WO2018089493A1 - Inhibiteurs pyrroles de mtorc et leurs utilisations - Google Patents

Inhibiteurs pyrroles de mtorc et leurs utilisations Download PDF

Info

Publication number
WO2018089493A1
WO2018089493A1 PCT/US2017/060640 US2017060640W WO2018089493A1 WO 2018089493 A1 WO2018089493 A1 WO 2018089493A1 US 2017060640 W US2017060640 W US 2017060640W WO 2018089493 A1 WO2018089493 A1 WO 2018089493A1
Authority
WO
WIPO (PCT)
Prior art keywords
mmol
methyl
piperazin
dimethyl
nitrogen
Prior art date
Application number
PCT/US2017/060640
Other languages
English (en)
Inventor
David John O'neill
Eddine Saiah
Seong Woo Anthony Kang
Andrew BREARLEY
Jonathan Bentley
Original Assignee
Navitor Pharmaceuticals, 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 Navitor Pharmaceuticals, Inc. filed Critical Navitor Pharmaceuticals, Inc.
Priority to US16/348,198 priority Critical patent/US20190389843A1/en
Publication of WO2018089493A1 publication Critical patent/WO2018089493A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/06Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a carbon chain containing only aliphatic carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D207/00Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom
    • C07D207/02Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom
    • C07D207/04Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members
    • C07D207/10Heterocyclic compounds containing five-membered rings not condensed with other rings, with one nitrogen atom as the only ring hetero atom with only hydrogen or carbon atoms directly attached to the ring nitrogen atom having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D207/16Carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals
    • 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
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • 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
    • C07D487/04Ortho-condensed systems

Definitions

  • the present invention relates to compounds and methods useful for modulating mTORCl activity.
  • the invention also provides pharmaceutically acceptable compositions comprising provided compounds of the present invention and methods of using such compositions in the treatment of various disorders.
  • rapamycin The mechanistic target of rapamycin (mTOR) signaling pathway integrates both intracellular and extracellular signals and serves as a central regulator of cell metabolism, growth, proliferation and survival.
  • mTOR rapamycin
  • mTOR inhibitors [rapamycin and its analogues (rapalogues)] in pathological settings, including the treatment of solid tumors, organ transplantation, coronary restenosis and rheumatoid arthritis.
  • mTOR complex 1 (mTORCl) positively regulates cell growth and proliferation by promoting many anabolic processes, including biosynthesis of proteins, lipids and organelles, and by limiting catabolic processes such as autophagy.
  • Much of the knowledge about mTORCl function comes from the use of the bacterial macrolide rapamycin. Upon entering the cell, rapamycin binds to FK506-binding protein of 12 kDa (FKBP12) and interacts with the FKBP12- rapamycin binding domain (FRB) of mTOR, thus inhibiting mTORCl functions (reviewed by Guertin and Sabatini, 2007).
  • mTORC2 mTOR complex 2
  • mTORC2 mTOR complex 2
  • mTORC2 mTORC2
  • this paradigm might not be entirely accurate, as chronic rapamycin treatment can, in some cases, inhibit mTORC2 activity by blocking its assembly (Sarbassov et al., 2006).
  • diseases are associated with abnormal cellular responses triggered by events as described above. These diseases include, but are not limited to, autoimmune diseases, inflammatory diseases, bone diseases, metabolic diseases, neurological and neurodegenerative diseases, cancer, cardiovascular diseases, allergies and asthma, Alzheimer's disease, and hormone-related diseases.
  • rapamycin complex 1 is a master growth regulator that senses diverse environmental cues, such as growth factors, cellular stresses, and nutrient and energy levels. When activated, mTORCl phosphorylates substrates that potentiate anabolic processes, such as mRNA translation and lipid synthesis, and limits catabolic ones, such as autophagy.
  • mTORCl dysregulation occurs in a broad spectrum of diseases, including diabetes, epilepsy, neurodegeneration, immune response, suppressed skeletal muscle growth, and cancer among others (Howell et al., (2013) Biochemical Society transactions 41, 906-912; Kim et al., (2013) Molecules and cells 35, 463-473; Laplante and Sabatini, (2012) Cell 149, 274-293). Accordingly, there remains a need to find protein kinase inhibitors useful as therapeutic agents.
  • Glucose transporters are a family of membrane proteins (GLUT1, 2, 3, 4, and 5) that facilitate the transport of glucose and other hexoses across cell membranes.
  • the transport of glucose into cells is one of the most important cellular transport events because of the role in maintaining normal cellular respiration and metabolism (Gould and Holman, (1993) Biochem J., 295, 329-341).
  • Dysfunction or dysregulation of glucose transporters may contribute to, or directly result in, disease states because of the central role the transporters play in cellular homeostasis and metabolism.
  • mutations in the GLUT1 gene are responsible for the rare autosomal disorder De Vivo disease, which is characterized by impaired glucose transport into the brain.
  • GLUT inhibition may normalize cellular metabolism and response in affected cells, including immune cells such as neutrophils. Therefore, GLUT inhibition would enable the treatment of cystic fibrosis, as well as autoimmune diseases characterized by abnormal GLUT expression or activity.
  • Compounds of the present invention, and pharmaceutically acceptable compositions thereof, are useful for treating a variety of diseases, disorders or conditions, associated with mTORCl .
  • diseases, disorders, or conditions include those described herein.
  • the present invention provides a compound of Formula I:
  • a 1 is N or CH
  • a 2 is N or CR', provided at least one of A 1 and A 2 comprises a nitrogen;
  • a 3 is N or CH
  • a 4 is N or CH
  • R' is H or C 1-6 aliphatic
  • R' and L 4 are optionally taken together with their intervening atoms to form a 5-8 membered saturated spiro-fused ring having 0-2 heteroatoms independently selected from nitrogen, oxygen or sulfur;
  • each R is independently hydrogen or an optionally substituted group selected from Ci-6 aliphatic, a 3-8 membered saturated or partially unsaturated monocyclic carbocyclic ring, phenyl, an 8-10 membered bicyclic aromatic carbocyclic ring, a 4-8 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 5-6 membered monocyclic heteroaromatic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or an 8-10 membered bicyclic heteroaromatic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
  • each of m, n, p, q, and x is independently 0, 1, or 2;
  • each of y and z is independently 0, 1, 2, 3, or 4;
  • each of R 1 and R 2 is independently R, or:
  • R 1 groups are optionally taken together to form a covalent bond or a bivalent Ci-4 alkylene chain
  • two R 2 groups are optionally taken together to form a covalent bond or a bivalent C1-4 alkylene chain
  • Ring A are optionally taken together with their intervening atoms to form a 5-8 membered aryl fused ring having 0-2 heteroatoms independently selected from nitrogen, oxygen or sulfur;
  • an R 2 group and Ring B are optionally taken together with their intervening atoms to form a 5-8 membered aryl fused ring having 0-2 heteroatoms independently selected from nitrogen, oxygen or sulfur; or
  • an R 2 group and L 3 are optionally taken together with their intervening atoms to form a 5-8 membered aryl fused ring having 0-2 heteroatoms independently selected from nitrogen, oxygen or sulfur;
  • each of R 3 is independently hydrogen, Ci-6 aliphatic, or -CN;
  • R 4 is hydrogen or an optionally substituted Ci-6 aliphatic group
  • Ring A is an optionally substituted ring selected from 6-membered aryl containing 0-2 nitrogen atoms, 5-membered heteroaryl with 1-4 heteroatoms independently selected from nitrogen, oxygen or sulfur, or 8-10 membered bicyclic aryl or heteroaryl with 1-4 heteroatoms independently selected from nitrogen, oxygen or sulfur;
  • Ring B is an optionally substituted ring selected from 6-membered aryl containing 0-2 nitrogen atoms, 5-membered heteroaryl with 1-4 heteroatoms independently selected from nitrogen, oxygen or sulfur, or 8-10 membered bicyclic aryl or heteroaryl with 1-4 heteroatoms independently selected from nitrogen, oxygen or sulfur;
  • L 1 is a covalent bond or a C1-3 bivalent straight or branched saturated or unsaturated hydrocarbon chain wherein 1-2 methylene units of the chain are independently and optionally replaced with -C(O)-, -N(R)-, or -CH(R)-;
  • L 2 is a covalent bond or a C1-3 bivalent straight or branched saturated or unsaturated hydrocarbon chain wherein 1-2 methylene units of the chain are independently and optionally replaced with -C(O)- or -CH(R)-;
  • L 3 is a covalent bond or -N(R)-
  • L 4 is a covalent bond or -N(R)-.
  • aliphatic or "aliphatic group”, as used herein, means a straight-chain (i.e., unbranched) or branched, substituted or unsubstituted hydrocarbon chain that is completely saturated or that contains one or more units of unsaturation, or a monocyclic hydrocarbon or bicyclic hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic (also referred to herein as "carbocycle,” “cycloaliphatic” or “cycloalkyl”), that has a single point of attachment to the rest of the molecule.
  • aliphatic groups contain 1-6 aliphatic carbon atoms.
  • aliphatic groups contain 1-5 aliphatic carbon atoms. In other embodiments, aliphatic groups contain 1-4 aliphatic carbon atoms. In still other embodiments, aliphatic groups contain 1-3 aliphatic carbon atoms, and in yet other embodiments, aliphatic groups contain 1-2 aliphatic carbon atoms.
  • cycloaliphatic (or “carbocycle” or “cycloalkyl”) refers to a monocyclic C3-C6 hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic, that has a single point of attachment to the rest of the molecule.
  • Suitable aliphatic groups include, but are not limited to, linear or branched, substituted or unsubstituted alkyl, alkenyl, alkynyl groups and hybrids thereof such as (cycloalkyl)alkyl, (cycloalkenyl)alkyl or (cycloalkyl)alkenyl.
  • heteroatom means one or more of oxygen, sulfur, nitrogen, phosphorus, or silicon (including, any oxidized form of nitrogen, sulfur, phosphorus, or silicon; the quaternized form of any basic nitrogen or; a substitutable nitrogen of a heterocyclic ring, for example N (as in 3,4-dihydro-2H-pyrrolyl), NH (as in pyrrolidinyl) or NR + (as in N-substituted pyrrolidinyl)).
  • Ci -8 saturated or unsaturated, straight or branched, hydrocarbon chain
  • bivalent Ci -8 or Ci-6 saturated or unsaturated, straight or branched, hydrocarbon chain
  • alkylene refers to a bivalent alkyl group.
  • An "alkylene chain” is a polymethylene group, i.e., -(CH2) n - wherein n is a positive integer, preferably from 1 to 6, from 1 to 4, from 1 to 3, from 1 to 2, or from 2 to 3.
  • a substituted alkylene chain is a polymethylene group in which one or more methylene hydrogen atoms are replaced with a substituent. Suitable substituents include those described below for a substituted aliphatic group.
  • alkenylene refers to a bivalent alkenyl group.
  • a substituted alkenylene chain is a polymethylene group containing at least one double bond in which one or more hydrogen atoms are replaced with a substituent. Suitable substituents include those described below for a substituted aliphatic group.
  • halogen means F, CI, Br, or I.
  • aryl used alone or as part of a larger moiety as in “aralkyl,” “aralkoxy,” or “aryloxyalkyl,” refers to monocyclic or bicyclic ring systems having a total of five to fourteen ring members, wherein at least one ring in the system is aromatic and wherein each ring in the system contains 3 to 7 ring members.
  • aryl may be used interchangeably with the term “aryl ring.”
  • aryl refers to an aromatic ring system which includes, but not limited to, phenyl, biphenyl, naphthyl, anthracyl and the like, which may bear one or more substituents.
  • aryl is a group in which an aromatic ring is fused to one or more non-aromatic rings, such as indanyl, phthalimidyl, naphthimidyl, phenanthridinyl, or tetrahydronaphthyl, and the like.
  • heteroaryl and “heteroar-,” used alone or as part of a larger moiety, e.g., “heteroaralkyl,” or “heteroaralkoxy,” refer to groups having 5 to 10 ring atoms, preferably 5, 6, or 9 ring atoms; having 6, 10, or 14 ⁇ electrons shared in a cyclic array; and having, in addition to carbon atoms, from one to five heteroatoms.
  • heteroatom refers to nitrogen, oxygen, or sulfur, and includes any oxidized form of nitrogen or sulfur, and any quaternized form of a basic nitrogen.
  • Heteroaryl groups include, without limitation, thienyl, furanyl, pyrrolyl, imidazolyl, pyrazolyl, triazolyl, tetrazolyl, oxazolyl, isoxazolyl, oxadiazolyl, thiazolyl, isothiazolyl, thiadiazolyl, pyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, indolizinyl, purinyl, naphthyridinyl, and pteridinyl.
  • heteroaryl and “heteroar-”, as used herein, also include groups in which a heteroaromatic ring is fused to one or more aryl, cycloaliphatic, or heterocyclyl rings, where the radical or point of attachment is on the heteroaromatic ring.
  • Nonlimiting examples include indolyl, isoindolyl, benzothienyl, benzofuranyl, dibenzofuranyl, indazolyl, benzimidazolyl, benzthiazolyl, quinolyl, isoquinolyl, cinnolinyl, phthalazinyl, quinazolinyl, quinoxalinyl, 4H-quinolizinyl, carbazolyl, acridinyl, phenazinyl, phenothiazinyl, phenoxazinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, and pyrido[2,3-b]-l,4-oxazin-3(4H)-one.
  • heteroaryl group may be mono- or bicyclic.
  • heteroaryl may be used interchangeably with the terms “heteroaryl ring,” “heteroaryl group,” or “heteroaromatic,” any of which terms include rings that are optionally substituted.
  • heteroarylkyl refers to an alkyl group substituted by a heteroaryl, wherein the alkyl and heteroaryl portions independently are optionally substituted.
  • heterocycle As used herein, the terms “heterocycle,” “heterocyclyl,” “heterocyclic radical,” and “heterocyclic ring” are used interchangeably and refer to a stable 5- to 7-membered monocyclic or 7-10-membered bicyclic heterocyclic moiety that is either saturated or partially unsaturated, and having, in addition to carbon atoms, one or more, preferably one to four, heteroatoms, as defined above.
  • nitrogen includes a substituted nitrogen.
  • the nitrogen may be N (as in 3,4-dihydro- 2H-pyrrolyl), H (as in pyrrolidinyl), or + R (as in N-substituted pyrrolidinyl).
  • a heterocyclic ring can be attached to its pendant group at any heteroatom or carbon atom that results in a stable structure and any of the ring atoms can be optionally substituted.
  • saturated or partially unsaturated heterocyclic radicals include, without limitation, tetrahydrofuranyl, tetrahydrothiophenyl pyrrolidinyl, piperidinyl, pyrrolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, decahydroquinolinyl, oxazolidinyl, piperazinyl, dioxanyl, dioxolanyl, diazepinyl, oxazepinyl, thiazepinyl, morpholinyl, and quinuclidinyl.
  • heterocycle used interchangeably herein, and also include groups in which a heterocyclyl ring is fused to one or more aryl, heteroaryl, or cycloaliphatic rings, such as indolinyl, 3H-indolyl, chromanyl, phenanthridinyl, or tetrahydroquinolinyl.
  • a heterocyclyl group may be mono- or bicyclic.
  • heterocyclylalkyl refers to an alkyl group substituted by a heterocyclyl, wherein the alkyl and heterocyclyl portions independently are optionally substituted.
  • partially unsaturated refers to a ring moiety that includes at least one double or triple bond.
  • partially unsaturated is intended to encompass rings having multiple sites of unsaturation, but is not intended to include aryl or heteroaryl moieties, as herein defined.
  • compounds of the invention may contain "optionally substituted” moieties.
  • substituted whether preceded by the term “optionally” or not, means that one or more hydrogens of the designated moiety are replaced with a suitable substituent.
  • an "optionally substituted” group may have a suitable substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position.
  • Combinations of substituents envisioned by this invention are preferably those that result in the formation of stable or chemically feasible compounds.
  • stable refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, and, in certain embodiments, their recovery, purification, and use for one or more of the purposes disclosed herein.
  • Suitable monovalent substituents on R° are independently halogen, -(CH 2 )o- 2 R*, -(haloR"), -(CH 2 )o- 2 OH, -(CH 2 ) 0 - 2 OR", -(CH 2 ) 0 - 2 CH(OR") 2 ; -O(haloR'), -CN, -N 3 , -(CH 2 ) 0 - 2 C(0)R', -(CH 2 )o- 2 C(0)OH, -(CH 2 )o- 2 C(0)OR', -(CH 2 ) 0 - 2 SR', -(CH 2 ) 0 - 2 SH, -(CH 2 ) 0 - 2 H 2 , - (CH 2 )o- 2 HR e , -(CH 2 )o- 2 R' 2 , -
  • Suitable divalent substituents that are bound to vicinal substitutable carbons of an "optionally substituted” group include: -0(CR* 2 ) 2 - 3 0-, wherein each independent occurrence of R* is selected from hydrogen, Ci-6 aliphatic which may be substituted as defined below, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • Suitable substituents on the aliphatic group of R * include halogen, -R", -(haloR"), -OH, -OR', -O(haloR'), -CN, -C(0)OH, -C(0)OR', - H 2 , -NHR", -NR' 2 , or -N0 2 , wherein each R" is unsubstituted or where preceded by "halo" is substituted only with one or more halogens, and is independently aliphatic, -CH 2 Ph, -0(CH 2 )o-iPh, or a 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • Suitable substituents on a substitutable nitrogen of an "optionally substituted" group include -R ⁇ , - R ⁇ 2 , -C(0)R ⁇ , -C(0)OR ⁇ , -C(0)C(0)R ⁇ , C(0)CH 2 C(0)R ⁇ , -S(0) 2 R ⁇ , -S(0) 2 R ⁇ 2 , -C(S) R ⁇ 2 , -C( H) R ⁇ 2 , or -N(R ⁇ )S(0) 2 R ⁇ ; wherein each R ⁇ is independently hydrogen, Ci-6 aliphatic which may be substituted as defined below, unsubstituted -OPh, or an unsubstituted 5-6-membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or, notwithstanding the definition above, two independent occurrences of R ⁇ , taken together with their intervening atom(s) form
  • Suitable substituents on the aliphatic group of R ⁇ are independently halogen, - R', -(haloR*), -OH, -OR', -O(haloR'), -CN, -C(0)OH, -C(0)OR', -NH 2 , -NHR', -NR' 2 , or -N0 2 , wherein each R* is unsubstituted or where preceded by "halo" is substituted only with one or more halogens, and is independently Ci- 4 aliphatic, -CH 2 Ph, -0(CH 2 )o-iPh, or a 5-6- membered saturated, partially unsaturated, or aryl ring having 0-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur.
  • the term "pharmaceutically acceptable salt” refers to those salts which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, allergic response and the like, and are commensurate with a reasonable benefit/risk ratio.
  • Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge et al., describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 1977, 66, 1-19, incorporated herein by reference.
  • Pharmaceutically acceptable salts of the compounds of this invention include those derived from suitable inorganic and organic acids and bases.
  • Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange.
  • inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid
  • organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange.
  • salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphor sulfonate, citrate, cyclopentanepropionate, digluconate, dodecyl sulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, gluconate, hemisulfate, heptanoate, hexanoate, hydroiodide, 2- hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, pamoate, pec
  • Salts derived from appropriate bases include alkali metal, alkaline earth metal, ammonium and salts.
  • Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium, and the like.
  • Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, loweralkyl sulfonate and aryl sulfonate.
  • structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, and geometric (or conformational)) forms of the structure; for example, the R and S configurations for each asymmetric center, Z and E double bond isomers, and Z and E conformational isomers. Therefore, single stereochemical isomers as well as enantiomeric, diastereomeric, and geometric (or conformational) mixtures of the present compounds are within the scope of the invention. Unless otherwise stated, all tautomeric forms of the compounds of the invention are within the scope of the invention.
  • structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms.
  • compounds having the present structures including the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13 C- or 14 C-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.
  • measurable affinity and “measurably inhibit,” as used herein, means a measurable change in mTORCl activity between a sample comprising a compound of the present invention, or composition thereof, and mTORCl, and an equivalent sample comprising mTORCl in the absence of said compound, or composition thereof.
  • the present invention provides a compound of Formula I:
  • a 1 is N or CH
  • a 2 is N or CR', provided at least one of A 1 and A 2 comprises a nitrogen;
  • a 3 is N or CH
  • a 4 is N or CH
  • R' is H or C i- 6 aliphatic
  • R' and L 4 are optionally taken together with their intervening atoms to form a 5-8 membered saturated spiro-fused ring having 0-2 heteroatoms independently selected from nitrogen, oxygen or sulfur;
  • each R is independently hydrogen or an optionally substituted group selected from Ci-6 aliphatic, a 3-8 membered saturated or partially unsaturated monocyclic carbocyclic ring, phenyl, an 8-10 membered bicyclic aromatic carbocyclic ring, a 4-8 membered saturated or partially unsaturated monocyclic heterocyclic ring having 1-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur, a 5-6 membered monocyclic heteroaromatic ring having 1-4 heteroatoms independently selected from nitrogen, oxygen, or sulfur, or an 8-10 membered bicyclic heteroaromatic ring having 1-5 heteroatoms independently selected from nitrogen, oxygen, or sulfur;
  • each of m, n, p, q, and x is independently 0, 1, or 2;
  • each of y and z is independently 0, 1, 2, 3, or 4;
  • each of R 1 and R 2 is independently R, or:
  • R 1 groups are optionally taken together to form a covalent bond or a bivalent Ci-4 alkylene chain;
  • R 2 groups are optionally taken together to form a covalent bond or a bivalent Ci-4 alkylene chain;
  • Ring A are optionally taken together with their intervening atoms to form a 5-8 membered aryl fused ring having 0-2 heteroatoms independently selected from nitrogen, oxygen or sulfur;
  • an R 2 group and Ring B are optionally taken together with their intervening atoms to form a 5-8 membered aryl fused ring having 0-2 heteroatoms independently selected from nitrogen, oxygen or sulfur; or
  • an R 2 group and L 3 are optionally taken together with their intervening atoms to form a 5-8 membered aryl fused ring having 0-2 heteroatoms independently selected from nitrogen, oxygen or sulfur;
  • each of R 3 is independently hydrogen, Ci-6 aliphatic, or -CN;
  • R 4 is hydrogen or an optionally substituted Ci-6 aliphatic group
  • Ring A is an optionally substituted ring selected from 6-membered aryl containing 0-2 nitrogen atoms, 5-membered heteroaryl with 1-4 heteroatoms independently selected from nitrogen, oxygen or sulfur, or 8-10 membered bicyclic aryl or heteroaryl with 1-4 heteroatoms independently selected from nitrogen, oxygen or sulfur;
  • Ring B is an optionally substituted ring selected from 6-membered aryl containing 0-2 nitrogen atoms, 5-membered heteroaryl with 1-4 heteroatoms independently selected from nitrogen, oxygen or sulfur, or 8-10 membered bicyclic aryl or heteroaryl with 1-4 heteroatoms independently selected from nitrogen, oxygen or sulfur;
  • L 1 is a covalent bond or a C1-3 bivalent straight or branched saturated or unsaturated hydrocarbon chain wherein 1-2 methylene units of the chain are independently and optionally replaced with -C(O)-, -N(R)-, or -CH(R)-;
  • L 2 is a covalent bond or a C1-3 bivalent straight or branched saturated or unsaturated hydrocarbon chain wherein 1-2 methylene units of the chain are independently and optionally replaced with -C(O)- or -CH(R)-;
  • L 3 is a covalent bond or -N(R)-
  • L 4 is a covalent bond or -N(R)-.
  • a 1 is N or CH. In some embodiments, A 1 is N. In some embodiments, A 1 is CH.
  • a 1 is selected from those depicted in Table 1, below.
  • a 2 is N or CR', provided at least one of A 1 and
  • a 1 comprises a nitrogen.
  • a 2 is N.
  • a 2 is CR' .
  • at least one of A 1 and A 2 comprises a nitrogen.
  • a 2 is selected from those depicted in Table 1, below.
  • each of m, and n is independently 0, 1, or 2.
  • m is 0.
  • n is 0.
  • m is 1.
  • n is 1.
  • m is 2.
  • n is 2.
  • each of p, q, and x is independently 0, 1, or 2.
  • p is 0. In some embodiments, q is 0. In some embodiments, x is 0. In some embodiments, p is 1. In some embodiments, q is 1. In some embodiments, x is 1. In some embodiments, p is 2. In some embodiments, q is 2. In some embodiments, x is 2.
  • each of y and z is independently 0, 1, 2, 3, or
  • y is 0. In some embodiments, z is 0. In some embodiments, y is 1. In some embodiments, z is 1. In some embodiments, y is 2. In some embodiments, z is 2. In some embodiments, y is 3. In some embodiments, z is 3. In some embodiments, y is 4. In some embodiments, z is 4.
  • a 3 is N or CH. In some embodiments, A 3 is N. In some embodiments, A 3 is CH.
  • a 3 is selected from those depicted in Table 1, below. [0044] As defined above and described herein, A 4 is N or CH. In some embodiments, A 4 is N. In some embodiments, A 4 is CH.
  • a 4 is selected from those depicted in Table 1, below.
  • R' is H or C i- 6 aliphatic, or R' and L 4 are optionally taken together with their intervening atoms to form a 5-8 membered saturated spiro- fused ring having 0-2 heteroatoms independently selected from nitrogen, oxygen or sulfur.
  • R' is H.
  • R' is Ci-6 aliphatic.
  • R' and L 4 are optionally taken together with their intervening atoms to form a 5-8 membered saturated spiro-fused ring having 0-2 heteroatoms independently selected from nitrogen, oxygen or sulfur.
  • R' and L 4 are optionally taken together with their intervening atoms to form a 5 membered saturated spiro-fused ring having one nitrogen.
  • R' is selected from those depicted in Table 1, below.
  • two R 2 groups are optionally taken together to form a covalent bond. In some embodiments, two R 2 groups are optionally taken together to form a bivalent C 1-4 alkylene chain. In some embodiments, an R 2 group and Ring B are optionally taken together with their intervening atoms to form a 5-8 membered fused ring having 0-2 heteroatoms independently selected from nitrogen, oxygen, or sulfur. In some embodiments, an R 2 group and L 3 are optionally taken together with their intervening atoms to form a 5-8 membered aryl fused ring having 0-2 heteroatoms independently selected from nitrogen, oxygen or sulfur.
  • each of R 1 and R 2 is independently selected from those depicted in Table 1, below.
  • each of R 3 is independently hydrogen, Ci- 6 aliphatic, or -CN.
  • R 3 is hydrogen.
  • R 3 is Ci-6 aliphatic.
  • R 3 is -CN.
  • R 3 is methyl.
  • R 3 is independently selected from those depicted in Table 1, below.
  • R 4 is hydrogen or an optionally substituted Ci- 6 aliphatic group. In some embodiments, R 4 is hydrogen. In some embodiments, R 4 is n optionally substituted Ci- 6 aliphatic group. In some embodiments, R 4 is methyl. In some embodiments, R 4 is eth embo some
  • R 4 is In some embodiments, R 4 is . In some embodiments, R 4 is OCH3 j n some embodiments , R 4 is
  • R 4 is
  • R 4 is is selected from those depicted in Table 1, below.
  • Ring A is an optionally substituted ring selected from 6-membered aryl containing 0-2 nitrogen atoms, 5-membered heteroaryl with 1-4 heteroatoms independently selected from nitrogen, oxygen or sulfur, or 8-10 membered bicyclic aryl or heteroaryl with 1-4 heteroatoms independently selected from nitrogen, oxygen or sulfur.
  • Ring B is an optionally substituted ring selected from 6-membered aryl containing 0-2 nitrogen atoms, 5-membered heteroaryl with 1-4 heteroatoms independently selected from nitrogen, oxygen or sulfur, or 8-10 membered bicyclic aryl or heteroaryl with 1-4 heteroatoms independently selected from nitrogen, oxygen or sulfur.
  • Ring B is phenyl, pyridyl, pyrimidinyl, pyrazinyl, pyridazinyl,
  • Ring B is
  • L 1 is a covalent bond or a C1-3 bivalent straight or branched saturated or unsaturated hydrocarbon chain wherein 1-2 methylene units of the chain are independently and optionally replaced with -C(O)-, -N(R)-, or -CH(R)-.
  • L 1 is a covalent bond.
  • L 1 is -CH 2 -.
  • L 1 is . In some embodiments, L 1 is . In some embodiments,
  • L 1 is selected from those depicted in Table 1 , below.
  • L 2 is a covalent bond or a C1-3 bivalent straight or branched saturated or unsaturated hydrocarbon chain wherein 1 -2 methylene units of the chain are independently and optionally replaced with -C(O)- or -CH(R)-.
  • L 2 is a covalent bond. In some embodiments, L 2 is -CH2-. In
  • L is '
  • L 2 is selected from those depicted in Table 1 , below.
  • L 3 is a covalent bond or -N(R)-.
  • L 3 is a covalent bond. In some embodiments, L 3 is -N(R)-. In some embodiments, L 3 is - H-.
  • L 3 is selected from those depicted in Table 1 , below.
  • L 4 is a covalent bond or -N(R)-.
  • L 4 is a covalent bond. In some embodiments, L 4 is -N(R)-. In some embodiments, L 4 is - H-.
  • L 4 is selected from those depicted in Table 1 , below.
  • the present invention provides a compound of Formulae la, I- b. or I-c:
  • a 1 , A 2 , Ring B, m, n, x, and y is defined above and described in embodiments herein.
  • the present invention provides a compound of Formula I-d:
  • Ring A, A 3 , A 4 , R 1 , R 2 , R 3 , R 4 , L 2 ,L 3 , L 4 , Ring B, p, q, x, y, and z is defined above and described in embodiments herein.
  • the present invention provides a compound set forth in Table 1, above, or a pharmaceutically acceptable salt thereof. It will be appreciated that the present invention also provides a compound set forth in Table 1, above, as a racemic mixture at the C7 position, or a pharmaceutically acceptable salt thereof.
  • compositions are provided.
  • the invention provides a composition comprising a compound of this invention or a pharmaceutically acceptable derivative thereof and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
  • the amount of compound in compositions of this invention is such that is effective to measurably inhibit mTORCl, in a biological sample or in a patient.
  • the amount of compound in compositions of this invention is such that is effective to measurably inhibit mTORCl, in a biological sample or in a patient.
  • a composition of this invention is formulated for administration to a patient in need of such composition.
  • a composition of this invention is formulated for oral administration to a patient.
  • patient means an animal, preferably a mammal, and most preferably a human.
  • compositions of this invention refers to a nontoxic carrier, adjuvant, or vehicle that does not destroy the pharmacological activity of the compound with which it is formulated.
  • Pharmaceutically acceptable carriers, adjuvants or vehicles that may be used in the compositions of this invention include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, cellulose-based substances, polyethylene glycol, sodium carboxymethylcellulose, polyacrylates, waxes, polyethylene- polyoxypropylene
  • compositions of the present invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir.
  • parenteral as used herein includes subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques.
  • the compositions are administered orally, intraperitoneally or intravenously.
  • Sterile injectable forms of the compositions of this invention may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a non-toxic parenterally acceptable diluent or solvent, for example as a solution in 1,3-butanediol.
  • a non-toxic parenterally acceptable diluent or solvent for example as a solution in 1,3-butanediol.
  • acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or di-glycerides.
  • Fatty acids such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxy ethyl ated versions.
  • These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersing agents that are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions.
  • Other commonly used surfactants such as Tweens, Spans and other emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation.
  • compositions of this invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions.
  • carriers commonly used include lactose and corn starch.
  • Lubricating agents such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried cornstarch.
  • aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added.
  • compositions of this invention may be administered in the form of suppositories for rectal administration.
  • suppositories for rectal administration.
  • suppositories can be prepared by mixing the agent with a suitable non-irritating excipient that is solid at room temperature but liquid at rectal temperature and therefore will melt in the rectum to release the drug.
  • suitable non-irritating excipient include cocoa butter, beeswax and polyethylene glycols.
  • compositions of this invention may also be administered topically, especially when the target of treatment includes areas or organs readily accessible by topical application, including diseases of the eye, the skin, or the lower intestinal tract. Suitable topical formulations are readily prepared for each of these areas or organs.
  • Topical application for the lower intestinal tract can be effected in a rectal suppository formulation (see above) or in a suitable enema formulation. Topically-transdermal patches may also be used.
  • compositions may be formulated in a suitable ointment containing the active component suspended or dissolved in one or more carriers.
  • Carriers for topical administration of compounds of this invention include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water.
  • provided pharmaceutically acceptable compositions can be formulated in a suitable lotion or cream containing the active components suspended or dissolved in one or more pharmaceutically acceptable carriers.
  • Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
  • compositions may be formulated as micronized suspensions in isotonic, pH adjusted sterile saline, or, preferably, as solutions in isotonic, pH adjusted sterile saline, either with or without a preservative such as benzylalkonium chloride.
  • the pharmaceutically acceptable compositions may be formulated in an ointment such as petrolatum.
  • compositions of this invention may also be administered by nasal aerosol or inhalation. Such compositions are prepared according to techniques well- known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other conventional solubilizing or dispersing agents. [0088] Most preferably, pharmaceutically acceptable compositions of this invention are formulated for oral administration. Such formulations may be administered with or without food. In some embodiments, pharmaceutically acceptable compositions of this invention are administered without food. In other embodiments, pharmaceutically acceptable compositions of this invention are administered with food.
  • compositions of the present invention that may be combined with the carrier materials to produce a composition in a single dosage form will vary depending upon the host treated, the particular mode of administration.
  • provided compositions should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of the inhibitor can be administered to a patient receiving these compositions.
  • a specific dosage and treatment regimen for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, and the judgment of the treating physician and the severity of the particular disease being treated.
  • the amount of a compound of the present invention in the composition will also depend upon the particular compound in the composition.
  • the activity of a compound utilized in this invention as an inhibitor of mTORCl may be assayed in vitro, in vivo or in a cell line.
  • In vitro assays include assays that determine the inhibition of mTORCl .
  • Detailed conditions for assaying a compound utilized in this invention as an inhibitor of mTORCl are well known to one of ordinary skill in the art. Such methods are described in detail by Liu et al., Cancer Research, 73(8), April 15, 2013 and Liu et al., J. Biological Chemistry, vol 287, no. 13, pp 9742-9752 (2012).
  • treatment refers to reversing, alleviating, delaying the onset of, or inhibiting the progress of a disease or disorder, or one or more symptoms thereof, as described herein.
  • treatment may be administered after one or more symptoms have developed.
  • treatment may be administered in the absence of symptoms.
  • treatment may be administered to a susceptible individual prior to the onset of symptoms (e.g., in light of a history of symptoms and/or in light of genetic or other susceptibility factors). Treatment may also be continued after symptoms have resolved, for example to prevent or delay their recurrence.
  • the present invention provides a method for treating an mTORCl -mediated disorder comprising the step of administering to a patient in need thereof a compound of the present invention, or pharmaceutically acceptable composition thereof.
  • mTORCl -mediated disorders, diseases, and/or conditions means any disease or other deleterious condition in which mTORCl, is known to play a role. Accordingly, another embodiment of the present invention relates to treating or lessening the severity of one or more diseases in which mTORCl is known to play a role.
  • an mTORCl -mediated disorder, disease, and/or condition is selected from those described by Matt Kaeberlin, Scientifica, vol. 2013, Article ID 849186.
  • the methods described herein include methods for the treatment of cancer in a subject.
  • to "treat” means to ameliorate or improve at least one symptom or clinical parameter of the cancer.
  • a treatment can result in a reduction in tumor size or growth rate.
  • a treatment need not cure the cancer or cause remission 100% of the time, in all subjects.
  • cancer refers to cells having the capacity for autonomous growth, i.e., an abnormal state or condition characterized by rapidly proliferating cell growth.
  • the term is meant to include all types of cancerous growths or oncogenic processes, metastatic tissues or malignantly transformed cells, tissues, or organs, irrespective of histopathologic type or stage of invasiveness.
  • tumor refers to cancerous cells, e.g., a mass of cancer cells.
  • Cancers that can be treated or diagnoses using the methods described herein include malignancies of the various organ systems, such as affecting lung, breast, thyroid, lymphoid, gastrointestinal, and genito-urinary tract, as well as adenocarcinomas which include malignancies such as most colon cancers, renal-cell carcinoma, prostate cancer and/or testicular tumors, non- small cell carcinoma of the lung, cancer of the small intestine and cancer of the esophagus.
  • the methods described herein are used for treating or diagnosing a carcinoma in a subject.
  • carcinoma is art recognized and refers to malignancies of epithelial or endocrine tissues including respiratory system carcinomas, gastrointestinal system carcinomas, genitourinary system carcinomas, testicular carcinomas, breast carcinomas, prostatic carcinomas, endocrine system carcinomas, and melanomas.
  • the cancer is renal carcinoma or melanoma.
  • Exemplary carcinomas include those forming from tissue of the cervix, lung, prostate, breast, head and neck, colon and ovary.
  • carcinosarcomas e.g., which include malignant tumors composed of carcinomatous and sarcomatous tissues.
  • An "adenocarcinoma” refers to a carcinoma derived from glandular tissue or in which the tumor cells form recognizable glandular structures.
  • sarcoma is art recognized and refers to malignant tumors of mesenchymal derivation.
  • the cancers that are treated by the methods described herein are cancers that have increased levels of mTORC 1 or an increased expression or activity of a mTORC 1 relative to normal tissues or to other cancers of the same tissues; methods known in the art and described herein can be used to identify those cancers.
  • the methods include obtaining a sample comprising cells of the cancer, determining the mTORC 1 activity in the sample, and administering a treatment as described herein (e.g., a provided inhibitor of mTORCl).
  • the cancer is one that is shown herein to have increased levels of mTORCl activity
  • the present invention provides a method for treating one or more disorders, diseases, and/or conditions wherein the disorder, disease, or condition includes, but is not limited to, a cellular proliferative disorder.
  • the present invention features methods and compositions for the diagnosis and prognosis of cellular proliferative disorders (e.g., cancer) and the treatment of these disorders by inhibiting mTORCl activity.
  • cellular proliferative disorders described herein include, e.g., cancer, obesity, and proliferation-dependent diseases. Such disorders may be diagnosed using methods known in the art.
  • Cancers include, without limitation, leukemias (e.g., acute leukemia, acute lymphocytic leukemia, acute myelocytic leukemia, acute myeloblastic leukemia, acute promyelocyte leukemia, acute myelomonocytic leukemia, acute monocytic leukemia, acute erythroleukemia, chronic leukemia, chronic myelocytic leukemia, chronic lymphocytic leukemia), polycythemia vera, lymphoma (e.g., Hodgkin's disease or non-Hodgkin's disease), Waldenstrom's macroglobulinemia, multiple myeloma, heavy chain disease, and solid tumors such as sarcomas and carcinomas (e.g., fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endot
  • Idiopathic Pulmonary Fibrosis IPF.
  • the PI3K pathway is activated in fibrotic foci, the cardinal lesions in IPF.
  • mTOR kinase inhibitor GSK2126458 reduces PI3K pathway signaling and functional responses in IPF-derived lung fibroblasts and mTOR inhibition reduces collagen expression in models of IPF patients.
  • rapamycin treatment is antifibrotic, and rapamycin also decreases expression of a-smooth muscle actin and fibronectin by fibroblasts in vitro.
  • the method of inhibiting mTORCl activity is used to treat idiopathic pulmonary fibrosis (IPF).
  • IPF idiopathic pulmonary fibrosis
  • the present invention provides a method of treating idiopathic pulmonary fibrosis (IPF), in a patient in need thereof, comprising the step of administering to said patient a provided compound or pharmaceutically acceptable salt thereof.
  • Kidney Fibrosis mTORCl is activated in myofibroblasts, a major pathogenic cell type in kidney fibrosis. Inhibition of mTOR with rapamycin in a murine model of kidney fibrosis (UUO), attenuated expression of markers of fibrosis and tubulointerstitial damage.
  • UUO murine model of kidney fibrosis
  • the method of inhibiting mTORCl activity is used to treat kidney fibrosis.
  • kidney fibrosis See J Am Soc Nephrol 2013, 24, pp. 1114-1126; Kidney International 2006, 69, pp. 2029-2036; PLoS 2012,7, Issue 3, e33626; Clin Invest Med 2014, Vol 37, no 3, E142).
  • the present invention provides a method of treating kidney fibrosis, in a patient in need thereof, comprising the step of administering to said patient a provided compound or pharmaceutically acceptable salt thereof.
  • the method of inhibiting mTORCl activity is used to treat scleroderma.
  • the present invention provides a method of treating scleroderma, in a patient in need thereof, comprising the step of administering to said patient a provided compound or pharmaceutically acceptable salt thereof.
  • the method of inhibiting mTORCl activity is used to treat hypertrophic scarring and keloid disease.
  • the present invention provides a method of treating hypertrophic scarring and keloid disease, in a patient in need thereof, comprising the step of administering to said patient a provided compound or pharmaceutically acceptable salt thereof.
  • the method of inhibiting mTORCl activity is used to treat cardiac fibrosis.
  • the present invention provides a method of treating cardiac fibrosis, in a patient in need thereof, comprising the step of administering to said patient a provided compound or pharmaceutically acceptable salt thereof.
  • proliferative diseases include, e.g., obesity, benign prostatic hyperplasia, psoriasis, abnormal keratinization, lymphoproliferative disorders (e.g., a disorder in which there is abnormal proliferation of cells of the lymphatic system), chronic rheumatoid arthritis, arteriosclerosis, restenosis, and diabetic retinopathy.
  • lymphoproliferative disorders e.g., a disorder in which there is abnormal proliferation of cells of the lymphatic system
  • chronic rheumatoid arthritis e.g., arteriosclerosis, restenosis, and diabetic retinopathy.
  • Proliferative diseases that are hereby incorporated by reference include those described in U.S. Pat. Nos. 5,639,600 and 7,087,648.
  • Other Disorders include those described in U.S. Pat. Nos. 5,639,600 and 7,087,648.
  • Other disorders include lysosomal storage diseases, including but not limited to Pompe disease, Gaucher disease, mucopolysaccharidosis, multiple sulfatase deficiency; neurodegenerative diseases such as Parkinson's disease, Alzheimer's disease, Huntington's disease, alphal -anti -trypsin deficiency, and spinal bulbar muscular atrophy.
  • the present invention provides compounds that were shown to cause translocation of TFEB to the nucleus. TFEB translocation to the nucleus promotes exocytosis and/or cellular clearance of accumulating substrates in the above-mentioned diseases.
  • the method of inhibiting mTORCl activity is used to treat asthma. (See Respirology 2015 Oct; 20(7): 1055-65). Accordingly, in some embodiments, the present invention provides a method of treating asthma, in a patient in need thereof, comprising the step of administering to said patient a provided compound or pharmaceutically acceptable salt thereof.
  • the method of inhibiting mTORCl activity is used to treat a lysosomal storage disease.
  • a lysosomal storage disease ⁇ See Annals of the New York Academy of Sciences, 2016, Volume 1371, Issue 1, pp. 3-14; Hum Mol Genet. 2015, 24(20), pp. 5775-88; EMBO Mol Med. 2013, 5(5), pp. 691-706; Medina, D.L., et al., Dev Cell. 2011 Sep 13, 21(3), pp. 421-30).
  • the present invention provides a method of treating a lysosomal storage disease, in a patient in need thereof, comprising the step of administering to said patient a provided compound or pharmaceutically acceptable salt thereof.
  • the method of inhibiting mTORCl activity is used to treat Parkinson's disease.
  • the present invention provides a method of treating Parkinson's disease, in a patient in need thereof, comprising the step of administering to said patient a provided compound or pharmaceutically acceptable salt thereof.
  • the method of inhibiting mTORCl activity is used to treat Alzheimer's disease. ⁇ See EMBO Mol Med. 2014, 6(9), pp. 1142-60). Accordingly, in some embodiments, the present invention provides a method of treating Alzheimer's disease, in a patient in need thereof, comprising the step of administering to said patient a provided compound or pharmaceutically acceptable salt thereof.
  • the method of inhibiting mTORCl activity is used to treat Huntington's disease. ⁇ See Sci Transl Med. 2012, 4(142): 142ra97). Accordingly, in some embodiments, the present invention provides a method of treating Huntingtons's disease, in a patient in need thereof, comprising the step of administering to said patient a provided compound or pharmaceutically acceptable salt thereof.
  • the method of inhibiting mTORCl activity is used to treat alphal -anti -trypsin deficiency.
  • the present invention provides a method of treating alphal -anti -trypsin deficiency, in a patient in need thereof, comprising the step of administering to said patient a provided compound or pharmaceutically acceptable salt thereof.
  • the method of inhibiting mTORC 1 activity is used to treat spinal bulbar muscular atrophy.
  • the present invention provides a method of treating spinal bulbar muscular atrophy, in a patient in need thereof, comprising the step of administering to said patient a provided compound or pharmaceutically acceptable salt thereof.
  • the present invention provides compounds that are inhibitors of mTORCl activity and were shown to selectively inhibit mTORCl over mTORC2 as measured by pS6K inhibition (a measure of mTORCl activity) and pAKT activation (a measure of mTORC2 activity).
  • a provided compound inhibits mTORCl selectively over mTORC2.
  • a provided compound does not measurably inhibit mTORC2.
  • a provided compound has a pAKT activation IC50 of >10 ⁇ .
  • a provided compound inhibits mTORCl with >10-fold selectivity over mTORC2.
  • a provided compound inhibits mTORCl with >20-fold selectivity over mTORC2. In some embodiments, a provided compound inhibits mTORCl with >50-fold selectivity over mTORC2. In some embodiments, a provided compound inhibits mTORCl with >100-fold selectivity over mTORC2. In some embodiments, a provided compound inhibits mTORCl with >150-fold selectivity over mTORC2. In some embodiments, a provided compound inhibits mTORCl with >200-fold selectivity over mTORC2. In some embodiments, a provided compound inhibits mTORCl with >500-fold selectivity over mTORC2.
  • a provided compound inhibits mTORCl with >1, 000-fold selectivity over mTORC2. Accordingly, in some embodiments, the present invention provides a method of treating a disorder associate with mTORCl comprising administering to patient a compound that inhibits mTORCl wherein said compound does not inhibit mTORC2.
  • Such compounds may be employed for indications where rapamycin and rapalogs demonstrated a benefit either in animal models or in a human disease setting. Such indications include:
  • the method of inhibiting mTORCl activity is used to treat metabolic disease (obesity and insulin resistance in type 2 diabetes).
  • metabolic disease ovalbuproliferative disease
  • the present invention provides a method of treating metabolic disease (obesity and insulin resistance in type 2 diabetes), in a patient in need thereof, comprising the step of administering to said patient a provided compound or pharmaceutically acceptable salt thereof.
  • Neurofibromatosis Neurofibromatosis type 1 (NF1) is caused by mutations in the NF1 gene. Its protein product, neurofibromin, functions as a tumor suppressor and ultimately produces constitutive upregulation of mTOR. mTOR inhibitors have been shown to reduce tumor size and induce anti-proliferative effect in NF1 -associated plexiform neurofibroma.
  • the method of inhibiting mTORCl activity is used to treat neurofibromatosis. ⁇ See Curr Neurol Neurosci Rep. 2012 Jun 12(3), pp. 294-301; Oncotarget. 2016 Jan 31). Accordingly, in some embodiments, the present invention provides a method of treating neurofibromatosis, in a patient in need thereof, comprising the step of administering to said patient a provided compound or pharmaceutically acceptable salt thereof.
  • LMNA Emery-Dreifuss muscular dystrophy model
  • Mutations in LMNA result in several human diseases including limb-girdle muscular dystrophy (LGMDIB), Emery-Dreifuss muscular dystrophy (EDMD2/3), dilated cardiomyopathy (DCM) and conduction-system disease (CMDIA), lipodystrophy, Charcot- Marie-Tooth disease, and Hutchinson-Gilford progeria syndrome (HGPS).
  • LGMDIB limb-girdle muscular dystrophy
  • EDMD2/3 Emery-Dreifuss muscular dystrophy
  • DCMDIA conduction-system disease
  • lipodystrophy Charcot- Marie-Tooth disease
  • HGPS Hutchinson-Gilford progeria syndrome
  • Lmna _/" mice have elevated mTORCl activity and short-term treatment with rapamycin in Lmna _/” mice results in reduced mTORCl signaling, improved cardiac and skeletal muscle function and enhanced survival
  • the method of inhibiting mTORCl activity is used to treat cardiomyopathy and skeletal muscle dystrophy. ⁇ See Sci Transl Med. 2012, 4(144): 144ral03; Handb Clin Neurol. 2013, 113, pp. 1367-76). Accordingly, in some embodiments, the present invention provides a method of treating cardiomyopathy and skeletal muscle dystrophy, in a patient in need thereof, comprising the step of administering to said patient a provided compound or pharmaceutically acceptable salt thereof.
  • Ndufs4 knockout mice are used as a model of Leigh syndrome and exhibit hyperactivation of mTORCl and metabolic defects. Treatment of Ndufs4 KO mice with rapamycin extended lifespan, improve metabolic and neurological defect associated with this disease.
  • the method of inhibiting mTORCl activity is used to treat Leigh syndrome. ⁇ See Science 2013, 342(6165), pp. 1524-8). Accordingly, in some embodiments, the present invention provides a method of treating Leigh syndrome, in a patient in need thereof, comprising the step of administering to said patient a provided compound or pharmaceutically acceptable salt thereof.
  • Oncology Inhibition of mTOR with rapalogs has been shown to have antitumor activity in murine cancer models and in cancer patients.
  • sensitive cancer types include, but are not limited to, hepatocellular carcinoma, breast cancers, mantle cell lymphomas, lung carcinoma, tuberous sclerosis and lymphangioleiomyomatosis.
  • the method of inhibiting mTORCl activity is used to treat cancer and oncologic disorders. ⁇ See Trends Cancer 2016; In press). Accordingly, in some embodiments, the present invention provides a method of treating cancer and oncologic disorders, in a patient in need thereof, comprising the step of administering to said patient a provided compound or pharmaceutically acceptable salt thereof.
  • Non-alcoholic steatohepatitis (NASH).
  • the present invention provides inhibitors that induce autophagy to clear degraded cytoplasmic proteins, and NASH disease is characterized by lipid deposits, inflammation and fibrosis in the liver.
  • the inhibition of mTORCl pathway induce autophagy and down regulate SREBP-1 to decrease lipid biosynthesis to reduce lipid storage.
  • the method of inhibiting mTORC 1 activity is used to treat nonalcoholic steatohepatitis (NASH).
  • NASH nonalcoholic steatohepatitis
  • the present invention provides a method of treating non-alcoholic steatohepatitis (NASH), in a patient in need thereof, comprising the step of administering to said patient a provided compound or pharmaceutically acceptable salt thereof.
  • TSC Tuberous sclerosis
  • LAM lymphangioleiomyomatosis
  • TSC inherited disorder tuberous sclerosis complex
  • LAM lymphangioleiomyomatosis
  • Both diseases are caused by mutations of TSC1 or TSC2 leading to inappropriate activity of signaling downstream of mTORCl .
  • TSC patients develop nonmalignant tumors in many organs, including the brain, while LAM patients, mostly women, accumulate abnormal, muscle-like cells in certain organs or tissues, especially the lungs, lymph nodes, and kidneys.
  • the rapalogs, Everolimus and Sirolimus are currently approved for the treatment of both TSC and LAM, respectively, by the US FDA.
  • the method of inhibiting mTORCl activity is used to treat tuberous sclerosis and lymphangioleiomyomatosis.
  • the present invention provides a method of treating tuberous sclerosis and lymphangioleiomyomatosis, in a patient in need thereof, comprising the step of administering to said patient a provided compound or pharmaceutically acceptable salt thereof.
  • Rapamycin suppresses the mammalian TORC1 complex, which regulates translation, and extends lifespan in diverse species, including mice. Rapamycin was shown to inhibit the pro-inflammatory phenotype of senescent cells. As senescent cells accumulate with age, the senescence-associated secretory phenotype (SASP) can disrupt tissues and contribute to age-related pathologies, including cancer. Inhibition of mTOR suppressed the secretion of inflammatory cytokines by senescent cells. Rapamycin reduced cytokine levels including IL6 and suppressed translation of the membrane-bound cytokine ILIA. Reduced ILIA diminishes F- ⁇ transcriptional activity, which controls the SASP. Thus, mTORCl inhibitors might ameliorate age-related pathologies, including late-life cancer, by suppressing senescence- associated inflammation.
  • SASP senescence-associated secretory phenotype
  • the method of inhibiting mTORCl activity is used to treat senescence and diseases of aging.
  • the present invention provides a method of treating senescence and diseases of aging, in a patient in need thereof, comprising the step of administering to said patient a provided compound or pharmaceutically acceptable salt thereof.
  • Additional therapeutic indications where mTORC inhibition may be beneficial are: cardiovascular disease (acute coronary syndrome), coronary occlusions with eluting stents, polycystic kidney disease, neurofibromatosis, epilepsy assoc.
  • TSC1 and/or TSC2 mutations polycystic liver, pachyonychia congenital, fragile x syndrome, Friedrich ataxia, Peutz-Jeghers syndrome, eye disease including neovascular age-related macular degeneration, uveitis, diabetic macular edema, fibroblast growth including pulmonary fibrosis, renal insufficiency/fibrosis, metabolic syndrome, diseases of the immune system including immune senescence, lupus nephritis, chronic immune thrombocytopenia, multiple sclerosis, cancer including lymphoma, tumors associated with TSCl/2 mutations, angiomyolipoma assoc. with TSCl/2 mutations, breast cancer, hepatocellular cancer, leukemia, glioma, adenoid cystic carcinoma, senescence, autism, and vascular rheumatoid arthritis.
  • the method of inhibiting mTORCl activity is used to treat cardiovascular disease (acute coronary syndrome), coronary occlusions with eluting stents, polycystic kidney disease, neurofibromatosis, epilepsy assoc.
  • cardiovascular disease acute coronary syndrome
  • coronary occlusions with eluting stents polycystic kidney disease
  • neurofibromatosis epilepsy assoc.
  • TSC1 and/or TSC2 mutations polycystic liver, pachyonychia congenital, fragile x syndrome, Friedrich ataxia, Peutz-Jeghers syndrome, eye disease including neovascular age-related macular degeneration, uveitis, diabetic macular edema, fibroblast growth including pulmonary fibrosis, renal insufficiency/fibrosis, metabolic syndrome, diseases of the immune system including immune senescence, lupus nephritis, chronic immune thrombocytopenia, multiple sclerosis, cancer including lymphoma, tumors associated with TSCl/2 mutations, angiomyolipoma assoc.
  • the present invention provides a method of treating cardiovascular disease (acute coronary syndrome), coronary occlusions with eluting stents, polycystic kidney disease, neurofibromatosis, epilepsy assoc.
  • cardiovascular disease acute coronary syndrome
  • coronary occlusions with eluting stents polycystic kidney disease
  • neurofibromatosis epilepsy assoc.
  • TSC1 and/or TSC2 mutations polycystic liver, pachyonychia congenital, fragile x syndrome, Friedrich ataxia, Peutz-Jeghers syndrome, eye disease including neovascular age-related macular degeneration, uveitis, diabetic macular edema, fibroblast growth including pulmonary fibrosis, renal insufficiency/fibrosis, metabolic syndrome, diseases of the immune system including immune senescence, lupus nephritis, chronic immune thrombocytopenia, multiple sclerosis, cancer including lymphoma, tumors associated with TSCl/2 mutations, angiomyolipoma assoc.
  • TSCl/2 mutations breast cancer, hepatocellular cancer, leukemia, glioma, adenoid cystic carcinoma, senescence, autism, and vascular rheumatoid arthritis, in a patient in need thereof, comprising the step of administering to said patient a provided compound or pharmaceutically acceptable salt thereof.
  • the present invention is an inhibitor of members of the glucose transporter (GLUT) family.
  • the present invention is a pan-glucose inhibitor, inhibiting GLUT subtypes 1, 2, 3, 4, and 5.
  • the present invention is an inhibitor of one or more GLUT subtypes, individually or severally.
  • compositions of this invention can be administered to humans and other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), bucally, as an oral or nasal spray, or the like, depending on the severity of the infection being treated.
  • the compounds of the invention may be administered orally or parenterally at dosage levels of about 0.01 mg/kg to about 50 mg/kg and preferably from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect.
  • Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • the oral compositions can also include adj
  • Injectable preparations for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S.P. and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil can be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid are used in the preparation of injectables.
  • Injectable formulations can be sterilized, for example, by filtration through a bacterial- retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
  • a compound of the present invention In order to prolong the effect of a compound of the present invention, it is often desirable to slow the absorption of the compound from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the compound then depends upon its rate of dissolution that, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered compound form is accomplished by dissolving or suspending the compound in an oil vehicle. Injectable depot forms are made by forming microencapsule matrices of the compound in biodegradable polymers such as polylactide- polyglycolide.
  • the rate of compound release can be controlled.
  • biodegradable polymers include poly(orthoesters) and poly(anhydrides).
  • Depot injectable formulations are also prepared by entrapping the compound in liposomes or microemulsions that are compatible with body tissues.
  • compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
  • suitable non-irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar—agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol
  • Solid compositions of a similar type may also be employed as fillers in soft and hard- filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polethylene glycols and the like.
  • the active compounds can also be in micro-encapsulated form with one or more excipients as noted above.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art.
  • the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch.
  • Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose.
  • the dosage forms may also comprise buffering agents. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner.
  • buffering agents include polymeric substances and waxes.
  • Dosage forms for topical or transdermal administration of a compound of this invention include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches.
  • the active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required.
  • Ophthalmic formulation, ear drops, and eye drops are also contemplated as being within the scope of this invention.
  • the present invention contemplates the use of transdermal patches, which have the added advantage of providing controlled delivery of a compound to the body.
  • Such dosage forms can be made by dissolving or dispensing the compound in the proper medium.
  • Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.
  • biological sample includes, without limitation, cell cultures or extracts thereof; biopsied material obtained from a mammal or extracts thereof; and blood, saliva, urine, feces, semen, tears, or other body fluids or extracts thereof.
  • the present invention provides a method for treating a disorder mediated by mTORCl in a patient in need thereof, comprising the step of administering to said patient a compound according to the present invention or pharmaceutically acceptable composition thereof.
  • a disorder mediated by mTORCl in a patient in need thereof, comprising the step of administering to said patient a compound according to the present invention or pharmaceutically acceptable composition thereof.
  • additional therapeutic agents that are normally administered to treat that condition may also be present in the compositions of this invention.
  • additional therapeutic agents that are normally administered to treat a particular disease, or condition are known as "appropriate for the disease, or condition, being treated.”
  • a compound of the current invention may also be used to advantage in combination with other antiproliferative compounds.
  • antiproliferative compounds include, but are not limited to aromatase inhibitors; antiestrogens; topoisomerase I inhibitors; topoisomerase II inhibitors; microtubule active compounds; alkylating compounds; histone deacetylase inhibitors; compounds which induce cell differentiation processes; cyclooxygenase inhibitors; MMP inhibitors; mTOR inhibitors; antineoplastic antimetabolites; platin compounds; compounds targeting/decreasing a protein or lipid kinase activity and further anti-angiogenic compounds; compounds which target, decrease or inhibit the activity of a protein or lipid phosphatase; gonadorelin agonists; anti-androgens; methionine aminopeptidase inhibitors; matrix metalloproteinase inhibitors; bisphosphonates; biological response modifiers; antiproliferative antibodies; heparanase inhibitors; inhibitor
  • aromatase inhibitor as used herein relates to a compound which inhibits estrogen production, for instance, the conversion of the substrates androstenedione and testosterone to estrone and estradiol, respectively.
  • the term includes, but is not limited to steroids, especially atamestane, exemestane and formestane and, in particular, non-steroids, especially aminoglutethimide, roglethimide, pyridoglutethimide, tnlostane, testolactone, ketokonazole, vorozole, fadrozole, anastrozole and letrozole.
  • Exemestane is marketed under the trade name AromasinTM.
  • Formestane is marketed under the trade name LentaronTM.
  • Fadrozole is marketed under the trade name AfemaTM.
  • Anastrozole is marketed under the trade name ArimidexTM.
  • Letrozole is marketed under the trade names FemaraTM or FemarTM.
  • Aminoglutethimide is marketed under the trade name OrimetenTM.
  • a combination of the invention comprising a chemotherapeutic agent which is an aromatase inhibitor is particularly useful for the treatment of hormone receptor positive tumors, such as breast tumors.
  • antiestrogen as used herein relates to a compound which antagonizes the effect of estrogens at the estrogen receptor level.
  • the term includes, but is not limited to tamoxifen, fulvestrant, raloxifene and raloxifene hydrochloride.
  • Tamoxifen is marketed under the trade name NolvadexTM.
  • Raloxifene hydrochloride is marketed under the trade name EvistaTM.
  • Fulvestrant can be administered under the trade name FaslodexTM.
  • a combination of the invention comprising a chemotherapeutic agent which is an antiestrogen is particularly useful for the treatment of estrogen receptor positive tumors, such as breast tumors.
  • anti-androgen as used herein relates to any substance which is capable of inhibiting the biological effects of androgenic hormones and includes, but is not limited to, bicalutamide (CasodexTM).
  • gonadorelin agonist as used herein includes, but is not limited to abarelix, goserelin and goserelin acetate. Goserelin can be administered under the trade name ZoladexTM.
  • topoisom erase I inhibitor includes, but is not limited to topotecan, gimatecan, irinotecan, camptothecian and its analogues, 9-nitrocamptothecin and the macromolecular camptothecin conjugate PNU-166148.
  • Irinotecan can be administered, e.g. in the form as it is marketed, e.g. under the trademark CamptosarTM.
  • Topotecan is marketed under the trade name HycamptinTM.
  • topoisomerase II inhibitor includes, but is not limited to the anthracyclines such as doxorubicin (including liposomal formulation, such as CaelyxTM), daunorubicin, epirubicin, idarubicin and nemorubicin, the anthraquinones mitoxantrone and losoxantrone, and the podophillotoxines etoposide and teniposide.
  • Etoposide is marketed under the trade name EtopophosTM.
  • Teniposide is marketed under the trade name VM 26-Bristol
  • Doxorubicin is marketed under the trade name AcriblastinTM or AdriamycinTM.
  • Epirubicin is marketed under the trade name FarmorubicinTM.
  • Idarubicin is marketed, under the trade name ZavedosTM.
  • Mitoxantrone is marketed under the trade name Novantron.
  • microtubule active agent relates to microtubule stabilizing, microtubule destabilizing compounds and microtublin polymerization inhibitors including, but not limited to taxanes, such as paclitaxel and docetaxel; vinca alkaloids, such as vinblastine or vinblastine sulfate, vincristine or vincristine sulfate, and vinorelbine; discodermolides; cochicine and epothilones and derivatives thereof.
  • Paclitaxel is marketed under the trade name TaxolTM.
  • Docetaxel is marketed under the trade name TaxotereTM.
  • Vinblastine sulfate is marketed under the trade name Vinblastin R.PTM.
  • Vincristine sulfate is marketed under the trade name FarmistinTM.
  • alkylating agent includes, but is not limited to, cyclophosphamide, ifosfamide, melphalan or nitrosourea (BCNU or Gliadel). Cyclophosphamide is marketed under the trade name CyclostinTM. Ifosfamide is marketed under the trade name HoloxanTM.
  • histone deacetylase inhibitors or "HDAC inhibitors” relates to compounds which inhibit the histone deacetylase and which possess antiproliferative activity. This includes, but is not limited to, suberoylanilide hydroxamic acid (SAHA).
  • SAHA suberoylanilide hydroxamic acid
  • antimetabolite includes, but is not limited to, 5-fluorouracil or 5-FU, capecitabine, gemcitabine, DNA demethylating compounds, such as 5-azacytidine and decitabine, methotrexate and edatrexate, and folic acid antagonists such as pemetrexed.
  • Capecitabine is marketed under the trade name XelodaTM.
  • Gemcitabine is marketed under the trade name GemzarTM.
  • platinum compound as used herein includes, but is not limited to, carboplatin, cis-platin, cisplatinum and oxaliplatin.
  • Carboplatin can be administered, e.g., in the form as it is marketed, e.g. under the trademark CarboplatTM.
  • Oxaliplatin can be administered, e.g., in the form as it is marketed, e.g. under the trademark EloxatinTM.
  • the term "compounds targeting/decreasing a protein or lipid kinase activity; or a protein or lipid phosphatase activity; or further anti-angiogenic compounds” as used herein includes, but is not limited to, protein tyrosine kinase and/or serine and/or threonine kinase inhibitors or lipid kinase inhibitors, such as a) compounds targeting, decreasing or inhibiting the activity of the platelet-derived growth factor-receptors (PDGFR), such as compounds which target, decrease or inhibit the activity of PDGFR, especially compounds which inhibit the PDGF receptor, such as an N-phenyl-2-pyrimidine-amine derivative, such as imatinib, SU101, SU6668 and GFB- 111; b) compounds targeting, decreasing or inhibiting the activity of the fibroblast growth factor- receptors (FGFR); c) compounds targeting, decreasing or inhibiting the activity of the insulin-like growth factor receptor I (IGF
  • BCR-Abl kinase and mutants, such as compounds which target decrease or inhibit the activity of c-Abl family members and their gene fusion products, such as an N-phenyl-2-pyrimidine-amine derivative, such as imatinib or nilotinib (AMN107); PD 180970; AG957; NSC 680410; PD173955 from ParkeDavis; or dasatinib (BMS-354825); j) compounds targeting, decreasing or inhibiting the activity of members of the protein kinase C (PKC) and Raf family of serine/threonine kinases, members of the MEK, SRC, JAK/pan-JAK, FAK, PDK1, PKB/Akt, Ras/MAPK, PI3K, SYK, TYK2, BTK and TEC family, and/or members of the cyclin- dependent kinase family (CDK) including staurosporine derivatives, such as midostaurin;
  • c-Met receptor compounds which target, decrease or inhibit the activity of c-Met, especially compounds which inhibit the kinase activity of c-Met receptor, or antibodies that target the extracellular domain of c-Met or bind to HGF
  • JAK family members J
  • PI3K inhibitor includes, but is not limited to compounds having inhibitory activity against one or more enzymes in the phosphatidylinositol-3 -kinase family, including, but not limited to ⁇ , ⁇ , ⁇ , ⁇ , PBK-C2a, PBK-C2p, PBK- C2y, Vps34, pi 10-a, pi 10- ⁇ , pi 10- ⁇ , pi 10- ⁇ , p85-a, ⁇ 85- ⁇ , ⁇ 55- ⁇ , pl50, plOl, and p87.
  • PBK inhibitors useful in this invention include but are not limited to ATU-027, SF-1126, DS- 7423, PBI-05204, GSK-2126458, ZSTK-474, buparlisib, pictrelisib, PF-4691502, BYL-719, dactolisib, XL-147, XL-765, and idelalisib.
  • Bcl-2 inhibitor includes, but is not limited to compounds having inhibitory activity against B-cell lymphoma 2 protein (Bcl-2), including but not limited to ABT-199, ABT-731, ABT-737, apogossypol, Ascenta's pan-Bcl-2 inhibitors, curcumin (and analogs thereof), dual Bcl-2/Bcl-xL inhibitors (Infinity Pharmaceuticals/Novartis Pharmaceuticals), Genasense (G3139), HA14-1 (and analogs thereof; see WO2008118802), navitoclax (and analogs thereof, see US7390799), H-1 (Shenayng Pharmaceutical University), obatoclax (and analogs thereof, see WO2004106328), S-001 (Gloria Pharmaceuticals), TW series compounds (Univ. of Michigan), and venetoclax.
  • the Bcl-2 inhibitor is a small molecule therapeutic.
  • the Bcl-2 is a small molecule therapeutic.
  • the Bcl-2
  • BTK inhibitor includes, but is not limited to compounds having inhibitory activity against Bruton's Tyrosine Kinase (BTK), including, but not limited to AVL-292 and ibrutinib.
  • SYK inhibitor includes, but is not limited to compounds having inhibitory activity against spleen tyrosine kinase (SYK), including but not limited to PRT- 062070, R-343, R-333, Excellair, PRT-062607, and fostamatinib
  • BTK inhibitory compounds and conditions treatable by such compounds in combination with compounds of this invention can be found in WO2008039218 and WO2011090760, the entirety of which are incorporated herein by reference.
  • SYK inhibitory compounds, and conditions treatable by such compounds in combination with compounds of this invention can be found in WO2003063794, WO2005007623, and WO2006078846, the entirety of which are incorporated herein by reference.
  • PI3K inhibitory compounds, and conditions treatable by such compounds in combination with compounds of this invention can be found in WO2004019973, WO2004089925, WO2007016176, US8138347, WO2002088112, WO2007084786, WO2007129161, WO2006122806, WO2005113554, and WO2007044729 the entirety of which are incorporated herein by reference.
  • JAK inhibitory compounds and conditions treatable by such compounds in combination with compounds of this invention can be found in WO2009114512, WO2008109943, WO2007053452, WO2000142246, and WO2007070514, the entirety of which are incorporated herein by reference.
  • Further anti-angiogenic compounds include compounds having another mechanism for their activity, e.g. unrelated to protein or lipid kinase inhibition e.g. thalidomide (ThalomidTM) and T P-470.
  • ThilomidTM thalidomide
  • T P-470 T P-470.
  • proteasome inhibitors useful for use in combination with compounds of the invention include, but are not limited to bortezomib, disulfiram, epigallocatechin-3-gallate (EGCG), salinosporamide A, carfilzomib, ONX-0912, CEP-18770, and MLN9708.
  • Compounds which target, decrease or inhibit the activity of a protein or lipid phosphatase are e.g. inhibitors of phosphatase 1, phosphatase 2A, or CDC25, such as okadaic acid or a derivative thereof.
  • Compounds which induce cell differentiation processes include, but are not limited to, retinoic acid, ⁇ - ⁇ - or ⁇ - tocopherol or a- ⁇ - or ⁇ -tocotrienol.
  • cyclooxygenase inhibitor as used herein includes, but is not limited to, Cox- 2 inhibitors, 5-alkyl substituted 2-arylaminophenylacetic acid and derivatives, such as celecoxib (CelebrexTM), rofecoxib (VioxxTM), etoricoxib, valdecoxib or a 5-alkyl-2- arylaminophenylacetic acid, such as 5-methyl-2-(2'-chloro-6'-fluoroanilino)phenyl acetic acid, lumiracoxib.
  • Cox- 2 inhibitors such as celecoxib (CelebrexTM), rofecoxib (VioxxTM), etoricoxib, valdecoxib or a 5-alkyl-2- arylaminophenylacetic acid, such as 5-methyl-2-(2'-chloro-6'-fluoroanilino)phenyl acetic acid, lumiracoxib.
  • bisphosphonates includes, but is not limited to, etridonic, clodronic, tiludronic, pamidronic, alendronic, ibandronic, risedronic and zoledronic acid.
  • Etridonic acid is marketed under the trade name DidronelTM.
  • Clodronic acid is marketed under the trade name BonefosTM.
  • Tiludronic acid is marketed under the trade name SkelidTM.
  • Pamidronic acid is marketed under the trade name ArediaTM.
  • Alendronic acid is marketed under the trade name FosamaxTM.
  • Ibandronic acid is marketed under the trade name BondranatTM.
  • Risedronic acid is marketed under the trade name ActonelTM.
  • Zoledronic acid is marketed under the trade name ZometaTM.
  • mTOR inhibitors relates to compounds which inhibit the mammalian target of rapamycin (mTOR) and which possess antiproliferative activity such as sirolimus (Rapamune®), everolimus (CerticanTM), CCI-779 and ABT578.
  • heparanase inhibitor refers to compounds which target, decrease or inhibit heparin sulfate degradation.
  • the term includes, but is not limited to, PI-88.
  • biological response modifier refers to a lymphokine or interferons.
  • inhibitor of Ras oncogenic isoforms such as H-Ras, K-Ras, or N-Ras, as used herein refers to compounds which target, decrease or inhibit the oncogenic activity of Ras; for example, a “farnesyl transferase inhibitor” such as L-744832, DK8G557 or Rl 15777 (ZarnestraTM).
  • telomerase inhibitor refers to compounds which target, decrease or inhibit the activity of telomerase. Compounds which target, decrease or inhibit the activity of telomerase are especially compounds which inhibit the telomerase receptor, such as telomestatin.
  • methionine aminopeptidase inhibitor refers to compounds which target, decrease or inhibit the activity of methionine aminopeptidase.
  • Compounds which target, decrease or inhibit the activity of methionine aminopeptidase include, but are not limited to, bengamide or a derivative thereof.
  • proteasome inhibitor refers to compounds which target, decrease or inhibit the activity of the proteasome.
  • Compounds which target, decrease or inhibit the activity of the proteasome include, but are not limited to, Bortezomib (VelcadeTM) and MLN 341.
  • matrix metalloproteinase inhibitor or (“MMP” inhibitor) as used herein includes, but is not limited to, collagen peptidomimetic and nonpeptidomimetic inhibitors, tetracycline derivatives, e.g. hydroxamate peptidomimetic inhibitor batimastat and its orally bioavailable analogue marimastat (BB-2516), prinomastat (AG3340), metastat (NSC 683551) BMS-279251 , BAY 12-9566, TAA211 , MMI270B or AAJ996.
  • MMP matrix metalloproteinase inhibitor
  • FMS-like tyrosine kinase inhibitors which are compounds targeting, decreasing or inhibiting the activity of FMS-like tyrosine kinase receptors (Flt-3R); interferon, ⁇ - ⁇ -D-arabinofuransylcytosine (ara-c) and bisulfan; and ALK inhibitors, which are compounds which target, decrease or inhibit anaplastic lymphoma kinase.
  • FMS-like tyrosine kinase receptors are especially compounds, proteins or antibodies which inhibit members of the Flt-3R receptor kinase family, such as PKC412, midostaurin, a staurosporine derivative, SU11248 and MLN518.
  • HSP90 inhibitors includes, but is not limited to, compounds targeting, decreasing or inhibiting the intrinsic ATPase activity of HSP90; degrading, targeting, decreasing or inhibiting the HSP90 client proteins via the ubiquitin proteosome pathway.
  • Compounds targeting, decreasing or inhibiting the intrinsic ATPase activity of HSP90 are especially compounds, proteins or antibodies which inhibit the ATPase activity of HSP90, such as 17-allylamino,17-demethoxygeldanamycin (17AAG), a geldanamycin derivative; other geldanamycin related compounds; radicicol and HDAC inhibitors.
  • antiproliferative antibodies includes, but is not limited to, trastuzumab (HerceptinTM), Trastuzumab-DMl, erbitux, bevacizumab (AvastinTM), rituximab (Rituxan ® ), PR064553 (anti-CD40) and 2C4 Antibody.
  • trastuzumab HerceptinTM
  • Trastuzumab-DMl erbitux
  • bevacizumab AvastinTM
  • rituximab Rasteran ®
  • PR064553 anti-CD40
  • compounds of the current invention can be used in combination with standard leukemia therapies, especially in combination with therapies used for the treatment of AML.
  • compounds of the current invention can be administered in combination with, for example, farnesyl transferase inhibitors and/or other drugs useful for the treatment of AML, such as Daunorubicin, Adriamycin, Ara-C, VP- 16, Teniposide, Mitoxantrone, Idarubicin, Carboplatinum and PKC412.
  • anti-leukemic compounds include, for example, Ara-C, a pyrimidine analog, which is the 2 -alpha-hydroxy ribose (arabinoside) derivative of deoxycytidine. Also included is the purine analog of hypoxanthine, 6-mercaptopurine (6-MP) and fludarabine phosphate.
  • HDAC hi stone deacetylase
  • SAHA suberoylanilide hydroxamic acid
  • HDAC inhibitors include MS275, SAHA, FK228 (formerly FR901228), Trichostatin A and compounds disclosed in US 6,552,065 including, but not limited to, N-hydroxy-3-[4-[[[2-(2-methyl-lH-indol-3-yl)-ethyl]- amino]methyl]phenyl]- 2E-2-propenamide, or a pharmaceutically acceptable salt thereof and N-hydroxy-3-[4-[(2- hydroxyethyl) ⁇ 2-(lH-indol-3-yl)ethyl]-amino]methyl]phenyl]-2E-2- propenamide, or a pharmaceutically acceptable salt thereof, especially the lactate salt.
  • Somatostatin receptor antagonists as used herein refer to compounds which target, treat or inhibit the somatostatin receptor such as octreotide, and SOM230.
  • Tumor cell damaging approaches refer to approaches such as ionizing radiation.
  • the term "ionizing radiation” referred to above and hereinafter means ionizing radiation that occurs as either electromagnetic rays (such as X-rays and gamma rays) or particles (such as alpha and beta particles). Ionizing radiation is provided in, but not limited to, radiation therapy and is known in the art. See Hellman, Principles of Radiation Therapy, Cancer, in Principles and Practice of Oncology, Devita et al., Eds., 4 th Edition, Vol. 1 , pp. 248-275 (1993).
  • EDG binders and ribonucleotide reductase inhibitors.
  • EDG binders refers to a class of immunosuppressants that modulates lymphocyte recirculation, such as FTY720.
  • ribonucleotide reductase inhibitors refers to pyrimidine or purine nucleoside analogs including, but not limited to, fludarabine and/or cytosine arabinoside (ara-C), 6-thioguanine, 5-fluorouracil, cladribine, 6-mercaptopurine (especially in combination with ara-C against ALL) and/or pentostatin.
  • Ribonucleotide reductase inhibitors are especially hydroxyurea or 2-hydroxy-lH-isoindole-l ,3-dione derivatives.
  • VEGF vascular endothelial growth factor
  • l-(4-chloroanilino)-4-(4-pyridylmethyl)phthalazine or a pharmaceutically acceptable salt thereof l-(4-chloroanilino)-4-(4-pyridylmethyl)phthalazine succinate
  • AngiostatinTM EndostatinTM
  • anthranilic acid amides ZD4190; ZD6474; SU5416; SU6668
  • bevacizumab or anti-VEGF antibodies or anti-VEGF receptor antibodies, such as rhuMAb and RHUFab
  • VEGF aptamer such as Macugon
  • FLT-4 inhibitors, FLT-3 inhibitors VEGFR-2 IgGI antibody
  • Angiozyme RI 4610)
  • Bevacizumab AvastinTM
  • Photodynamic therapy refers to therapy which uses certain chemicals known as photosensitizing compounds to treat or prevent cancers.
  • Examples of photodynamic therapy include treatment with compounds, such as VisudyneTM and porfimer sodium.
  • Angiostatic steroids refers to compounds which block or inhibit angiogenesis, such as, e.g., anecortave, triamcinolone, hydrocortisone, 11-a-epihydrocotisol, cortex ol one, 17a-hydroxyprogesterone, corticosterone, desoxycorticosterone, testosterone, estrone and dexamethasone.
  • angiogenesis such as, e.g., anecortave, triamcinolone, hydrocortisone, 11-a-epihydrocotisol, cortex ol one, 17a-hydroxyprogesterone, corticosterone, desoxycorticosterone, testosterone, estrone and dexamethasone.
  • Implants containing corticosteroids refers to compounds, such as fluocinolone and dexamethasone.
  • chemotherapeutic compounds include, but are not limited to, plant alkaloids, hormonal compounds and antagonists; biological response modifiers, preferably lymphokines or interferons; antisense oligonucleotides or oligonucleotide derivatives; shRNA or siRNA; or miscellaneous compounds or compounds with other or unknown mechanism of action.
  • a compound of the current invention may also be used in combination with known therapeutic processes, for example, the administration of hormones or radiation.
  • a provided compound is used as a radiosensitizer, especially for the treatment of tumors which exhibit poor sensitivity to radiotherapy.
  • a compound of the current invention can be administered alone or in combination with one or more other therapeutic compounds, possible combination therapy taking the form of fixed combinations or the administration of a compound of the invention and one or more other therapeutic compounds being staggered or given independently of one another, or the combined administration of fixed combinations and one or more other therapeutic compounds.
  • a compound of the current invention can besides or in addition be administered especially for tumor therapy in combination with chemotherapy, radiotherapy, immunotherapy, phototherapy, surgical intervention, or a combination of these. Long-term therapy is equally possible as is adjuvant therapy in the context of other treatment strategies, as described above. Other possible treatments are therapy to maintain the patient's status after tumor regression, or even chemopreventive therapy, for example in patients at risk.
  • Those additional agents may be administered separately from an inventive compound- containing composition, as part of a multiple dosage regimen. Alternatively, those agents may be part of a single dosage form, mixed together with a compound of this invention in a single composition. If administered as part of a multiple dosage regime, the two active agents may be submitted simultaneously, sequentially or within a period of time from one another normally within five hours from one another.
  • the term "combination,” “combined,” and related terms refers to the simultaneous or sequential administration of therapeutic agents in accordance with this invention.
  • a compound of the present invention may be administered with another therapeutic agent simultaneously or sequentially in separate unit dosage forms or together in a single unit dosage form.
  • the present invention provides a single unit dosage form comprising a compound of the current invention, an additional therapeutic agent, and a pharmaceutically acceptable carrier, adjuvant, or vehicle.
  • compositions of this invention should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of an inventive compound can be administered.
  • compositions which comprise an additional therapeutic agent that additional therapeutic agent and the compound of this invention may act synergistically. Therefore, the amount of additional therapeutic agent in such compositions will be less than that required in a monotherapy utilizing only that therapeutic agent. In such compositions a dosage of between 0.01 - 1,000 ⁇ g/kg body weight/day of the additional therapeutic agent can be administered.
  • the amount of additional therapeutic agent present in the compositions of this invention will be no more than the amount that would normally be administered in a composition comprising that therapeutic agent as the only active agent.
  • the amount of additional therapeutic agent in the presently disclosed compositions will range from about 50% to 100% of the amount normally present in a composition comprising that agent as the only therapeutically active agent.
  • the compounds of this invention, or pharmaceutical compositions thereof, may also be incorporated into compositions for coating an implantable medical device, such as prostheses, artificial valves, vascular grafts, stents and catheters.
  • an implantable medical device such as prostheses, artificial valves, vascular grafts, stents and catheters.
  • Vascular stents for example, have been used to overcome restenosis (re-narrowing of the vessel wall after injury).
  • patients using stents or other implantable devices risk clot formation or platelet activation. These unwanted effects may be prevented or mitigated by pre-coating the device with a pharmaceutically acceptable composition comprising a kinase inhibitor.
  • Implantable devices coated with a compound of this invention are another embodiment of the present invention.
  • BINAP (2,2'-bi s(diplienylpliosphino)- 1 , 1 -binaphthyl)
  • CHIRAL-HPLC chiral high performance liquid chromatography
  • CMBP (cyanom ethyl ene)tributylphosphorane
  • DIBAL-H diisobutylaluminium hydride
  • DIPEA N,N-diisopropylethylamine
  • EDTA ethylenediaminetetraacetic acid
  • HATU ⁇ , ⁇ , ⁇ ' , ⁇ ' -tetramethyl-0-(7-azabenzotriazol- 1 -yl)uranium
  • FiPLC high performance liquid chromatography
  • JackiePhos 2- ⁇ Bis[3,5-bis(trifluoromethyl)phenyl]phosphino ⁇ -3,6-dimethoxy - 2',4',6'-triisopropyl-l, l '-biphenyl, Bis(3,5-bis(trifluoromethyl)phenyl)(2',4',6'- triisopropyl-3,6-dimethoxybiphenyl-2-yl)phosphine
  • MgO magnesium oxide
  • NMP N-methylpyrrolidine MR: Nuclear Magnetic Resonance
  • PBS phosphate buffered saline
  • PE petroleum ether
  • TEBAC Benzyltriethylammonium chloride
  • TfAA trifluoromethanesulfonic anhydride
  • TIPS triisopropylsilyl
  • Method A SunFire C18, 4.6*50mm, 3.5um column Xbridge C18 3.5 ⁇ 4.6x50mm column.
  • the elution system used was a gradient of 5%-95% over 1.5 min at 2 ml/min and the solvent was acetonitrile/0.01% aqueous TFA.
  • Method B Xbridge C18 3.5 ⁇ 4.6> ⁇ 50mm column, the elution system used was a gradient of 5%-95% over 1.5 min at 2 ml/min and the solvent was acetonitrile/10 mM ammonium acetate aqueous solution.
  • Samples were purified by prep-HPLC using the following two methods : Method C: the crude samples were dissolved in DMF before purification, unless otherwise noted. Boston C18 21 *250mm ⁇ column. The mobile phase was acetonitrile/0.01% aqueous TFA (or 0.01% aqueous HC1).
  • Method D the crude samples were dissolved in DMF before purification, unless otherwise noted. Boston C18 21 *250mm ⁇ column. The mobile phase was acetonitrile/10 mM ammonium acetate aqueous solution.
  • Step 2 Methyl-5-methyl-lH-pyrrole-2-carboxylate [00209] A mixture of 2,2,2-trichloro-l -(5 -methyl- lH-pyrrol-2-yl)ethanone (40 g, 176.6 mmol, crude) in MeONa/MeOH (100 mL) and MeOH (400 mL) was stirred at rt for 2 hrs. The reaction mixture was concentrated, cooled to rt, poured into ice-water, then extracted with EtOAc (500 mL x 2).
  • Step 6 4-((4-(2-Chlorophenyl)piperazin-l-yl)methyl)-l,5-dimethyl-lH-pyrrole-2-carboxylic acid
  • Step 4 4-((4-(2-Chlorophenyl)piperazin-l-yl)methyl)-l-methyl-lH-pyrrole-2-carboxylic acid
  • Step 1 (4-((4-(2-Chlorophenyl)piperazin-l-yl)methyl)-l,5-dimethyl-lH-pyrrol-2-yl)(4- phenylpiperazin-l-yl)methanone
  • Step 3 (4-((4-(2-Chlorophenyl) piperazin-l-yl) methyl)-l, 5-dimethyl-lH-pyrrol-2-yl) (4-(3, 5-difluorophenyl) piperazin-l-yl) methanone
  • Step 3 (S)-(4-((4-(2-chlorophenyl) piperazin-l-yl) methyl)-!, 5-dimethyl-lH-pyrrol-2-yl) (4- (4-fluorophenyl)-2-methylpiperazin-l-yl) methanone
  • Step 1 (S)-tert-butyl 4-(3,5-difluorophenyl)-2-methylpiperazine-l-carboxylate [00234] A solution of (S)-tert-butyl-2-methylpiperazine-l-carboxylate (1.2 g, 6 mmol), 1,3- difluoro-5-iodobenzene (1.685 g, 7.2 mmol), t-BuONa (865 mg, 9 mmol), BINAP (150 mg, 0.24 mmol), Pd 2 (dba)3 (110 mg, 0.12 mmol) in dry toluene (40 mL) was stirred under N 2 at 80°C for 16 hrs.
  • Step 3 (S)-(4-((4-(2-chlorophenyl)piperazin-l-yl)methyl)-l,5-dimethyl-lH-pyrrol-2-yl)(4- (3,5-difluorophenyl)-2-methylpiperazin-l-yl)methanone
  • Step 3 (4-(3-Chloro-4-fluorophenyl)piperazin-l-yl)(4-((4-(2-chlorophenyl)piperazin-l- yl)methyl)-l,5-dimethyl-lH-pyrrol-2-yl)methanone
  • Step 2 tert-butyl 4-(2-nitrophenyl)piperazine-l-carboxylate
  • Step 3 tert-butyl 4-(2-aminophenyl)piperazine-l-carboxylate
  • Step-4 tert-butyl 4-(2-azidophenyl)piperazine-l-carboxylate
  • Step 6 (4-((4-(2-azidophenyl)piperazin-l-yl)methyl)-l,5-dimethyl-lH-pyrrol-2-yl)(4- phenylpiperazin-l-yl)methanone
  • Step 4 (S)-(4-((4-(2-azidophenyl)piperazin-l-yl)methyl)-l,5-dimethyl-lH-pyrrol-2-yl)(2- methyl-4-phenylpiperazin-l-yl)methanone
  • Step 2 (S)-teri-butyl-2-methyl-4-(l-((2-(trimethylsilyl)ethoxy)methyl)-lH-indol-7- yl)piperazine-l-carboxylate
  • Step 5 (S)-(4-(lH-indol-7-yl)-2-methylpiperazin-l-yl)(4-((4-(2-chlorophenyl)piperazin-l- yl)methyl)-l,5-dimethyl-lH-pyrrol-2-yl)methanone trifluoroacetate
  • Step 2 l-(4-fluorophenyl)-l,4-diazepane hydrochloride salt
  • Example 17 (4-((4-(2-Chlorophenyl)piperazin-l-yl)methyl)-l,5-dimethyl-lH-pyrrol-2-yl)(3- phenyl-6,7-dihydropyrazolo[ -a]pyrazin-5( H)-yl)methanone, 1-35
  • Step 4 ieri-Butyl-2-(5-(hydroxymethyl)-4-phenyl-lH-pyrazol-l-yl)ethylcarbamate
  • Step 5 ieri-Butyl-2-(5-(chloromethyl)-4-phenyl-lH-pyrazol-l-yl)ethylcarbamate
  • Step 6 terf-Butyl 3-phenyl-6,7-dihydropyrazolo[l,5-a]pyrazine-5( H)-carboxylate [00272] To a solution of crude tert-butyl-2-(5-(chloromethyl)-4-phenyl-lH-pyrazol-l- yl)ethylcarbamate (200 mg) in dry DMF (5 mL) was added 60% NaH (71 mg, 1.78 mmol) at 0°C. The mixture was allowed to warm to rt and stirred for 2 hrs, then aq. H4CI was added and the mixture was extracted with EtOAc (20 mL).
  • Step 7 3-Phenyl-4,5,6,7-tetrahydropyrazolo[l,5-a]pyrazine hydrochloride salt
  • Step 8 (4-((4-(2-Chlorophenyl)piperazin-l-yl)methyl)-l,5-dimethyl-lH-pyrrol-2-yl)(3- phenyl-6,7-dihydropyrazolo [1 ,5-a] pyrazin-5(4H)-yl)methanone
  • Example 18 (4-((4-(2-chlorophenyl)piperazin-l-yl)methyl)-l,5-dimethyl-lH-pyrrol-2- yl)(3,4-dihydrobenzo[4,5]imidazo[l,2-a]pyrazin-2(lH)-yl)methanone, 1-91
  • Step 3 (4-((4-(2-chlorophenyl)piperazin-l-yl)methyl)-l,5-dimethyl-lH-pyrrol-2-yl)(3,4- dihydrobenzo[4,5]imidazo[l,2-a]pyrazin-2(lH)-yl)methanone
  • reaction mixture was purified by prep-HPLC to afford (4-((4-(2-chlorophenyl)piperazin- 1 -yl)methyl)- 1 ,5-dimethyl- lH-pyrrol-2-yl)(3 ,4- dihydrobenzo[4,5]imidazo[l,2-a]pyrazin-2(lH)-yl)methanone 1-91 (28.5 mg, 0.057 mmol, 27%) as a white solid.
  • Step 2 (S)-tert-butyl 2-methyl-4-(l-((2-(trimethylsilyl)ethoxy)methyl)-lH-indol-5- yl)piperazine-l-carboxylate
  • Step 5 (S)-2-(4-(5-(4-(li -indol-5-yl)-2-methylpiperazine-l-carbonyl)-2,4-di- methylbenzyl)piperazin-l-yl)benzonitrile trifluoroacetate salt
  • Step 3 (S)-ieri-butyl-3-(3-(4-(4-((4-(2-chlorophenyl)piperazin-l-yl)methyl)-l,5-dimethyl- lH-pyrrole-2-carbonyl)-3-methylpiperazin-l-yl)phenyl)propylcarbamate
  • Step 4 (S)-(4-(3-(3-aminopropyl)phenyl)-2-methylpiperazin-l-yl)(4-((4-(2- chlorophenyl)piperazin-l-yl)methyl)-l,5-dimethyl-lH-pyrrol-2-yl)methanone
  • Step 2 (R)-tert-butyl 3-(2-(4-((l,2-dimethyl-5-(2-methyl-4-phenylpiperazine-l-carbonyl)- lH-pyrrol-3-yl)methyl)piperazin-l-yl)phenyl)propyl(methoxymethyl) carbamate
  • Step 3 (R)-(4-((4-(2-(3-aminopropyl)phenyl)piperazin-l-yl)methyl)-l,5-dimethyl-lH- pyrrol-2-yl)(2-methyl-4-phenylpiperazin-l-yl)methanone ditrifluoroacetate salt
  • (R)-tert-butyl-3-(2-(4-((l,2-dimethyl-5-(2-methyl-4-phenyl piperazine- l-carbonyl)-lH-pyrrol-3-yl)methyl)piperazin-l-yl)phenyl)propyl(methoxy methyl) carbamate 180 mg, 0.27 mmol
  • TFA 4 mL
  • Step 2 2-(4-(4-((4-(2-chlorophenyl)piperazin-l-yl)methyl)-l,5-dimethyl-lH-pyrrole-2- carbonyl)piperazin-l-yl)-6-fluorobenzonitrile
  • Step 3 Synthesis of (S)-(4-((4-(2-chlorophenyl)piperazin-l-yl)methyl)-l,5-dimethyl-lH- pyrrol-2-yl)(2- methyl-4- (pyrimidin-2-yl)piperazin-l-yl)methanone
  • Example 35 (4-((4-(2-Chlorophenyl)piperazin-l-yl)methyl)-l,5-dimethyl-lH-pyrrol-2-yl)(2- phenyl-6,7-dihydropyrazolo 1 ,5-a] pyrazin-5(4H)-yl)methanone 2,2,2-trifluoroacetate, 1-81
  • Step 2 4-((4-(3-Chloropyridin-2-yl)piperazin-l-yl)methyl)-l,5-dimethyl-lH-pyrrole-2- carboxylic acid
  • Step 3 (S)-(4-((4-(3-chloropyridin-2-yl)piperazin-l-yl)methyl)-l,5-dimethyl-lH-pyrrol-2- yl)(4-(4-fluorophenyl)-2-methylpiperazin-l-yl)methanone
  • Step 1 Synthesis of (4-((4-(3-chloropyridin-2-yl) piperazin-l-yl) methyl)-!, 5 -dimethyl-lH- pyrrol-2-yl)(4-(3,4-difluorop henyl) piperazin-l-yl)methanone:
  • Example 39 (4-((4-(3-Chloropyridin-2-yl)piperazin-l-yl)methyl)-l,5-dimethyl-lH-pyrrol-2- yl)(4-(4-fluorophenyl)piperazin-l-yl)methanone, 1-80
  • Example 40 (S)-(4-((4-(3-chloropyridin-2-yl)piperazin-l-yl)methyl)-l,5-dimethyl-lH- pyrrol-2-yl)(4-(3,4-difluoro henyl)-2-methylpiperazin-l-yl)methanone, 1-78
  • Step 3 (S)-(4-(3-chlorophenyl)-2-methylpiperazin-l-yl)(4-((4-(2-chlorophenyl)piperazin-l- yl)methyl)-l,5-dimethyl-lH-pyrrol-2-yl)methanone
  • Step 3 5-(4-(4-((4-(2-Chlorophenyl)piperazin-l-yl)methyl)-l,5-dimethyl-lH-pyrrole-2- carbonyl)piperazin-l-yl)-2-fluorobenzonitrile
  • Step 1 l,2-Dimethyl-5-(4-(pyridin-2-yl)piperazine-l-carbonyl)-lH-pyrrole-3-carbaldehyde
  • Step 2 tert-Butyl 3-(2-cyanophenyl)-3,8-diazabicyclo[3.2.1]octane-8-carboxylate
  • Step 4 2-(8-((l,2-Dimethyl-5-(4-(pyridin-2-yl)piperazine-l-carbonyl)-lH-pyrrol-3- yl)methyl)-3,8-diazabicyclo[3.2.1]octan-3-yl)benzonitrile
  • Step 2 (S)-4-(2-chlorophenyl)-l-((5-(4-(4-fluorophenyl)-2-methylpiperazine-l-carbonyl)- l,2-dimethyl-2H-pyrrol-3-yl)methylene)piperazin-l-ium perchlorate
  • Step 3 (4-(l-(4-(2-Chlorophenyl)piperazin-l-yl)ethyl)-l,5-dimethyl-lH-pyrrol-2-yl)((S)-4- (4-fluorophenyl)-2-methylpiperazin-l-yl)methanone
  • Step 1 tert-Butyl 4-(2-chlorophenyl)-l,4-diazepane-l-carboxylate
  • Step 3 (S)-(4-((4-(2-chlorophenyl)-l ,4-diazepan- l-yl)methyl)- 1 ,5-dimethyl- lH-pyrrol-2- yl)(4-(4-fluorophenyl)-2-methylpiperazin-l-yl)methanone
  • Example 48 (S)-(4-(3-chlorophenyl)-2-methylpiperazin-l-yl)(4-((4-(3-chloropyridin-2- yl)piperazin-l-yl)methyl)-l,5-dimethyl-lH-pyrrol-2-yl)methanone, 1-90

Abstract

La présente invention concerne des composés, des compositions de ces composés, et des méthodes d'utilisation correspondantes.
PCT/US2017/060640 2016-11-08 2017-11-08 Inhibiteurs pyrroles de mtorc et leurs utilisations WO2018089493A1 (fr)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US16/348,198 US20190389843A1 (en) 2016-11-08 2017-11-08 PYRROLE mTORC INHIBITORS AND USES THEREOF

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201662419016P 2016-11-08 2016-11-08
US62/419,016 2016-11-08

Publications (1)

Publication Number Publication Date
WO2018089493A1 true WO2018089493A1 (fr) 2018-05-17

Family

ID=62109678

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2017/060640 WO2018089493A1 (fr) 2016-11-08 2017-11-08 Inhibiteurs pyrroles de mtorc et leurs utilisations

Country Status (1)

Country Link
WO (1) WO2018089493A1 (fr)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020142748A1 (fr) 2019-01-03 2020-07-09 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Procédés et matériaux pour augmenter les niveaux de polypeptides eb de facteur de transcription
WO2022233782A1 (fr) 2021-05-03 2022-11-10 Lead Discovery Center Gmbh Composition comprenant un inhibiteur de la transcription mitochondriale

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6258813B1 (en) * 1996-09-13 2001-07-10 Merck Patent Gesellschaft Mit D4 receptor selectivity piperazine derivatives
US20080317671A1 (en) * 2004-03-31 2008-12-25 Carruthers Nicholas I Non-imidazole heterocyclic compounds
US20140378429A1 (en) * 2012-02-01 2014-12-25 Rigel Pharmaceuticals, Inc. Carboxamide, Sulfonamide and Amine Compounds and Methods for Using Them

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6258813B1 (en) * 1996-09-13 2001-07-10 Merck Patent Gesellschaft Mit D4 receptor selectivity piperazine derivatives
US20080317671A1 (en) * 2004-03-31 2008-12-25 Carruthers Nicholas I Non-imidazole heterocyclic compounds
US20140378429A1 (en) * 2012-02-01 2014-12-25 Rigel Pharmaceuticals, Inc. Carboxamide, Sulfonamide and Amine Compounds and Methods for Using Them

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
BARRON, C., INVESTIGATION OF THE EXPRESSION OF GLUCOSE TRANSPORTER PROTEINS IN HUMAN CANCEL CELLS, June 2012 (2012-06-01), pages 1 - 132, Retrieved from the Internet <URL:http://www.dr.library.brocku.ca/bitstream/handle/10464/4087/Brock_Barron_Carly_2012.pdf?sequence=1&isAllowed=y> [retrieved on 20171206] *
BRENNAN, AL ET AL.: "Clinical importance of cystic fibrosis-related diabetes", JOURNAL OF CYSTIC FIBROSIS, vol. 3, 27 October 2004 (2004-10-27), pages 209 - 222, XP004732150 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020142748A1 (fr) 2019-01-03 2020-07-09 University Of Pittsburgh - Of The Commonwealth System Of Higher Education Procédés et matériaux pour augmenter les niveaux de polypeptides eb de facteur de transcription
CN113316453A (zh) * 2019-01-03 2021-08-27 匹兹堡大学联邦***高等教育 用于提高转录因子eb多肽水平的方法和材料
EP3906033A4 (fr) * 2019-01-03 2022-11-09 University of Pittsburgh - Of the Commonwealth System of Higher Education Procédés et matériaux pour augmenter les niveaux de polypeptides eb de facteur de transcription
WO2022233782A1 (fr) 2021-05-03 2022-11-10 Lead Discovery Center Gmbh Composition comprenant un inhibiteur de la transcription mitochondriale

Similar Documents

Publication Publication Date Title
WO2018089433A1 (fr) Inhibiteurs de mtorc phényle et leurs utilisations
US10683308B2 (en) Rapamycin analogs and uses thereof
JP5580330B2 (ja) Vegf−r2のピリジルオキシインドール類阻害剤および疾患の処置のためのその使用
WO2017023894A1 (fr) Inhibiteurs de mthfd2 et utilisation desdits inhibiteurs
US11944605B2 (en) Rapamycin analogs and uses thereof
US11819476B2 (en) Rapamycin analogs and uses thereof
WO2016040449A1 (fr) Inhibiteurs de la 3-phosphoglycérate déshydrogénase) et leurs utilisations
US10844013B2 (en) Phenyl amino piperidine mTORC inhibitors and uses thereof
WO2018191146A1 (fr) Inhibiteurs de rheb à base d&#39;hétéroaryle et leurs utilisations
JP2007511575A (ja) タンパク質キナーゼ阻害剤としての1h−イミダゾキノリン誘導体
WO2019126106A1 (fr) Inhibiteurs acycliques de cxcr4 et leurs utilisations
US20220267352A1 (en) Rapamycin analogs and uses thereof
WO2018089493A1 (fr) Inhibiteurs pyrroles de mtorc et leurs utilisations
US20190389843A1 (en) PYRROLE mTORC INHIBITORS AND USES THEREOF
US20240131012A1 (en) Rapamycin analogs and uses thereof
EA045993B1 (ru) Аналоги рапамицина и их применения

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: 17870317

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: 17870317

Country of ref document: EP

Kind code of ref document: A1