein, for example, a halogen, a hydroxyl, a carbonyl (such as a carboxyl, an alkoxy carbonyl, a formyl, or an acyl), a thiocarbonyl (such as a thioester, a thioacetate, or a thioformate), an alkoxy, a phosphoryl, a phosphate, a phosphonate, a phosphinate, an amino, an ami do, an amidine, an imine, a cyano, a nitro, an azido, a sulfhydryl, an alkylthio, a sulfate, a sulfonate, a sulfamoyl, a sulfonamido, a sulfonyl, a heterocyclyl, an aralkyl, or an aromatic or heteroaromatic moiety. In preferred embodiments, the substituents on substituted alkyls are selected from Ci-6 alkyl, C3-6 cycloalkyl, halogen, carbonyl, cyano, or hydroxyl. In more preferred embodiments, the substituents on substituted alkyls are selected from fluoro, carbonyl, cyano, or hydroxyl. It will be understood by those skilled in the art that substituents can themselves be substituted, if appropriate. Unless specifically stated as “unsubstituted,” references to chemical moieties herein are understood to include substituted variants. For example, reference to an “aryl” group or moiety implicitly includes both substituted and unsubstituted variants.
As used herein, the definition of each expression, e.g., alkyl, m, n, etc., when it occurs more than once in any structure, is intended to be independent of its definition elsewhere in the same structure.
As used herein, “small molecules” refers to small organic or inorganic molecules of molecular weight below about 3,000 Daltons. In general, small molecules useful for the invention have a molecular weight of less than 3,000 Daltons (Da). The small molecules can be, e.g., from at least about 100 Da to about 3,000 Da (e.g., between about 100 to about 3,000 Da, about 100 to about 2500 Da, about 100 to about 2,000 Da, about 100 to about 1,750 Da, about 100 to about 1,500 Da, about 100 to about 1,250 Da, about 100 to about 1,000 Da, about 100 to about 750 Da, about 100 to about 500 Da, about 200 to about 1500, about 500 to about 1000, about 300 to about 1000 Da, or about 100 to about 250 Da).
In some embodiments, a “small molecule” refers to an organic, inorganic, or organometallic compound typically having a molecular weight of less than about 1000. In some embodiments, a small molecule is an organic compound, with a size on the order of 1 nm. In some embodiments, small molecule drugs of the invention encompass oligopeptides and other biomolecules having a molecular weight of less than about 1000.
An “effective amount” is an amount sufficient to effect beneficial or desired results. For example, a therapeutic amount is one that achieves the desired therapeutic effect. This amount can be the same or different from a prophylactically effective amount, which is an amount necessary to prevent onset of disease or disease symptoms. An effective amount can be administered in one or more administrations, applications or dosages. A therapeutically effective amount of a composition depends on the composition selected. The compositions can be administered from one or more times per day to one or more times per week; including once every other day. The skilled artisan will appreciate that certain factors may influence the dosage and timing required to effectively treat a subject, including but not limited to the severity of the disease or disorder, previous treatments, the general health and/or age of the subject, and other diseases present. Moreover, treatment of a subject with a therapeutically effective amount of the compositions described herein can include a single treatment or a series of treatments.
The terms “decrease,” “reduce,” “reduced”, “reduction”, “decrease,” and “inhibit” are all used herein generally to mean a decrease by a statistically significant amount relative to a reference. However, for avoidance of doubt, “reduce,” “reduction” or “decrease” or “inhibit” typically means a decrease by at least 10% as compared to a reference level and can include, for example, a decrease by at least about 20%, at least about 25%, at least about 30%, at least about 35%, at least about 40%, at least about 45%, at least about 50%, at least about 55%, at least about 60%, at least about 65%, at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 90%, at least about 95%, at least about 98%, at least about 99%, up to and including, for example, the complete absence of the given entity or parameter ascompared to the reference level, or any decrease between 10-99% as compared to the absence of a given treatment. The terms “increased”, “increase” or “enhance” or “activate” are all used herein to generally mean an increase by a statically significant amount; for the avoidance of any doubt, the terms “increased”, “increase” or “enhance” or “activate” means an increase of at least 10% as compared to a reference level, for example an increase of at least about 20%, or at least about 30%, or at least about 40%, or at least about 50%, or at least about 60%, or at least about 70%, or at least about 80%, or at least about 90% or up to and including a 100% increase or any increase between 10-100% as compared to a reference level, or at least about a 2-fold, or at least about a 3-fold, or at least about a 4-fold, or at least about a 5-fold or at least about a 10-fold increase, or any increase between 2-fold and 10-fold or greater as compared to a reference level. As used herein, the term “modulate” includes up-regulation and down-regulation, e.g., enhancing or inhibiting a response. A “radiopharmaceutical agent,” as defined herein, refers to a pharmaceutical agent which contains at least one radiation-emitting radioisotope. Radiopharmaceutical agents are routinely used in nuclear medicine for the diagnosis and/or therapy of various diseases. The radiolabelled pharmaceutical agent, for example, a radiolabelled antibody, contains a radioisotope (RI) which serves as the radiation source. As contemplated herein, the term “radioisotope” includes metallic and non-metallic radioisotopes. The radioisotope is chosen based on the medical application of the radiolabeled pharmaceutical agents. When the radioisotope is a metallic radioisotope, a chelator is typically employed to bind the metallic radioisotope to the rest of the molecule. When the radioisotope is a non-metallic radioisotope, the non-metallic radioisotope is typically linked directly, or via a linker, to the rest of the molecule. For purposes of this invention, the chemical elements are identified in accordance with the Periodic Table of the Elements, CAS version, Handbook of Chemistry and Physics, 67th Ed., 1986-87, inside cover. Compounds of the Invention One aspect of the invention relates to a compound of Formula (I):
Figure imgf000018_0001
wherein: A and A' are each independently selected from C-R' and N; A'' is selected from C-R'' and N; provided that at least one of A, A', and A'' is N; or A and A' are each independently selected from C-R' and N, and A'' is C-R'', wherein R'' taken together with R and the ring atoms to which they are bonded form a fused five-membered heteroaryl ring, such that formula I is a substituted indole or azaindole;
Figure imgf000019_0001
n is 0, 1, 2, or 3; m is 1; R1, R2, R3, R4, R5 and R6 are independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, aryl, heteroaryl, -C(O)OR11, -C(O)NR12R13 and –C(O)R14; R7, R8, R9, and R10 are independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, aryl, heteroaryl, -C(O)OR11, -C(O)NR12R13, and –C(O)R14; Z is selected from -H, halogen, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O- cycloheteroalkyl, -O-halocycloalkyl, -NH-alkyl, -NH-heteroalkyl, -NH-haloalkyl, -NH- cycloalkyl, -NH-cycloheteroalkyl, -NH-halocycloalkyl, aryl, -O-aryl, -NH-aryl, heteroaryl, - O-heteroaryl, -NH-heteroaryl, -C(O)OR11, -C(O)NR12R13, -NR12R13, and –C(O)R14; R is selected from alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O-cycloheteroalkyl, - O-halocycloalkyl, aryl, -O-aryl, heteroaryl, -O-heteroaryl -OSO2R11, and -NR15R16; R' is selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O- cycloalkyl, -O-cycloheteroalkyl, -O-halocycloalkyl, -O-aryl, -O-heteroaryl, -OSO2R11, and – NR12R13; R'' is selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O- cycloalkyl, -O-cycloheteroalkyl, -O-halocycloalkyl, -O-aryl, -O-heteroaryl, -OSO2R11, and – NR12R13; each occurrence of R11, R12 and R13 is independently selected from H, alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl; each occurrence of R14 is independently selected from alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl; R15 and R16 are independently selected from -H, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -C(O)R11, -C(O)OR11, and - C(O)NR12R13; or R15 and R16 taken together with the nitrogen atom to which they are bonded form an unsubstituted or substituted C4-C8 cycloheteroalkyl; and provided that if A is N, A' and A'' are each
Figure imgf000020_0001
is selected from phenyl and 2-methoxy-5-fluorophenyl, then R is not , ,
Figure imgf000020_0003
In some embodiments, R' is selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, and heteroaryl; and R'' is selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, and heteroaryl. In some embodiments, Y is selected from
Figure imgf000020_0002
In some embodiments, R1, R2, R3, R4, R5, R6, R7 and R8 are -H. In some embodiments, one of R1, R2, R3, R4, R5, R6, R7 and R8 is not -H. In some embodiments, two of R1, R2, R3, R4, R5, R6, R7 and R8 are not -H. In some embodiments, three of R1, R2, R3, R4, R5, R6, R7 and R8 are not -H.
Figure imgf000021_0002
In some embodiments, Y is
Figure imgf000021_0001
. In some embodiments, R7 and R8 are -H. In some embodiments, R9 and R10 are independently selected from -H and alkyl. In some embodiments, R7 and R8 are -H; and wherein R9 and R10 are independently selected from -H and alkyl. In some embodiments, R7, R8, R9, and R10 are each -H In some embodiments, Z is aryl or heteroaryl. In some embodiments, Z is selected from unsubstituted or substituted phenyl, pyridine, pyrimidine, pyrazole, and triazole. In some embodiments, Z is selected from
Figure imgf000022_0001
Figure imgf000022_0003
Ri7, R18, Rig, R20, and R21 are independently selected from -H, halogen, CN, CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, -O- heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O-cycloheteroalkyl, -O-halocycloalkyl, -NH-alkyl, -NH-heteroalkyl, -NH-haloalkyl, -NH-cycloalkyl, -NH-cycloheteroalkyl, -NH- halocycloalkyl, aryl, and heteroaryl; and Rmis haloalkyl or alkyl.
In some embodiments, Z is
Figure imgf000022_0002
Ri7 and Rix are independently selected from -H, halogen, CN, CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, -O-heteroalkyl, -O- haloalkyl, -O-cycloalkyl, -O-cycloheteroalkyl, -O-halocycloalkyl, -NH-alkyl, -NH- heteroalkyl, -NH-haloalkyl, -NH-cycloalkyl, -NH-cycloheteroalkyl, -NH-halocycloalkyl, aryl, and heteroaryl, or R17 taken together with Rix and the ring atoms to which they are bonded form a fused ring, such that Z is a benzimidazole, dihydrobenzofuran, benzodioxole, benzooxazole, or benzooxazolone; R19 and R20 are independently selected from -H, halogen, CN, CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O-cycloheteroalkyl, -O- halocycloalkyl, -NH-alkyl, -NH-heteroalkyl, -NH-haloalkyl, -NH-cycloalkyl, -NH- cycloheteroalkyl, -NH-halocycloalkyl, aryl, and heteroaryl, or R19 taken together with R20 and the ring atoms to which they are bonded form a fused ring, such that Z is a benzimidazole, dihydrobenzofuran, benzodioxole, benzooxazole, or benzooxazolone; and R211S selected from -H, halogen, CN, CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O- cycloheteroalkyl, -O-halocycloalkyl, -NH-alkyl, -NH-heteroalkyl, -NH-haloalkyl, -NH- cycloalkyl, -NH-cycloheteroalkyl, -NH-halocycloalkyl, aryl, and heteroaryl. In some embodiments, the compound having the structure selected from
Figure imgf000023_0001
In some embodiments, the compound having the structure selected from
Figure imgf000023_0002
In some embodiments, R is selected from aryl, heteroaryl, and -NR15R16; and R15 and R16 taken together with the nitrogen atom to which they are bonded form an unsubstituted or substituted C4-C8 cycloheteroalkyl. In some embodiments, R is selected from aryl and heteroaryl. In some embodiments, R is phenyl. In some embodiments, R is -NR15R16; and R15 and R16 taken together with the nitrogen atom to which they are bonded form an unsubstituted or substituted piperidine, piperazine, pyrrolidine, morpholine, or azetidine. In some embodiments,
Figure imgf000023_0003
R22 is selected from alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -SO2R23, -C(O)OR23, -C(O)NR24R25, and -C(O)R26; Ra, Rb, Rc, Rd, Rg, Rh, Ri, and Rj are independently selected from -H, halogen, -CN, - CF3, alkyl, heteroalkyl, haloalkyl, hydroxyalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -SO2R23, -C(O)OR23, -C(O)NR24R25, and -C(O)R26; or Ra taken together with Rb and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, ^-lactam, or ^-lactam; or Rc taken together with Rd and the carbon atom to which they are bonded form an unsubstituted or substituted C3 C6 cycloalkyl ^ lactam or ^ lactam; each occurrence of R23, R24 and R25 is independently selected from H, alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl; and each occurrence of R26 is selected from alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl. In some embodiments,
Figure imgf000024_0001
Ra, Rb, Ri and Rj are independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, - C(O)OR23, -C(O)NR24R25, and -C(O)R26; or Ra taken together with Rb and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, ^-lactam, or ^- lactam; Rc, Rd, Re, Rf, Rg and Rh are independently selected from -H, halogen, -CN, -CF3, - OH, -NH2, alkyl, heteroalkyl, haloalkyl, hydroxyalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O-cycloheteroalkyl, - O-halocycloalkyl, -NH-alkyl, -NH-heteroalkyl, -NH-haloalkyl, -NH-cycloalkyl, -NH- cycloheteroalkyl, -NH-halocycloalkyl, aryl, -O-aryl, -NH-aryl, heteroaryl, -O-heteroaryl, - NH-heteroaryl, -SO2R23, -OSO2R23, -C(O)OR23, -NR24C(O)R25, -NR24SO2R25, - C(O)NR24R25, -OSO2(N26)2, -SO2(N26)2, -N(R26)2, -C(O)R26, and alkyl-NHSO2-R26; or Rc taken together with Rd and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, ^-lactam, or ^-lactam; or Re taken together with Rf and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, ^-lactam, or ^-lactam; each occurrence of R23, R24 and R25 is independently selected from -H, alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl; and each occurrence of R26 is selected from alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl. In some embodiments,
Figure imgf000024_0002
Ra, Rb, Rc, Rd, Rg, Rh Ri and Rj are independently selected from -H, halogen, -CN, CF3, - alkyl, heteroalkyl, haloalkyl, hydroxyalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -SO2R23, -C(O)OR23, -C(O)NR24R25, -OSO2(N26)2, - SO2(N26)2, –C(O)R26 , and alkyl-NHSO2-R26; or Ra taken together with Rb and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, ^- lactam, or ^-lactam; or Rc taken together with Rd and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, ^-lactam, or ^-lactam; each occurrence of R23, R24 and R25 is independently selected from -H, alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl; and each occurrence of R26 is selected from alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl.
Figure imgf000025_0001
Ra, Rb, Rg and Rh are independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, hydroxyalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -SO2R23, -C(O)OR23, -C(O)NR24R25, -SO2(N26)2, and -C(O)R26; or Ra taken together with Rb and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, ^-lactam, or ^-lactam; Rc, Rd, Re and Rf are independently selected from -H, halogen, -CN, -CF3, -OH, -NH2, alkyl, heteroalkyl, haloalkyl, hydroxyalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O- alkyl, -O-heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O-cycloheteroalkyl, -O-halocycloalkyl, - NH-alkyl, -NH-heteroalkyl, -NH-haloalkyl, -NH-cycloalkyl, -NH-cycloheteroalkyl, -NH- halocycloalkyl, aryl, -O-aryl, -NH-aryl, heteroaryl, -O-heteroaryl, -NH-heteroaryl, -SO2R23, - OSO2R23, -C(O)OR23, -NR24C(O)R25, -NR24SO2R25, -C(O)NR24R25, -OSO2(N26)2, - SO2(N26)2, -N(R26)2, and -C(O)R26; or Rc taken together with Rd and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, ^-lactam, or ^- lactam; and each occurrence of R23, R24 and R25 is independently selected from -H, alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl; and each occurrence of R26 is selected from alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl. In some embodiments, R is
Figure imgf000026_0003
Ra, Rb, Re and Rf are independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, hydroxy alkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -SO2R23, -C(0)0R23, and -C(0)NR24R25; or Ra taken together with Rb and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, γ-lactam, or δ-lactam;
Rc and Rd are independently selected from -H, halogen, -CN, -CF3, -OH, -NH2, alkyl, heteroalkyl, haloalkyl, hydroxy alkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, - O-heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O-cycloheteroalkyl, -O-halocycloalkyl, -NH- alkyl, -NH-heteroalkyl, -NH-haloalkyl, -NH-cycloalkyl, -NH-cycloheteroalkyl, -NH- halocycloalkyl, aryl, heteroaryl- SO2R23, -OSO2R23, -C(0)0R23, -NR24C(0)R25, -NR24SO2R25, -C(0)NR24R25, -0S02(N26)2, -S02(N26)2, -N(R26)2, and -C(0)R26; or Rc taken together with Rdand the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, γ-lactam, or δ-lactam; each occurrence of R23, R24 and R25 is independently selected from -H, alkyl, heteroalkyl, haloalkyl, aryl or heteroaryl; and each occurrence of R26 is selected from alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl.
Figure imgf000026_0001
In some embodiments, R is selected from
Figure imgf000026_0002
Figure imgf000026_0004
each occurrence of R22 is selected from alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -SO2R19, -C(0)Ri9, -C(0)0Ri9, and - C(0)NRI9R2O; each occurrence of Ra, Rb, Rc, Rd, Rg, Rh Ri and Rj is independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, hydroxy alkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -SO2R23, -C(0)0R23, -C(0)NR24R25, and -C(0)R26; or Ra taken together with Rb and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, ^-lactam, or ^-lactam; each occurrence of R23, R24 and R25 is independently selected from H, alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl; and each occurrence of R26 is selected from alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl.
Figure imgf000027_0001
each occurrence of Ra, Rb, Ri and Rj is independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -C(O)OR23, -C(O)NR24R25, and -C(O)R26; or Ra taken together with Rb and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, ^-lactam, or ^-lactam; each occurrence of Rc, Rd, Re, Rf, Rg and Rh is independently selected from -H, halogen, -CN, -CF3, -OH, -NH2, alkyl, heteroalkyl, haloalkyl, hydroxyalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O- cycloheteroalkyl, -O-halocycloalkyl, -NH-alkyl, -NH-heteroalkyl, -NH-haloalkyl, -NH- cycloalkyl, -NH-cycloheteroalkyl, -NH-halocycloalkyl, aryl, -O-aryl, -NH-aryl, heteroaryl, - O-heteroaryl, -NH-heteroaryl, -SO2R23, -OSO2R23, -C(O)OR23, -NR24C(O)R25, -NR24SO2R25, -C(O)NR25R26, -OSO2(N26)2, -SO2(N26)2, -N(R26)2, -C(O)R26, and alkyl-NHSO2-R26; each occurrence of R23, R24 and R25 is independently selected from -H, alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl; and each occurrence of R26 is selected from alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl. In some embodiments, R is selected from
Figure imgf000028_0001
Figure imgf000028_0004
each occurrence of Ra, Rb, Rc, Rd, Rg, Rh Ri and Rj is independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, hydroxyalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -SO2R23, -C(0)0R23, -C(0)NR24R25, -0S02(N26)2, - S02(N26)2, -C(0)R26, and alkyl-NHS02-R26; or Ra taken together with Rb and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, γ- lactam, or δ -lactam; each occurrence of R23, R24 and R25 is independently selected from -H, alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl; and each occurrence of R26 is selected from alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl.
In some embodiments, R is selected from , and
Figure imgf000028_0002
Figure imgf000028_0003
each occurrence of Ra, Rb, Rg and Rh is independently selected from -H, halogen, - CN, -CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, hydroxyalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -SO2R23, -C(0)0R23, -C(0)NR24R25, -S02(N26)2, and - C(0)R26; or Ra taken together with Rb and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, γ-lactam, or δ-lactam; each occurrence of Rc, Rd, Re and Rr is independently selected from -H, halogen, -CN, -CF3, -OH, -NH2, alkyl, heteroalkyl, haloalkyl, hydroxyalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O-cycloheteroalkyl, - O-halocycloalkyl, -NH-alkyl, -NH-heteroalkyl, -NH-haloalkyl, -NH-cycloalkyl, -NH- cycloheteroalkyl, -NH-halocycloalkyl, aryl, -O-aryl, -NH-aryl, heteroaryl, -O-heteroaryl, - NH-heteroaryl, -SO2R23, -OSO2R23, -C(O)OR23, -NR24C(O)R25, -NR24SO2R25, - C(O)NR24R25, -OSO2(N26)2, -SO2(N26)2, -N(R26)2, and -C(O)R26; and each occurrence of R23, R24 and R25 is independently selected from -H, alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl; and each occurrence of R26 is selected from alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl. In some embodiments,
Figure imgf000029_0001
each occurrence Ra and Rb is independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, hydroxyalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -SO2R23, -C(O)OR23, and -C(O)NR24R25; or Ra taken together with Rb and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, ^-lactam, or ^-lactam; each occurrence Rc and Rd is independently selected from -H, halogen, -CN, -CF3, - OH, -NH2, alkyl, heteroalkyl, haloalkyl, hydroxyalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O-cycloheteroalkyl, - O-halocycloalkyl, -NH-alkyl, -NH-heteroalkyl, -NH-haloalkyl, -NH-cycloalkyl, -NH- cycloheteroalkyl, -NH-halocycloalkyl, aryl, heteroaryl-SO2R23, -OSO2R23, -C(O)OR23, - NR24C(O)R25, -NR24SO2R25, -C(O)NR24R25, -OSO2(N26)2, -SO2(N26)2, -N(R26)2, and - C(O)R26; and each occurrence of R23, R24 and R25 is independently selected from -H, alkyl, heteroalkyl, haloalkyl, aryl or heteroaryl; and each occurrence of R26 is selected from alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl. In some embodiments, each occurrence of R26 is independently selected from -H, alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl. In some embodiments, the compound is selected from the following table:
Figure imgf000029_0002
Figure imgf000030_0001
Figure imgf000031_0001
Figure imgf000032_0001
Figure imgf000033_0001
Figure imgf000034_0001
Figure imgf000035_0001
Figure imgf000036_0001
Figure imgf000037_0001
Figure imgf000038_0001
Figure imgf000039_0001
Figure imgf000040_0001
Figure imgf000041_0001
Figure imgf000042_0001
In some embodiments, R taken together with R'' and the ring atoms to which they are bonded form a fused five-membered heteroaryl ring, such that formula I is a substituted indole or azaindole.
Figure imgf000043_0001
In some embodiments, the compound having the structure:
Figure imgf000043_0002
Figure imgf000043_0003
, wherein each occurrence of R27 is selected from alkyl, heteroalkyl, haloalkyl, hydroxyalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, and heteroaryl. In some embodiments, the compound is selected from the following table:
Figure imgf000043_0004
In some embodiments, the compounds are atropisomers. Additionally, unless otherwise stated, structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, compounds produced by the replacement of a hydrogen with deuterium or tritium, or of a carbon with a 13C- or 14C-enriched carbon are within the scope of this invention. Such compounds are useful, for example, as analytical tools, as probes in biological assays, or as therapeutic agents in accordance with the present invention. For example, in the case of variable R1, the (C1-C4)alkyl or the -O-(C1-C4)alkyl can be suitably deuterated (e.g., -CD3, -OCD3). Any compound of the invention can also be radiolabed for the preparation of a radiopharmaceutical agent. Methods of Treatment Another aspect of the invention relates to methods of treating or preventing a disease or disorder associated with a SLC6A8 mutation, treating an inflammatory disease, increasing transport of guanidinoacetic acid or a salt thereof across the blood-brain barrier, improving the function of a cellular creatine transporter, or decreasing the accumulation or the concentration of guanidinoacetic acid or a salt thereof in a cell, in a subject in need thereof comprising administering to the subject an effective amount of a compound of Formula (I):
Figure imgf000044_0002
wherein: A and A' are each independently selected from C-R' and N; A'' is selected from C-R'' and N; provided that at least one of A, A', and A'' is N; or A and A' are each independently selected from C-R' and N, and A'' is C-R'', wherein R'' taken together with R and the ring atoms to which they are bonded form a fused five-membered heteroaryl ring, such that formula I is a substituted indole or azaindole; X is selected from
Figure imgf000044_0001
Y is selected from
Figure imgf000045_0001
« is 0, 1, 2, or 3; m is 1;
Ri, R2, R3, R4, RS and Re are independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, aryl, heteroaryl, -C(0)0Rn, -C(0)NRi2Ri3, and -C(0)Ri4;
Rv, Rs, R9, and Rio are independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, aryl, heteroaryl, -C(0)0Rn, -C(0)NRi2Ri3, and -C(0)Ri4;
Z is selected from -H, halogen, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O- cycloheteroalkyl, -O-halocycloalkyl, -NH-alkyl, -NH-heteroalkyl, -NH-haloalkyl, -NH- cycloalkyl, -NH-cycloheteroalkyl, -NH-halocycloalkyl, aryl, -O-aryl, -NH-aryl, heteroaryl, - O-heteroaryl, -NH-heteroaryl, -C(0)0Rn, -C(0)NRi2Ri3, -NR12R13, and -C(0)Ri4;
R is selected from alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O-cycloheteroalkyl, - O-halocycloalkyl, aryl, -O-aryl, heteroaryl, -O-heteroaryl -OSO2R11, and -NRisRie;
R is selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O- cycloalkyl, -O-cycloheteroalkyl, -O-halocycloalkyl, -O-aryl, -O-heteroaryl, -OSO2R11, and -
NR12R13;
R" is selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O- cycloalkyl, -O-cycloheteroalkyl, -O-halocycloalkyl, -O-aryl, -O-heteroaryl, -OSO2R11, and -
NR12R13; each occurrence of R11, R12 and R13 is independently selected from H, alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl; each occurrence of Ri4is independently selected from alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl;
Ri5 and Rie are independently selected from -H, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -C(0)Rn, -C(0)0Rn, and - C(0)NRI2RI3; or R15 and Rie taken together with the nitrogen atom to which they are bonded form an un substituted or substituted C4-C8 cycloheteroalkyl. In some embodiments, the above compounds wherein if A is N, A' and A'' are each C- H, X is
Figure imgf000046_0001
, , and Z is selected from phenyl and 2-methoxy-5- fluorophenyl, then R is not
Figure imgf000046_0002
. Another aspect of the invention relates to methods of treating or preventing a disease or disorder associated with a SLC6A8 mutation, treating an inflammatory disease, increasing transport of guanidinoacetic acid or a salt thereof across the blood-brain barrier, improving the function of a cellular creatine transporter, or decreasing the accumulation or the concentration of guanidinoacetic acid or a salt thereof in a cell, in a subject in need thereof comprising administering to the subject an effective amount of any of the compounds disclosed herein. Another aspect of the invention relates to methods of treating or preventing a disease or disorder associated with a SLC6A8 mutation, treating an inflammatory disease, increasing transport of guanidinoacetic acid or a salt thereof across the blood-brain barrier, improving the function of a cellular creatine transporter, or decreasing the accumulation or the concentration of guanidinoacetic acid or a salt thereof in a cell, in a subject in need thereof comprising administering to the subject an effective amount of any of the pharmaceutical compositions disclosed herein. Creatine transporter deficiency (CTD) CTD is an inborn error of creatine metabolism in which creatine is not properly transported to the brain and muscles due to defective creatine transporters. CTD is an X- linked disorder caused by mutations in the SLC6A8 gene. The SLC6A8 gene is located on the short arm of the sex chromosome, Xq28. Hemizygous males with CTD express speech and behavior abnormalities, intellectual disabilities, development delay, seizures, and autistic behavior. Heterozygous females with CTD generally express fewer, less severe symptoms.CTD is one of three different types of cerebral creatine deficiency (CCD). The other two types of CCD are guanidinoacetate methyltransferase (GAMT) deficiency and L- arginine:glycine amidinotransferase (AGAT) deficiency. Clinical presentation of CTD is similar to that of GAMT and AGAT deficiency. CTD was first identified in 2001 with the presence of a hemizygous nonsense mutation in the SLC6A8 gene in a male patient. CTD is difficult to treat because the actual transporter responsible for transporting creatine to the brain and muscles is defective. Studies in which oral creatine monohydrate supplements were given to patients with CTD found that patients did not respond to treatment. However, similar studies conducted in which patients that had GAMT or AGAT deficiency were given oral creatine monohydrate supplements found that patient’s clinical symptoms improved. Patients with CTD are unresponsive to oral creatine monohydrate supplements because regardless of the amount of creatine they ingest, the creatine transporter is still defective, and therefore creatine is incapable of being transported across the BBB.
Accordingly, in certain embodiments, the invention provides methods of treating or preventing a disease or disorder associated with a SLC6A8 mutation in a subjet in need thereof, comprising administering to the subject an effective amount of a compound disclosed herein.
In some embodiments, the disease or disorder is creatine transporter deficiency. In some embodiments, the disease or disorder is motor dysfunction. In some embodiments, the disease or disorder is intellectual disability. In some embodiments, the disease or disorder is language delay or speech delay. In some embodiments, the disease or disorder is hypotonia.
In some embodiments, the disease or disorder is seizures. In some embodiments, the disease or disorder is behaviours associated with autism and attention deficit hyperactivity disorder.
In some embodiments, the disease or disorder is fatigue. In some embodiments, the disease or disorder is muscular hypotonia. In some embodiments, the disease or disorder is low weight gain. In some embodiments, the disease or disorder is gastrointestinal disorders. In some embodiments, the disease or disorder is cardiac disorders.
In certain embodiments, the invention provides a method of improving the function of a cellular creatine transporter in a subject in need thereof, comprising administering to the subject an effective amount of a compound disclosed herein.
In certain embodiments, the invention provides a method of increasing cellular trafficking of a cellular creatine transporter in a subject in need thereof, comprising administering to the subject an effective amount of a compound disclosed herein.
In certain embodiments, the invention provides a method of correcting a defect in a cellular creatine transporter function in a subject in need thereof, comprising administering to the subject an effective amount of a compound disclosed herein.
In some embodiments, the method wherein the cellular creatine transporter is SLC6A8. In some embodiments, the method wherein cellular creatine transporter is a mutant creatine transporter. In some embodiments, the method wherein mutant creatine transporter is mutant SLC6A8. In some embodiments, the method wherein the cellular concentration of creatine is increased.
In some embodiments, the invention provides a method of decreasing accumulation or the concentration of guanidinoacetic acid or a salt thereof in a cell of a subject in need thereof, comprising administering to the subject an effective amount of a compound dislosed herein.
In some embodiments, the invention provides a method of decreasing accumulation or the concentration of guanidinoacetic acid or a salt thereof in a cell of a subject in need thereof, comprising administering to the subject an effective amount of a compound dislosed herein that increases transport of guanidinoacetic acid or a salt thereof by a mutant creatine transporter.
In some embodiments, the method wherein compound decreases the intracellular concentration of guanidinoacetic acid or a salt thereof. In some embodiments, the method wherein compound decreases the intracellular accumulation of guanidinoacetic acid or a salt thereof.
In some embodiments, the invention provides a method of increasing transport of guanidinoacetic acid or a salt thereof across the blood-brain barrier in the subject in need thereof, comprising administering to the subject aneffective amount of a compound dislosed herein.
In some embodiments, the invention provides a method of treating an inflammatory disease in a subject in need thereof, comprising administering to the subject aneffective amount of a compound dislosed herein.
In some embodiments, the method wherein the inflammatory disease is acute. In some embodiments, the method wherein the inflammatory disease is chronic.
In some embodiments, the method wherein the inflammatory disease is selected from inflammatory bowel diseases (for example, ulcerative colitis or Crohn's disease), multiple sclerosis, psoriasis, arthritis, rheumatoid arthritis, osteoarthritis, juvenile arthritis, psoriatic arthritis, reactive arthritis, ankylosing spondylitis, cryopyrin associated periodic syndromes, Muckle- Wells syndrome, familial cold auto-inflammatory syndrome, neonatal-onset multisystem inflammatory disease, TNF receptor associated periodic syndrome, acute and chronic pancreatitis, atherosclerosis, gout, ankylosing spondylitis, fibrotic disorders (for example, hepatic fibrosis or idiopathic pulmonary fibrosis), nephropathy, sarcoidosis, scleroderma, anaphylaxis, diabetes (for example, diabetes mellitus type 1 or diabetes mellitus type 2), diabetic retinopathy, Still's disease, vasculitis, sarcoidosis, pulmonary inflammation, acute respiratory distress syndrome, wet and dry age-related macular degeneration, autoimmune hemolytic syndromes, autoimmune and inflammatory hepatitis, autoimmune neuropathy, autoimmune ovarian failure, autoimmune orchitis, autoimmune thrombocytopenia, silicone implant associated autoimmune disease, Sjogren's syndrome, familial Mediterranean fever, systemic lupus erythematosus, vasculitis syndromes (for example, temporal, Takayasu's and giant cell arteritis, Behçet's disease or Wegener's granulomatosis), vitiligo, secondary hematologic manifestation of autoimmune diseases (for example, anemias), drug-induced autoimmunity, Hashimoto's thyroiditis, hypophysitis, idiopathic thrombocytic pupura, metal-induced autoimmunity, myasthenia gravis, pemphigus, autoimmune deafness (for example, Meniere's disease), Goodpasture's syndrome, Graves' disease, HW-related autoimmune syndromes, Gullain-Barre disease, Addison's disease, anti- phospholipid syndrome, asthma, atopic dermatitis, Celiac disease, Cushing's syndrome, dermatomyositis, idiopathic adrenal adrenal atrophy, idiopathic thrombocytopenia, Kawasaki syndrome, Lambert-Eaton Syndrome, pernicious anemia, pollinosis, polyarteritis nodosa, primary biliary cirrhosis, primary sclerosing cholangitis, Raynaud's, Reiter's Syndrome, relapsing polychondritis, Schmidt's syndrome, thyrotoxidosis, sepsis, septic shock, endotoxic shock, exotoxin-induced toxic shock, gram negative sepsis, toxic shock syndrome, glomerulonephritis, peritonitis, interstitial cystitis, hyperoxia-induced inflammations, chronic obstructive pulmonary disease (COPD), vasculitis, graft vs. host reaction (for example, graft vs. host disease), allograft rejections (for example, acute allograft rejection or chronic allograft rejection), early transplantation rejection (for example, acute allograft rejection), reperfusion injury, pain (for example, acute pain, chronic pain, neuropathic pain, or fibromyalgia), chronic infections, meningitis, encephalitis, myocarditis, gingivitis, post- surgical trauma, tissue injury, traumatic brain injury, enterocolitis, sinusitis, uveitis, ocular inflammation, optic neuritis, gastric ulcers, esophagitis, peritonitis, periodontitis, dermatomyositis, gastritis, myositis, polymyalgia, pneumonia, and bronchitis. In some embodiments of any of the disclosed methods, the cell is a brain cell. In some embodiments of any of the disclosed methods, wherein the subject is a mammal. In some embodiments of any of the disclosed methods, the mammal is a male. In some embodiments of any of the disclosed methods, the mammal is a female. In some embodiments of any of the disclosed methods, the mammal is a primate, equine, bovine, ovine, feline, or canine. In some embodiments of any of the disclosed methods, the mammal is a human. Pharmaceutical Compositions, Routes of Administration, and Dosing In certain embodiments, the invention is directed to a pharmaceutical composition, comprising a compound of the invention and a pharmaceutically acceptable carrier. In certain embodiments, the pharmaceutical composition comprises a plurality of compounds of the invention and a pharmaceutically acceptable carrier. In certain embodiments, a pharmaceutical composition of the invention further comprises at least one additional pharmaceutically active agent other than a compound of the invention. The at least one additional pharmaceutically active agent can be an agent useful in the treatment of ischemia-reperfusion injury. Pharmaceutical compositions of the invention can be prepared by combining one or more compounds of the invention with a pharmaceutically acceptable carrier and, optionally, one or more additional pharmaceutically active agents. As stated above, an “effective amount” refers to any amount that is sufficient to achieve a desired biological effect. Combined with the teachings provided herein, by choosing among the various active compounds and weighing factors such as potency, relative bioavailability, patient body weight, severity of adverse side-effects and mode of administration, an effective prophylactic or therapeutic treatment regimen can be planned which does not cause substantial unwanted toxicity and yet is effective to treat the particular subject. The effective amount for any particular application can vary depending on such factors as the disease or condition being treated, the particular compound of the invention being administered, the size of the subject, or the severity of the disease or condition. One of ordinary skill in the art can empirically determine the effective amount of a particular compound of the invention and/or other therapeutic agent without necessitating undue experimentation. A maximum dose may be used, that is, the highest safe dose according to some medical judgment. Multiple doses per day may be contemplated to achieve appropriate systemic levels of compounds. Appropriate systemic levels can be determined by, for example, measurement of the patient’s peak or sustained plasma level of the drug. “Dose” and “dosage” are used interchangeably herein. In certain embodiments, intravenous administration of a compound may typically be from 0.1 mg/kg/day to 20 mg/kg/day. In one embodiment, intravenous administration of a compound may typically be from 0.1 mg/kg/day to 2 mg/kg/day. In one embodiment, intravenous administration of a compound may typically be from 0.5 mg/kg/day to 5 mg/kg/day. In one embodiment, intravenous administration of a compound may typically be from 1 mg/kg/day to 20 mg/kg/ day. In one embodiment, intravenous administration of a compound may typically be from 1 mg/kg/day to 10 mg/kg/day.
Generally, daily oral doses of a compound will be, for human subjects, from about 0.01 milligrams/kg per day to 1000 milligrams/kg per day. It is expected that oral doses in the range of 0.5 to 50 milligrams/kg, in one or more administrations per day, will yield therapeutic results. Dosage may be adjusted appropriately to achieve desired drug levels, local or systemic, depending upon the mode of administration. For example, it is expected that intravenous administration would be from one order to several orders of magnitude lower dose per day. In the event that the response in a subject is insufficient at such doses, even higher doses (or effective higher doses by a different, more localized delivery route) may be employed to the extent that patient tolerance permits. Multiple doses per day are contemplated to achieve appropriate systemic levels of the compound.
For any compound described herein the therapeutically effective amount can be initially determined from animal models. A therapeutically effective dose can also be determined from human data for compounds which have been tested in humans and for compounds which are known to exhibit similar pharmacological activities, such as other related active agents. Higher doses may be required for parenteral administration. The applied dose can be adjusted based on the relative bioavailability and potency of the administered compound. Adjusting the dose to achieve maximal efficacy based on the methods described above and other methods as are well-known in the art is well within the capabilities of the ordinarily skilled artisan.
The formulations of the invention can be administered in pharmaceutically acceptable solutions, which may routinely contain pharmaceutically acceptable concentrations of salt, buffering agents, preservatives, compatible carriers, adjuvants, and optionally other therapeutic ingredients.
For use in therapy, an effective amount of the compound can be administered to a subject by any mode that delivers the compound to the desired surface. Administering a pharmaceutical composition may be accomplished by any means known to the skilled artisan. Routes of administration include but are not limited to intravenous, intramuscular, intraperitoneal, intravesical (urinary bladder), oral, subcutaneous, direct injection (for example, into a tumor or abscess), mucosal (e.g., topical to eye), inhalation, and topical.
For intravenous and other parenteral routes of administration, a compound of the invention can be formulated as a lyophilized preparation, as a lyophilized preparation of liposome-intercalated or -encapsulated active compound, as a lipid complex in aqueous suspension, or as a salt complex. Lyophilized formulations are generally reconstituted in suitable aqueous solution, e.g., in sterile water or saline, shortly prior to administration.
For oral administration, the compounds can be formulated readily by combining the active compound(s) with pharmaceutically acceptable carriers well known in the art. Such carriers enable the compounds of the invention to be formulated as tablets, pills, dragees, capsules, liquids, gels, syrups, slurries, suspensions and the like, for oral ingestion by a subject to be treated. Pharmaceutical preparations for oral use can be obtained as solid excipient, optionally grinding a resulting mixture, and processing the mixture of granules, after adding suitable auxiliaries, if desired, to obtain tablets or dragee cores. Suitable excipients are, in particular, fillers such as sugars, including lactose, sucrose, mannitol, or sorbitol; cellulose preparations such as, for example, maize starch, wheat starch, rice starch, potato starch, gelatin, gum tragacanth, methyl cellulose, hydroxypropylmethyl-cellulose, sodium carboxymethylcellulose, and/or polyvinylpyrrolidone (PVP). If desired, disintegrating agents may be added, such as the cross-linked polyvinyl pyrrolidone, agar, or alginic acid or a salt thereof such as sodium alginate. Optionally the oral formulations may also be formulated in saline or buffers, e.g., EDTA for neutralizing internal acid conditions or may be administered without any carriers.
Also specifically contemplated are oral dosage forms of the above component or components. The component or components may be chemically modified so that oral delivery of the derivative is efficacious. Generally, the chemical modification contemplated is the attachment of at least one moiety to the component molecule itself, where said moiety permits (a) inhibition of acid hydrolysis; and (b) uptake into the blood stream from the stomach or intestine. Also desired is the increase in overall stability of the component or components and increase in circulation time in the body. Examples of such moieties include: polyethylene glycol, copolymers of ethylene glycol and propylene glycol, carboxymethyl cellulose, dextran, polyvinyl alcohol, polyvinyl pyrrolidone and polyproline. Abuchowski and Davis, “Soluble Polymer-Enzyme Adducts”, In: Enzymes as Drugs, Hocenberg and Roberts, eds., Wiley-Interscience, New York, N.Y., pp. 367-383 (1981); Newmark et ah, J Appl Biochem 4:185-9 (1982). Other polymers that could be used are poly- 1 ,3 -dioxolane and poly-1, 3, 6-tioxocane. For pharmaceutical usage, as indicated above, polyethylene glycol moieties are suitable.
For the component (or derivative) the location of release may be the stomach, the small intestine (the duodenum, the jejunum, or the ileum), or the large intestine. One skilled in the art has available formulations which will not dissolve in the stomach, yet will release the material in the duodenum or elsewhere in the intestine. Preferably, the release will avoid the deleterious effects of the stomach environment, either by protection of the compound of the invention (or derivative) or by release of the biologically active material beyond the stomach environment, such as in the intestine. To ensure full gastric resistance a coating impermeable to at least pH 5.0 is essential. Examples of the more common inert ingredients that are used as enteric coatings are cellulose acetate trimellitate (CAT), hydroxypropylmethylcellulose phthalate (HPMCP), HPMCP 50, HPMCP 55, polyvinyl acetate phthalate (PVAP), Eudragit L30D, Aquateric, cellulose acetate phthalate (CAP), Eudragit L, Eudragit S, and shellac. These coatings may be used as mixed films. A coating or mixture of coatings can also be used on tablets, which are not intended for protection against the stomach. This can include sugar coatings, or coatings which make the tablet easier to swallow. Capsules may consist of a hard shell (such as gelatin) for delivery of dry therapeutic (e.g., powder); for liquid forms, a soft gelatin shell may be used. The shell material of cachets could be thick starch or other edible paper. For pills, lozenges, molded tablets or tablet triturates, moist massing techniques can be used. The therapeutic can be included in the formulation as fine multi-particulates in the form of granules or pellets of particle size about 1 mm. The formulation of the material for capsule administration could also be as a powder, lightly compressed plugs or even as tablets. The therapeutic could be prepared by compression. Colorants and flavoring agents may all be included. For example, the compound of the invention (or derivative) may be formulated (such as by liposome or microsphere encapsulation) and then further contained within an edible product, such as a refrigerated beverage containing colorants and flavoring agents. One may dilute or increase the volume of the therapeutic with an inert material. These diluents could include carbohydrates, especially mannitol, ^-lactose, anhydrous lactose, cellulose, sucrose, modified dextrans and starch. Certain inorganic salts may be also be used as fillers including calcium triphosphate, magnesium carbonate and sodium chloride. Some commercially available diluents are Fast-Flo, Emdex, STA-Rx 1500, Emcompress and Avicell. Disintegrants may be included in the formulation of the therapeutic into a solid dosage form. Materials used as disintegrates include but are not limited to starch, including the commercial disintegrant based on starch, Explotab. Sodium starch glycolate, Amberlite, sodium carboxymethylcellulose, ultramylopectin, sodium alginate, gelatin, orange peel, acid carboxymethyl cellulose, natural sponge and bentonite may all be used. Another form of the disintegrants are the insoluble cationic exchange resins. Powdered gums may be used as disintegrants and as binders and these can include powdered gums such as agar, Karaya or tragacanth. Alginic acid and its sodium salt are also useful as disintegrants.
Binders may be used to hold the therapeutic agent together to form a hard tablet and include materials from natural products such as acacia, tragacanth, starch and gelatin. Others include methyl cellulose (MC), ethyl cellulose (EC) and carboxymethyl cellulose (CMC). Polyvinyl pyrrolidone (PVP) and hydroxypropylmethyl cellulose (HPMC) could both be used in alcoholic solutions to granulate the therapeutic.
An anti-frictional agent may be included in the formulation of the therapeutic to prevent sticking during the formulation process. Lubricants may be used as a layer between the therapeutic and the die wall, and these can include but are not limited to; stearic acid including its magnesium and calcium salts, polytetrafluoroethylene (PTFE), liquid paraffin, vegetable oils and waxes. Soluble lubricants may also be used such as sodium lauryl sulfate, magnesium lauryl sulfate, polyethylene glycol of various molecular weights, Carbowax 4000 and 6000.
Glidants that might improve the flow properties of the drug during formulation and to aid rearrangement during compression might be added. The glidants may include starch, talc, pyrogenic silica and hydrated silicoaluminate.
To aid dissolution of the therapeutic into the aqueous environment a surfactant might be added as a wetting agent. Surfactants may include anionic detergents such as sodium lauryl sulfate, dioctyl sodium sulfosuccinate and dioctyl sodium sulfonate. Cationic detergents which can be used and can include benzalkonium chloride and benzethonium chloride. Potential non-ionic detergents that could be included in the formulation as surfactants include lauromacrogol 400, polyoxyl 40 stearate, polyoxyethylene hydrogenated castor oil 10, 50 and 60, glycerol monostearate, polysorbate 40, 60, 65 and 80, sucrose fatty acid ester, methyl cellulose and carboxy methyl cellulose. These surfactants could be present in the formulation of the compound of the invention or derivative either alone or as a mixture in different ratios.
Pharmaceutical preparations which can be used orally include push-fit capsules made of gelatin, as well as soft, sealed capsules made of gelatin and a plasticizer, such as glycerol or sorbitol. The push-fit capsules can contain the active ingredients in admixture with filler such as lactose, binders such as starches, and/or lubricants such as talc or magnesium stearate and, optionally, stabilizers. In soft capsules, the active compounds may be dissolved or suspended in suitable liquids, such as fatty oils, liquid paraffin, or liquid polyethylene glycols. In addition, stabilizers may be added. Microspheres formulated for oral administration may also be used. Such microspheres have been well defined in the art. All formulations for oral administration should be in dosages suitable for such administration. For buccal administration, the compositions may take the form of tablets or lozenges formulated in conventional manner. For topical administration, the compound may be formulated as solutions, gels, ointments, creams, suspensions, etc. as are well-known in the art. Systemic formulations include those designed for administration by injection, e.g., subcutaneous, intravenous, intramuscular, intrathecal or intraperitoneal injection, as well as those designed for transdermal, transmucosal oral or pulmonary administration. For administration by inhalation, compounds for use according to the present invention may be conveniently delivered in the form of an aerosol spray presentation from pressurized packs or a nebulizer, with the use of a suitable propellant, e.g., dichlorodifluoromethane, trichlorofluoromethane, dichlorotetrafluoroethane, carbon dioxide or other suitable gas. In the case of a pressurized aerosol the dosage unit may be determined by providing a valve to deliver a metered amount. Capsules and cartridges of e.g., gelatin for use in an inhaler or insufflator may be formulated containing a powder mix of the compound and a suitable powder base such as lactose or starch. Also contemplated herein is pulmonary delivery of the compounds disclosed herein (or salts thereof). The compound is delivered to the lungs of a mammal while inhaling and traverses across the lung epithelial lining to the blood stream. Other reports of inhaled molecules include Adjei et al., Pharm Res 7:565-569 (1990); Adjei et al., Int J Pharmaceutics 63:135-144 (1990) (leuprolide acetate); Braquet et al., J Cardiovasc Pharmacol 13(suppl. 5):143-146 (1989) (endothelin-1); Hubbard et al., Annal Int Med 3:206-212 (1989) ( ^1- antitrypsin); Smith et al., 1989, J Clin Invest 84:1145-1146 (a-1-proteinase); Oswein et al., 1990, "Aerosolization of Proteins", Proceedings of Symposium on Respiratory Drug Delivery II, Keystone, Colorado, March, (recombinant human growth hormone); Debs et al., 1988, J Immunol 140:3482-3488 (interferon-gamma and tumor necrosis factor alpha) and Platz et al., U.S. Pat. No.5,284,656 (granulocyte colony stimulating factor; incorporated by reference). A method and composition for pulmonary delivery of drugs for systemic effect is described in U.S. Pat. No.5,451,569 (incorporated by reference), issued Sep.19, 1995 to Wong et al. Contemplated for use in the practice of this invention are a wide range of mechanical devices designed for pulmonary delivery of therapeutic products, including but not limited to nebulizers, metered dose inhalers, and powder inhalers, all of which are familiar to those skilled in the art. Some specific examples of commercially available devices suitable for the practice of this invention are the Ultravent nebulizer, manufactured by Mallinckrodt, Inc., St. Louis, Mo.; the Acorn II nebulizer, manufactured by Marquest Medical Products, Englewood, Colo.; the Ventolin metered dose inhaler, manufactured by Glaxo Inc., Research Triangle Park, North Carolina; and the Spinhaler powder inhaler, manufactured by Fisons Corp., Bedford, Mass. All such devices require the use of formulations suitable for the dispensing of the compounds of the invention. Typically, each formulation is specific to the type of device employed and may involve the use of an appropriate propellant material, in addition to the usual diluents, adjuvants and/or carriers useful in therapy. Also, the use of liposomes, microcapsules or microspheres, inclusion complexes, or other types of carriers is contemplated. Chemically modified compound of the invention may also be prepared in different formulations depending on the type of chemical modification or the type of device employed. Formulations suitable for use with a nebulizer, either jet or ultrasonic, will typically comprise a compound of the invention (or derivative) dissolved in water at a concentration of about 0.1 to 25 mg of biologically active compound of the invention per mL of solution. The formulation may also include a buffer and a simple sugar (e.g., for inhibitor stabilization and regulation of osmotic pressure). The nebulizer formulation may also contain a surfactant, to reduce or prevent surface induced aggregation of the compound of the invention caused by atomization of the solution in forming the aerosol. Formulations for use with a metered-dose inhaler device will generally comprise a finely divided powder containing the compound of the invention (or derivative) suspended in a propellant with the aid of a surfactant. The propellant may be any conventional material employed for this purpose, such as a chlorofluorocarbon, a hydrochlorofluorocarbon, a hydrofluorocarbon, or a hydrocarbon, including trichlorofluoromethane, dichlorodifluoromethane, dichlorotetrafluoroethanol, and 1,1,1,2-tetrafluoroethane, or combinations thereof. Suitable surfactants include sorbitan trioleate and soya lecithin. Oleic acid may also be useful as a surfactant. Formulations for dispensing from a powder inhaler device will comprise a finely divided dry powder containing compound of the invention (or derivative) and may also include a bulking agent, such as lactose, sorbitol, sucrose, or mannitol in amounts which facilitate dispersal of the powder from the device, e.g., 50 to 90% by weight of the formulation. The compound of the invention (or derivative) should advantageously be prepared in particulate form with an average particle size of less than 10 micrometers ( ^m), most preferably 0.5 to 5 ^m, for most effective delivery to the deep lung. Nasal delivery of a pharmaceutical composition of the present invention is also contemplated. Nasal delivery allows the passage of a pharmaceutical composition of the present invention to the blood stream directly after administering the therapeutic product to the nose, without the necessity for deposition of the product in the lung. Formulations for nasal delivery include those with dextran or cyclodextran. For nasal administration, a useful device is a small, hard bottle to which a metered dose sprayer is attached. In one embodiment, the metered dose is delivered by drawing the pharmaceutical composition of the present invention solution into a chamber of defined volume, which chamber has an aperture dimensioned to aerosolize and aerosol formulation by forming a spray when a liquid in the chamber is compressed. The chamber is compressed to administer the pharmaceutical composition of the present invention. In a specific embodiment, the chamber is a piston arrangement. Such devices are commercially available. Alternatively, a plastic squeeze bottle with an aperture or opening dimensioned to aerosolize an aerosol formulation by forming a spray when squeezed is used. The opening is usually found in the top of the bottle, and the top is generally tapered to partially fit in the nasal passages for efficient administration of the aerosol formulation. Preferably, the nasal inhaler will provide a metered amount of the aerosol formulation, for administration of a measured dose of the drug. The compounds, when it is desirable to deliver them systemically, may be formulated for parenteral administration by injection, e.g., by bolus injection or continuous infusion. Formulations for injection may be presented in unit dosage form, e.g., in ampoules or in multi-dose containers, with an added preservative. The compositions may take such forms as suspensions, solutions or emulsions in oily or aqueous vehicles, and may contain formulatory agents such as suspending, stabilizing and/or dispersing agents. Pharmaceutical formulations for parenteral administration include aqueous solutions of the active compounds in water-soluble form. Additionally, suspensions of the active compounds may be prepared as appropriate oily injection suspensions. Suitable lipophilic solvents or vehicles include fatty oils such as sesame oil, or synthetic fatty acid esters, such as ethyl oleate or triglycerides, or liposomes. Aqueous injection suspensions may contain substances which increase the viscosity of the suspension, such as sodium carboxymethylcellulose, sorbitol, or dextran. Optionally, the suspension may also contain suitable stabilizers or agents which increase the solubility of the compounds to allow for the preparation of highly concentrated solutions.
Alternatively, the active compounds may be in powder form for constitution with a suitable vehicle, e.g., sterile pyrogen-free water, before use.
The compounds may also be formulated in rectal or vaginal compositions such as suppositories or retention enemas, e.g., containing conventional suppository bases such as cocoa butter or other glycerides.
In addition to the formulations described above, a compound may also be formulated as a depot preparation. Such long acting formulations may be formulated with suitable polymeric or hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, or as sparingly soluble derivatives, for example, as a sparingly soluble salt.
The pharmaceutical compositions also may comprise suitable solid or gel phase carriers or excipients. Examples of such carriers or excipients include but are not limited to calcium carbonate, calcium phosphate, various sugars, starches, cellulose derivatives, gelatin, and polymers such as polyethylene glycols.
Suitable liquid or solid pharmaceutical preparation forms are, for example, aqueous or saline solutions for inhalation, microencapsulated, encochleated, coated onto microscopic gold particles, contained in liposomes, nebulized, aerosols, pellets for implantation into the skin, or dried onto a sharp object to be scratched into the skin. The pharmaceutical compositions also include granules, powders, tablets, coated tablets, (micro)capsules, suppositories, syrups, emulsions, suspensions, creams, drops or preparations with protracted release of active compounds, in whose preparation excipients and additives and/or auxiliaries such as disintegrants, binders, coating agents, swelling agents, lubricants, flavorings, sweeteners or solubilizers are customarily used as described above. The pharmaceutical compositions are suitable for use in a variety of drug delivery systems. For a brief review of methods for drug delivery, see Langer R, Science 249:1527-33 (1990).
The compound of the invention and optionally other therapeutics may be administered per se (neat) or in the form of a pharmaceutically acceptable salt or cocrystal. When used in medicine the salts or cocrystals should be pharmaceutically acceptable, but non- pharmaceutically acceptable salts or cocrystals may conveniently be used to prepare pharmaceutically acceptable salts or cocrystals thereof. Such salts include, but are not limited to, those prepared from the following acids: hydrochloric, hydrobromic, sulphuric, nitric, phosphoric, maleic, acetic, salicylic, p -toluene sulphonic, tartaric, citric, methane sulphonic, formic, malonic, succinic, naphthal ene-2- sulphoni c, and benzene sulphonic. Also, such salts can be prepared as alkaline metal or alkaline earth salts, such as sodium, potassium or calcium salts of the carboxylic acid group.
Suitable buffering agents include: acetic acid and a salt (1-2% w/v); citric acid and a salt (1-3% w/v); boric acid and a salt (0.5-2.5% w/v); and phosphoric acid and a salt (0.8-2% w/v). Suitable preservatives include benzalkonium chloride (0.003-0.03% w/v); chlorobutanol (0.3-0.9% w/v); parabens (0.01-0.25% w/v) and thimerosal (0.004-0.02% w/v).
Pharmaceutical compositions of the invention contain an effective amount of a compound as described herein and optionally therapeutic agents included in a pharmaceutically acceptable carrier. The term “pharmaceutically acceptable carrier” means one or more compatible solid or liquid filler, diluents or encapsulating substances which are suitable for administration to a human or other vertebrate animal. The term “carrier” denotes an organic or inorganic ingredient, natural or synthetic, with which the active ingredient is combined to facilitate the application. The components of the pharmaceutical compositions also are capable of being commingled with the compounds of the present invention, and with each other, in a manner such that there is no interaction which would substantially impair the desired pharmaceutical efficiency.
The therapeutic agent(s), including specifically but not limited to a compound of the invention, may be provided in particles. Particles as used herein means nanoparticles or microparticles (or in some instances larger particles) which can consist in whole or in part of the compound of the invention or the other therapeutic agent(s) as described herein. The particles may contain the therapeutic agent(s) in a core surrounded by a coating, including, but not limited to, an enteric coating. The therapeutic agent(s) also may be dispersed throughout the particles. The therapeutic agent(s) also may be adsorbed into the particles.
The particles may be of any order release kinetics, including zero-order release, first-order release, second-order release, delayed release, sustained release, immediate release, and any combination thereof, etc. The particle may include, in addition to the therapeutic agent(s), any of those materials routinely used in the art of pharmacy and medicine, including, but not limited to, erodible, nonerodible, biodegradable, or nonbiodegradable material or combinations thereof. The particles may be microcapsules which contain the compound of the invention in a solution or in a semi-solid state. The particles may be of virtually any shape.
Both non-biodegradable and biodegradable polymeric materials can be used in the manufacture of particles for delivering the therapeutic agent(s). Such polymers may be natural or synthetic polymers. The polymer is selected based on the period of time over which release is desired. Bioadhesive polymers of particular interest include bioerodible hydrogels described in Sawhney H S et al. (1993 ) Macromolecules 26:581-7, the teachings of which are incorporated herein. These include polyhyaluronic acids, casein, gelatin, glutin, polyanhydrides, polyacrylic acid, alginate, chitosan, poly(methyl methacrylates), poly(ethyl methacrylates), poly(butylmethacrylate), poly (isobutyl methacrylate), poly(hexylmethacrylate), poly(isodecyl methacrylate), poly(lauryl methacrylate), poly(phenyl methacrylate), poly(methyl acrylate), poly (isopropyl acrylate), poly (isobutyl acrylate), and poly(octadecyl acrylate).
The therapeutic agent(s) may be contained in controlled release systems. The term “controlled release” is intended to refer to any drug-containing formulation in which the manner and profile of drug release from the formulation are controlled. This refers to immediate as well as non-immediate release formulations, with non-immediate release formulations including but not limited to sustained release and delayed release formulations. The term “sustained release” (also referred to as “extended 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 preferably, although not necessarily, results in substantially constant blood levels of a drug over an extended time period. The term “delayed release” is used in its conventional sense to refer to a drug formulation in which there is a time delay between administration of the formulation and the release of the drug there from. “Delayed release” may or may not involve gradual release of drug over an extended period of time, and thus may or may not be “sustained release.”
Use of a long-term sustained release implant may be particularly suitable for treatment of chronic conditions. “Long-term” release, as used herein, means that the implant is constructed and arranged to deliver therapeutic levels of the active ingredient for at least 7 days, and preferably 30-60 days. Long-term sustained release implants are well-known to those of ordinary skill in the art and include some of the release systems described above.
It will be understood by one of ordinary skill in the relevant arts that other suitable modifications and adaptations to the compositions and methods described herein are readily apparent from the description of the invention contained herein in view of information known to the ordinarily skilled artisan, and may be made without departing from the scope of the invention or any embodiment thereof. Having now described the present invention in detail, the same will be more clearly understood by reference to the following examples, which are included herewith for purposes of illustration only and are not intended to be limiting of the invention. EXAMPLES The invention is further described in the following examples, which do not limit the scope of the invention described in the claims. Example 1: PathHunter MEM-EA Pharmacotrafficking Assay for SLC6A8 CTD mutants Cell Lines Preparation of Cells  U-2 OS MEM-EA cells were purchased from Eurofins (catalog #93-1101C3). From these parental cells, stable cell lines expressing SLC6A8 CTD mutants were made using standard cell culture protocols, involving transfections of plasmids followed by antibiotic selection. These plasmids encoded CTD mutant SLC6A8 proteins with a C-terminal ProLink2 tag. U-2 OS MEM-EA cells and derived stable cell lines were grown in RPMI medium 1640 (Thermo Fisher Scientific, catalog #A10491-01) supplemented with 10% Fetal Bovine Serum (FBS), 200 ug/mL hygromycin B (Thermo Fisher Scientific, catalog #10687010), 100 mg/mL streptomycin, and 100 U/mL penicillin. Cells were grown at 37°C in a humidified CO2 incubator. Assay U-2 OS MEM-EA cells stably expressing SLC6A8 CTD mutants were plated into white-walled 96-well plates (Corning, catalog #3903) at a density of 20,000 cells per well. For background subtraction, the parental U-2 OS MEM-EA cells were also plated. After 24 hrs, compounds were dispensed directly into the plated cells using the Tecan D300e Digital Dispenser. After an additional 24 hrs, the media with compound was again removed and white covers (Thermo Fisher Scientific, catalog #236272) were placed on the bottoms of the 96-well plates. Luminescence indicative of SLC6A8 CTD mutant cell surface localization was measured according to the manufacturer’s protocol, using the PathHunter Detection kit (Eurofins catalog #93-0001L) and an EnVision plate reader (PerkinElmer, 2104 multilabel reader). Data were analyzed in Excel. Background signal from wells containing parental cells was subtracted, and then fold-changes were computed with respect to DMSO. Example 2: Corrector Assay for SLC6A8 CTD mutant Cell Lines Preparation of Cells A number of SLC6A8 CTD mutant cell lines were made in U-2 OS MEM-EA cells, 293T cells, HeLa cells, and CHO cells. All cells lines were generated as described above for U-2 OS MEM-EA cells, namely stable cell lines expressing SLC6A8 CTD mutants were made using standard cell culture protocols involving transfections of plasmids followed by antibiotic selection. Assay Stable cell lines expressing CTD mutants were plated into 96-well plates (Corning, catalog #3595) at a density of 40,000 cells per well. After 24 hrs, compounds were dispensed directly into the plated cells using a Tecan D300e Digital Dispenser. After an additional 24 hrs, the media with compound was removed. Cells were then incubated with a solution of 100 uM D3-creatine (SIGMA, 616249-1G) in media (without FBS). This solution was incubated with the cells for a 30 min incubation at 37°C. After the incubation, the media was removed, and the cells were washed once with 180 uL of phosphate buffered solution (PBS). To extract metabolites, water was added to the cells for 1 hour with vigorous shaking at 700 rpm. Cell extracts were analyzed on an ABSciex-4000 triple quad mass spectrometer coupled with a RapidFire sample desalting/injection system with a graphitic carbon desalting column and a basic buffer system in reverse phase. Abundances of D3- creatine were analyzed in Excel, and then fold-changes were computed with respect to DMSO. Example 3: General procedures for the synthesis of representative compounds of the invention
General procedure 1:
Figure imgf000063_0001
Step 1: Synthesis of compound 2 PBr3 (0.8 eq.) was slowly added at 0oC over 30 minutes to a solution of compound 1 (1 eq.) in anhydrous DCM (0.3 M), and the resulting mixture was stirred at room temperature for 2 hours. The mixture was then poured into a saturated NaHCO3 solution and washed twice with DCM. The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was then purified via flash column chromatography (eluted with PE/EtOAc) to give pure compound 2. Step 2: Synthesis of compound 3 1M TBAF in THF (1.5 eq.) was added dropwise at 0oC over 30 minutes to a mixture of compound 2 (1 eq.) and TMSCN (1.5 eq.) in acetonitrile (0.3M). The resulting mixture was stirred at room temperature for 1-4 hours and monitored by TLC. The mixture was then diluted with water and extracted with EtOAc (x3). The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified via flash column chromatography (eluted with PE/EtOAc) to give pure compound 3. Step 3: Synthesis of compound 4 EtMgBr (2.2 eq., 3 M in Et2O) was added dropwise at -78oC under N2 atmosphere to a solution of Ti(O-iPr)4 (1.2 eq.) in anhydrous THF (0.15M). After stirring at -78oC for 1 hour, a solution of compound 3(1 eq) in anhydrous THF (15M) was added to the above mixture and the resulting mixture was slowly warmed to room temperature (2-4 hours). Boron trifluoride diethyl etherate (1.5-2 eq.) was then added, and the mixture was stirred at room temperature for 1hour. After completion, the mixture was quenched with 1 Maq. HCl solution and extracted twice with MTBE.The aqueous phase was then basified with 1M aq. NaOH to pH>12 and extracted twice with EtOAc. The combined organic layers were dried over anhydrous Na2SO4, filtered, and concentrated under vacuum to give compound 4, which was used for the next step without any further purification. Step 4: Synthesis of compound 6 EDCI (1.2 eq.), HOBt (1.2 eq.) and NMM (3 eq.) were added to a mixture of compound 4 (1 eq.) and compound 5 (1.1 eq.) in DMF (0.2M), and the resulting mixture was stirred at room temperature for 16 hours. The mixture was then diluted with saturated aq. NH4Cl solution and extracted twice with EtOAc. The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified via flash column chromatography (eluted with DCM/MeOH) to give pure compound 6. Step 5: Synthesis of compound 7 The appropriate amine (2 eq.) was added to a mixture of compound 6 (1 eq.) and DIEA (3 eq.) in NMP (0.2M), and the resulting mixture was stirred in a sealed tube at 160oC for 6-10 hours. The mixture was then cooled down, diluted with water, and extracted twice with EtOAc. The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified via prep-HPLC (Gemini 5μm C18 250*21.2mm, H2O/MeCN (5-95 %)/0.1% NH3 .H2O) to give pure compound 7.
Example: Synthesis of M1
Figure imgf000065_0001
Step 1: Synthesis of M1-int-2 PBr3 (2.14 g, 8 mmol) was added dropwise at 0oC over 30 minutes to a solution of M1-int-1 (1.52 g, 10 mmol) in anhydrous DCM (30 mL), and the resulting mixture was stirred at room temperature for 2 hours. The mixture was then poured into saturated NaHCO3 solution (60 mL) and washed twice with DCM (30 mL). The combined organic layers were dried over anhydrous Na2SO4, filtered, and concentrated under vacuum. The residue was purified by flash column chromatography (eluted with PE/EtOAc= 100:0 to 40:1) to give M1-int-2(1.82 g, 84.6% yield) as a light-yellow oil.LC/MS (ESI) m/z: 215 (M+H) +. Step 2: Synthesis of M1-int-3 TBAF (12.7 mL, 1 M in THF) was added dropwise at 0oC over 30 minutes to a mixture of M1- int-2 (1.82 g, 8.46 mmol) and TMSCN (1.27 g, 12.7 mmol) in acetonitrile (30 mL), andthe resulting mixture was stirred at room temperature for 3 hours. The reaction wasthen diluted with water (80 mL) and extracted twice with EtOAc (40 mL). The combined organic layers were dried over anhydrous Na2SO4, filtered, and concentrated under vacuum. The residue was purified by flash column chromatography (eluted with PE/EtOAc= 100:0 to 30:1) to give M1- int-3 (810 mg, 59.5% yield) as a colorless oil. LC/MS (ESI) m/z: 162 (M+H) +. Step 3: Synthesis of M1-int-4 EtMgBr (3.7 mL, 3 M in Et2O) was added dropwise at -78oC under N2 atmosphere to a solution of Ti(O-iPr)4 (1.72 g, 6.04 mmol) in anhydrous THF (40 mL). The reaction mixture was then stirred at -78oC for 1 hour before a solution of M1-int-3(810 mg, 5.03 mmol) in anhydrous THF (3 mL) was added. The resulting mixture was then slowly warmed to room temperature (3 hours). Boron trifluoride diethyl etherate (1.07 g, 7.55 mmol) was then added and the mixture was stirred at room temperature for 1 hour. After completion, the mixture was quenched with 1 M aq. HCl solution (15 mL) and extracted twice with MTBE(40 mL).The aqueous phase was then basified with 1 M aq. NaOH to pH>12 and extracted with EtOAc(40mL x3). The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated under vacuum to give crude M1-int-4(210 mg, ~60% purity, 13.1% yield) as a light-yellow oil without any further purification.LC/MS (ESI) m/z: 192 (M+H) +. Step 4: Synthesis of M1-int-6 EDCI (152 mg, 0.8 mmol), HOBt (108 mg, 0.8 mmol) and NMM (202 mg, 2 mmol) were added to a mixture of M1-int-4 (210 mg, 0.66 mmol) and M1-int-5(114 mg, 0.73 mmol) in DMF (4 mL), and the resulting mixture was stirred at room temperature for 16 hours. The mixture was diluted with saturated aq. NH4Cl (40 mL) and extracted twice with EtOAc (40 mL). The combined organic layers werewashed with brine, dried over anhydrous Na2SO4, filtered, and concentrated under vacuum. The residue was purified by flash column chromatography (eluted with DCM/MeOH= 100:0 to 40:1) to give M1-int-6 (72 mg, 33.0 % yield) as a white solid. LC/MS (ESI) m/z: 331 (M+H) +. Step 5: Synthesis of M1 Cis-2,6-dimethylmorpholine (51 mg, 0.44 mmol) was added to a mixture of M1-int-6 (72 mg, 0.22 mmol) and DIEA (86 mg, 0.66 mmol) in NMP (3 mL), and the resulting mixture was stirred in a sealed tube at 160oC for 8 hours. The reaction mixture was then cooled down, diluted with water (50 mL), and extracted twice with EtOAc (40 mL). The combined organic layers were dried over anhydrous Na2SO4, filtered, and concentrated under vacuum. The residue was purified via prep-HPLC (Gemini 5μm C18 250*21.2mm, H2O/MeCN (5-95 %)/0.1% NH3.H2O) to give M1 (14 mg, 15.5 % yield) as a white solid. LC/MS (ESI) m/z: 410 (M+H) +.1H NMR (400 MHz, DMSO-d6) δ 13.65 (br s, 1H), 9.40 (s, 1H), 8.21 (d, J = 7.3 Hz, 1H), 7.68 (d, J = 2.4 Hz, 1H), 7.31 – 7.25 (m, 1H), 7.04 – 6.96 (m, 2H), 6.81 (d, J = 8.3 Hz, 1H), 4.18 (d, J = 13.1 Hz, 2H), 3.66 – 3.59 (m, 2H), 3.56 (s, 3H), 2.84 – 2.74 (m, 2H), 2.20 (s, 3H), 1.17 (d, J = 6.2 Hz, 6H), 0.86 (t, J = 5.9 Hz, 2H), 0.80 (t, J = 5.7 Hz, 2H). The compounds in the tables below were prepared from the appropriate starting materials, described above or commercially available, using the above general procedure.
Figure imgf000067_0001
Figure imgf000068_0001
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
Figure imgf000072_0001
Figure imgf000073_0001
Figure imgf000074_0001
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
Figure imgf000080_0001
Figure imgf000081_0001
Figure imgf000082_0001
Figure imgf000083_0001
Figure imgf000084_0001
Figure imgf000085_0001
Figure imgf000086_0001
Figure imgf000087_0001
Figure imgf000088_0001
Figure imgf000089_0001
Figure imgf000090_0001
Figure imgf000091_0001
Figure imgf000092_0001
Figure imgf000093_0002
General procedure 2
Figure imgf000093_0001
Step 1: Synthesis of compound 2 The appropriate secondary amine (2 eq.) was added to a mixture of compound 1 (1 eq.) and DIEA (3 eq.) in NMP (0.3M), and the resulting mixture was stirred in a sealed tube at 120oC for 16 hours. The mixture was then cooled down, diluted with water and extracted twice with EtOAc. The combined organic layers were dried over anhydrous Na2S04, filtered and concentrated under vacuum. The residue was purified by column chromatography on silica gel (eluted with DCM/MeOH = 100: 1 to 30:1) to give compound 2.
Step 2: Synthesis of compound 5 Method A:
PBn (2 eq.) was added dr op wise at 0°C over 30 minutes to a solution of compound 3 (1 eq.) in anhydrous DCM (0.2M), and the resulting mixture was stirred at room temperature for 2-6 hours. The mixture was then poured into saturated aq. NaHCCb solution and washed twice with DCM. The combined organic layers were dried over anhydrous NaiSCri, filtered and concentrated under vacuum. The residue was purified by flash column chromatography (eluted with PE/EtOAc) to give pure compound 5.
Method B:
MBS (1.05 eq.) was added to a mixture of compound 4 (1 eq.) and AIBN (0.1 eq.) in CCU, and the resulting mixture was refluxed for 16 hours under N2 atmosphere. The reaction mixture was then cooled down, diluted with DCM and filtered. The filtrate was evaporated under vacuum and the residue was purified by flash column chromatography (eluted with PE/EtOAc) to give pure compound 5.
Step 3: Synthesis of compound 6
1M TBAF in THF (1.5 eq.) was added dropwise at 0°C over 30 minutes to a mixture of compound 5 (1 eq.) and TMSCN (1.5 eq.) in acetonitrile (0.15M), and the resulting mixture was then stirred at room temperature for 14 hours. The mixture was diluted with water and extracted twice with EtOAc. The combined organic layers were dried over anhydrous NaiSCri, filtered, and concentrated under vacuum. The residue was purified by flash column chromatography (eluted with PE/EtOAc) to give pure compound 6.
Step 4: Synthesis of compound 7
EtMgBr (2.2-5.8 eq., 3 M in EtiO) was added dropwise at -78°C under N2 atmosphere to a solution of Ti(0-/Pr)4 (1.2-3 eq.) in anhydrous THE (0.25M). The reaction mixture was stirred at -78°C for 1 hour before a solution of compound 6 (1 eq.) in anhydrous THF (0.5M) was added. The resulting mixture was slowly warmed to room temperature (2-4 hours), and BF3- Et2O (1.5 ~ 2 eq.) was added. The reaction mixture was then stirred at room temperature for 1 hour. After completion, the mixture was quenched with 1 M aq. HCl and extracted twice with MTBE. The aqueous phase was then basified with 1 M aq. NaOH to pH>12 and extracted with EtOAc (x3). The combined organic layers were dried over anhydrous Na2SO4, filtered, and concentrated under vacuum to give crude compound 7, which was used for next step without any further purification. Step 5: Synthesis of compound 8 EDCI (1.2 eq), HOBt (1.2 eq.) and NMM (3 eq) were added to a mixture of compound 7 (1 eq.) and compound 2 (1.1 eq.) in DMF (0.1M), and the resulting mixture was stirred at room temperature for 16 hours. The mixture was then diluted with saturated aq. NH4Cl and extracted twice with EtOAc. The combined organic layers were dried over anhydrous Na2SO4, filtered, and concentrated under vacuum. The residue was purified via prep-HPLC (Gemini 5μm C18 250*21.2mm, H2O/MeCN (5-95 %)/0.1% NH3.H2O) to give pure compound 8. Example 1: Synthesis of M2
Figure imgf000095_0001
Step 1: Synthesis of M2-int-2 Cis-2,6-dimethylmorpholine (23 g, 200 mmol) was added to a mixture of M2-int-1 (15.7 g, 100 mmol) and DIEA (39 g, 300 mmol) in NMP (300 mL), and the resulting mixture was stirred in a sealed tube at 120oC for 16 hours. The mixture was then cooled down, diluted with water (2L) and extracted twice with EtOAc (1L). The combined organic layers were dried over anhydrous Na2S04, filtered and concentrated under vacuum. The residue was purified by column chromatography on silica gel (eluted with DCM/MeOH = 100:1 to 30:1) to give M2- int-2 (12.1 g, 51.3 % yield) as a white solid. LC/MS (ESI) m/z: 235 (M-H) +.
Step 1: Synthesis of M2-int-4
PBn (1.1 mL, 10.78 mmol) was added dropwise at 0°C over 30 minutes to a solution of M2- int-3 (750 mg, 5.39 mmol) in anhydrous DCM (30 mL), and the resulting mixture was stirred at room temperature for 6 hours. The mixture was then poured into a saturated aq. NaHCCb solution (60 mL) and washed twice with DCM (30 mL). The combined organic layers were dried over anhydrous NaiSCL, filtered, and concentrated under vacuum. The residue was purified by flash column chromatography (eluted with PE/EtOAc= 100:0 to 40:1) to give pure M2-int-4 (720 mg, 66.5 % yield) as a light-yellow oil. LC/MS (ESI) m/z: 202 (M+H) +.
Step 2: Synthesis of M2-int-5
TBAF (7 mL, 1 M in THF) was added dropwise at 0°C over 30 minutes to a mixture of M2- int-4 (700 mg, 3.48 mmol) and TMSCN (690 mg, 6.97 mmol) in acetonitrile (25 mL), and the reaction mixture was stirred at room temperature for 2 hours. The mixture was then diluted with water and extracted twice with EtOAc (40 mL). The combined organic layers were dried over anhydrous Na2$04, filtered, and concentrated under vacuum. The residue was purified by flash column chromatography (eluted with PE/EtOAc= 30:1 to 4:1) to give M2-int-5 (430 mg, 83.5 % yield) as a light-yellow solid. LC/MS (ESI) m/z: 149 (M+H) +.
Step 3: Synthesis of M2-int-6
EtMgBr (4.5 mL, 3 M in EtiO) was added dropwise at -78°C under N2 atmosphere to a solution of Ti(0-/Pr)4 (0.79 mL, 7.25 mmol) in anhydrous THF (30 mL). The reaction mixture was stirred at -78°C for 1 hour before a solution of M2-int-5 (430 mg, 2.9 mmol) in anhydrous THF (5 mL) was added. The resulting mixture was slowly warmed to room temperature over 3 hours, then BF3-Et20 (618 mg, 4.35 mmol) was added, and the mixture was stirred at room temperature for 1 hour. After completion, the mixture was quenched with 1 M aq. HC1 (10 mL) and extracted twice with MTBE (50 mL). The aqueous phase was then basified with 1 M aq. NaOH to pH>12 and extracted with EtOAc (40 mL x3). The combined organic layers were dried over anhydrous Na2$04, filtered, and concentrated under vacuum to give crude M2-int- 6(60 mg, -50% purity, 5.8 % yield) as a light-yellow oil without any further purification. LC/MS (ESI) m/z: 179 (M+H) +. Step 4: Synthesis of M2 EDCI (53 mg, 0.27 mmol), HOBt (37 mg, 0.27 mmol) and NMM (51 mg, 0.5 mmol) were added to a mixture of M2-int-6 (60 mg, 0.17 mmol) and M2-int-2 (44 mg, 0.19 mmol) in DMF (2 mL), andthe resulting mixture was stirred at room temperature for 16 hours. The mixture was then diluted with saturated aq. NH4Cl (30 mL) and extracted twice with EtOAc (30 mL). The combined organic layers were dried over anhydrous Na2SO4, filtered, and concentrated under vacuum. The residue was purified via prep-HPLC (Gemini 5μm C18 250*21.2mm, H2O/MeCN (5-95 %)/0.1% NH3 .H2O) to give M2 (8 mg, 11.9 % yield) as a white solid. LC/MS (ESI) m/z: 397 (M+H)+.1H NMR (400 MHz, DMSO-d6) δ 8.61 (s, 1H), 8.12 (d, J = 5.9 Hz, 1H), 7.57 (dd, J = 8.2, 7.3 Hz, 1H), 7.45 (d, J = 2.6 Hz, 1H), 6.97 (dd, J = 6.0, 2.7 Hz, 1H), 6.84 (d, J = 7.0 Hz, 1H), 6.61 (d, J = 8.0 Hz, 1H), 3.86 - 3.82 (m, 2H), 3.75 (s, 3H), 3.66 - 3.60 (m, 2H), 2.99 (s, 2H), 2.46 - 2.40 (m, 2H), 1.17 (d, J = 6.2 Hz, 6H), 0.91 – 0.82 (m, 4H). The compounds in the table below were prepared from the appropriate starting materials, described previously or commercially available, using the above general procedure.
Figure imgf000097_0001
Figure imgf000098_0001
Figure imgf000099_0001
Figure imgf000100_0001
Figure imgf000101_0001
Figure imgf000102_0001
Figure imgf000103_0001
Figure imgf000104_0001
Figure imgf000105_0001
Figure imgf000106_0001
Step 1: Synthesis of compound 2 n-BuLi (1.2 eq., 1.6 M in hexane) was added dropwise at -78°C under N2 atmosphere to a solution of compound 1 (1 eq.) in anhydrous THF (0.1M). After stirring at-78°C for 1 hour, BF3 Et20 (2 eq.) and the corresponding epoxide (2 eq.) were added to the above mixture. The resulting mixture was then stirred at -78°C for an additional hour, quenched with iced saturated aq. NHrCl, and extracted twice with EtOAc. The combined organic layers were washed with brine, dried over anhydrous Na2SOr, filtered, and concentrated under vacuum. The residue was purified by column chromatography on silica gel (eluted with PE/EtOAc) to give pure compound 2.
Step 2: Synthesis of compound 3
MsCI (1.5 eq.) was added at 0°C under N2 atmosphere to a mixture of compound 2 (1 eq.) and DIEA (3 eq.) in anhydrous DCM (0.15M), and the resulting mixture was then allowed to warm to room temperature over 2 hours. The reaction was then quenched with ice/ThO, and extracted twice with DCM. The combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered, and concentrated under vacuum to give compound 3, which was used for next step without any further purification. Step 3: Synthesis of compound 4 NaN3 (2 eq.) was added to a solution of compound 3 (1 eq.) in DMF (0.2M), and the resulting mixture was stirred at 60°C for 8 hours. The mixture was then diluted with H2O and extracted twice with EtOAc. The combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered and concentrated under vacuum to give compound 4, which was used for next reaction without any further purification. Step 4: Synthesis of compound 5 Pd/C (10%, w/w) was added to a solution of compound 4 (1 eq.) in EtOAc(0.05M), and the resulting mixture was stirred under 25 psi H2 atmosphere for 16 hours at room temperature. The mixture was then diluted with EtOAc, filtered and concentrated under vacuum. The residue was purified by column chromatography on silica gel (eluted with PE/EtOAc) to give pure compound 5. Step 5: Synthesis of compound 7 EDCI (1.2 eq.), HOBt (1.3 eq.) and NMM (3 eq.) were added under N2 atmosphere to a mixture of compound 5 (1 eq.) and compound 6 (1.1 eq.) in DMF (0.1M), and the resulting mixture was stirred at room temperature for 8 hours. The mixture was then diluted with saturated aq. NH4Cl and extracted twice with EtOAc. The combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified via prep-HPLC (Gemini 5μm C18 250*21.2mm, H2O/MeCN (5-95 %)/0.1% NH3 .H2O) to give pure compound 7.
Example: Synthesis of M3
Figure imgf000108_0001
Step 1: Synthesis of M3-int-2 n-BuLi (3.8 mL, 1.6 M in hexane) was slowly added at -78°C under N2 atmosphere to a solution of 1-bromo-2-chlorobenzene (1 g, 5.29 mmol) in anhydrous THF (50 mL). The mixture was stirred at -78°C for 1 hour, then BF3 .Et2O (3mL, 10.58 mmol) and (R)-2-methyloxirane (614 mg, 10.58 mmol) were added. The resulting mixture was stirred at -78°C for an additional hour. The mixture was then quenched with iced saturated aq. NH4Cl and extracted twice with EtOAc. The combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered, and concentrated under vacuum. The residue was purified by column chromatography on silica gel (eluted with PE: EtOAc= 30:1 to 5:1) to give M3-int-2 (520 mg, 57.8 % yield) as a yellow oil. LC/MS (ESI) m/z: 171 (M+H)+. Step 2: Synthesis of M3-int-3 MsCl (528 mg, 4.6 mmol) was added at 0°C under N2 atmosphere to a mixture of M3-int-2 (520 mg, 3.06 mmol) and DIEA (1.2 g, 9.18 mmol) in anhydrous DCM (20 mL), and the resulting mixture was allowed to warm to room temperature over 1 hour. The mixture was then quenched with saturated aq. NaHCO3 and extracted twice with DCM. The combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered, and concentrated under vacuum to give crude M3-int-3 (660 mg, 87.0 % yield) as a yellow oil without any further purification. LC/MS (ESI) m/z: 249 (M+H)+. Step 3: Synthesis of M3-int-4 NaN3 (278 mg, 4.26 mmol) was added to a solution of M3-int-3 (660 mg, 2.66 mmol) in DMF (15 mL) and the resulting mixture was stirred at 60°C for 6 hours The reaction mixture was then cooled down, diluted with H2O, and extracted twice with EtOAc. The combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered and concentrated under vacuum to give M3-int-4 (220 mg, 42.4 % yield) as a yellow oil without any further purification. LC/MS (ESI) m/z: 196 (M+H)+. Step 4: Synthesis of M3-int-5 Pd/C (22 mg) was added to a solution of M3-int-4 (220 mg, 1.13 mmol) in EtOAc (15 mL), and the resulting mixture was stirred under 25 psi H2 atmosphere at room temperature for 16 hours. The mixture was then diluted with EtOAc (20 mL), filtered, and concentrated under vacuum. The residue was purified by column chromatography on silica gel (PE/EtOAc= 80:1 to 40:1) to give M3-int-5(90 mg, 47.1 % yield) as a yellow oil. LC/MS (ESI) m/z: 170 (M+H)+. Step 5: Synthesis of M3 EDCI (122 mg, 0.64 mmol), HOBt (87 mg, 0.64 mmol) and NMM (160 mg, 1.6 mmol) were added under N2 atmosphere to a mixture of M3-int-5 (90 mg, 0.53 mmol) and M3-int-6 (138 mg, 0.59 mmol) in DMF (4 mL), and the resulting mixture was stirred at room temperature for 8 hours. The mixture was then diluted with saturated aq. NH4Cl (20 mL) and extracted twice with EtOAc (20 mL). The combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered, and concentrated under vacuum. The residue was purified via prep-HPLC (Gemini 5μm C18250*21.2mm, H2O/MeCN (5-95 %)/0.1% NH3.H2O) to give M3 (22 mg, 10.7 % yield) as a white solid.LC/MS (ESI) m/z: 388 (M+H)+.1H NMR (400 MHz, DMSO-d6) δ 8.67 (s, 1H), 8.19 (d, J = 6.2 Hz, 1H), 7.45 (s, 1H), 7.41 – 7.38 (m, 1H), 7.36 – 7.32 (m, 1H), 7.23 – 7.20 (m, 2H), 7.08 – 6.99 (m, 1H), 4.41 – 4.33 (m, 1H), 3.95 – 3.83 (m, 2H), 3.66 – 3.61 (m, 2H), 3.49 – 3.45 (m, 2H), 3.06 – 2.94 (m, 2H), 1.22 (d, J = 6.6 Hz, 3H), 1.17 (d, J = 6.2 Hz, 6H). The compounds in the table below were prepared from the appropriate starting materials, described previously or commercially available, using the general procedure described above.
Figure imgf000109_0001
Figure imgf000110_0002
General procedure 4
Figure imgf000110_0001
Step 1: Synthesis of compound 3 The appropriate compound 1 (1 eq.) and 4A MS (100 %, w/w) were added to a solution of compound 2 (1 eq.) in anhydrous DCM, and the resulting mixture was stirred under N2 atmosphere at room temperature overnight. The mixture was then diluted with DCM, filtered, and the filtrate was concentrated under vacuum to give the crude compound 3, which was used for next step without any further purification. Step 2: Synthesis of compound 5 The appropriate Grignard reagent (2 eq.) was added dropwise at 0oC under N2 atmosphere to a mixture of compound 3 (1 eq.) and BF3 .Et2O (2eq.) in anhydrous THF. The resulting mixture was stirred at 0oC for 30 minutes and then warmed to room temperature over 2 hours. The mixture was then poured into iced aq. NH4Cl and extracted twice with EtOAc. The combined organic layers were washed with brine, dried over Na2SO4, filtered and concentrated under vacuum. The residue was purified by column chromatography on silica gel (eluted with petroleum ether/EtOAc) to give compound 5. Step 3: Synthesis of compound 6 Pd/C (15%, w/w) was added to a solution of compound 5 (1 eq.) in MeOH, and the resulting mixture was stirred under 25 psi H2 atmosphere at room temperature for 16 hours. The mixture was then filtered, and the filtrate was concentrated under vacuum to give crude compound 6, which was used for next step without any further purification. Step 4: Synthesis of compound 8 EDCI (2 eq.), HOBt (2 eq.) and NMM (3 eq.) were added to a mixture of compound 6 (1 eq.) and compound 7 (1.1 eq.) in DMF, and the resulting mixture was stirred at room temperature for 16 hours. The mixture was then diluted with saturated aq. NaHCO3 and extracted twice with EtOAc. The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified via prep-HPLC (Gemini 5μm C18 250*21.2mm, H2O/MeCN (5-95 %)/0.1% NH3.H2O) to give compound 8.
Example: Synthesis of M4
Figure imgf000112_0001
Step 1: Synthesis of N-benzyloxetan-3-imine (M4-int-2) Oxetan-3-one (1 g, 13.88 mmol) and 4A MS (1 g) were added to a solution of benzylamine (1.48 g, 13.88 mmol) in anhydrous DCM (25 mL), and the resulting mixture was stirred under N2 atmosphere at room temperature overnight. The mixture was then diluted with DCM, filtered, and the filtrate was concentrated under vacuum to give crude M4-int-2 (1.1 g, 49.2 % yield) as a colorless oil, which was used for next step without any further purification. Step 2: Synthesis of N,3-dibenzyloxetan-3-amine (M4-int-3) Benzyl magnesium bromide (3.7 mL, 1 M in THF) was added dropwise at 0º C under N2 atmosphere to a solution of M4-int-2 (300 mg, 1.86 mmol) and BF3.Et2O (529 mg, 3.72 mmol) in THF (20 mL). The resulting mixture was then stirred at 0oC for 30 minutes and warmed to room temperature over 2 hours. The mixture was then poured into iced aq. NH4Cl (30 mL) and extracted twice with EtOAc (30 mL). The combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified by column chromatography on silica gel (eluted with petroleum ether/EtOAc=10: 1 to 5: 1) to give M4-int-3 (400 mg, 85.0 % yield) as a colorless oil.LC/MS (ESI) m/z: 254 (M+H)+. Step 3: Synthesis of 3-benzyloxetan-3-amine (M4-int-4) Pd/C (30 mg) was added to a solution of N,3-dibenzyloxetan-3-amine (200 mg, 0.47 mmol) in MeOH (10 mL), and the resulting mixture was stirred under 25 psi H2 atmosphere at room temperature for 16 hours. The mixture was then filtered and the filtrate was concentrated under vacuum to give crude M4-int-4 (50 mg, 65.3 % yield) as a yellow oil, which was used for next step without any further purification. LC/MS (ESI) m/z: 164 (M+H)+. Step 4: Synthesis of N-(3-benzyloxetan-3-yl)-4-((2S,6R)-2,6-dimethylmorpholino) picolinamide (M4) EDCI (76 mg, 0.49 mmol), HOBt (66 mg, 0.49 mmol) and NMM (74 mg, 0.75mmol) were added to a mixture of 3-benzyloxetan-3-amine (40 mg, 0.25 mmol) and M4-int-5 (69 mg, 0.29 mmol) in DMF (8 mL), and the resulting mixture was stirred at room temperature for 16 hours. The mixture was then diluted with saturated aq. NH4Cl (30 mL) and extracted twice with EtOAc (30 mL). The combined organic layerswere dried over Na2SO4, filtered and concentrated under vacuum. The residue was purified via prep-HPLC (Gemini 5μm C18 250*21.2mm, H2O/MeCN (5-95 %)/0.1% NH3 .H2O) to give M4 (10 mg, 10.5 % yield) as a white solid.LC/MS (ESI) m/z: 382 (M+H)+.1H NMR (400 MHz, CD3OD) δ 8.17 (d, J = 5.9 Hz, 1H), 7.52 (d, J = 2.6 Hz, 1H), 7.27 – 7.12 (m, 5H), 6.93 (dd, J = 5.9, 2.7 Hz, 1H), 4.79 (d, J = 6.7 Hz, 2H), 4.74 (d, J = 6.8 Hz, 2H), 3.82 (d, J = 12.6 Hz, 2H), 3.77 – 3.68 (m, 2H), 3.42 (s, 2H), 2.56 - 2.50 (m, 2H), 1.25 (d, J = 6.2 Hz, 6H). The compound in the table below was prepared from the appropriate starting materials, described previously or commercially available, using the general procedure described above.
Figure imgf000113_0001
General procedure 5
Figure imgf000114_0001
Step 1: Synthesis of 2-(5-fluoro-2-methoxyphenyl)acetonitrile (2) TMSCN (6.75 g, 68 mmol) and TBAF (68 mL, 1 M in THF) were added at room temperature to a mixture of compound 1 (10 g, 45.6 mmol) in MeCN (100 mL), and the resulting mixture was stirred at 80oC for 16 hours. The reaction was then cooled down, poured into water (200 mL) and extracted twice with EtOAc (150 mL). The combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified by column chromatography on silica gel (PE/EtOAc= 30: 1 to 5: 1) to give compound 2 (4.0 g, 53.2 % yield) as a white solid. LC/MS (ESI) m/z: 166 (M+H)+. Step 2: Synthesis of 1-(5-fluoro-2-methoxybenzyl)cyclopropan-1-amine (3) Ethylmagnesium bromide (16 mL, 3M in ether) was slowly added at -60oC under N2 atmosphere to a stirred mixture of 2-(5-fluoro-2-methoxyphenyl)acetonitrile (4 g, 24 mmol) and Ti(Oi-Pr)4 (7.8 g, 27 mmol) in anhydrous THF (40 mL). After stirring from -60oC to 0oC over 3 hours, BF3-Et2O (6.82 g, 48 mmol) was added, and the resulting mixture was stirred for additional 2 hours. The mixture was then poured into 1 N aq. NaOH (20 mL) and extracted twice with EtOAc (20 mL). The combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered and concentrated under vacuum to give crude compound 3 (1.2 g, 25.6 % yield) as a yellow oil, which was used for next step without any further purification. LC/MS (ESI) m/z: 196 (M+H)+. Step 3: Synthesis of compound 4 EDCI (1.2 eq), HOBt (1.2 eq) and NMM (3.0 eq) were added under N2 atmosphere to a mixture of 1-(5-fluoro-2-methoxybenzyl)cyclopropan-1-amine (1.0 eq) and the appropriate acid (1.1 eq.) in DMF, and the reaction was stirred at room temperature overnight. The reaction was then diluted with saturated aq. NH4Cl and extracted twice with EtOAc. The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified via prep-HPLC (Gemini 5μm C18 250*21.2mm, H2O/MeCN (5-95 %)/0.1% NH3.H2O) to give compound 4. Example: Synthesis of M5
Figure imgf000115_0001
Step 1: Synthesis of M5-int-2 NaH (240 mg, 6 mmol) was added at 0oC over 30 minutes to a solution of M5-int-1 (704 mg, 4 mmol) in anhydrous DMF (25 mL), and the reaction mixture was stirred at 0oC for 45 minutes. MeI (682 mg, 4.8 mmol) was then added to the above mixture and the resulting mixture was stirred at room temperature for 16 hours. The mixture was then quenched with saturated aq. NH4Cl (50 mL) and extracted twice with EtOAc (50 mL). The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified by column chromatography (eluted with PE/EtOAc= 40:1 to 2:1) to give M5-int- 2 (670 mg, 88.2 % yield) as a yellow solid. LC/MS (ESI) m/z: 191 (M+H) +. Step 2: Synthesis of M5-int-3 A solution of NaOH (240 mg, 6 mmol) in H2O (12 mL) was added at 0oC over 15 minutes to a solution of M5-int-2 (380 mg, 2 mmol) in MeOH (12 mL), and the resulting mixture was stirred at room temperature for 16 hours. The mixture was then diluted with water (15 mL) and extracted twice with MTBE (50 mL). The aqueous layer was separated, acidified with 1N aq. HCl to pH=5 and extracted with EtOAc. The organic layer was dried over anhydrous Na2SO4, filtered and concentrated under vacuum to give crude M5-int-3 (310 mg, 88.6 % yield) as a yellow solid, which was used for next step without any further purification. LC/MS (ESI) m/z: 175 (M-H)+. Step 3: Synthesis of M5 EDCI (92 mg, 0.48 mmol), HOBt (65 mg, 0.48 mmol) and NMM (122 mg, 1.2 mmol) were added to a mixture of M5-int-4 (78 mg, 0.44 mmol) and M5-int-3 (78 mg, 0.4 mmol) in DMF (8 mL), and the resulting mixture was stirred at room temperature for 16 hours. The mixture was then diluted with saturated aq. NH4Cl (30 mL) and extracted twice with EtOAc (30 mL). The combined organic layers were dried over Na2SO4, filtered and concentrated under vacuum. The residue was purified via prep-HPLC (Gemini 5μm C18250*21.2mm, H2O/MeCN (5-95 %)/0.1% NH3.H2O) to give M5 (23 mg, 16.3 % yield) as a white solid.LC/MS (ESI) m/z: 354 (M+H)+.1H NMR (400 MHz, CDCl3) δ 8.74 (s, 1H), 8.43 (s, 1H), 7.31 (s, 1H), 6.90 – 6.86 (m, 1H), 6.84 – 6.72 (m, 2H), 6.67 – 6.63 (m, 1H), 3.91 (s, 3H), 3.62 (s, 3H), 3.07 (s, 2H), 0.97 - 0.93 (m, 2H), 0.87 – 0.83 (m, 2H). The compounds in table below were prepared from the appropriate starting materials, described previously or commercially available, using the general procedure described above.
Figure imgf000116_0001
Figure imgf000117_0001
General procedure 6
Figure imgf000118_0001
Step 1: Synthesis of compound 2 Compound 2 was prepared from compound 1 using Suzuki or Ullmann reactions. Method A: K2CO3 (2.5 eq.) and Pd(PPh3)4 (0.08 eq.) were added to a mixture of compound 1 (1 eq.) and the appropriate boronic acid (1.3 eq.) in 1,4-dioxane/H2O (9:1, V/V), and the resulting mixture was stirred at 90oC under N2 atmosphere for 16 hours. The mixture was then cooled to room temperature and diluted with EtOAc. The mixture was filtered and the filtrate was washed with saturated aq. NaHCO3 and brine. The organic layer was then dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified by flash column chromatography (eluted with PE/EtOAc) to give compound 2. Method B: K2CO3 (2.5 eq.), L-proline (0.1 eq.) and CuI (0.1 eq.) were added to a mixture of compound 1 (1 eq.) and the corresponding heterocycle (1.3 eq.) in DMSO, and the resulting mixture was stirred at 100oC under N2 atmosphere for 16 hours. The mixture was then cooled to room temperature and diluted with EtOAc. The mixture was filtered and the filtrate was washed with saturated aq. NaHCO3. The organic layer was then dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified by flash column chromatography (eluted with PE/EtOAc) to give compound 2. Step 2: Synthesis of compound 4 AlMe3 (1 eq., 2M in hexane) was slowly added at 0 ºC under N2 atmosphere to a mixture of compound 2 (1 eq.) and compound 3 (1.5 eq.) in anhydrous toluene, and the resulting mixture was stirred at 80oC under N2 atmosphere for 16 hours. The mixture was then cooled to room temperature, quenched with saturated aq. NaHCO3 and extracted twice with EtOAc. The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified via prep-HPLC (Gemini 5μm C18 250*21.2mm, H2O/MeCN (5-95 %)/0.1% NH3 .H2O) to give compound 4. Example: Synthesis of M6
Figure imgf000119_0001
Step 1: Synthesis of M6-int-2 K2CO3 (1.73 g, 12.5 mmol) and Pd(PPh3)4 (462 mg, 0.4 mmol) were added to a mixture of methyl 4-bromopicolinate (1.08 g, 5 mmol) and phenylboronic acid (793 mg, 6.5 mmol) in1,4- dioxane/H2O (30 mL, 9:1, V/V), and the resulting mixture was stirred at 90oC under N2 atmosphere for 16 hours. The mixture was then cooled to room temperature and diluted with EtOAc (30 mL). The mixture was filtered and the filtrate was washed with saturated aq. NaHCO3 (30 mL) and brine. The organic layer was then dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified by flash column chromatography (eluted with PE: EA= 20:1 to 5:1) to give compound M6-int-2 (780 mg, 73.2 % yield) as a white solid. LC/MS (ESI) m/z: 214 (M+H)+. Step 2: Synthesis of M6 AlMe3 (1.9 mL, 2M in hexane) was slowly added at 0oC under N2 atmosphere to a mixture of M6-int-2 (780 mg, 3.66 mmol) and M6-int-3 (1.07 g, 5.5 mmol) in anhydrous toluene (40 mL), and the resulting mixture was stirred at 80oC under N2 atmosphere for 16 hours. The mixture was then cooled to room temperature, quenched with saturated aq. NaHCO3 (40 mL) and extracted twice with EtOAc (30 mL). The combined organic layerswere dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified via prep- HPLC (Gemini 5μm C18250*21.2mm, H2O/MeCN (5-95 %)/0.1% NH3.H2O) to give M6 (86 mg, 6.3 % yield) as a white solid. LC/MS (ESI) m/z: 377 (M+H)+.1H NMR (400 MHz, CDCl3) δ 8.50 (d, J = 5.1 Hz, 1H), 8.47 (d, J = 1.3 Hz, 1H), 8.26 (s, 1H), 7.74 – 7.70 (m, 2H), 7.62 (dd, J = 5.1, 1.9 Hz, 1H), 7.54 – 7.43 (m, 3H), 6.93 – 6.83 (m, 2H), 6.74 (dd, J = 8.9, 4.5 Hz, 1H), 3.65 (s, 3H), 3.08 (s, 2H), 0.96 – 0.94 (m, 2H), 0.94 – 0.92 (m, 2H). The compounds in the table below were prepared from the appropriate starting materials, described previously or commercially available, using the general procedure described above.
Figure imgf000120_0001
Figure imgf000121_0002
General procedure 7
Figure imgf000121_0001
Step 1: Synthesis of compound 3 KHMDS (1.54 eq.) was added at 0oC under N2 atmosphere to a solution of compound 2 (1.1 eq.) in anhydrous toluene, and the reaction was stirred at 0oC for 2 hours before the appropriate compound 1 (1 eq.) was added. The resulting mixture was stirred at room temperature under N2 atmosphere overnight. The mixture was then poured into aq. NH4Cl and extracted with EtOAc. The organic layer was dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified via flash column chromatography (eluted with PE: EtOAc) to give compound 3. Step 2: Synthesis of compound 4 Hydroxylamine hydrochloride (2 eq.) and TEA (3 eq.) were added at 0oC to a mixture of compound 3 (1 eq.) in EtOH, and the resulting mixture was stirred at 75oC for 18 hours. The mixture was then diluted with water and extracted with EtOAc. The organic layer was washed with brine, dried over Na2SO4, filtered and concentrated under vacuumto give crude compound 4 without any further purification. Step 3: Synthesis of compound 6 CDI (1.5 eq.) was added to a mixture of compound 4 (1.0 eq.) and compound 5 (1.0 eq.) in pyridine, and the resulting mixture was stirred at 100oC for 3 hours. The reaction mixture was cooled to room temperature, diluted with ethyl acetate and washed with water. The organic layer was dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified via prep-HPLC (Gemini 5μm C18250*21.2mm, H2O/MeCN (5-95 %)/0.1% NH3.H2O) to give compound 6. Example: Synthesis of M7
Figure imgf000122_0001
Step 1: Synthesis of 1-benzylcyclopropane-l-carbonitrile (M7-int-01)
KHMDS (128 mL, 1 M in THF) was added at 0 °C under N2 atmosphere to a solution of cyclopropanecarbonitrile (6.1 g, 90.95 mmol) in anhydrous toluene (150 mL), and the reaction was stirred at 0°C for 2 hours before benzyl bromide (14 g, 81.85 mmol) was added. The resulting mixture was stirred at room temperature under N2 atmosphere overnight. The mixture was then poured into aq. NLLCl (100 mL) and extracted with EtOAc (100 mL). The organic layer was separated, dried over anhydrous Na2$04, filtered and concentrated under vacuum. The residue was purified via flash column chromatography (eluted with PE/EtOAc= 100:0 to 20:1) to give 1 -benzylcyclopropane- 1 -carbonitrile (10.5 g, 81.7 % yield) as a colorless oil. LC/MS (ESI) m/z: 158(M+H)+.
Step 2: Synthesis of N-hydroxy-l-[(2-methoxyphenyl)methyl]cyclopropane-l- carboximidamide (M7-int-02)
Hydroxylamine hydrochloride (1.78g, 25.64 mmol) and TEA (5.345 mL, 38.45 mmol) were added at 0°C to a mixture of 1 -[(2-methoxyphenyl)methyl]cyclopropane- 1 -carbonitrile (2.4 g, 12.82 mmol) in EtOH (60 mL), and the resulting mixture was stirred at 75°C for 18 hours. The mixture was then diluted with water (50 mL) and extracted with EtOAc (50 mL). The organic layer was washed with brine, dried over anhydrous NaiSCri, filtered and concentrated under vacuum to give crude N-hydroxy- 1 -[(2-methoxyphenyl)methyl]cyclopropane- 1 - carboximidamide (1.2 g, 49.3 % yield) as a colorless oil without any further purification. LC/MS (ESI) m/z: 191 (M+H)+.
Step 3: Synthesis of compound M7
CDI (111 mg, 0.681 mmol) was added to a mixture of M7-int-02 (100 mg, 0.454 mmol) and M7-int-03 (94.53 mg, 0.454 mmol) in pyridine (4 mL), and the resulting mixture was stirred at 100°C for 3 hours. After cooling, the mixture was diluted with EtOAc (20 mL) and washed with water (20 mL). The organic layer was then washed with brine, dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified via prep-HPLC (Gemini 5μm C18250*21.2mm, H2O/MeCN (5-95 %)/0.1% NH3 .H2O) to give M7 (10 mg, 6.1 % yield) as a white solid. LC/MS (ESI) m/z: 363(M+H)+.1H NMR (400 MHz, DMSO-d6) δ 8.24 (d, J = 5.8 Hz, 1H), 7.31 - 7.24 (m, 4H), 7.19 - 7.16 (m, 2H), 6.67 (dd, J = 5.8, 2.4 Hz, 1H), 5.08 (d, J = 3.6 Hz, 1H), 4.45 - .4.41 (m, 1H), 3.50 - 3.41 (m, 3H), 3.22 - 3.18 (m, 3H), 2.08 - 1.96 (m, 2H), 1.28 – 1.23 (m, 2H), 1.12 - 1.09 (m, 2H). General procedure 8
Figure imgf000123_0001
Step 1: Synthesis of compound 2 KHMDS (3.6 eq.) was added at -78oC under N2 atmosphere to a solution of compound 1 (1 eq.) in anhydrous THF (40 mL), and the resulting mixture was stirred at -78oC for 1 hour. The mixture was then quenched with aq. NH4Cl and extracted twice with EtOAc. The combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified via flash column chromatography on silica gel (eluted with PE/EtOAc) to give compound 2. Step 2: Synthesis of compound 4 NaI (0.2 eq.) and NaHCO3 (2 eq.) were added to a mixture of compound 2 and compound 3 (1 eq.) in a mixture of THF/H2O (15:1, V/V), and the resulting mixture was stirred at 80 ºC for 2 hours. The reaction was then cooled down to room temperature, diluted with EtOAc, and washed with H2O and brine. The organic layer was dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified via flash column chromatography (eluted with DCM/MeOH) to give compound 4. Step 3: Synthesis of compound 6 Compound 5 (2.0 eq.) and DIEA (3.0 eq.) were added to a mixture of compound 4 (1.0 eq.) in NMP, and the resulting mixture was stirred in a sealed tube at 160 ºC for 7 hours. The reaction mixture was then cooled down to room temperature, diluted with water and extracted with EtOAc. The organic layer was dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified via prep-HPLC (Gemini 5μm C18250*21.2mm, H2O/MeCN (5-95 %)/0.1% NH3 .H2O) to give compound 6. Example: Synthesis of M8
Figure imgf000124_0001
Step 1: Synthesis of 4-chloropyridine-2-carboximidamide (M8-int-2) KHMDS (20.87 mL, 1M in THF) was added at -78 ºC under N2 atmosphere to a solution of 4- chloropyridine-2-carbonitrile (800 mg, 5.8 mmol) in anhydrous THF (40 mL), and the resulting mixture was stirred at -78oC for 1 hour. The mixture was then quenched with aq. NH4Cl (20 mL) and extracted with EtOAc (2 x 30 mL). The combined organic layers were washed with brine (30 mL), dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified via flash column chromatography on silica gel (eluted with PE: EtOAc= 30:1 to 10:1) to give 4-chloropyridine-2-carboximidamide (400 mg, 44.5% yield) as a white solid.LC/MS (ESI) m/z: 156 (M+H)+. Step 2: Synthesis of 2-[4-(1-benzylcyclopropyl)-1H-imidazol-2-yl]-4-chloropyridine (M8- int-3) NaI (43 mg, 0.383 mmol) and NaHCO3 (322 mg, 3.833 mmol) were added to a mixture of 1- (1-benzylcyclopropyl)-2-chloroethan-1-one (400 mg, 1.917 mmol) and 4-chloropyridine-2- carboximidamide (298 mg, 1.917 mmol) in a mixture of THF (15 mL) and H2O (1 mL). The resulting mixture was stirred at 80oC for 2 hours. The reaction mixture was then cooled down to room temperature, diluted with EtOAc (40 mL), and washed with H2O (40 mL) and brine (40 mL). The organic layer was then dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified by flash column chromatography (eluted with DCM/MeOH=100:0 to 20:1) to give M8-int-3 (354 mg, 59.8% yield) as a colorless oil.LC/MS (ESI) m/z: 310(M+H)+. Step 2: Synthesis of M8 Cis-2,6-dimethylmorpholine (75 mg, 0.648 mmol) and DIEA (127 mg, 0.972 mmol) were added to a mixture of M8-int-3 (100 mg, 0.324mmol) in NMP (5 mL), and the resulting mixture was stirred in a sealed tube at 160oC for 7 hours. The reaction mixture was cooled to room temperature, diluted with water (30 mL) and extracted twice with EtOAc (30 mL). The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified via prep-HPLC (Gemini 5μm C18 250*21.2mm, H2O/MeCN (5-95 %)/0.1% NH3.H2O) to give M8 (20 mg, 15.9 % yield) as a white solid. LC/MS (ESI) m/z: 389(M+H)+.1HNMR (400 MHz, CD3OD) δ 8.08 (d, J = 7.2 Hz, 1H), 7.51 (d, J = 2.8 Hz, 1H), 7.23 – 7.13 (m, 5H),7.07 (dd, J = 7.3, 2.8 Hz, 1H), 6.97 (s, 1H), 4.08 - 4.04 (m, 2H), 3.78 – 3.69 (m, 2H), 3.07 (s, 2H), 2.86 - 2.80 (m, 2H), 1.28 (d, J = 6.2 Hz, 6H), 1.09 - 1.05 (m, 2H), 0.99 - 0.95 (m, 2H). The compound in the table below was prepared from the appropriate starting materials, described previously or commercially available, using the general procedure described above.
Figure imgf000125_0002
General procedure 9
Figure imgf000125_0001
Step 1: Synthesis of compound 3 The appropriate amine 2 (2 eq.) was added to a mixture of compound 1 (1 eq.) and DIEA (3 eq.) in n-BuOH, and the resulting mixture was stirred in a sealed tube at 120 ºC for 16 hours. After cooling, the mixture was concentrated under vacuum, and the residue was purified by column chromatography on silica gel (eluted with DCM/MeOH) to give compound 3. Step 2: Synthesis of compound 5 EDCI (1.2 eq.), HOBt (1.2 eq.) and NMM (3 eq.) were added to a mixture of the appropriate compound 4 (1 eq.) and compound 3 (1.1 eq.) in DMF, and the resulting mixture was stirred at room temperature for 16 hours. The mixture was then diluted with saturated aq. NH4Cl and extracted twice with EtOAc. The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified via prep-HPLC (Gemini 5μm C18250*21.2mm, H2O/MeCN (5-95 %)/0.1% NH3 .H2O) to give compound 5. Compounds of the formula of 4 were prepared using the procedure in Organic & Biomolecular Chemistry (2016), 14(8), 2498-2503. Example: Synthesis of M9
Figure imgf000126_0001
Step 1: Synthesis of compound M9-int-2 Cis-2,6-dimethylmorpholine (2.3 g, 20 mmol) was added to a mixture of M9-int-1 (1.57 g, 10 mmol) and DIEA (3.9 g, 30 mmol) in n-BuOH (50 mL), and the resulting mixture was stirred in a sealed tube at 120oC for 16 hours. After cooling, the mixture was concentrated under vacuum and the residue was purified by column chromatography on silica gel (eluted with DCM: MeOH= 40:1 to 12:1) to give M9-int-2 (730 mg, 30.9 % yield) as a white solid. LC/MS (ESI) m/z: 235 (M-H)+. Step 2: Synthesis of M9 EDCI (230 mg, 1.2 mmol), HOBt (162 mg, 1.2 mmol) and NMM (303 mg, 3 mmol) were added to a mixture of M9-int-3 (133 mg, 1 mmol) and M9-int-2 (260 mg, 1.1 mmol) in DMF (3 mL), and the resulting mixture was stirred at room temperature for 16 hours. The mixture was then diluted with saturated aq. NH4Cl (20 mL) and extracted twice with EtOAc (20 mL). The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified via prep-HPLC (Gemini 5μm C18 250*21.2mm, H2O/MeCN (5-95 %)/0.1% NH3 .H2O) to give M9 (41 mg, 11.7 % yield) as a white solid. LC/MS (ESI) m/z: 352 (M+H)+.1H NMR (400 MHz, DMSO-d6) δ 8.86 (d, J = 5.3 Hz, 1H), 8.21 (d, J = 5.9 Hz, 1H), 7.45 (d, J = 2.6 Hz, 1H), 7.30 – 7.23 (m, 2H), 7.19 – 7.13 (m, 3H), 7.03 – 6.99 (m, 1H), 3.89 – 3.79 (m, 2H), 3.69 – 3.59 (m, 2H), 3.07 – 2.99 (m, 1H), 2.47 – 2.39 (m, 2H), 2.19 – 2.11 (m, 1H), 1.49 – 1.40 (m, 1H), 1.27 – 1.19 (m, 1H), 1.17 (d, J = 6.2 Hz, 6H). The compounds in the table below were prepared from the appropriate starting materials, described previously or commercially available, using the general procedure described above.
Figure imgf000127_0001
Figure imgf000128_0001
Figure imgf000129_0001
Figure imgf000130_0001
Figure imgf000131_0001
Figure imgf000132_0001
Figure imgf000133_0001
Figure imgf000134_0002
General procedure 10
Figure imgf000134_0001
Step 1: Synthesis of compound 3 LDA (1.2 eq.) was added at -78oC under N2 atmosphere to a solution of compound 2 (1 eq.) in anhydrous THF, and the mixture was stirred at -78 ºC for 1 hour. A solution of compound 1 (1 eq.) in THF was then added dropwise over 30 minutes and the resulting mixture was allowed to warm to room temperature over 3 hours under N2 atmosphere. The mixture was then quenched with saturated aq. NH4Cl and extracted twice with EtOAc. The combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified via flash column chromatography on silica gel (PE/EtOAc) to give compound 3. Step 2: Synthesis of compound 4 A solution of KOH (3 eq.) in H2O was added to a solution of compound 3 (1 eq.) in ethylene glycol, and the resulting mixture was stirred at 100 ºC for 16 hours. After cooling, the mixture was diluted with water and extracted twice with MTBE. The aqueous layer was then acidified with 1 N aq. HCl to pH=5 and extracted with EtOAc. The organic layer was dried over anhydrous Na2SO4, filtered and concentrated under vacuum to give crude compound 4, which was used for next step without any further purification. Step 3: Synthesis of compound 5 DPPA (1.5 eq.) was added to a mixture of compound 4 (1 eq.) and Et3N (3 eq.) in t-BuOH, and the resulting mixture was stirred at 90 ºC under N2 atmosphere for 16 hours. After cooling, the mixture was diluted with DCM and washed with water. The organic layer was dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified via flash column chromatography on silica gel (PE/EtOAc) to give compound 5, which was used for next step without any further purification. Step 4: Synthesis of compound 6 Compound 5 (1 eq.) was added into a solution of TFA in DCM (1:3, V/V) at 0oC and the resulting mixture was stirred at room temperature under N2 atmosphere for 3 hours. The mixture was then concentrated under vacuum to give crude compound 6, which was used for next step without any further purification. Step 5: Synthesis of compound 8 EDCI (1.2 eq.), HOBt (1.2 eq.) and NMM (3 eq.) were added under N2 atmosphere to a mixture of compound 6 (1 eq.) and compound 7 (1.1 eq.) in DMF, and the resulting mixture was stirred at room temperature for 8 hours. The mixture was then diluted with saturated aq. NH4Cl and extracted twice with EtOAc. The combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified via prep-HPLC (Gemini 5μm C18250*21.2mm, H2O/MeCN (5-95 %)/0.1% NH3 .H2O) to give compound 8.
Example: Synthesis of compound M10
Figure imgf000136_0001
Step 1: Synthesis of M10-int-2 LDA (6 mL, 2 M in THF) was added at -78oC under N2 atmosphere to a solution of cyclobutane carbonitrile (810 mg, 10 mmol) in anhydrous THF (40 mL), and the resulting mixture was stirred at -78 ºC for 1 hour. A solution of M10-int-1 (2.19 g, 10 mmol) in THF (8 mL) was then added dropwise over 30 minutes, and the resulting mixture was allowed to warm to room temperature over 3 hours under N2 atmosphere. The mixture was then quenched with saturated aq. NH4Cl (40 mL) and extracted twice with EtOAc. The combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified by column chromatography on silica gel (PE: EtOAc = 80:1 to 12:1) to give M10- int-2 (1.1 g, 50.2 % yield) as a colorless oil. LC/MS (ESI) m/z: 220 (M+H)+. Step 2: Synthesis of M10-int-3 A solution of KOH (840 mg, 15 mmol) in H2O (15 mL) was added to a solution of M10-int-2 (1.1 g, 5.02 mmol) in ethylene glycol (30 mL), and the resulting mixture was stirred at 100 ºC for 16 hours. The reaction mixture was cooled down, diluted with water (20 mL), and extracted twice with MTBE (30 mL). The aqueous layer was then acidified with 1 M aq. HCl to pH=5 and extracted with EtOAc. The organic layer was dried over anhydrous Na2SO4, filtered and concentrated under vacuum to give M10-int-3 (620 mg, 52.1 % yield) as colorless oil without any further purification. LC/MS (ESI) m/z: 237 (M-H)+. Step 3: Synthesis of M10-int-4 DPPA (1.15 g, 4.17 mmol) was added to a mixture of M10-int-3 (620 mg, 2.61 mmol) and Et3N (790 mg, 7.82 mmol) in t-BuOH (40 mL), and the resulting mixture was stirred under N2 atmosphere at 90 ºC for 16 hours. The reaction mixture was cooled down, diluted with DCM (60 mL), and washed with water (60 mL). The organic layer was then dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified by column chromatography on silica gel (PE: EtOAc = 80:1 to 40:1) to give M10-int-4 (210 mg, 26.0 % yield) as a white solid. LC/MS (ESI) m/z: 310 (M+H)+. Step 4: Synthesis of M10-int-5 M10-int-4 (210 mg, 0.68 mmol) was added at 0 ºC into a solution of TFA (2 mL) in DCM (6 mL), and the resulting mixture was stirred under N2 atmosphere at room temperature for 3 hours. The mixture was then concentrated under vacuum to give M10-int-5 (110 mg, 77.4 % yield) as a yellow oil without any further purification. LC/MS (ESI) m/z: 210 (M+H)+. Step 5: Synthesis of M10 EDCI (122 mg, 0.64 mmol), HOBt (87 mg, 0.64 mmol) and NMM (160 mg, 1.6 mmol) were added under N2 atmosphere to a mixture of M10-int-5 (110 mg, 0.53 mmol) and M10-int-6 (139 mg, 0.59 mmol) in DMF (6 mL), and the resulting mixture was stirred at room temperature for 8 hours. The mixture was then diluted with saturated aq. NH4Cl (30 mL) and extracted twice with EtOAc (30 mL). The combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified via prep- HPLC (Gemini 5 μm C18250*21.2mm, H2O/MeCN (5-95 %)/0.1% NH3 .H2O) to give M10 (41 mg, 18.1 % yield) as a white solid. LC/MS (ESI) m/z: 428 (M+H)+.1H-NMR (400 MHz, DMSO-d6) δ 8.65 (s, 1H), 8.18 (d, J = 5.9 Hz, 1H), 7.47 (d, J = 2.5 Hz, 1H), 7.11 – 6.84 (m, 4H), 3.87 (t, J = 11.5 Hz, 2H), 3.74 (s, 3H), 3.66 - 3.62 (m, 2H), 3.15 - 3.11 (m, 2H), 2.62 – 2.51 (m, 2H), 2.50 – 2.37 (m, 2H), 2.17 – 2.04 (m, 2H), 1.92 – 1.77 (m, 2H), 1.17 (d, J = 6.2 Hz, 6H). General procedure 11
Figure imgf000138_0001
Step 1: Synthesis of compound 3 EDCI (1.2 eq.), HOBt (1.2 eq.) and NMM (3 eq.) were added to a mixture of compound 2 (1 eq.) and compound1 (1.1 eq.) in DMF, and the resulting mixture was stirred at room temperature for 16 hours. The mixture was then diluted with saturated aq. NH4Cl and extracted twice with EtOAc. The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified via flash column chromatography (eluted with PE/EtOAc) to give compound 3. Step 2: Synthesis of compound 4 Ethylmagnesium bromide (2.2 eq.) was slowly added at -60 oC under N2 atmosphere to a stirred mixture of compound 3 (1 eq.) and Ti(O-iPr)4 (1.4 eq.) in anhydrous THF. The reaction mixture was then warmed to 0 ºC over 3 hours and BF3-Et2O (1.2 eq.) was added. The resulting mixture was stirred for 2 hours and then poured into 1N aq. NaOH and extracted twice with EtOAc (20 mL). The combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified via flash column chromatography (eluted with PE/EtOAc) to give compound 4. Step 3: Synthesis of compound 5 Pd/C (15%, w/w) was added to a solution of compound 4 (1 eq.) in MeOH, and the resulting mixture was stirred under 25 psi H2 atmosphere at room temperature for 16 hours. The vacuum to give crude compound 5, which was used for next step without any further purification. Step 4: Synthesis of compound 7 EDCI (1.2 eq.), HOBt (1.2 eq.) and NMM (3 eq.) were added to a mixture of compound 5 (1 eq.) and compound 6 (1.1 eq.) in DMF, and the resulting mixture was stirred at room temperature for 16 hours. The mixture was then diluted with saturated aq. NaHCO3 and extracted twice with EtOAc. The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified via prep-HPLC (Gemini 5μm C18250*21.2mm, H2O/MeCN (5-95 %)/0.1% NH3 .H2O) to give compound 7. General procedure 12
Figure imgf000139_0001
Step 1: Synthesis of compound 3 PPh3 (1.3 eq.) and DIAD (2 eq.) were added at 0 ºC under N2 atmosphere to a mixture of compound 1 (1 eq.) and compound 2 (1.1 eq.) in anhydrous THF, and the resulting mixture was stirred at room temperature for 16 hours. The mixture was then diluted with saturated aq. NaHCO3 and extracted twice with EtOAc. The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified via flash column chromatography (eluted with PE/EtOAc) to give compound 3. Step 2: Synthesis of compound 4 Compound 3 (1 eq.) was added into a mixture of TFA/DCM (1:4, V/V) at 0 ºC under N2 atmosphere, and the resulting mixture was stirred at room temperature for 2 hours. The mixture was then concentrated under vacuum and the residue was diluted with DCM and washed with saturated aq. NaHCO3. The organic layer was separated, dried over anhydrous Na2SO4, filtered and concentrated under vacuum to give crude compound 3, which was used for next step without any further purification. Step 3: Synthesis of compound 6 EDCI (1.2 eq.), HOBt (1.2 eq.) and NMM (3 eq.) were added to a mixture of compound 4 (1 eq.) and compound 5 (1.1 eq.) in DMF, and the resulting mixture was stirred at room temperature for 16 hours. The mixture was then diluted with saturated aq. NaHCO3 and extracted twice with EtOAc. The combined organic layers were dried over Na2SO4, filtered and concentrated under vacuum. The residue was purified via prep-HPLC (Gemini 5μm C18 250*21.2mm, H2O/MeCN (5-95 %)/0.1% NH3.H2O) to give compound 6. General procedure 13
Figure imgf000140_0001
Step 1: Synthesis of compound 2 A solution of compound 1 (1 eq.) in DCM was added dropwise at 0 ºC under N2 atmosphere into a solution of HCl/1,4-dioxane (4 M), and the resulting mixture was stirred at room temperature for 5 hours. The mixture was then concentrated under vacuum to give crude compound 2, which was used for next step without any further purification. Step 2: Synthesis of compound 4 EDCI (1.2 eq.), HOBt (1.2 eq.) and NMM (3 eq.) were added to a mixture of compound 2 (1 eq.) and compound 3 (1.1 eq.) in DMF, and the resulting mixture was stirred at room temperature for 16 hours. The mixture was then diluted with saturated aq. NH4Cl and extracted twice with EtOAc. The combined organic layers were dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified by flash column chromatography (eluted with PE/EtOAc) to give compound 4. Step 3: Synthesis of compound 5 Anhydrous DMSO (1.5 eq.) in dry DCM was added dropwise at -65°C under N2 atmosphere to a solution of oxalyl chloride (1.5 eq.) in dry DCM. After stirring for 30 minutes, a solution of compound 4 (1 eq.) in dry DCM was added dropwise, and the reaction mixture was stirred for 2 hours before Et3N (3 eq.) was added dropwise. The mixture was then slowly warmed to room temperature until completion. Water was added and the mixture was extracted twice with DCM. The combined organic layers were washed with brine, dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified by flash column chromatography (eluted with PE/EtOAc) to give compound 5. Step 4: Synthesis of compound 7 4A MS and compound 6 (1 eq.) were added to a mixture of compound 5 (1 eq.) in DCE, and the mixture was stirred at room temperature for 2 hours before NaBH(OAc)3 (2 eq.) was added. The resulting mixture was stirred at room temperature for 16 hours. The mixture was then diluted with DCM and washed with saturated aq. NaHCO3. The organic layer was dried over anhydrous Na2SO4, filtered and concentrated under vacuum. The residue was purified via prep-HPLC (Gemini 5μm C18250*21.2mm, H2O/MeCN (5-95 %)/0.1% NH3.H2O) to give compound 7. INCORPORATION BY REFERENCE All of the U.S. patents and U.S. patent application publications cited herein are hereby incorporated by reference. EQUIVALENTS Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.

Claims

We claim: 1. A compound of Formula (I):
Figure imgf000142_0002
wherein: A and A' are each independently selected from C-R' and N; A'' is selected from C-R'' and N; provided that at least one of A, A', and A'' is N; or A and A' are each independently selected from C-R' and N, and A'' is C-R'', wherein R'' taken together with R and the ring atoms to which they are bonded form a fused five-membered heteroaryl ring, such that formula I is a substituted indole or azaindole; X is selected from
Figure imgf000142_0003
Y is selected from
Figure imgf000142_0001
n is 0, 1, 2, or 3; m is 1; R1, R2, R3, R4, R5 and R6 are independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, aryl, heteroaryl, -C(O)OR11, -C(O)NR12R13 and -C(O)R14; R7, R8, R9, and R10 are independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, aryl, heteroaryl, -C(O)OR11, -C(O)NR12R13, and -C(O)R14; Z is selected from -H, halogen, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O- cycloheteroalkyl, -O-halocycloalkyl, -NH-alkyl, -NH-heteroalkyl, -NH-haloalkyl, -NH- cycloalkyl, -NH-cycloheteroalkyl, -NH-halocycloalkyl, aryl, -O-aryl, -NH-aryl, heteroaryl, - O-heteroaryl, -NH-heteroaryl, -C(O)OR11, -C(O)NR12R13, -NR12R13, and -C(O)R14; R is selected from alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O-cycloheteroalkyl, - O-halocycloalkyl, aryl, -O-aryl, heteroaryl, -O-heteroaryl -OSO2R11, and -NR15R16; R' is selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O- cycloalkyl, -O-cycloheteroalkyl, -O-halocycloalkyl, -O-aryl, -O-heteroaryl, -OSO2R11, and -NR12R13; R'' is selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O- cycloalkyl, -O-cycloheteroalkyl, -O-halocycloalkyl, -O-aryl, -O-heteroaryl, -OSO2R11, and -NR12R13; each occurrence of R11, R12 and R13 is independently selected from H, alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl; each occurrence of R14 is independently selected from alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl; R15 and R16 are independently selected from -H, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -C(O)R11, -C(O)OR11, and - C(O)NR12R13; or R15 and R16 taken together with the nitrogen atom to which they are bonded form an unsubstituted or substituted C4-C8 cycloheteroalkyl; and provided that if A is N, A' and A'' are each
Figure imgf000143_0001
is selected from phenyl and 2-methoxy-5-fluorophenyl, then R is not , ,
Figure imgf000143_0002
2. The compound of claim 1, wherein X is . 3. The compound of claim 1, wherein X is selected from
Figure imgf000143_0003
.
4. The compound of any one of claims 1-3, wherein: Y is selected from
Figure imgf000144_0001
5. The compound of claim 4, wherein R1, R2, R3, R4, R5, R6, R7 and R8 are -H. 6. The compound of claim 4 or 5, wherein
Figure imgf000144_0002
7. The compound of claim 4 or 5, wherein
Figure imgf000144_0003
Figure imgf000144_0004
8. The compound of claim 4 or 5, wherein Y is . 9. The compound of any one of claims 1-3, wherein Y is selected from
Figure imgf000144_0005
,
Figure imgf000144_0006
10. The compound of any one of claims 1-3, wherein Y is . 11. The compound of claim 10, wherein R7 and R8 are -H. 12. The compound of claim 10 or 11, wherein R9 and R10 are independently selected from -H and alkyl.
13. The compound of claim 10, wherein Y is
Figure imgf000145_0001
. 14. The compound of any one of claims 1-13, wherein Z is aryl or heteroaryl. 15. The compound of claim 14, wherein Z is selected from unsubstituted or substituted phenyl, pyridine, pyrimidine, pyrazole, and triazole.
Figure imgf000145_0002
R17, R18, R19, R20, and R21 are independently selected from -H, halogen, CN, CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, -O- heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O-cycloheteroalkyl, -O-halocycloalkyl, -NH-alkyl, -NH-heteroalkyl, -NH-haloalkyl, -NH-cycloalkyl, -NH-cycloheteroalkyl, -NH- halocycloalkyl, aryl, and heteroaryl; and R19a is haloalkyl or alkyl. 17. The compound of claim 14, wherein
Figure imgf000145_0003
R17 and R18 are independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O-cycloheteroalkyl, -O-halocycloalkyl, -NH-alkyl, -NH- heteroalkyl, -NH-haloalkyl, -NH-cycloalkyl, -NH-cycloheteroalkyl, -NH-halocycloalkyl, aryl, and heteroaryl, or R17 taken together with R18 and the ring atoms to which they are bonded form a fused ring, such that Z is a benzimidazole, dihydrobenzofuran, benzodioxole, benzooxazole, or benzooxazolone; R19 and R20 are independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O-cycloheteroalkyl, - O-halocycloalkyl, -NH-alkyl, -NH-heteroalkyl, -NH-haloalkyl, -NH-cycloalkyl, -NH- cycloheteroalkyl, -NH-halocycloalkyl, aryl, and heteroaryl, or R19 taken together with R20 and the ring atoms to which they are bonded form a fused ring, such that Z is a benzimidazole, dihydrobenzofuran, benzodioxole, benzooxazole, or benzooxazolone; and R21 is selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O- cycloheteroalkyl, -O-halocycloalkyl, -NH-alkyl, -NH-heteroalkyl, -NH-haloalkyl, -NH- cycloalkyl, -NH-cycloheteroalkyl, -NH-halocycloalkyl, aryl, and heteroaryl. 18. The compound of any one of claims 1-17 having the structure selected from
Figure imgf000146_0001
19. The compound of claim 18, having the structure selected from
Figure imgf000146_0002
20. The compound of claim 18 or 19, wherein: R is selected from aryl, heteroaryl, and -NR15R16; and R15 and R16 taken together with the nitrogen atom to which they are bonded form an unsubstituted or substituted C4-C8 cycloheteroalkyl.
21. The compound of claim 20, wherein R is selected from aryl and heteroaryl. 22. The compound of claim 21, wherein R is phenyl. 23. The compound of claim 20, wherein R is -NR15R16; and R15 and R16 taken together with the nitrogen atom to which they are bonded form an unsubstituted or substituted piperidine, piperazine, pyrrolidine, morpholine, or azetidine. 24. The compound of claim 20,
Figure imgf000147_0001
R22 is selected from alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -SO2R23, -C(O)OR23, -C(O)NR24R25, and -C(O)R26; Ra, Rb, Rc, Rd, Rg, Rh, Ri, and Rj are independently selected from -H, halogen, -CN, - CF3, alkyl, heteroalkyl, haloalkyl, hydroxyalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -SO2R23, -C(O)OR23, -C(O)NR24R25, and -C(O)R26; or Ra taken together with Rb and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, ^-lactam, or ^-lactam; or Rc taken together with Rd and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, ^-lactam, or ^-lactam; each occurrence of R23, R24 and R25 is independently selected from H, alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl; and each occurrence of R26 is selected from alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl. 25. The compound of claim 20, wherein
Figure imgf000147_0002
Ra, Rb, Ri and Rj are independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, - C(O)OR23, -C(O)NR24R25, and -C(O)R26, or Ra taken together with Rb and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, ^-lactam, or ^- lactam; Rc, Rd, Re, Rf, Rg and Rh are independently selected from -H, halogen, -CN, -CF3, - OH, -NH2, alkyl, heteroalkyl, haloalkyl, hydroxyalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O-cycloheteroalkyl, - O-halocycloalkyl, -NH-alkyl, -NH-heteroalkyl, -NH-haloalkyl, -NH-cycloalkyl, -NH- cycloheteroalkyl, -NH-halocycloalkyl, aryl, -O-aryl, -NH-aryl, heteroaryl, -O-heteroaryl, - NH-heteroaryl, -SO2R23, -OSO2R23, -C(O)OR23, -NR24C(O)R25, -NR24SO2R25, - C(O)NR24R25, -OSO2(N26)2, -SO2(N26)2, -N(R26)2, -C(O)R26, and alkyl-NHSO2-R26; or Rc taken together with Rd and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, ^-lactam, or ^-lactam; or Re taken together with Rf and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, ^-lactam, or ^-lactam; each occurrence of R23, R24 and R25 is independently selected from -H, alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl; and each occurrence of R26 is selected from alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl. 26. The compound of claim 20,
Figure imgf000148_0001
Ra, Rb, Rc, Rd, Rg, Rh Ri and Rj are independently selected from -H, halogen, -CN, CF3, - alkyl, heteroalkyl, haloalkyl, hydroxyalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -SO2R23, -C(O)OR23, -C(O)NR24R25, -OSO2(N26)2, - SO2(N26)2, –C(O)R26 , and alkyl-NHSO2-R26; or Ra taken together with Rb and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, ^- lactam, or ^-lactam; or Rc taken together with Rd and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, ^-lactam, or ^-lactam; each occurrence of R23, R24 and R25 is independently selected from -H, alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl; and each occurrence of R26 is selected from alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl.
27. The compound of claim 20, wherein
Figure imgf000149_0001
Ra, Rb, Rg and Rh are independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, hydroxyalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -SO2R23, -C(O)OR23, -C(O)NR24R25, -SO2(N26)2, and -C(O)R26; or Ra taken together with Rb and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, ^-lactam, or ^-lactam; Rc, Rd, Re and Rf are independently selected from -H, halogen, -CN, -CF3, -OH, -NH2, alkyl, heteroalkyl, haloalkyl, hydroxyalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O- alkyl, -O-heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O-cycloheteroalkyl, -O-halocycloalkyl, - NH-alkyl, -NH-heteroalkyl, -NH-haloalkyl, -NH-cycloalkyl, -NH-cycloheteroalkyl, -NH- halocycloalkyl, aryl, -O-aryl, -NH-aryl, heteroaryl, -O-heteroaryl, -NH-heteroaryl, -SO2R23, - OSO2R23, -C(O)OR23, -NR24C(O)R25, -NR24SO2R25, -C(O)NR24R25, -OSO2(N26)2, - SO2(N26)2, -N(R26)2, and -C(O)R26; or Rc taken together with Rd and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, ^-lactam, or ^- lactam; and each occurrence of R23, R24 and R25 is independently selected from -H, alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl; and each occurrence of R26 is selected from alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl. 28. The compound of claim 20, wherein
Figure imgf000149_0002
Ra, Rb, Re and Rf are independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, hydroxyalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -SO2R23, -C(O)OR23, and -C(O)NR24R25; or Ra taken together with Rb and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, γ-lactam, or ^-lactam; Rc and Rd are independently selected from -H, halogen, -CN, -CF3, -OH, -NH2, alkyl, heteroalkyl, haloalkyl, hydroxyalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, - O-heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O-cycloheteroalkyl, -O-halocycloalkyl, -NH- alkyl, -NH-heteroalkyl, -NH-haloalkyl, -NH-cycloalkyl, -NH-cycloheteroalkyl, -NH- halocycloalkyl, aryl, heteroaryl-SO2R23, -OSO2R23, -C(O)OR23, -NR24C(O)R25, -NR24SO2R25, -C(O)NR24R25, -OSO2(N26)2, -SO2(N26)2, -N(R26)2, and -C(O)R26; or Rc taken together with Rd and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, ^-lactam, or ^-lactam; each occurrence of R23, R24 and R25 is independently selected from -H, alkyl, heteroalkyl, haloalkyl, aryl or heteroaryl; and each occurrence of R26 is selected from alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl.
Figure imgf000150_0001
each occurrence of R22 is selected from alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -SO2R19, -C(O)R19, -C(O)OR19, and - C(O)NR19R20; each occurrence of Ra, Rb, Rc, Rd, Rg, Rh Ri and Rj is independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, hydroxyalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -SO2R23, -C(O)OR23, -C(O)NR24R25, and -C(O)R26; or Ra taken together with Rb and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, ^-lactam, or ^-lactam; each occurrence of R23, R24 and R25 is independently selected from H, alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl; and each occurrence of R26 is selected from alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl.
Figure imgf000151_0001
each occurrence of Ra, Rb, Ri and Rj is independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -C(O)OR23, -C(O)NR24R25, and -C(O)R26; or Ra taken together with Rb and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, ^-lactam, or ^-lactam; each occurrence of Rc, Rd, Re, Rf, Rg and Rh is independently selected from -H, halogen, -CN, -CF3, -OH, -NH2, alkyl, heteroalkyl, haloalkyl, hydroxyalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O- cycloheteroalkyl, -O-halocycloalkyl, -NH-alkyl, -NH-heteroalkyl, -NH-haloalkyl, -NH- cycloalkyl, -NH-cycloheteroalkyl, -NH-halocycloalkyl, aryl, -O-aryl, -NH-aryl, heteroaryl, - O-heteroaryl, -NH-heteroaryl, -SO2R23, -OSO2R23, -C(O)OR23, -NR24C(O)R25, -NR24SO2R25, -C(O)NR25R26, -OSO2(N26)2, -SO2(N26)2, -N(R26)2, -C(O)R26, and alkyl-NHSO2-R26; each occurrence of R23, R24 and R25 is independently selected from -H, alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl; and each occurrence of R26 is selected from alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl.
Figure imgf000151_0002
each occurrence of Ra, Rb, Rc, Rd, Rg, Rh Ri and Rj is independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, hydroxyalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -SO2R23, -C(O)OR23, -C(O)NR24R25, -OSO2(N26)2, - SO2(N26)2, -C(O)R26, and alkyl-NHSO2-R26; or Ra taken together with Rb and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, ^- lactam, or ^-lactam; each occurrence of R23, R24 and R25 is independently selected from -H, alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl; and each occurrence of R26 is selected from alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl.
Figure imgf000152_0001
each occurrence of Ra, Rb, Rg and Rh is independently selected from -H, halogen, - CN, -CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, hydroxyalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -SO2R23, -C(O)OR23, -C(O)NR24R25, -SO2(N26)2, and - C(O)R26; or Ra taken together with Rb and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, ^-lactam, or ^-lactam; each occurrence of Rc, Rd, Re and Rf is independently selected from -H, halogen, -CN, -CF3, -OH, -NH2, alkyl, heteroalkyl, haloalkyl, hydroxyalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O-cycloheteroalkyl, - O-halocycloalkyl, -NH-alkyl, -NH-heteroalkyl, -NH-haloalkyl, -NH-cycloalkyl, -NH- cycloheteroalkyl, -NH-halocycloalkyl, aryl, -O-aryl, -NH-aryl, heteroaryl, -O-heteroaryl, - NH-heteroaryl, -SO2R23, -OSO2R23, -C(O)OR23, -NR24C(O)R25, -NR24SO2R25, - C(O)NR24R25, -OSO2(N26)2, -SO2(N26)2, -N(R26)2, and -C(O)R26; and each occurrence of R23, R24 and R25 is independently selected from -H, alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl; and each occurrence of R26 is selected from alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl. 33. The compound of claim 20, wherein
Figure imgf000153_0001
each occurrence Ra and Rb is independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, hydroxyalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -SO2R23, -C(O)OR23, and -C(O)NR24R25; or Ra taken together with Rb and the carbon atom to which they are bonded form an unsubstituted or substituted C3-C6 cycloalkyl, ^-lactam, or ^-lactam; each occurrence Rc and Rd is independently selected from -H, halogen, -CN, -CF3, - OH, -NH2, alkyl, heteroalkyl, haloalkyl, hydroxyalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O-cycloheteroalkyl, - O-halocycloalkyl, -NH-alkyl, -NH-heteroalkyl, -NH-haloalkyl, -NH-cycloalkyl, -NH- cycloheteroalkyl, -NH-halocycloalkyl, aryl, heteroaryl-SO2R23, -OSO2R23, -C(O)OR23, - NR24C(O)R25, -NR24SO2R25, -C(O)NR24R25, -OSO2(N26)2, -SO2(N26)2, -N(R26)2, and - C(O)R26; and each occurrence of R23, R24 and R25 is independently selected from -H, alkyl, heteroalkyl, haloalkyl, aryl or heteroaryl; and each occurrence of R26 is selected from alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl. 34. The compound of claim 1, selected from the following table:
Figure imgf000153_0002
Figure imgf000154_0001
Figure imgf000155_0001
Figure imgf000156_0001
Figure imgf000157_0001
Figure imgf000158_0001
Figure imgf000159_0001
Figure imgf000160_0001
Figure imgf000161_0001
Figure imgf000162_0001
Figure imgf000163_0001
Figure imgf000164_0001
Figure imgf000165_0001
Figure imgf000166_0001
35. The compound of any one of claims 1-17, wherein R taken together with R'' and the ring atoms to which they are bonded form a fused five-membered heteroaryl ring, such that formula I is a substituted indole or azaindole.
36. The compound of claim 35 having the structure:
Figure imgf000167_0001
,
Figure imgf000167_0002
37. The compound of claim 36 having the structure:
Figure imgf000167_0003
, wherein each occurrence of R27 is selected from alkyl, heteroalkyl, haloalkyl, hydroxyalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, and heteroaryl. 38. The compound of claim 1 selected from the following table:
Figure imgf000167_0004
39. A pharmaceutical composition, comprising a compound of any one of claims 1-38; and a pharmaceutical acceptable excipient. 40. A method of treating or preventing a disease or disorder associated with a SLC6A8 mutation, treating an inflammatory disease, increasing transport of guanidinoacetic acid or a salt thereof across the blood-brain barrier, improving the function of a cellular creatine transporter, or decreasing the accumulation or the concentration of guanidinoacetic acid or a salt thereof in a cell, in a subject in need thereof comprising administering to the subject an effective amount of a compound of Formula (I): wherein:
Figure imgf000168_0001
A and A' are each independently selected from C-R' and N; A'' is selected from C-R'' and N; provided that at least one of A, A', and A'' is N; or A and A' are each independently selected from C-R' and N, and A'' is C-R'', wherein R'' taken together with R and the ring atoms to which they are bonded form a fused five-membered heteroaryl ring, such that formula I is a substituted indole or azaindole;
Figure imgf000168_0002
n is 0, 1, 2, or 3; m is 1; R1, R2, R3, R4, R5 and R6 are independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, aryl, heteroaryl, -C(O)OR11, -C(O)NR12R13, and –C(O)R14; R7, R8, R9, and R10 are independently selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, aryl, heteroaryl, -C(O)OR11, -C(O)NR12R13, and –C(O)R14; Z is selected from -H, halogen, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O- cycloheteroalkyl, -O-halocycloalkyl, -NH-alkyl, -NH-heteroalkyl, -NH-haloalkyl, -NH- cycloalkyl, -NH-cycloheteroalkyl, -NH-halocycloalkyl, aryl, -O-aryl, -NH-aryl, heteroaryl, - O-heteroaryl, -NH-heteroaryl, -C(O)OR11, -C(O)NR12R13, -NR12R13, and –C(O)R14; R is selected from alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O-cycloalkyl, -O-cycloheteroalkyl, - O-halocycloalkyl, aryl, -O-aryl, heteroaryl, -O-heteroaryl -OSO2R11, and -NR15R16; R' is selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O- cycloalkyl, -O-cycloheteroalkyl, -O-halocycloalkyl, -O-aryl, -O-heteroaryl, -OSO2R11, and – NR12R13;; R'' is selected from -H, halogen, -CN, -CF3, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -O-alkyl, -O-heteroalkyl, -O-haloalkyl, -O- cycloalkyl, -O-cycloheteroalkyl, -O-halocycloalkyl, -O-aryl, -O-heteroaryl, -OSO2R11, and – NR12R13;; each occurrence of R11, R12 and R13 is independently selected from H, alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl; each occurrence of R14 is independently selected from alkyl, heteroalkyl, haloalkyl, aryl, and heteroaryl; R15 and R16 are independently selected from -H, alkyl, heteroalkyl, haloalkyl, cycloalkyl, cycloheteroalkyl, halocycloalkyl, aryl, heteroaryl, -C(O)R11, -C(O)OR11, and - C(O)NR12R13; or R15 and R16 taken together with the nitrogen atom to which they are bonded form an unsubstituted or substituted C4-C8 cycloheteroalkyl. 41. The method of claim 40, wherein if A is N, A' and A'' are each C-H, X is
Figure imgf000169_0001
Y
Figure imgf000169_0002
, and Z is selected from phenyl and 2-methoxy-5-fluorophenyl, then R is not
Figure imgf000169_0003
42. The method of claim 40 or 41comprising treating or preventing a disease or disorder associated with a SLC6A8 mutation in the subject in need thereof. 43. The method of claim 42, wherein the disease or disorder is creatine transporter deficiency. 44. The method of claim 42, wherein the disease or disorder is motor dysfunction. 45. The method of claim 42, wherein the disease or disorder is intellectual disability. 46. The method of claim 42, wherein the disease or disorder is language delay or speech delay. 47. The method of claim 42, wherein the disease or disorder is hypotonia. 48. The method of claim 40 or 41 comprising improving the function of a cellular creatine transporter in the subject in need thereof. 49. The method of claim 48, wherein the cellular creatine transporter is SLC6A8. 50. The method of claim 48 or 49, wherein the cellular creatine transporter is a mutant cellular creatine transporter. 51. The method of claim 50, wherein the mutant cellular creatine transporter is mutant SLC6A8. 52. The method of claim 40 or 41 comprising decreasing the accumulation or the concentration of guanidinoacetic acid or a salt thereof in a cell of the subject in need thereof. 53. The method of claim 53, wherein the subject is a mammal.
54. The method of claim 52 or 53, wherein the compound decreases intracellular accumulation of guanidinoacetic acid or a salt thereof. 55. The method of claim 52 or 53, wherein the compound decreases the intracellular concentration of guanidinoacetic acid or a salt thereof. 56. The method of any one of claims 52-55, wherein the cell is a brain cell. 57. The method of any one of claims 53-56, wherein the mammal is a male. 58. The method of any one of claims 53-56, wherein the mammal is a female. 59. The method of any one of claims 53-58, wherein the mammal is a primate, equine, bovine, ovine, feline, or canine. 60. The method of any one of claims 53-58, wherein the mammal is a human. 61. The method of claim 40 or 41 comprising increasing transport of guanidinoacetic acid or a salt thereof across the blood-brain barrier in the subject in need thereof. 62. The method of claim 61, wherein the subject is a mammal. 63. The method of claim 62, wherein the mammal is a male. 64. The method of claim 62, wherein the mammal is a female. 65. The method of any one of claims 62-64, wherein the mammal is a primate, equine, bovine, ovine, feline, or canine. 66. The method of any one of claims 62-64, wherein the mammal is a human. 67. The method of claim 40 or 41 comprising treating an inflammatory disease in the subject in need thereof.
68. The method of claim 67, wherein the subject is a mammal.
69. The method of claim 67 or 68, wherein the inflammatory disease is acute.
70. The method of claim 67 or 68, wherein the inflammatory disease is chronic.
71. The method of any one of claims 67-69, wherein the inflammatory disease is selected from inflammatory bowel diseases (for example, ulcerative colitis or Crohn's disease), multiple sclerosis, psoriasis, arthritis, rheumatoid arthritis, osteoarthritis, juvenile arthritis, psoriatic arthritis, reactive arthritis, ankylosing spondylitis, cryopyrin associated periodic syndromes, Muckle-Wells syndrome, familial cold auto- inflammatory syndrome, neonatal-onset multisystem inflammatory disease, TNF receptor associated periodic syndrome, acute and chronic pancreatitis, atherosclerosis, gout, ankylosing spondylitis, fibrotic disorders (for example, hepatic fibrosis or idiopathic pulmonary fibrosis), nephropathy, sarcoidosis, scleroderma, anaphylaxis, diabetes (for example, diabetes mellitus type 1 or diabetes mellitus type 2), diabetic retinopathy, Still's disease, vasculitis, sarcoidosis, pulmonary inflammation, acute respiratory distress syndrome, wet and dry age-related macular degeneration, autoimmune hemolytic syndromes, autoimmune and inflammatory hepatitis, autoimmune neuropathy, autoimmune ovarian failure, autoimmune orchitis, autoimmune thrombocytopenia, silicone implant associated autoimmune disease, Sjogren's syndrome, familial Mediterranean fever, systemic lupus erythematosus, vasculitis syndromes (for example, temporal, Takayasu's and giant cell arteritis, Behgefs disease or Wegener's granulomatosis), vitiligo, secondary hematologic manifestation of autoimmune diseases (for example, anemias), drug-induced autoimmunity, Hashimoto's thyroiditis, hypophysitis, idiopathic thrombocytic pupura, metal-induced autoimmunity, myasthenia gravis, pemphigus, autoimmune deafness (for example, Meniere's disease), Goodpasture's syndrome, Graves' disease, HW- r elated autoimmune syndromes, Gullain-Barre disease, Addison's disease, antiphospholipid syndrome, asthma, atopic dermatitis, Celiac disease, Cushing's syndrome, dermatomyositis, idiopathic adrenal adrenal atrophy, idiopathic thrombocytopenia, Kawasaki syndrome, Lambert-Eaton Syndrome, pernicious anemia, pollinosis, polyarteritis nodosa, primary biliary cirrhosis, primary sclerosing cholangitis, Raynaud's, Reiter's Syndrome, relapsing polychondritis, Schmidt's syndrome, thyrotoxidosis, sepsis, septic shock, endotoxic shock, exotoxin-induced toxic shock, gram negative sepsis, toxic shock syndrome, glomerulonephritis, peritonitis, interstitial cystitis, hyperoxia-induced inflammations, chronic obstructive pulmonary disease (COPD), vasculitis, graft vs. host reaction (for example, graft vs. host disease), allograft rejections (for example, acute allograft rejection or chronic allograft rejection), early transplantation rejection (for example, acute allograft rejection), reperfusion injury, pain (for example, acute pain, chronic pain, neuropathic pain, or fibromyalgia), chronic infections, meningitis, encephalitis, myocarditis, gingivitis, post-surgical trauma, tissue injury, traumatic brain injury, enterocolitis, sinusitis, uveitis, ocular inflammation, optic neuritis, gastric ulcers, esophagitis, peritonitis, periodontitis, dermatomyositis, gastritis, myositis, polymyalgia, pneumonia, and bronchitis. 72. The method of any one of claims 68-71, wherein the mammal is a male. 73. The method of any one of claims 68-71, wherein the mammal is a female. 74. The method of any one of claims 68-71, wherein the mammal is a primate, equine, bovine, ovine, feline, or canine. 75. The method of any one of claims 68-71, wherein the mammal is a human. 76. The method of any one of claims 40-75, wherein the compound is selected from the following table:
Figure imgf000173_0001
Figure imgf000174_0001
Figure imgf000175_0001
Figure imgf000176_0001
Figure imgf000177_0001
Figure imgf000178_0001
Figure imgf000179_0001
Figure imgf000180_0001
Figure imgf000181_0001
Figure imgf000182_0001
Figure imgf000183_0001
Figure imgf000184_0001
Figure imgf000185_0001
Figure imgf000186_0001
Figure imgf000187_0002
77. The method of any one of claims 40 or 42-75, wherein the compound is selected from the following table:
Figure imgf000187_0001
PCT/US2021/042208 2020-07-20 2021-07-19 Small molecule correctors of mammalian slc6a8 function WO2022020247A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US202063054000P 2020-07-20 2020-07-20
US63/054,000 2020-07-20

Publications (1)

Publication Number Publication Date
WO2022020247A1 true WO2022020247A1 (en) 2022-01-27

Family

ID=79729807

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2021/042208 WO2022020247A1 (en) 2020-07-20 2021-07-19 Small molecule correctors of mammalian slc6a8 function

Country Status (1)

Country Link
WO (1) WO2022020247A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115010620A (en) * 2022-02-21 2022-09-06 上海应用技术大学 Polyacid catalytic preparation method of phenylacetonitrile

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160130230A1 (en) * 2010-06-30 2016-05-12 Bayer Intellectual Property Gmbh Substituted dicyanopyridines and use thereof

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20160130230A1 (en) * 2010-06-30 2016-05-12 Bayer Intellectual Property Gmbh Substituted dicyanopyridines and use thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
DATABASE PubChem 27 January 2016 (2016-01-27), "SUBSTANCE RECORD SID 296117630", XP055901674, retrieved from NCBI Database accession no. 296117630 *
DATABASE Pubchem Substance 25 May 2018 (2018-05-25), "SUBSTANCE RECORD SID 369797099", XP055901676, retrieved from NCBI Database accession no. 369797099 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115010620A (en) * 2022-02-21 2022-09-06 上海应用技术大学 Polyacid catalytic preparation method of phenylacetonitrile
CN115010620B (en) * 2022-02-21 2023-05-30 上海应用技术大学 Polyacid catalytic preparation method of benzyl cyanide

Similar Documents

Publication Publication Date Title
US8710086B2 (en) Substituted di-arylhydantoin and di-arylthiohydantoin compounds and methods of use thereof
KR101081149B1 (en) A process for the manufacture of snac (salcaprozate sodium)
US9126962B2 (en) Substituted phenylcarbamoyl alkylamino arene compounds and N,N′-BIS-arylurea compounds
US20240043421A1 (en) Amino acid compounds and methods of use
TW200806633A (en) S1P receptor modulating compounds and use thereof
SI9112003A (en) (-)-//4-(1,4,5,6-tetrahydro-4-methyl-6-oxo-3-pyridazinyl) phenyl/hydrazono/propanedinitrile
US20230286904A1 (en) Inhibitors of yap/taz-tead oncoproteins, synthesis and use thereof
WO2024060911A1 (en) Benzoisothiazole compound, and pharmaceutical composition and use thereof
US20240043417A1 (en) Compounds for the treatment of sars
JP2022548250A (en) Thyroid hormone receptor beta agonist compounds
WO2022109598A1 (en) Integrin inhibitor and uses thereof
WO2022192370A1 (en) Small molecule inhibitors of mammalian slc6a19 function
US20160075683A1 (en) Novel heterocycle compounds and uses thereof
WO2022020247A1 (en) Small molecule correctors of mammalian slc6a8 function
TW200407310A (en) Compounds
US20210371403A1 (en) Small molecules targeting mutant mammalian proteins
WO2020061476A1 (en) Small molecules targeting mutant mammalian proteins
JP2005533088A (en) Production of naphthalimide containing amonafide and its pharmaceutical preparation
US20230303494A1 (en) Benzylamine derivative, preparation method therefor and use thereof
WO2024059005A1 (en) Treating pku with correctors of mammalian slc6a19 function
WO2024015569A2 (en) Small molecule inhibitors of mammalian slc6a19 function
WO2024015574A1 (en) Small molecule inhibitors of mammalian slc6a19 function
WO2023122267A2 (en) Small molecule inhibitors of mammalian slc6a19 function
US20140135399A1 (en) Novel polymorphs and salts
WO2022221182A1 (en) Small molecule inhibitors of mammalian slc34a1 function

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: 21846017

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21846017

Country of ref document: EP

Kind code of ref document: A1