WO2012034526A1 - Fused heteroaryls and their uses - Google Patents

Fused heteroaryls and their uses Download PDF

Info

Publication number
WO2012034526A1
WO2012034526A1 PCT/CN2011/079684 CN2011079684W WO2012034526A1 WO 2012034526 A1 WO2012034526 A1 WO 2012034526A1 CN 2011079684 W CN2011079684 W CN 2011079684W WO 2012034526 A1 WO2012034526 A1 WO 2012034526A1
Authority
WO
WIPO (PCT)
Prior art keywords
optionally substituted
alkyl
heteroaryl
aryl
heterocycle
Prior art date
Application number
PCT/CN2011/079684
Other languages
French (fr)
Inventor
Weiguo Su
Weihan Zhang
Haibin Yang
Original Assignee
Hutchison Medipharma Limited
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 Hutchison Medipharma Limited filed Critical Hutchison Medipharma Limited
Priority to EP11824596.8A priority Critical patent/EP2616468A4/en
Priority to MX2013002784A priority patent/MX2013002784A/en
Priority to AU2011301518A priority patent/AU2011301518B2/en
Priority to NZ60752711A priority patent/NZ607527A/en
Priority to US13/823,604 priority patent/US9181264B2/en
Priority to KR1020137009341A priority patent/KR20130087020A/en
Priority to SG2013014162A priority patent/SG188302A1/en
Priority to JP2013528507A priority patent/JP5634609B2/en
Priority to RU2013108348/04A priority patent/RU2552114C2/en
Priority to CN201180044818.8A priority patent/CN103124731B/en
Priority to BR112013005855A priority patent/BR112013005855A2/en
Priority to CA2810708A priority patent/CA2810708A1/en
Priority to TW101107297A priority patent/TW201311682A/en
Publication of WO2012034526A1 publication Critical patent/WO2012034526A1/en

Links

Classifications

    • 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
    • C07D487/12Heterocyclic 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 three hetero rings
    • C07D487/14Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4375Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a six-membered ring having nitrogen as a ring heteroatom, e.g. quinolizines, naphthyridines, berberine, vincamine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/12Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains three hetero rings
    • C07D471/14Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems

Definitions

  • Phosphoinositide 3-kinases are a family of enzymes that may be involved in cellular functions such as cell growth, proliferation, differentiation, motility, survival and intracellular trafficking, which in turn can be involved in cancer.
  • the PI3K family may include four distinct classes defined by structural and functional characteristics and termed as Classes l-IV.
  • the most fully characterized class may be the Class l-PI3Ks.
  • Class I comprises three class I a isoforms - PI3Ka, ⁇ 3 ⁇ and ⁇ 3 ⁇ .
  • PI3Ka appears to be highly relevant in human cancers and malignancies. PI3Ka can be overexpressed in human cancers.
  • mTOR Mammalian target of rapamycin
  • mTOR is the downstream kinase of PI3K family. Inhibition of mTOR can inhibit the activity of PI3K. Therefore, the PI3K/mTOR pathway can be exploited for new cancer drug discovery.
  • a 1 is N or CH
  • a 4 and A 5 are independently N or CR 2 ;
  • a 2 and A 3 , together with B ring are a 5-membered heteroaryl or heterocycli containing 1 to 4 heteroatoms selected from N, O, and S, and said 5-membered heteroaryl or heterocycle is optionally substituted by one or more groups
  • R 1 is heteroaryl, optionally substituted by one or more groups independently chosen from alkyl, alkenyl, alkynyl, aryl, cycloalkyl, oxo, -C(O)R a , -C(O)OR b , -CN, - C(O)NR c R d , -NR c R d , -NR c C(O)R a , -NR c S(O) n R e , -NR c S(O) n f R g , -NR c C(O)OR b , - NR c C(O)NR d R e , -NO 2 , -OR b , -S(O) n R e , -S(O) n NR c R d , halo, haloalkyi, heteroaryl, and heterocycle;
  • R and R 2 are independently chosen from H, alkyl, alkenyl, alkynyl, aryl, cycloalkyl, -C(O)R a , -C(O)OR b , -CN, -C(O)NR c R d , -NR c R d , -NR c C(O)R a , -NR c S(O) n R e , -NR c S(O) n NR f R g , -NR c C(O)OR b , -NR c C(O)NR d R e , -NO 2 , -OR b , -OC(O)R a , - OC(O)NR c R d , -S(O) n R e , -S(O) n NR c R d , halo, haloalkyi, heteroaryl, and heterocycle
  • R a , R b , R c , R d , R e , R f and R g are each independently chosen from H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl optionally substituted aryl, optionally substituted cycloalkyi, optionally substituted haloalkyl, optionally substituted heteroaryl, and optionally substituted heterocycle,
  • R a and R c , and/or R c and R d , and/or R c and R e , and/or R c and R f , and/or R d and R e , and /or R g and R f together with the atom(s) to which they are attached, form a 3-10 membered optionally substituted heterocycle ring;
  • n is independently 0, 1 , or 2;
  • each optionally substituted group above can be unsubstituted or independently substituted with one or more, such as one, two, or three, substituents independently selected from Ci-C 4 alkyl, cycloalkyi, oxo, aryl, heterocycle, heteroaryl, aryl-Ci-C 4 alkyl-, heteroaryl-Ci-C 4 alkyl-, Ci-C 4 haloalkyl-, -OCi-C 4 alkyl,
  • -OC1-C4 alkylphenyl -d-C 4 alkyl-OH, -C1-C4 alkyl-O-d-C 4 alkyl, -OC1-C4 haloalkyl, halo, -OH, -NH 2 , -d-C 4 alkyl-NH 2 , -N(d-C 4 alkyl)(Ci-C 4 alkyl), -NH(Ci-C 4 alkyl), -N(Ci-C 4 alkyl)(Ci-C 4 alkylphenyl), -NH(Ci-C 4 alkylphenyl), cyano, nitro, oxo, -CO2H, -C(O)OCi-C 4 alkyl, -C(O)Ocycloalkyl, -C(O)Oaryl, -C(O)Oheteroaryl, - C(O)Oheterocycle, -CON(Ci-C
  • -C(O)(cycloalkyl), -C(O)(heterocycle), -C(O)(aryl) such as -C(O) phenyl, - C(O)(heteroaryl), -C(O)d-C 4 haloalkyl, -OC(O)d-C 4 alkyl, -OC(O)(cycloalkyl), - OC(O)(heterocycle), - - OC(O)(heteroaryl), OC(O)(aryl), such as -OC(O)phenyl, - SO 2 (Ci-C 4 alkyl), - SO 2 (cycloalkyl), -SO 2 (heterocycle), -SO 2 (aryl) such as SO 2 (phenyl), -SO 2 (heteroaryl), -SO 2 (Ci-C 4 haloalkyl), -SO 2 NH 2 , -SO 2 NR'R" where
  • -SO 2 NH(cycloalkyl), -SO 2 NH(heterocycle), -SO 2 NH(aryl) such as -SO 2 NH(phenyl), -SO 2 NH(heteroaryl), -NHSO 2 (Ci-C 4 alkyl), NHSO 2 (cycloalkyl), NHSO 2 (heterocycle), NHSO 2 (aryl) such as -NHSO 2 (phenyl), NHSO 2 (heteroaryl), and
  • -NHSO 2 (Ci-C 4 haloalkyl), in which each of alkyl, phenyl, aryl, cycloalkyl, heterocycle, and heteroaryl is optionally substituted by one or more groups independently chosen from -OH, halo, cycloalkyl, heterocycle, Ci-C 4 alkyl, Ci-C 4 haloalkyl-, -OCi-C 4 alkyl, C 1 -C4 alkyl-OH, -d-C 4 alkyl-O-d-C 4 alkyl, -OC 1 -C4 haloalkyl, cyano, nitro, -NH 2 , - CO 2 H, -C(O)OCi-C 4 alkyl, -CON(C C 4 alkyl )(Ci-C 4 alkyl), -CONH(C C 4 alkyl), -CONH 2 , -NHC(O)(Ci-C 4 alkyl), and -N
  • composition comprising at least one compound and/or at least one pharmaceutically acceptable salt thereof described herein and at least one pharmaceutically acceptable carrier.
  • Also provided is a method of inhibiting the activity of PI3K and/or mTOR comprising contacting the enzyme with an effective amount of at least one compound and/or at least one pharmaceutically acceptable salt thereof described herein.
  • Also provided is a method of treating cancer responsive to inhibition of PI3K and/or mTOR comprising administering to a subject in need of treating for said cancer an effective amount of at least one compound and/or at least one pharmaceutically acceptable salt thereof described herein.
  • a dash (“-") that is not between two letters or symbols is used to indicate a point of attachment for a substituent.
  • -CONH 2 is attached through the carbon atom.
  • alkyl refers to a straight or branched hydrocarbon, containing 1 -18, such as 1 -12, further such as 1 -6 carbon atoms.
  • alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, /-propyl, n-butyl, /-butyl, and /-butyl.
  • “Lower alkyl” refers to a straight or branched hydrocarbon, containing 1 -6, such as 1 -4 carbon atoms.
  • alkoxy herein refers to a straight or branched alkyl group of the indicated number of carbon atoms attached through an oxygen bridge such as, for example, methoxy, ethoxy, propoxy, isopropoxy, n-butoxy, sec-butoxy, tert-butoxy, pentoxy, 2-pentyloxy, isopentoxy, neopentoxy, hexoxy, 2-hexoxy, 3-hexoxy, 3- methyl pentoxy, and the like.
  • Alkoxy groups will usually have from 1 to 6 carbon atoms attached through the oxygen bridge.
  • ""Lower alkoxy" refers to a straight or branched alkoxy, wherein the alkyl portion contains 1 -4 carbon atoms.
  • alkenyl groups include, but are not limited to, vinyl, 2-propenyl, and 2- butenyl.
  • alkynyl herein refers to a straight or branched hydrocarbon, containing one or more C ⁇ C triple bonds and 2-10, such as 2-6 carbon atoms.
  • alkynyl groups include, but are not limited to, ethynyl, 2-propynyl, and 2- butynyl.
  • cycloalkyl refers to saturated and partially unsaturated cyclic hydrocarbon groups having 3 to 12, such as 3 to 8 carbon atoms.
  • examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloheptyl, and cyclooctyl.
  • the ring may be saturated or have one or more double bonds (i.e. partially unsaturated), but not fully conjugated, and not aromatic, as defined herein.
  • bicyclic ring systems wherein at least one ring is carbocyclic and aromatic, for example, naphthalene, indane, and 1 ,2,3,4-tetrahydroquinoline; and
  • tricyclic ring systems wherein at least one ring is carbocyclic and aromatic, for example, fluorene.
  • aryl includes 5 and 6-membered carbocyclic aromatic rings fused to a 5- to 7-membered cycloalkyl or heterocyclic ring containing zero or more heteroatoms selected from N, O, and S, provided that the point of attachment is at the carbocyclic aromatic ring when the carbocyclic aromatic ring is fused with a
  • heterocyclic ring and the point of attachment can be at the carbocyclic aromatic ring or at the cycloalkyl when the carbocyclic aromatic ring fused with a cycloalkyl.
  • Bivalent radicals formed from substituted benzene derivatives and having the free valences at ring atoms are named as substituted phenylene radicals.
  • Bivalent radicals derived from univalent polycyclic hydrocarbon radicals whose names end in "-yl” by removal of one hydrogen atom from the carbon atom with the free valence are named by adding "-idene" to the name of the corresponding univalent radical, e.g., a naphthyl group with two points of attachment is termed naphthylidene.
  • Aryl does not encompass or overlap in any way with heteroaryl, separately defined below.
  • halo includes fluoro, chloro, bromo, and iodo, and the term
  • halogen includes fluorine, chlorine, bromine, and iodine.
  • heteroaryl refers to aryl
  • heteroatoms selected from N, O, and S, with the remaining ring atoms being carbon and wherein at least one ring is aromatic and at least one heteroatom is present in the aromatic ring;
  • 1 1 - to 14-membered tricyclic rings containing one or more, for example, from 1 to 4, or in some embodiments, from 1 to 3, heteroatoms selected from N, O, and S, with the remaining ring atoms being carbon and wherein at least one ring is aromatic and at least one heteroatom is present in an aromatic ring.
  • heteroaryl includes a 5- to 7-membered heterocyclic aromatic ring fused to a 5- to 7-membered cycloalkyl ring.
  • bicyclic heteroaryl ring systems wherein only one of the rings contains one or more heteroatoms, the point of attachment may be at the heteroaromatic ring or at the cycloalkyl ring.
  • the total number of S and O atoms in the heteroaryl group exceeds 1 , those heteroatoms are not adjacent to one another. In some embodiments, the total number of S and O atoms in the heteroaryl group is not more than 2. In some embodiments, the total number of S and O atoms in the aromatic heterocycle is not more than 1 .
  • heteroaryl groups include, but are not limited to, (as numbered from the linkage position assigned priority 1 ), pyridyl (such as 2-pyridyl, 3-pyridyl, or 4- pyridyl), pyrazinyl, 2,4-pyrimidinyl, 3,5-pyrimidinyl, 2,4-imidazolyl, isoxazolyl, oxazolyl, thiazolyl, thiadiazolyl, tetrazolyl, thienyl, benzothienyl, furyl, benzofuryl, benzoimidazolyl, indolyl, indolinyl, pyridizinyl, triazolyl, quinolinyl, pyrazolyl,
  • pyrrolopyridinyl such as 1 H-pyrrolo[2,3-b]pyridin-5-yl
  • pyrazolopyridinyl such as1 H- pyrazolo[3,4-b]pyridin-5-yl
  • benzoxazolyl such as benzo[d]oxazol-6-yl
  • benzothiazolyl such as benzo[d]thiazol-6-yl
  • indazolyl such as 1 H-indazol-5-yl
  • 5,6,7,8-tetrahydroisoquinoline
  • Bivalent radicals derived from univalent heteroaryl radicals whose names end in "-yl” by removal of one hydrogen atom from the atom with the free valence are named by adding "-idene" to the name of the corresponding univalent radical, e.g., a pyridyl group with two points of attachment is a pyridylidene.
  • Heteroaryl does not encompass or overlap with aryl as defined above.
  • Substituted heteroaryl also includes ring systems substituted with one or more oxide (-O " ) substituents, such as pyridinyl N-oxides.
  • heterocycle or “heterocyclic ring” is meant a 4- to 12-membered
  • Heterocycle also refers to 5- to 7-membered
  • heterocyclic ring containing one or more heteroatoms selected from N, O, and S fused with 5-, 6-, and/or 7-membered cycloalkyl, carbocyclic aromatic or heteroaromatic ring, provided that the point of attachment is at the heterocyclic ring when the heterocyclice ring is fused with a carbocyclic aromatic or a heteroaromatic ring, and that the point of attachment can be at the cycloalkyl or heterocyclic ring when the heterocylic ring is fused with cycloalkyl.
  • Heterocycle also refers to an aliphatic spirocyclic ring containing one or more heteroatoms selected from N, O, and S, provided that the point of attachment is at the heterocyclic ring.
  • the rings may be saturated or have one or more double bonds (i.e. partially unsaturated).
  • the heterocycle can be substituted by oxo.
  • the point of the attachment may be carbon or heteroatom in the heterocyclic ring.
  • a heterocyle is not a heteroaryl as defined herein.
  • Suitable heterocycles include, for example (as numbered from the linkage position assigned priority 1 ), 1 -pyrrolidinyl, 2-pyrrolidinyl, 2,4-imidazolidinyl, 2,3- pyrazolidinyl, 1 -piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-piperidinyl, 2,5-piperazinyl, pyranyl, 2-morpholinyl and 3-morpholinyl.
  • Substituted heterocycle also includes ring systems substituted with one or more oxo moieties, such as piperidinyl N-oxide, morpholinyl-N-oxide, 1 -oxo-1 -thiomorpholinyl and 1 ,1 -dioxo-1 -thiomorpholinyl.
  • oxo moieties such as piperidinyl N-oxide, morpholinyl-N-oxide, 1 -oxo-1 -thiomorpholinyl and 1 ,1 -dioxo-1 -thiomorpholinyl.
  • substituted means that any one or more hydrogen atoms on the designated atom or group is replaced with a selection from the indicated group, provided that the designated atom's normal valence is not exceeded.
  • a stable compound or stable structure is meant to imply a compound that is sufficiently robust to survive isolation from a reaction mixture, and subsequent formulation as an agent having at least practical utility.
  • substituents are named into the core structure. For example, it is to be understood that when (cycloalkyl)alkyl is listed as a possible substituent, the point of attachment of this substituent to the core structure is in the alkyl portion. [030] In some embodiments, "substituted with one or more groups" refers to two hydrogen atoms on the designated atom or group being independently replaced with two selections from the indicated group of substituents.
  • substituted with one or more groups refers to three hydrogen atoms on the designated atom or group being independently replaced with three selections from the indicated group of substituents. In some embodiments, “substituted with one or more groups” refers to four hydrogen atoms on the designated atom or group being independently replaced with four selections from the indicated group of substituents.
  • enantiomers including racemates, mixtures of diastereomers, and other mixtures thereof, to the extent they can be made by one of ordinary skill in the art by routine experimentation.
  • the single enantiomers or diastereomers i.e., optically active forms, can be obtained by asymmetric synthesis or by resolution of the racemates or mixtures of diastereomers.
  • Resolution of the racemates or mixtures of diastereomers can be accomplished, for example, by conventional methods such as crystallization in the presence of a resolving agent, or
  • Such compounds include Z- and E- forms (or cis- and trans- forms) of compounds with carbon-carbon double bonds.
  • the term "compound” is intended to include, to the extent they can be made without undue experimentation, all tautomeric forms of the compound.
  • Such compounds also include crystal forms including polymorphs and clathrates, to the extent they can be made by one of ordinary skill in the art by routine experimentation.
  • salt is intended to include all isomers, racemates, other mixtures, Z- and E-forms, tautomeric forms and crystal forms of the salt of the compound, to the extent they can be made by one of ordinary skill in the art without undue experimentation.
  • “Pharmaceutically acceptable salts” include, but are not limited to salts with inorganic acids, such as hydrochlorate, phosphate, diphosphate, hydrobromate, sulfate, sulfinate, nitrate, and like salts; as well as salts with an organic acid, such as malate, maleate, fumarate, tartrate, succinate, citrate, lactate, methanesulfonate, p-toluenesulfonate, 2-hydroxyethylsulfonate, benzoate, salicylate, stearate, alkanoate such as acetate, and salts with HOOC-(CH 2 ) n -COOH where n is 0-4, and like salts.
  • pharmaceutically acceptable cations include, but are not limited to sodium, potassium, calcium, aluminum, lithium, and ammonium.
  • the free base can be obtained by basifying a solution of the acid salt.
  • an addition salt particularly a pharmaceutically acceptable addition salt, may be produced by dissolving the free base in a suitable organic solvent and treating the solution with an acid, in accordance with conventional procedures for preparing acid addition salts from base compounds.
  • a "solvate,” such as a “hydrate,” is formed by the interaction of a solvent and a compound.
  • the term “compound” is intended to include solvates, including hydrates, of compounds, to the extent they can be made by one of ordinary skill in the art by routine experimentation.
  • “salts” includes solvates, such as hydrates, of salts, to the extent they can be made by one of ordinary skill in the art by routine
  • Suitable solvates are pharmaceutically acceptable solvates, such as hydrates, including monohydrates and hemi-hydrates, to the extent they can be made by one of ordinary skill in the art by routine experimentation.
  • a "chelate” is formed by the coordination of a compound to a metal ion at two (or more) points.
  • the term “compound” is intended to include chelates of compounds.
  • salts includes chelates of salts.
  • a "non-covalent complex” is formed by the interaction of a compound and another molecule wherein a covalent bond is not formed between the compound and the molecule. For example, complexation can occur through van der Waals
  • hydrogen bond refers to a form of association between an electronegative atom (also known as a hydrogen bond acceptor) and a hydrogen atom attached to a second, relatively electronegative atom (also known as a hydrogen bond donor).
  • Suitable hydrogen bond donor and acceptors are well understood in medicinal chemistry (G. C. Pimentel and A. L. McClellan, The Hydrogen Bond, Freeman, San Francisco, 1960; R. Taylor and O. Kennard, "Hydrogen Bond Geometry in Organic Crystals", Accounts of Chemical Research, 17, pp. 320-326 (1984)).
  • group As used herein the terms "group”, “radical” or “fragment” are synonymous and are intended to indicate functional groups or fragments of molecules attachable to a bond or other fragments of molecules.
  • active agent is used to indicate a chemical substance which has biological activity.
  • an “active agent” is a chemical substance having pharmaceutical utility.
  • Treating refers to administering at least one compound and/or at least one pharmaceutically acceptable salt thereof described herein to a subject that has cancer, or has a symptom of cancer, or has a
  • the term "effective amount” refers to an amount of at least one compound and/or at least one pharmaceutically acceptable salt thereof described herein effective to "treat,” as defined above, a disease or disorder in a subject. In the case of cancer, the effective amount may cause any of the changes observable or measurable in a subject as described in the definition of "treating,” “treat,” “treatment” and “alleviation” above.
  • the effective amount can reduce the number of cancer or tumor cells; reduce the tumor size; inhibit or stop tumor cell infiltration into peripheral organs including, for example, the spread of tumor into soft tissue and bone; inhibit and stop tumor metastasis; inhibit and stop tumor growth; relieve to some extent one or more of the symptoms associated with the cancer, reduce morbidity and mortality; improve quality of life; or a combination of such effects.
  • An effective amount may be an amount sufficient to decrease the symptoms of a disease responsive to inhibition of PI3K/mTOR activity.
  • efficacy in vivo can, for example, be measured by assessing the duration of survival, time to disease progression (TTP), the response rates (RR), duration of response, and/or quality of life. Effective amounts may vary, as recognized by those skilled in the art, depending on route of administration, excipient usage, and co-usage with other agents.
  • inhibitortion indicates a decrease in the baseline activity of a biological activity or process.
  • “Inhibition of PI3K and/or mTOR activity” refers to a decrease in the activity of PI3K and/or mTOR as a direct or indirect response to the presence of at least one compound and/or at least one pharmaceutically acceptable salt described herein, relative to the activity of PI3K and/or mTOR in the absence of the at least one compound and/or the at least one pharmaceutically acceptable salt thereof.
  • the decrease in activity is not bound by theory and may be due to the direct interaction of the at least one compound and/or at least one pharmaceutically acceptable salt thereof described herein with PI3K and/or mTOR, or due to the interaction of the at least one compound and/or at least one pharmaceutically acceptable salt described herein, with one or more other factors that in turn affect PI3K and/or mTOR activity.
  • the presence of at least one compound and/or at least one pharmaceutically acceptable salt thereof described herein may decrease PI3K and/or mTOR activity by directly binding to the PI3K and/or mTOR, by causing (directly or indirectly) another factor to decrease PI3K and/or mTOR activity, or by (directly or indirectly) decreasing the amount of PI3K and/or mTOR present in the cell or organism.
  • a 1 is N or CH
  • a 4 and A 5 are independently N or CR 2 ;
  • a 2 and A 3 together with B ring, are a 5-membered heteroaryl or heterocycle containing 1 to 4 heteroatoms selected from N, O, and S, and said 5-membered heteroaryl or heterocycle is optionally substituted by one or more groups
  • a and A , together with the B ring, are not is a single bond or a double bond;
  • R 1 is heteroaryl, optionally substituted by one or more groups independently chosen from alkyl, alkenyl, alkynyl, aryl, cycloalkyl, oxo, -C(O)R a , -C(O)OR b , -CN, - C(O)NR c R d , -NR c R d , -NR c C(O)R a , -NR c S(O) n R e , -NR c S(O) n f R g , -NR c C(O)OR b , - NR c C(O)NR d R e , -NO 2 , -OR b , -S(O) n R e , -S(O) n NR c R d , halo, haloalkyl, heteroaryl, and heterocycle;
  • R and R 2 are independently chosen from H, alkyl, alkenyl, alkynyl, aryl, cycloalkyl, -C(O)R a , -C(O)OR b , -CN, -C(O)NR c R d , -NR c R d , -NR c C(O)R a , -NR c S(O) n R e , -NR c S(O) n NR f R g , -NR c C(O)OR b , -NR c C(O)NR d R e , -NO 2 , -OR b , -OC(O)R a , - OC(O)NR c R d , -S(O) n R e , -S(O) n NR c R d , halo, haloalkyl, heteroaryl, and heterocycle
  • R a , R b , R c , R d , R e , R f and R g are each independently chosen from H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl optionally substituted aryl, optionally substituted cycloalkyl, optionally substituted haloalkyl, optionally substituted heteroaryl, and optionally substituted heterocycle,
  • R a and R c , and/or R c and R d , and/or R c and R e , and/or R c and R f , and/or R d and R e , and /or R g and R f together with the atom(s) to which they are attached, form a 3-10 membered optionally substituted heterocycle ring;
  • n is independently 0, 1 , or 2; wherein each optionally substituted group above can be unsubstituted or independently substituted with one or more, such as one, two, or three, substituents independently selected from, Ci-C 4 alkyl, cycloalkyi, oxo, aryl, heterocycle, heteroaryl, aryl-Ci-C 4 alkyl-, heteroaryl-Ci-C 4 alkyl-, Ci-C 4 haloalkyl-, -OCi-C 4 alkyl,
  • -OC1-C4 alkylphenyl -d-C 4 alkyl-OH, -C1-C4 alkyl-O-d-C 4 alkyl, -OC1-C4 haloalkyl, halo, -OH, -NH 2 , -d-C 4 alkyl-NH 2 , -N(d-C 4 alkyl)(Ci-C 4 alkyl), -NH(d-C 4 alkyl), -N(Ci-C 4 alkyl)(Ci-C 4 alkylphenyl), -NH(Ci-C 4 alkylphenyl), cyano, nitro, oxo, -CO2H, -C(O)OCi-C 4 alkyl, -C(O)Ocycloalkyl, -C(O)Oaryl, -C(O)Oheteroaryl, - C(O)Oheterocycle, -CON(Ci-C 4
  • -C(O)(cycloalkyl), -C(O)(heterocycle), -C(O)(aryl) such as -C(O) phenyl, - C(O)(heteroaryl), -C(O)d-C 4 haloalkyl, -OC(O)d-C 4 alkyl, -OC(O)(cycloalkyl), - OC(O)(heterocycle), - - OC(O)(heteroaryl), OC(O)(aryl) such as -OC(O)phenyl, - SO 2 (Ci-C 4 alkyl), - SO 2 (cycloalkyl), -SO 2 (heterocycle), -SO 2 (aryl) such as SO 2 (phenyl), -SO 2 (heteroaryl), -SO 2 (Ci-C 4 haloalkyl), -SO 2 NH 2 , -SO 2 NR'R" where
  • -SO 2 NH(cycloalkyl), -SO 2 NH(heterocycle), -SO 2 NH(aryl) such as -SO 2 NH(phenyl), -SO 2 NH(heteroaryl), -NHSO 2 (Ci-C 4 alkyl), NHSO 2 (cycloalkyl), NHSO 2 (heterocycle), NHSO 2 (aryl) such as -NHSO 2 (phenyl), -NHSO 2 (heteroaryl), and
  • -NHSO 2 (Ci-C 4 haloalkyl), in which each of alkyl, phenyl, aryl, cycloalkyi, heterocycle, and heteroaryl is optionally substituted by one or more groups independently chosen from -OH, halo, cycloalkyi, heterocycle, Ci-C 4 alkyl, Ci-C 4 haloalkyl-, -OCi-C 4 alkyl, C1-C4 alkyl-OH, -C1-C4 alkyl-O-d-C 4 alkyl, -OC1-C4 haloalkyl, cyano, nitro, -NH 2 , - CO 2 H, -C(O)OCi-C 4 alkyl, -CON(d-C 4 alkyl )(Ci-C 4 alkyl), -CONH(d-C 4 alkyl),
  • a 2 and A 3 together with B ring, are a 5-membered heteroaryl or heterocycle containing 1 to 3 heteroatoms selected from N, O and S. In some embodiments, A 2 and A 3 , together with B ring, are a 5-membered heteroaryl or heterocycle containing 1 to 3 nitrogen heteroatoms.
  • a 2 and A 3 together with B ring, can be selected from structures (2)-(6)
  • t is 1 , 2 or 3;
  • R 3 is independently chosen from H, C1-C6 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C 6 -Ci 4 aryl, C3-C9 cycloalkyi, -C(O)OR b , -CN, -C(O)NR c R d , -NR c R d , -NR c C(O)R a , -
  • a 4 is not CR 2 , wherein R 2 is as defined above.
  • R 3 is independently chosen from H, OH, CN, NO2, halo, C1-C6 alkyl,C2-Cs alkenyl, C2-C8 alkynyl, C6-Ci 4 aryl, C3-C9 cydoalkyi, heteroaryl, and heterocyde, wherein each of the alkyl, alkenyl, alkynyl, aryl, cydoalkyi, heteroaryl, and heterocyde can be optionally substituted by one or more groups independently chosen from optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted cydoalkyi, -OH, oxo, -C(O)R a , -C(O)OR b , -CN, -C(O)NR c R d , -NR c R d , -NR
  • a 2 and A 3 together with B ring are chosen from the following structures (2)-(5), wherein R 3 and t are as defined above.
  • a 2 , A 3 and together with the B ring are chosen from structure (3)-(4), wherein R 3 and t are as defined above.
  • a 4 is N or CH.
  • a 5 is N or CH.
  • Ai, A4, and A 5 are CH.
  • Ai and A 5 are CH, and A4 is N.
  • R 1 is a heteroaryl that is chosen from the following structures
  • R 1 is a heteroaryl chosen from
  • each of which is optionally substituted by one or more groups independently chosen from alkyi, alkenyl, alkynyl, aryl, cydoalkyi, haloalkyi, heteroaryl, heterocyde, oxo, -C(O)R a , -C(O)OR b , -CN, -C(O)NR c R d , -NR c R d , -
  • optionally substituted alkyi optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted cydoalkyi, -OH, oxo, - C(O)R a , -C(O)OR b , -CN, -C(O)NR c R d , -NR c R d , -NR c C(O)R a , -NR c S(O) n R e , - NR c S(O) n NR f R g , -NR c C(O)OR b , -NR c C(O)NR d R e , -NO 2 , OR b , -S(O) n R e , -S(O) n NR c R d , halo, optionally substituted haloalkyi, optionally substituted heteroaryl, and optional
  • R a , R b , R c , R d , R e , R f and R g are each independently chosen from H, alkyi, aryl, cydoalkyi, haloalkyi, heteroaryl, and heterocyde, wherein each of the alkyi, aryl, cydoalkyi, heteroaryl, and heterocyde in R a , R b , R c , R d , R e , R f and R g is optionally substituted by one or more, such as one or two or three, substitutents independently selected from halo and alkyi.
  • R 1 is
  • alkyl alkenyl, and alkynyl, wherein each of which can be optionally substituted by groups independently chosen from optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted cycloalkyi, -OH, oxo, -C(O)R a , -C(O)OR b , -CN, -C(O)NR c R d , -NR c R d , -
  • R a , R b , R c , R d , R e , R f and R 9 are each independently chosen from H, alkyl, aryl, cycloalkyi, haloalkyi, heteroaryl, and heterocycle, for example, R a , R b , R c , R d , R e , R f and R 9 are each independently chosen from H, C1-C6 alkyl, phenyl, C3-C6 cycloalkyi, C1-C3 haloalkyi, heteroaryl, and heterocycle, wherein each of the alkyl, aryl, cycloalkyi, heteroaryl, and heterocycle in R a , R b , R c , R d , R e , R f and R 9 is optionally substituted by one or more, such as one or two or three, substitutents independently selected from halo and alkyl.
  • At least one compound selected from compounds 1 to 184 and/or at least one pharmaceutically acceptable salt described herein are also provided.
  • the compounds described herein, and/or the pharmaceutically acceptable salts thereof, can be synthesized from commercially available starting materials by methods well known in the art, taken together with the disclosure in this patent application. The following schemes illustrate methods for preparation of some of the compounds disclosed herein.
  • the compounds thus obtained can be further modified at their peripheral positions to provide the desired compounds.
  • Synthetic chemistry transformations are described, for example, in R. Larock, Comprehensive Organic Transformations, VCH Publishers (1989); T.W. Greene and P.G.M. Wuts, Protective Groups in Organic Synthesis, 3 rd Ed., John Wiley and Sons (1999); L. Fieser and M. Fieser, Fieser and Fieser's Reagents for Organic Synthesis, John Wiley and Sons (1994); and L.
  • the at least one compound and/or at least one pharmaceutically acceptable salt described herein can be purified by column chromatography, high performance liquid chromatography, crystallization, or other suitable methods.
  • composition comprising at least one compound and/or at least one pharmaceutically acceptable salt described herein, and at least one pharmaceutically acceptable carrier.
  • a composition comprising at least one compound and/or at least one pharmaceutically acceptable salt described herein can be administered in various known manners, such as orally, parenterally, by inhalation spray, or via an implanted reservoir.
  • parenteral as used herein includes subcutaneous,
  • An oral composition can be any orally acceptable dosage form including, but not limited to, tablets, capsules, emulsions, and aqueous suspensions, dispersions and solutions.
  • Commonly used carriers for tablets include lactose and corn starch.
  • Lubricating agents, such as magnesium stearate, are also typically added to tablets.
  • useful diluents include lactose and dried corn starch.
  • the active ingredient can be suspended or dissolved in an oily phase combined with emulsifying or suspending agents. If desired, certain sweetening, flavoring, or coloring agents can be added.
  • a sterile injectable composition e.g., aqueous or oleaginous suspension
  • a sterile injectable composition can be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents.
  • the sterile injectable preparation can also be a sterile injectable solution or suspension in a non- toxic parenterally acceptable diluent or solvent, for example, as a solution in 1 ,3- butanediol.
  • suitable dispersing or wetting agents such as, for example, Tween 80
  • the sterile injectable preparation can also be a sterile injectable solution or suspension in a non- toxic parenterally acceptable diluent or solvent, for example, as a solution in 1 ,3- butanediol.
  • a non- toxic parenterally acceptable diluent or solvent for example, as a solution in 1 ,3- butanediol.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium (e.g., synthetic mono- or di-glycerides).
  • Fatty acids such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • These oil solutions or suspensions can also contain a long-chain alcohol diluent or dispersant, or carboxymethyl cellulose or similar dispersing agents.
  • An inhalation composition can be prepared according to techniques well known in the art of pharmaceutical formulation and can be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other solubilizing or dispersing agents known in the art.
  • a topical composition can be formulated in form of oil, cream, lotion, ointment and the like.
  • Suitable carriers for the composition include vegetable or mineral oils, white petrolatum (white soft paraffin), branched chain fats or oils, animal fats and high molecular weight alcohols (greater than C12).
  • the composition includes vegetable or mineral oils, white petrolatum (white soft paraffin), branched chain fats or oils, animal fats and high molecular weight alcohols (greater than C12).
  • pharmaceutically acceptable carrier is one in which the active ingredient is soluble.
  • Emulsifiers, stabilizers, humectants and antioxidants may also be included as well as agents imparting color or fragrance, if desired.
  • transdermal penetration enhancers may be employed in those topical formulations. Examples of such enhancers can be found in U.S. Patents 3,989,816 and 4,444,762.
  • Creams may be formulated from a mixture of mineral oil, self-emulsifying beeswax and water in which mixture the active ingredient, dissolved in a small amount of an oil, such as almond oil, is admixed.
  • An example of such a cream is one which includes about 40 parts water, about 20 parts beeswax, about 40 parts mineral oil and about 1 part almond oil.
  • Ointments may be formulated by mixing a solution of the active ingredient in a vegetable oil, such as almond oil, with warm soft paraffin and allowing the mixture to cool.
  • An example of such an ointment is one which includes about 30% by weight almond oil and about 70% by weight white soft paraffin.
  • a pharmaceutically acceptable carrier refers to a carrier that is compatible with active ingredients of the composition (and in some embodiments, capable of stabilizing the active ingredients) and not deleterious to the subject to be treated.
  • solubilizing agents such as cyclodextrins (which form specific, more soluble complexes with the at least one compound and/or at least one pharmaceutically acceptable salt described herein), can be utilized as pharmaceutical excipients for delivery of the active ingredients.
  • examples of other carriers include colloidal silicon dioxide, magnesium stearate, cellulose, sodium lauryl sulfate, and pigments such as D&C Yellow # 10.
  • Suitable in vitro assays can be used to preliminarily evaluate the efficacy of the at least one compound and/or at least one pharmaceutically acceptable salt described herein, in inhibiting the activity of PI3K and/or mTOR.
  • the at least one compound and/or at least one pharmaceutically acceptable salt described herein can further be examined for efficacy in treating cancer by in vivo assays.
  • the compounds described herein, and/or the pharmaceutically acceptable salts thereof can be administered to an animal (e.g., a mouse model) having cancer and its therapeutic effects can be accessed. Positve results in one or more of such tests are sufficient to increase the scientific storehouse of knowledge and hence sufficient to demonstrate practical utility of the compounds and/or salts tested. Based on the results, an appropriate dosage range and administration route for animals, such as humans, can also be determined.
  • the method comprises contacting the enzyme with at least one compound and/or at least one pharmaceutically acceptable salt described herein in an amount effective to inhibit the activity of PI3K and/or mTOR.
  • the at least one compound and/or at least one pharmaceutically acceptable salt described herein can be used to achieve a beneficial therapeutic or prophylactic effect, for example, in subjects with cancer.
  • cancer refers to a cellular disorder characterized by uncontrolled or disregulated cell proliferation, decreased cellular differentiation, inappropriate ability to invade surrounding tissue, and/or ability to establish new growth at ectopic sites.
  • cancer includes, but is not limited to, solid tumors and bloodborne tumors.
  • cancer encompasses diseases of skin, tissues, organs, bone, cartilage, blood, and vessels.
  • cancer further encompasses primary and metastatic cancers.
  • Non-limiting examples of solid tumors include pancreatic cancer; bladder cancer; colorectal cancer; breast cancer, including metastatic breast cancer; prostate cancer, including androgen-dependent and androgen-independent prostate cancer; renal cancer, including, e.g., metastatic renal cell carcinoma; hepatocellular cancer; lung cancer, including, e.g., non-small cell lung cancer (NSCLC), bronchioloalveolar carcinoma (BAC), and adenocarcinoma of the lung; ovarian cancer, including, e.g., progressive epithelial or primary peritoneal cancer; cervical cancer; gastric cancer; esophageal cancer; head and neck cancer, including, e.g., squamous cell carcinoma of the head and neck; skin cancer, including e.g., malignant melanoma;
  • NSCLC non-small cell lung cancer
  • BAC bronchioloalveolar carcinoma
  • ovarian cancer including, e.g., progressive epithelial or primary peritoneal cancer
  • neuroendocrine cancer including metastatic neuroendocrine tumors; brain tumors, including, e.g., glioma, anaplastic oligodendroglioma, adult glioblastoma multiforme, and adult anaplastic astrocytoma; bone cancer; soft tissue sarcoma; and thyroid carcinoma.
  • brain tumors including, e.g., glioma, anaplastic oligodendroglioma, adult glioblastoma multiforme, and adult anaplastic astrocytoma
  • bone cancer soft tissue sarcoma
  • thyroid carcinoma include pancreatic cancer; bladder cancer; colorectal cancer; breast cancer, and ovarian cancer.
  • Non-limiting examples of hematologic malignancies include acute myeloid leukemia (AML); chronic myelogenous leukemia (CML), including accelerated CML and CML blast phase (CML-BP); acute lymphoblastic leukemia (ALL); chronic lymphocytic leukemia (CLL); Hodgkin's disease (HD); non-Hodgkin's lymphoma (NHL), including follicular lymphoma and mantle cell lymphoma; B-cell lymphoma; T-cell lymphoma; multiple myeloma (MM); Waldenstrom's macroglobulinemia;
  • AML acute myeloid leukemia
  • CML chronic myelogenous leukemia
  • ALL acute lymphoblastic leukemia
  • CLL chronic lymphocytic leukemia
  • HD Hodgkin's disease
  • NHL non-Hodgkin's lymphoma
  • B-cell lymphoma including follicular lymphoma and mantle
  • MDS myelodysplastic syndromes
  • RA refractory anemia
  • RARS refractory anemia with ringed siderblasts
  • RAEB refractory anemia with excess blasts
  • RAEB-T myelodysplastic syndromes
  • the examples of the cancer to be treated include, but are not limited to, lung cancer, head and neck cancer, colorectal cancer, pancreatic cancer, colon cancer, breast cancer, ovarian cancer, prostate cancer, stomach cancer, kidney cancer, liver cancer, brain cancer, bone cancer, and leukemia.
  • the at least one compound and/or at least one pharmaceutically acceptable salt described herein is administered in conjunction with another therapeutic agent.
  • the other therapeutic agent is one that is normally administered to patients with the disease or condition being treated.
  • the at least one compound and/or at least one pharmaceutically acceptable salt described herein may be administered with the other therapeutic agent in a single dosage form or as a separate dosage form.
  • the other therapeutic agent may be administered prior to, at the same time as, or following administration of the at least one compound and/or at least one
  • anti-neoplastic agent refers to any agent that is administered to a subject with cancer for purposes of treating the cancer.
  • Nonlimiting examples of anti-neoplastic agents include: radiotherapy; immunotherapy; DNA damaging chemotherapeutic agents; and chemotherapeutic agents that disrupt cell replication.
  • Non-limiting examples of DNA damaging chemotherapeutic agents include topoisomerase I inhibitors (e.g., irinotecan, topotecan, camptothecin and analogs or metabolites thereof, and doxorubicin); topoisomerase II inhibitors (e.g., etoposide, teniposide, and daunorubicin); alkylating agents (e.g., melphalan, chlorambucil, busulfan, thiotepa, ifosfamide, carmustine, lomustine, semustine, streptozocin, decarbazine, methotrexate, mitomycin C, and cyclophosphamide); DNA intercalators (e.g., cisplatin, oxaliplatin, and carboplatin); DNA intercalators and free radical generators such as bleomycin; and nucleoside mimetics (e.g., 5-fluorouracil, capec
  • Chemotherapeutic agents that disrupt cell replication include: paclitaxel, docetaxel, and related analogs; vincristine, vinblastin, and related analogs;
  • thalidomide and related analogs e.g., CC-5013 and CC-4047
  • protein tyrosine kinase inhibitors e.g., imatinib mesylate and gefitinib
  • proteasome inhibitors e.g., bortezomib
  • NF-kappa B inhibitors including inhibitors of I kappa B kinase
  • antibodies which bind to proteins overexpressed in cancers and thereby downregulate cell replication e.g., trastuzumab, rituximab, cetuximab, and bevacizumab
  • other inhibitors of proteins or enzymes known to be upregulated, over-expressed or activated in cancers, the inhibition of which downregulates cell replication e.g., trastuzumab, rituximab, cetuximab, and bevacizumab
  • 6-chloro-1 ,5-naphthyridin-4-ol(1 .45 g, 8 mmol) was added to ice-cooled cone.
  • H 2 SO 4 (15 ml_) and followed by the addition of KNO 3 (1 .62 g, 16 mmol) slowly at 0°C.
  • the mixture was heated to 100°C for 1 h, and then was poured into ice-water.
  • the precipitate was collected and dried in vacuo to afford 6-chloro-3-nitro-1 ,5-naphthyridin- 4-ol as a solid in 90% yield (1 .97 g).
  • Fluorescence polarization was used in this assay.
  • the final conditions for kinase assay are 10 ⁇ of ATP, 0.2 ng/ L of PI3Ka kinase, 30 ⁇ /L of lipid substrate and assay buffer (50 mmol/L of HEPES (pH 7.5),100 mmol/L of NaCI, 1 mmol/L of EGTA, 3 mmol/L of MgCI 2 , 1 mmol/L of DTT and 0.03% CHAPS and 2% DMSO).
  • test compounds in 10% DMSO and10 pL of 0.5 ng/ L PI3K kinase (Invitrogen, PV4788) in assay buffer are put into a 96-well plate (Greiner, Cat.
  • a standard curve for ADP is obtained in a parallel way by replacing compound and PI3Ka kinase with DMSO and assay buffer, respectively. Different concentration of ADP, 0 -10 ⁇ , and ATP, 10-0 ⁇ ( ATP+ADP equal to 10 ⁇ ) are applied instead of a fixed concentration of ATP in this standard curve assay. Other conditions are the same as described above.
  • the standard curve is plotted using Origin 8.0 software.
  • the inhibition of test compound on ADP production is calculated based on ADP concentration from standard curve.
  • IC50 is obtained using XLfit 2.0 software.
  • PC3 human prostate cancer cell line
  • F-12 F-12 medium with 10% fetal bovine serum.
  • PC3 cells are seeded at a density of 5000 cells/90 ⁇ _ in Poly-D-Lysine 96-well plate (BD, 356692). After incubation for 24 hours, different concentrations of test compounds (10 ⁇ _) is added and cells are incubated for another 2 hours. 100 ⁇ _ of 4% prewarmed paraformaldehyde is added, and cells are fixed for 45min at room temperature.
  • 35 ⁇ _ of Alexa Fluor 488 goat anti-rabbit IgG (Invitrogen, A1 1034), in a 1 :1000 dilution buffer (0.1 % BSA in PBS), is added, and the reaction mixture is incubated for 1 .5 hours in the dark. It is washed twice with 160 ⁇ _ of PBS, and then 35 ⁇ _ of 1 .5 ⁇ propidium iodide (Sigma, P4170) is added to each well, and reaction plate is incubated at 37 °C, 5% CO2 for 30min. Finally, the plate is loaded into the Acumen eX3 (TTP LabTech) and scanned with the appropriate instrument settings. [0133] The inhibition of test compound is calculated based on the ratio of compound treated and untreated cells. IC50 is generated using XLfit 2.0 software.
  • Each compound specifically exemplified in the invention inhibited the PI3Ka kinase receptor with IC50 ⁇ 1 -0 ⁇ .
  • conditions for kinase assay are: 10 ⁇ /L of ATP, 0.2 ng/ L of mTOR kinase, 0.4 ⁇ /L of GFP-4EBP1 substrate and assay buffer (50 mmol/L of HEPES,pH 7.5, 0.01 % of Tween 20, 1 mmol/L of EGTA, 10 mmol/L of MnCI 2 , 2 mmol/L of DTT and 1 % DMSO).
  • test compounds in 4% DMSO and 2.5 ⁇ of 0.8 ng/ ⁇ mTOR kinase (Invitrogen, PV4753) diluted in assay buffer are added.
  • the reaction is initiated by the addition of 5 ⁇ mixture of 0.8 ⁇ /L GFP-4EBP1 Substrate
  • test compound The inhibition of test compound is calculated based on the ratio of 528 nm/490 nm.
  • IC50 of test compound is obtained using XLfit 2.0 software.
  • PC3 human prostate cancer cell line
  • F-12 F-12 medium with 10% fetal bovine serum.
  • PC3 cells are seeded at density of 5000 cells/90 ⁇ _ in Poly-D-Lysine 96-well plate (BD, 356692). After incubation for 24 hours, 10 ⁇ _ of different concentration of test compounds is added and cells are incubated for 2 hours, followed by the addition of 100 ⁇ _ of 4% pre-warmed paraformaldehyde. The cells are fixed at room temperature for 45min.
  • the inhibition of the test compound is calculated based on the ratio of compound treated and untreated cells.
  • IC50 is generated using XLfit 2.0 software.
  • Each compound specifically exemplified in the invention inhibited mTOR with IC50 ⁇ 10.0 ⁇ .

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Oxygen Or Sulfur (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)
  • Plural Heterocyclic Compounds (AREA)

Abstract

Provided are certain fused heteroaryls, compositions thereof and methods of use therefor.

Description

FUSED HETEROARYLS AND THEIR USES
[001] Phosphoinositide 3-kinases (PI 3-kinases or PI3Ks) are a family of enzymes that may be involved in cellular functions such as cell growth, proliferation, differentiation, motility, survival and intracellular trafficking, which in turn can be involved in cancer.
[002] The PI3K family may include four distinct classes defined by structural and functional characteristics and termed as Classes l-IV. The most fully characterized class may be the Class l-PI3Ks. Class I comprises three class I a isoforms - PI3Ka, ΡΙ3Κβ and ΡΙ3Κδ. PI3Ka appears to be highly relevant in human cancers and malignancies. PI3Ka can be overexpressed in human cancers.
[003] Mammalian target of rapamycin (mTOR) is the downstream kinase of PI3K family. Inhibition of mTOR can inhibit the activity of PI3K. Therefore, the PI3K/mTOR pathway can be exploited for new cancer drug discovery.
[004] Provided is at least one compound of formula 1 :
Figure imgf000002_0001
(1 )
and/or at least one pharmaceutically acceptable salt thereof wherein
A1 is N or CH;
A4 and A5 are independently N or CR2;
A2 and A3, together with B ring are a 5-membered heteroaryl or heterocycli containing 1 to 4 heteroatoms selected from N, O, and S, and said 5-membered heteroaryl or heterocycle is optionally substituted by one or more groups
independently chosen from alkyl, alkenyl, alkynyl, aryl, cycloalkyl, oxo, -C(O)Ra, - C(O)ORb, -CN, -C(O)NRcRd, -NRcRd, -NRcC(O)Ra, -NRcS(O)nRe, -NRcS(O)nNRfRg, - NRcC(O)ORb, -NRcC(O)NRdRe, -NO2, -ORb, -S(O)nRe, -S(O)nNRcRd, halo, haloalkyi, heteroaryl, and heterocycle;
provided that A2 and A3, together with the B ring are not
Figure imgf000003_0001
is a single bond or a double bond;
R1 is heteroaryl, optionally substituted by one or more groups independently chosen from alkyl, alkenyl, alkynyl, aryl, cycloalkyl, oxo, -C(O)Ra, -C(O)ORb, -CN, - C(O)NRcRd, -NRcRd, -NRcC(O)Ra, -NRcS(O)nRe, -NRcS(O)nNRfRg, -NRcC(O)ORb, - NRcC(O)NRdRe, -NO2, -ORb, -S(O)nRe, -S(O)nNRcRd, halo, haloalkyi, heteroaryl, and heterocycle;
R and R2 are independently chosen from H, alkyl, alkenyl, alkynyl, aryl, cycloalkyl, -C(O)Ra, -C(O)ORb, -CN, -C(O)NRcRd, -NRcRd, -NRcC(O)Ra, -NRcS(O)nRe, -NRcS(O)nNRfRg, -NRcC(O)ORb, -NRcC(O)NRdRe, -NO2, -ORb, -OC(O)Ra, - OC(O)NRcRd, -S(O)nRe, -S(O)nNRcRd, halo, haloalkyi, heteroaryl, and heterocycle; and each of said above alkyl, alkenyl, alkynyl, aryl, cycloalkyl, haloalkyi, heteroaryl and heterocycle can be optionally substituted by one or more groups independently chosen from optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted cycloalkyl, -OH, oxo, -C(O)Ra, -C(O)ORb, -CN, -C(O)NRcRd, -NRcRd, -NRcC(O)Ra, - NRcS(O)nRe, -NRcS(O)nNRfRg, -NRcC(O)ORb, -NRcC(O)NRdRe, -NO2, ORb, -S(O)nRe, - S(O)nNRcRd, halo, optionally substituted haloalkyi, optionally substituted heteroaryl, and optionally substituted heterocycle;
Ra, Rb, Rc, Rd, Re, Rf and Rg are each independently chosen from H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl optionally substituted aryl, optionally substituted cycloalkyi, optionally substituted haloalkyl, optionally substituted heteroaryl, and optionally substituted heterocycle,
or Ra and Rc, and/or Rc and Rd, and/or Rc and Re, and/or Rc and Rf, and/or Rd and Re, and /or Rg and Rf together with the atom(s) to which they are attached, form a 3-10 membered optionally substituted heterocycle ring; and
for each occurrence, n is independently 0, 1 , or 2;
wherein each optionally substituted group above can be unsubstituted or independently substituted with one or more, such as one, two, or three, substituents independently selected from Ci-C4 alkyl, cycloalkyi, oxo, aryl, heterocycle, heteroaryl, aryl-Ci-C4 alkyl-, heteroaryl-Ci-C4 alkyl-, Ci-C4 haloalkyl-, -OCi-C4 alkyl,
-OC1-C4 alkylphenyl, -d-C4 alkyl-OH, -C1-C4 alkyl-O-d-C4 alkyl, -OC1-C4 haloalkyl, halo, -OH, -NH2, -d-C4 alkyl-NH2, -N(d-C4 alkyl)(Ci-C4 alkyl), -NH(Ci-C4 alkyl), -N(Ci-C4 alkyl)(Ci-C4 alkylphenyl), -NH(Ci-C4 alkylphenyl), cyano, nitro, oxo, -CO2H, -C(O)OCi-C4 alkyl, -C(O)Ocycloalkyl, -C(O)Oaryl, -C(O)Oheteroaryl, - C(O)Oheterocycle, -CON(Ci-C4 alkyl)(Ci-C4 alkyl), -CONR'R" wherein R' and R" with the N to which they are attached form a heterocycle, -CON(cycloalkyl)(cycloalkyl), - CON(heterocycle)(heterocycle), -CONH(Ci-C4 alkyl), -CONH(cycloalkyl), - CONH(heterocycle), -CONH2, -NHC(0)(Ci-C4 alkyl), -NHC(O)(cycloalkyl), - NHC(O)(heterocycle), -NHC(O)(aryl) such as -NHC(O)(phenyl), -NHC(O)(heteroaryl), -N(Ci-C4 alkyl)C(O)(Ci-C4 alkyl), -N(d-C4 alkyl)C(O)(cycloalkyl),
-N(Ci-C4 alkyl )C(O)(heterocycle), -N(Ci-C4 alkyl )C(O)(aryl) such as
-N(Ci-C4 alkyl)C(O)(phenyl), N(Ci-C4 alkyl )C(O)(heteroaryl), -C(O)d-C4 alkyl,
-C(O)(cycloalkyl), -C(O)(heterocycle), -C(O)(aryl) such as -C(O) phenyl, - C(O)(heteroaryl), -C(O)d-C4 haloalkyl, -OC(O)d-C4 alkyl, -OC(O)(cycloalkyl), - OC(O)(heterocycle), - - OC(O)(heteroaryl), OC(O)(aryl), such as -OC(O)phenyl, - SO2(Ci-C4 alkyl), - SO2(cycloalkyl), -SO2(heterocycle), -SO2(aryl) such as SO2(phenyl), -SO2(heteroaryl), -SO2(Ci-C4 haloalkyl), -SO2NH2, -SO2NR'R" wherein R' and R" with the N to which they are attached form a heterocycle, -SO2NH(Ci-C4 alkyl),
-SO2NH(cycloalkyl), -SO2NH(heterocycle), -SO2NH(aryl) such as -SO2NH(phenyl), -SO2NH(heteroaryl), -NHSO2(Ci-C4 alkyl), NHSO2(cycloalkyl), NHSO2(heterocycle), NHSO2(aryl) such as -NHSO2(phenyl), NHSO2(heteroaryl), and
-NHSO2(Ci-C4 haloalkyl), in which each of alkyl, phenyl, aryl, cycloalkyl, heterocycle, and heteroaryl is optionally substituted by one or more groups independently chosen from -OH, halo, cycloalkyl, heterocycle, Ci-C4 alkyl, Ci-C4 haloalkyl-, -OCi-C4 alkyl, C1 -C4 alkyl-OH, -d-C4 alkyl-O-d-C4 alkyl, -OC1 -C4 haloalkyl, cyano, nitro, -NH2, - CO2H, -C(O)OCi-C4 alkyl, -CON(C C4 alkyl )(Ci-C4 alkyl), -CONH(C C4 alkyl), -CONH2, -NHC(O)(Ci-C4 alkyl), and -N(d-C4 alkyl)C(0)(Ci-C4 alkyl).
[005] Also provided is a pharmaceutical composition comprising at least one compound and/or at least one pharmaceutically acceptable salt thereof described herein and at least one pharmaceutically acceptable carrier.
[006] Also provided is a method of inhibiting the activity of PI3K and/or mTOR comprising contacting the enzyme with an effective amount of at least one compound and/or at least one pharmaceutically acceptable salt thereof described herein.
[007] Also provided is a method of treating cancer responsive to inhibition of PI3K and/or mTOR comprising administering to a subject in need of treating for said cancer an effective amount of at least one compound and/or at least one pharmaceutically acceptable salt thereof described herein.
[008] Also provided is a use of at lease one compound and/or at least one pharmaceutically acceptable salt thereof described herein in the manufacture of a medicament for inhibiting the activity of PI3K and/or mTOR.
[009] Also provided is a use of at lease one compound and/or at least one pharmaceutically acceptable salt thereof described herein in the manufacture of a medicament for treating cancer. [010] As used in the present specification, the following words, phrases and symbols are generally intended to have the meanings as set forth below, except to the extent that the context in which they are used indicates otherwise. The following abbreviations and terms have the indicated meanings throughout:
[011] A dash ("-") that is not between two letters or symbols is used to indicate a point of attachment for a substituent. For example, -CONH2 is attached through the carbon atom.
[012] The term "alkyl" herein refers to a straight or branched hydrocarbon, containing 1 -18, such as 1 -12, further such as 1 -6 carbon atoms. Examples of alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, /-propyl, n-butyl, /-butyl, and /-butyl. "Lower alkyl" refers to a straight or branched hydrocarbon, containing 1 -6, such as 1 -4 carbon atoms.
[013] The term "alkoxy" herein refers to a straight or branched alkyl group of the indicated number of carbon atoms attached through an oxygen bridge such as, for example, methoxy, ethoxy, propoxy, isopropoxy, n-butoxy, sec-butoxy, tert-butoxy, pentoxy, 2-pentyloxy, isopentoxy, neopentoxy, hexoxy, 2-hexoxy, 3-hexoxy, 3- methyl pentoxy, and the like. Alkoxy groups will usually have from 1 to 6 carbon atoms attached through the oxygen bridge. ""Lower alkoxy" refers to a straight or branched alkoxy, wherein the alkyl portion contains 1 -4 carbon atoms.
[014] The term "alkenyl" herein refers to a straight or branched hydrocarbon, containing one or more C=C double bonds and 2-10, such as 2-6 carbon atoms.
Examples of alkenyl groups include, but are not limited to, vinyl, 2-propenyl, and 2- butenyl.
[015] The term "alkynyl" herein refers to a straight or branched hydrocarbon, containing one or more C≡C triple bonds and 2-10, such as 2-6 carbon atoms.
Examples of alkynyl groups include, but are not limited to, ethynyl, 2-propynyl, and 2- butynyl. [016] The term "cycloalkyl" refers to saturated and partially unsaturated cyclic hydrocarbon groups having 3 to 12, such as 3 to 8 carbon atoms. Examples of cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cycloheptyl, and cyclooctyl. The ring may be saturated or have one or more double bonds (i.e. partially unsaturated), but not fully conjugated, and not aromatic, as defined herein.
[017] "Aryl" encompasses:
5- and 6-membered carbocyclic aromatic rings, for example, phenyl;
bicyclic ring systems wherein at least one ring is carbocyclic and aromatic, for example, naphthalene, indane, and 1 ,2,3,4-tetrahydroquinoline; and
tricyclic ring systems wherein at least one ring is carbocyclic and aromatic, for example, fluorene.
[018] For example, aryl includes 5 and 6-membered carbocyclic aromatic rings fused to a 5- to 7-membered cycloalkyl or heterocyclic ring containing zero or more heteroatoms selected from N, O, and S, provided that the point of attachment is at the carbocyclic aromatic ring when the carbocyclic aromatic ring is fused with a
heterocyclic ring, and the point of attachment can be at the carbocyclic aromatic ring or at the cycloalkyl when the carbocyclic aromatic ring fused with a cycloalkyl.
Bivalent radicals formed from substituted benzene derivatives and having the free valences at ring atoms are named as substituted phenylene radicals. Bivalent radicals derived from univalent polycyclic hydrocarbon radicals whose names end in "-yl" by removal of one hydrogen atom from the carbon atom with the free valence are named by adding "-idene" to the name of the corresponding univalent radical, e.g., a naphthyl group with two points of attachment is termed naphthylidene. Aryl, however, does not encompass or overlap in any way with heteroaryl, separately defined below. Hence, if one or more carbocyclic aromatic rings are fused with a heterocyclic aromatic ring, the resulting ring system is heteroaryl, not aryl, as defined herein. [019] The term "halo" includes fluoro, chloro, bromo, and iodo, and the term
"halogen" includes fluorine, chlorine, bromine, and iodine.
[020] The term "heteroaryl" refers to
5- to 7-membered aromatic, monocyclic rings containing one or more, for example, from 1 to 4, or, in some embodiments, from 1 to 3, heteroatoms selected from N, O, and S, with the remaining ring atoms being carbon;
8- to 12-membered bicyclic rings containing one or more, for example, from 1 to
4, or, in some embodiments, from 1 to 3, heteroatoms selected from N, O, and S, with the remaining ring atoms being carbon and wherein at least one ring is aromatic and at least one heteroatom is present in the aromatic ring; and
1 1 - to 14-membered tricyclic rings containing one or more, for example, from 1 to 4, or in some embodiments, from 1 to 3, heteroatoms selected from N, O, and S, with the remaining ring atoms being carbon and wherein at least one ring is aromatic and at least one heteroatom is present in an aromatic ring.
[021] For example, heteroaryl includes a 5- to 7-membered heterocyclic aromatic ring fused to a 5- to 7-membered cycloalkyl ring. For such fused, bicyclic heteroaryl ring systems wherein only one of the rings contains one or more heteroatoms, the point of attachment may be at the heteroaromatic ring or at the cycloalkyl ring.
[022] When the total number of S and O atoms in the heteroaryl group exceeds 1 , those heteroatoms are not adjacent to one another. In some embodiments, the total number of S and O atoms in the heteroaryl group is not more than 2. In some embodiments, the total number of S and O atoms in the aromatic heterocycle is not more than 1 .
[023] Examples of heteroaryl groups include, but are not limited to, (as numbered from the linkage position assigned priority 1 ), pyridyl (such as 2-pyridyl, 3-pyridyl, or 4- pyridyl), pyrazinyl, 2,4-pyrimidinyl, 3,5-pyrimidinyl, 2,4-imidazolyl, isoxazolyl, oxazolyl, thiazolyl, thiadiazolyl, tetrazolyl, thienyl, benzothienyl, furyl, benzofuryl, benzoimidazolyl, indolyl, indolinyl, pyridizinyl, triazolyl, quinolinyl, pyrazolyl,
pyrrolopyridinyl (such as 1 H-pyrrolo[2,3-b]pyridin-5-yl), pyrazolopyridinyl (such as1 H- pyrazolo[3,4-b]pyridin-5-yl), benzoxazolyl (such as benzo[d]oxazol-6-yl),
benzothiazolyl (such as benzo[d]thiazol-6-yl), indazolyl (such as 1 H-indazol-5-yl) and 5,6,7,8-tetrahydroisoquinoline.
[024] Bivalent radicals derived from univalent heteroaryl radicals whose names end in "-yl" by removal of one hydrogen atom from the atom with the free valence are named by adding "-idene" to the name of the corresponding univalent radical, e.g., a pyridyl group with two points of attachment is a pyridylidene. Heteroaryl does not encompass or overlap with aryl as defined above.
[025] Substituted heteroaryl also includes ring systems substituted with one or more oxide (-O") substituents, such as pyridinyl N-oxides.
[026] By "heterocycle" or "heterocyclic ring" is meant a 4- to 12-membered
monocyclic, bicyclic or tricyclic saturated or partially unsaturated ring containing at least 2 carbon atoms in addition to 1 -3 heteroatoms independently selected from oxygen, sulfur, and nitrogen. "Heterocycle" also refers to 5- to 7-membered
heterocyclic ring containing one or more heteroatoms selected from N, O, and S fused with 5-, 6-, and/or 7-membered cycloalkyl, carbocyclic aromatic or heteroaromatic ring, provided that the point of attachment is at the heterocyclic ring when the heterocyclice ring is fused with a carbocyclic aromatic or a heteroaromatic ring, and that the point of attachment can be at the cycloalkyl or heterocyclic ring when the heterocylic ring is fused with cycloalkyl. "Heterocycle" also refers to an aliphatic spirocyclic ring containing one or more heteroatoms selected from N, O, and S, provided that the point of attachment is at the heterocyclic ring. The rings may be saturated or have one or more double bonds (i.e. partially unsaturated). The heterocycle can be substituted by oxo. The point of the attachment may be carbon or heteroatom in the heterocyclic ring. A heterocyle is not a heteroaryl as defined herein. [027] Suitable heterocycles include, for example (as numbered from the linkage position assigned priority 1 ), 1 -pyrrolidinyl, 2-pyrrolidinyl, 2,4-imidazolidinyl, 2,3- pyrazolidinyl, 1 -piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-piperidinyl, 2,5-piperazinyl, pyranyl, 2-morpholinyl and 3-morpholinyl. Substituted heterocycle also includes ring systems substituted with one or more oxo moieties, such as piperidinyl N-oxide, morpholinyl-N-oxide, 1 -oxo-1 -thiomorpholinyl and 1 ,1 -dioxo-1 -thiomorpholinyl.
[028] By "optional" or "optionally" is meant that the subsequently described event or circumstance may or may not occur, and that the description includes instances where the event or circumstance occurs and instances in which it does not. For example, "optionally substituted alkyl" encompasses both "alkyl" and "substituted alkyl" as defined below. It will be understood by those skilled in the art, with respect to any group containing one or more substituents, that such groups are not intended to introduce any substitution or substitution patterns that are sterically impractical, synthetically non-feasible and/or inherently unstable.
[029] The term "substituted", as used herein, means that any one or more hydrogen atoms on the designated atom or group is replaced with a selection from the indicated group, provided that the designated atom's normal valence is not exceeded. When a substituent is oxo (i.e., =O) then 2 hydrogen atoms on the atom are replaced.
Combinations of substituents and/or variables are permissible only if such
combinations result in stable compounds or useful synthetic intermediates. A stable compound or stable structure is meant to imply a compound that is sufficiently robust to survive isolation from a reaction mixture, and subsequent formulation as an agent having at least practical utility. Unless otherwise specified, substituents are named into the core structure. For example, it is to be understood that when (cycloalkyl)alkyl is listed as a possible substituent, the point of attachment of this substituent to the core structure is in the alkyl portion. [030] In some embodiments, "substituted with one or more groups" refers to two hydrogen atoms on the designated atom or group being independently replaced with two selections from the indicated group of substituents. In some embodiments, "substituted with one or more groups" refers to three hydrogen atoms on the designated atom or group being independently replaced with three selections from the indicated group of substituents. In some embodiments, "substituted with one or more groups" refers to four hydrogen atoms on the designated atom or group being independently replaced with four selections from the indicated group of substituents.
[031] Compounds described herein include, but are not limited to, when possible, their optical isomers, such as enantiomers and diastereomers, mixtures of
enantiomers, including racemates, mixtures of diastereomers, and other mixtures thereof, to the extent they can be made by one of ordinary skill in the art by routine experimentation. In those situations, the single enantiomers or diastereomers, i.e., optically active forms, can be obtained by asymmetric synthesis or by resolution of the racemates or mixtures of diastereomers. Resolution of the racemates or mixtures of diastereomers, if possible, can be accomplished, for example, by conventional methods such as crystallization in the presence of a resolving agent, or
chromatography, using, for example a chiral high-pressure liquid chromatography (HPLC) column. In addition, such compounds include Z- and E- forms (or cis- and trans- forms) of compounds with carbon-carbon double bonds. Where compounds described herein exist in various tautomeric forms, the term "compound" is intended to include, to the extent they can be made without undue experimentation, all tautomeric forms of the compound. Such compounds also include crystal forms including polymorphs and clathrates, to the extent they can be made by one of ordinary skill in the art by routine experimentation. Similarly, the term "salt" is intended to include all isomers, racemates, other mixtures, Z- and E-forms, tautomeric forms and crystal forms of the salt of the compound, to the extent they can be made by one of ordinary skill in the art without undue experimentation.
[032] "Pharmaceutically acceptable salts" include, but are not limited to salts with inorganic acids, such as hydrochlorate, phosphate, diphosphate, hydrobromate, sulfate, sulfinate, nitrate, and like salts; as well as salts with an organic acid, such as malate, maleate, fumarate, tartrate, succinate, citrate, lactate, methanesulfonate, p-toluenesulfonate, 2-hydroxyethylsulfonate, benzoate, salicylate, stearate, alkanoate such as acetate, and salts with HOOC-(CH2)n-COOH where n is 0-4, and like salts. Similarly, pharmaceutically acceptable cations include, but are not limited to sodium, potassium, calcium, aluminum, lithium, and ammonium.
[033] In addition, if a compound described herein is obtained as an acid addition salt, the free base can be obtained by basifying a solution of the acid salt. Conversely, if the product is a free base, an addition salt, particularly a pharmaceutically acceptable addition salt, may be produced by dissolving the free base in a suitable organic solvent and treating the solution with an acid, in accordance with conventional procedures for preparing acid addition salts from base compounds. Those skilled in the art will recognize various synthetic methodologies that may be used without undue experimentation to prepare non-toxic pharmaceutically acceptable addition salts.
[034] A "solvate," such as a "hydrate," is formed by the interaction of a solvent and a compound. The term "compound" is intended to include solvates, including hydrates, of compounds, to the extent they can be made by one of ordinary skill in the art by routine experimentation. Similarly, "salts" includes solvates, such as hydrates, of salts, to the extent they can be made by one of ordinary skill in the art by routine
experimentation. Suitable solvates are pharmaceutically acceptable solvates, such as hydrates, including monohydrates and hemi-hydrates, to the extent they can be made by one of ordinary skill in the art by routine experimentation. [035] A "chelate" is formed by the coordination of a compound to a metal ion at two (or more) points. The term "compound" is intended to include chelates of compounds. Similarly, "salts" includes chelates of salts.
[036] A "non-covalent complex" is formed by the interaction of a compound and another molecule wherein a covalent bond is not formed between the compound and the molecule. For example, complexation can occur through van der Waals
interactions, hydrogen bonding, and electrostatic interactions (also called ionic bonding). Such non-covalent complexes are included in the term "compound."
[037] The term "hydrogen bond" refers to a form of association between an electronegative atom (also known as a hydrogen bond acceptor) and a hydrogen atom attached to a second, relatively electronegative atom (also known as a hydrogen bond donor). Suitable hydrogen bond donor and acceptors are well understood in medicinal chemistry (G. C. Pimentel and A. L. McClellan, The Hydrogen Bond, Freeman, San Francisco, 1960; R. Taylor and O. Kennard, "Hydrogen Bond Geometry in Organic Crystals", Accounts of Chemical Research, 17, pp. 320-326 (1984)).
[038] As used herein the terms "group", "radical" or "fragment" are synonymous and are intended to indicate functional groups or fragments of molecules attachable to a bond or other fragments of molecules.
[039] The term "active agent" is used to indicate a chemical substance which has biological activity. In some embodiments, an "active agent" is a chemical substance having pharmaceutical utility.
[040] "Treating," "treat," or "treatment" or "alleviation" refers to administering at least one compound and/or at least one pharmaceutically acceptable salt thereof described herein to a subject that has cancer, or has a symptom of cancer, or has a
predisposition toward cancer, with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve, or affect cancer, the symptoms of cancer, or the predisposition toward cancer. [041] The term "effective amount" refers to an amount of at least one compound and/or at least one pharmaceutically acceptable salt thereof described herein effective to "treat," as defined above, a disease or disorder in a subject. In the case of cancer, the effective amount may cause any of the changes observable or measurable in a subject as described in the definition of "treating," "treat," "treatment" and "alleviation" above. For example, the effective amount can reduce the number of cancer or tumor cells; reduce the tumor size; inhibit or stop tumor cell infiltration into peripheral organs including, for example, the spread of tumor into soft tissue and bone; inhibit and stop tumor metastasis; inhibit and stop tumor growth; relieve to some extent one or more of the symptoms associated with the cancer, reduce morbidity and mortality; improve quality of life; or a combination of such effects. An effective amount may be an amount sufficient to decrease the symptoms of a disease responsive to inhibition of PI3K/mTOR activity. For cancer therapy, efficacy in vivo can, for example, be measured by assessing the duration of survival, time to disease progression (TTP), the response rates (RR), duration of response, and/or quality of life. Effective amounts may vary, as recognized by those skilled in the art, depending on route of administration, excipient usage, and co-usage with other agents.
[042] The term "inhibition" indicates a decrease in the baseline activity of a biological activity or process. "Inhibition of PI3K and/or mTOR activity" refers to a decrease in the activity of PI3K and/or mTOR as a direct or indirect response to the presence of at least one compound and/or at least one pharmaceutically acceptable salt described herein, relative to the activity of PI3K and/or mTOR in the absence of the at least one compound and/or the at least one pharmaceutically acceptable salt thereof. The decrease in activity is not bound by theory and may be due to the direct interaction of the at least one compound and/or at least one pharmaceutically acceptable salt thereof described herein with PI3K and/or mTOR, or due to the interaction of the at least one compound and/or at least one pharmaceutically acceptable salt described herein, with one or more other factors that in turn affect PI3K and/or mTOR activity. For example, the presence of at least one compound and/or at least one pharmaceutically acceptable salt thereof described herein, may decrease PI3K and/or mTOR activity by directly binding to the PI3K and/or mTOR, by causing (directly or indirectly) another factor to decrease PI3K and/or mTOR activity, or by (directly or indirectly) decreasing the amount of PI3K and/or mTOR present in the cell or organism.
[043] The details of one or more embodiments of the invention are set forth below.
[044] Provided herein is at least one compound of formula 1 :
Figure imgf000015_0001
(1 )
and/or at least one pharmaceutically acceptable salt thereof wherein
A1 is N or CH;
A4 and A5 are independently N or CR2;
A2 and A3, together with B ring, are a 5-membered heteroaryl or heterocycle containing 1 to 4 heteroatoms selected from N, O, and S, and said 5-membered heteroaryl or heterocycle is optionally substituted by one or more groups
independently chosen from alkyl, alkenyl, alkynyl, aryl, cycloalkyl, oxo, -C(O)Ra, - C(O)ORb, -CN, -C(O)NRcRd, -NRcRd, -NRcC(O)Ra, -NRcS(O)nRe, -NRcS(O)nNRfRg, - NRcC(O)ORb, -NRcC(O)NRdRe, -NO2, -ORb, -S(O)nRe, -S(O)nNRcRd, halo, haloalkyl, heteroaryl, and heterocycle;
provided that A and A , together with the B ring, are not
Figure imgf000015_0002
is a single bond or a double bond;
R1 is heteroaryl, optionally substituted by one or more groups independently chosen from alkyl, alkenyl, alkynyl, aryl, cycloalkyl, oxo, -C(O)Ra, -C(O)ORb, -CN, - C(O)NRcRd, -NRcRd, -NRcC(O)Ra, -NRcS(O)nRe, -NRcS(O)nNRfRg, -NRcC(O)ORb, - NRcC(O)NRdRe, -NO2, -ORb, -S(O)nRe, -S(O)nNRcRd, halo, haloalkyl, heteroaryl, and heterocycle;
R and R2 are independently chosen from H, alkyl, alkenyl, alkynyl, aryl, cycloalkyl, -C(O)Ra, -C(O)ORb, -CN, -C(O)NRcRd, -NRcRd, -NRcC(O)Ra, -NRcS(O)nRe, -NRcS(O)nNRfRg, -NRcC(O)ORb, -NRcC(O)NRdRe, -NO2, -ORb, -OC(O)Ra, - OC(O)NRcRd, -S(O)nRe, -S(O)nNRcRd, halo, haloalkyl, heteroaryl, and heterocycle; and each of said above alkyl, alkenyl, alkynyl, aryl, cycloalkyl, haloalkyl, heteroaryl and heterocycle can be optionally substituted by one or more groups independently chosen from optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted cycloalkyl, -OH, oxo, -C(O)Ra, -C(O)ORb, -CN, -C(O)NRcRd, -NRcRd, -NRcC(O)Ra, - NRcS(O)nRe, -NRcS(O)nNRfRg, -NRcC(O)ORb, -NRcC(O)NRdRe, -NO2, ORb, -S(O)nRe, - S(O)nNRcRd, halo, optionally substituted haloalkyl, optionally substituted heteroaryl, and optionally substituted heterocycle;
Ra, Rb, Rc, Rd, Re, Rf and Rg are each independently chosen from H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl optionally substituted aryl, optionally substituted cycloalkyl, optionally substituted haloalkyl, optionally substituted heteroaryl, and optionally substituted heterocycle,
or Ra and Rc, and/or Rc and Rd, and/or Rc and Re, and/or Rc and Rf, and/or Rd and Re, and /or Rg and Rf together with the atom(s) to which they are attached, form a 3-10 membered optionally substituted heterocycle ring; and
for each occurrence, n is independently 0, 1 , or 2; wherein each optionally substituted group above can be unsubstituted or independently substituted with one or more, such as one, two, or three, substituents independently selected from, Ci-C4 alkyl, cycloalkyi, oxo, aryl, heterocycle, heteroaryl, aryl-Ci-C4 alkyl-, heteroaryl-Ci-C4 alkyl-, Ci-C4 haloalkyl-, -OCi-C4 alkyl,
-OC1-C4 alkylphenyl, -d-C4 alkyl-OH, -C1-C4 alkyl-O-d-C4 alkyl, -OC1-C4 haloalkyl, halo, -OH, -NH2, -d-C4 alkyl-NH2, -N(d-C4 alkyl)(Ci-C4 alkyl), -NH(d-C4 alkyl), -N(Ci-C4 alkyl)(Ci-C4 alkylphenyl), -NH(Ci-C4 alkylphenyl), cyano, nitro, oxo, -CO2H, -C(O)OCi-C4 alkyl, -C(O)Ocycloalkyl, -C(O)Oaryl, -C(O)Oheteroaryl, - C(O)Oheterocycle, -CON(Ci-C4 alkyl)(Ci-C4 alkyl), -CONR'R" wherein R' and R" with the N to which they are attached form a heterocycle, -CON(cycloalkyl)(cycloalkyl), - CON(heterocycle)(heterocycle), -CONH(Ci-C4 alkyl), -CONH(cycloalkyl), - CONH(heterocycle), -CONH2, -NHC(0)(Ci-C4 alkyl), -NHC(O)(cycloalkyl), - NHC(O)(heterocycle), -NHC(O)(aryl) such as -NHC(O)(phenyl), -NHC(O)(heteroaryl), -N(Ci-C4 alkyl)C(O)(Ci-C4 alkyl), -N(Ci-C4 alkyl)C(O)(cycloalkyl),
-N(Ci-C4 alkyl )C(O)(heterocycle), -N(Ci-C4 alkyl )C(O)(aryl) such as
-N(Ci-C4 alkyl)C(O)(phenyl), -N(Ci-C4 alkyl)C(O)(heteroaryl), -C(O)d-C4 alkyl,
-C(O)(cycloalkyl), -C(O)(heterocycle), -C(O)(aryl) such as -C(O) phenyl, - C(O)(heteroaryl), -C(O)d-C4 haloalkyl, -OC(O)d-C4 alkyl, -OC(O)(cycloalkyl), - OC(O)(heterocycle), - - OC(O)(heteroaryl), OC(O)(aryl) such as -OC(O)phenyl, - SO2(Ci-C4 alkyl), - SO2(cycloalkyl), -SO2(heterocycle), -SO2(aryl) such as SO2(phenyl), -SO2(heteroaryl), -SO2(Ci-C4 haloalkyl), -SO2NH2, -SO2NR'R" wherein R' and R" with the N to which they are attached form a heterocycle, -SO2NH(Ci-C4 alkyl),
-SO2NH(cycloalkyl), -SO2NH(heterocycle), -SO2NH(aryl) such as -SO2NH(phenyl), -SO2NH(heteroaryl), -NHSO2(Ci-C4 alkyl), NHSO2(cycloalkyl), NHSO2(heterocycle), NHSO2(aryl) such as -NHSO2(phenyl), -NHSO2(heteroaryl), and
-NHSO2(Ci-C4 haloalkyl), in which each of alkyl, phenyl, aryl, cycloalkyi, heterocycle, and heteroaryl is optionally substituted by one or more groups independently chosen from -OH, halo, cycloalkyi, heterocycle, Ci-C4 alkyl, Ci-C4 haloalkyl-, -OCi-C4 alkyl, C1-C4 alkyl-OH, -C1-C4 alkyl-O-d-C4 alkyl, -OC1-C4 haloalkyl, cyano, nitro, -NH2, - CO2H, -C(O)OCi-C4 alkyl, -CON(d-C4 alkyl )(Ci-C4 alkyl), -CONH(d-C4 alkyl),
-CONH2, -NHC(O)(Ci-C4 alkyl), and -N(d-C4 alkyl)C(0)(Ci-C4 alkyl).
[045] In some embodiments, A2 and A3, together with B ring, are a 5-membered heteroaryl or heterocycle containing 1 to 3 heteroatoms selected from N, O and S. In some embodiments, A2 and A3, together with B ring, are a 5-membered heteroaryl or heterocycle containing 1 to 3 nitrogen heteroatoms.
[046] In some embodiments, A2 and A3, together with B ring, can be selected from structures (2)-(6)
Figure imgf000018_0001
wherein,
t is 1 , 2 or 3; and
R3 is independently chosen from H, C1-C6 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, C6-Ci4aryl, C3-C9 cycloalkyi, -C(O)ORb, -CN, -C(O)NRcRd, -NRcRd, -NRcC(O)Ra, -
NRcS(O)nRe, -NRcS(O)nNRfRg, -NRcC(O)ORb, -NRcC(O)NRdRe, -NO2, -ORb, -OC(O)Ra, -OC(O)NRcRd, -S(O)nRe, -S(O)nNRcRd, halo, haloalkyl, heteroaryl, and heterocycle; and each of said above alkyl, alkenyl, alkynyl, aryl, cycloalkyi, heteroaryl and heterocycle can be optionally substituted by one or more groups independently chosen from optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted cycloalkyi, -OH, oxo, -C(O)Ra, -C(O)ORb, -CN, -C(O)NRcRd, -NRcRd, -NRcC(O)Ra, -NRcS(O)nRe, -NRcS(O)nNRfRg, - NRcC(O)ORb, -NRcC(O)NRdRe, -NO2, ORb, -S(O)nRe, -S(O)nNRcRd, halo, optionally substituted haloalkyi, optionally substituted heteroaryl, and optionally substituted heterocyde, and Ra, Rb, Rc, Rd, Re, Rf and Rg are as defined above.
provided that, when A2 and A3, together with B ring, are structure (4), A4 is not CR2, wherein R2 is as defined above.
[047] For example, R3 is independently chosen from H, OH, CN, NO2, halo, C1-C6 alkyl,C2-Cs alkenyl, C2-C8 alkynyl, C6-Ci4 aryl, C3-C9 cydoalkyi, heteroaryl, and heterocyde, wherein each of the alkyl, alkenyl, alkynyl, aryl, cydoalkyi, heteroaryl, and heterocyde can be optionally substituted by one or more groups independently chosen from optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted cydoalkyi, -OH, oxo, -C(O)Ra, -C(O)ORb, -CN, -C(O)NRcRd, -NRcRd, -NRcC(O)Ra, -NRcS(O)nRe, - NRcS(O)nNRfRg, -NRcC(O)ORb, -NRcC(O)NRdRe, -NO2, ORb, -S(O)nRe, -S(O)nNRcRd, halo, optionally substituted haloalkyi, optionally substituted heteroaryl, and optionally substituted heterocyde;
[048] In some embodiments, A2 and A3 together with B ring, are chosen from the following structures (2)-(5), wherein R3 and t are as defined above.
Figure imgf000019_0001
(2) (3) (4)
[049] In some embodiments, A2, A3 and together with the B ring, are chosen from structure (3)-(4), wherein R3 and t are as defined above.
Figure imgf000019_0002
(3) (4) [050] In some embodiments, A4 is N or CH.
[051] In some embodiments, A5 is N or CH.
[052] In some embodiments, Ai, A4, and A5 are CH.
[053] In some embodiments, Ai and A5 are CH, and A4 is N.
[054] In some embodiments, R1 is a heteroaryl that is chosen from the following structures
Figure imgf000020_0001
r example, R1 is a heteroaryl chosen from
Figure imgf000021_0001
wherein each of which is optionally substituted by one or more groups independently chosen from alkyi, alkenyl, alkynyl, aryl, cydoalkyi, haloalkyi, heteroaryl, heterocyde, oxo, -C(O)Ra, -C(O)ORb, -CN, -C(O)NRcRd, -NRcRd, -
NRcC(O)Ra, -NRcS(O)nRe, -NRcS(O)nNRfRg, -NRcC(O)ORb, -NRcC(O)NRdRe, -NO2, - ORb, -OC(O)Ra, -OC(O)NRcRd, -S(O)nRe, -S(O)nNRcRd and halo, wherein each of the alkyi, alkenyl, alkynyl, aryl, cydoalkyi, haloalkyi, heteroaryl and heterocyde can be optionally substituted by one or more groups independently chosen from
optionally substituted alkyi, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted cydoalkyi, -OH, oxo, - C(O)Ra, -C(O)ORb, -CN, -C(O)NRcRd, -NRcRd, -NRcC(O)Ra, -NRcS(O)nRe, - NRcS(O)nNRfRg, -NRcC(O)ORb, -NRcC(O)NRdRe, -NO2, ORb, -S(O)nRe, -S(O)nNRcRd, halo, optionally substituted haloalkyi, optionally substituted heteroaryl, and optionally substituted heterocyde;
wherein Ra, Rb, Rc, Rd, Re, Rf and Rg are each independently chosen from H, alkyi, aryl, cydoalkyi, haloalkyi, heteroaryl, and heterocyde, wherein each of the alkyi, aryl, cydoalkyi, heteroaryl, and heterocyde in Ra, Rb, Rc, Rd, Re, Rf and Rg is optionally substituted by one or more, such as one or two or three, substitutents independently selected from halo and alkyi.
[055] In some embodiments, R1 is
Figure imgf000022_0001
which is optionally substituted with one or more groups independently chosen from:
- alkyl, alkenyl, and alkynyl, wherein each of which can be optionally substituted by groups independently chosen from optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted cycloalkyi, -OH, oxo, -C(O)Ra, -C(O)ORb, -CN, -C(O)NRcRd, -NRcRd, -
NRcC(O)Ra, -NRcS(O)nRe, -NRcS(O)nNRfR9, -NRcC(O)ORb, -NRcC(O)NRdRe, -NO2,
ORb, -S(O)nRe, -S(O)nNRcRd, halo, optionally substituted haloalkyi, optionally substituted heteroaryl, andoptionally substituted heterocycle;
-C(O)NRcRd;
-NRcRd;
-ORb;
-halo;
-cyano;
-NRcS(O)nRe,
wherein Ra, Rb, Rc, Rd, Re, Rf and R9 are each independently chosen from H, alkyl, aryl, cycloalkyi, haloalkyi, heteroaryl, and heterocycle, for example, Ra, Rb, Rc, Rd, Re, Rf and R9 are each independently chosen from H, C1-C6 alkyl, phenyl, C3-C6 cycloalkyi, C1-C3 haloalkyi, heteroaryl, and heterocycle, wherein each of the alkyl, aryl, cycloalkyi, heteroaryl, and heterocycle in Ra, Rb, Rc, Rd, Re, Rf and R9 is optionally substituted by one or more, such as one or two or three, substitutents independently selected from halo and alkyl.
[056] Also provided is at least one compound selected from compounds 1 to 184 and/or at least one pharmaceutically acceptable salt described herein. [057] The compounds described herein, and/or the pharmaceutically acceptable salts thereof, can be synthesized from commercially available starting materials by methods well known in the art, taken together with the disclosure in this patent application. The following schemes illustrate methods for preparation of some of the compounds disclosed herein.
Scheme I
Figure imgf000023_0001
Scheme II
Figure imgf000023_0002
[058] The compounds thus obtained can be further modified at their peripheral positions to provide the desired compounds. Synthetic chemistry transformations are described, for example, in R. Larock, Comprehensive Organic Transformations, VCH Publishers (1989); T.W. Greene and P.G.M. Wuts, Protective Groups in Organic Synthesis, 3rd Ed., John Wiley and Sons (1999); L. Fieser and M. Fieser, Fieser and Fieser's Reagents for Organic Synthesis, John Wiley and Sons (1994); and L.
Paquette, ed., Encyclopedia of Reagents for Organic Synthesis, John Wiley and Sons (1995) and subsequent editions thereof.
[059] Before use, the at least one compound and/or at least one pharmaceutically acceptable salt described herein, can be purified by column chromatography, high performance liquid chromatography, crystallization, or other suitable methods.
[060] Also provided is a composition comprising at least one compound and/or at least one pharmaceutically acceptable salt described herein, and at least one pharmaceutically acceptable carrier.
[061] A composition comprising at least one compound and/or at least one pharmaceutically acceptable salt described herein, can be administered in various known manners, such as orally, parenterally, by inhalation spray, or via an implanted reservoir. The term "parenteral" as used herein includes subcutaneous,
intracutaneous, intravenous, intramuscular, intraarticular, intraarterial, intrasynovial, intrasternal, intrathecal, intralesional and intracranial injection or infusion techniques.
[062] An oral composition can be any orally acceptable dosage form including, but not limited to, tablets, capsules, emulsions, and aqueous suspensions, dispersions and solutions. Commonly used carriers for tablets include lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added to tablets. For oral administration in a capsule form, useful diluents include lactose and dried corn starch. When aqueous suspensions or emulsions are administered orally, the active ingredient can be suspended or dissolved in an oily phase combined with emulsifying or suspending agents. If desired, certain sweetening, flavoring, or coloring agents can be added. [063] A sterile injectable composition (e.g., aqueous or oleaginous suspension) can be formulated according to techniques known in the art using suitable dispersing or wetting agents (such as, for example, Tween 80) and suspending agents. The sterile injectable preparation can also be a sterile injectable solution or suspension in a non- toxic parenterally acceptable diluent or solvent, for example, as a solution in 1 ,3- butanediol. Among the pharmaceutically acceptable vehicles and solvents that can be employed are mannitol, water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium (e.g., synthetic mono- or di-glycerides). Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions can also contain a long-chain alcohol diluent or dispersant, or carboxymethyl cellulose or similar dispersing agents.
[064] An inhalation composition can be prepared according to techniques well known in the art of pharmaceutical formulation and can be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other solubilizing or dispersing agents known in the art.
[065] A topical composition can be formulated in form of oil, cream, lotion, ointment and the like. Suitable carriers for the composition include vegetable or mineral oils, white petrolatum (white soft paraffin), branched chain fats or oils, animal fats and high molecular weight alcohols (greater than C12). In some embodiments, the
pharmaceutically acceptable carrier is one in which the active ingredient is soluble. Emulsifiers, stabilizers, humectants and antioxidants may also be included as well as agents imparting color or fragrance, if desired. Additionally, transdermal penetration enhancers may be employed in those topical formulations. Examples of such enhancers can be found in U.S. Patents 3,989,816 and 4,444,762.
[066] Creams may be formulated from a mixture of mineral oil, self-emulsifying beeswax and water in which mixture the active ingredient, dissolved in a small amount of an oil, such as almond oil, is admixed. An example of such a cream is one which includes about 40 parts water, about 20 parts beeswax, about 40 parts mineral oil and about 1 part almond oil. Ointments may be formulated by mixing a solution of the active ingredient in a vegetable oil, such as almond oil, with warm soft paraffin and allowing the mixture to cool. An example of such an ointment is one which includes about 30% by weight almond oil and about 70% by weight white soft paraffin.
[067] A pharmaceutically acceptable carrier refers to a carrier that is compatible with active ingredients of the composition (and in some embodiments, capable of stabilizing the active ingredients) and not deleterious to the subject to be treated. For example, solubilizing agents, such as cyclodextrins (which form specific, more soluble complexes with the at least one compound and/or at least one pharmaceutically acceptable salt described herein), can be utilized as pharmaceutical excipients for delivery of the active ingredients. Examples of other carriers include colloidal silicon dioxide, magnesium stearate, cellulose, sodium lauryl sulfate, and pigments such as D&C Yellow # 10.
[068] Suitable in vitro assays can be used to preliminarily evaluate the efficacy of the at least one compound and/or at least one pharmaceutically acceptable salt described herein, in inhibiting the activity of PI3K and/or mTOR. The at least one compound and/or at least one pharmaceutically acceptable salt described herein, can further be examined for efficacy in treating cancer by in vivo assays. For example, the compounds described herein, and/or the pharmaceutically acceptable salts thereof, can be administered to an animal (e.g., a mouse model) having cancer and its therapeutic effects can be accessed. Positve results in one or more of such tests are sufficient to increase the scientific storehouse of knowledge and hence sufficient to demonstrate practical utility of the compounds and/or salts tested. Based on the results, an appropriate dosage range and administration route for animals, such as humans, can also be determined.
[069] Also provided is a method of inhibiting the activity of PI3K and/or mTOR. The method comprises contacting the enzyme with at least one compound and/or at least one pharmaceutically acceptable salt described herein in an amount effective to inhibit the activity of PI3K and/or mTOR.
[070] The at least one compound and/or at least one pharmaceutically acceptable salt described herein can be used to achieve a beneficial therapeutic or prophylactic effect, for example, in subjects with cancer. As used herein, the term "cancer" refers to a cellular disorder characterized by uncontrolled or disregulated cell proliferation, decreased cellular differentiation, inappropriate ability to invade surrounding tissue, and/or ability to establish new growth at ectopic sites. The term "cancer" includes, but is not limited to, solid tumors and bloodborne tumors. The term "cancer" encompasses diseases of skin, tissues, organs, bone, cartilage, blood, and vessels. The term
"cancer" further encompasses primary and metastatic cancers.
[071] Non-limiting examples of solid tumors include pancreatic cancer; bladder cancer; colorectal cancer; breast cancer, including metastatic breast cancer; prostate cancer, including androgen-dependent and androgen-independent prostate cancer; renal cancer, including, e.g., metastatic renal cell carcinoma; hepatocellular cancer; lung cancer, including, e.g., non-small cell lung cancer (NSCLC), bronchioloalveolar carcinoma (BAC), and adenocarcinoma of the lung; ovarian cancer, including, e.g., progressive epithelial or primary peritoneal cancer; cervical cancer; gastric cancer; esophageal cancer; head and neck cancer, including, e.g., squamous cell carcinoma of the head and neck; skin cancer, including e.g., malignant melanoma;
neuroendocrine cancer, including metastatic neuroendocrine tumors; brain tumors, including, e.g., glioma, anaplastic oligodendroglioma, adult glioblastoma multiforme, and adult anaplastic astrocytoma; bone cancer; soft tissue sarcoma; and thyroid carcinoma. For example, those solid tumors include pancreatic cancer; bladder cancer; colorectal cancer; breast cancer, and ovarian cancer.
[072] Non-limiting examples of hematologic malignancies include acute myeloid leukemia (AML); chronic myelogenous leukemia (CML), including accelerated CML and CML blast phase (CML-BP); acute lymphoblastic leukemia (ALL); chronic lymphocytic leukemia (CLL); Hodgkin's disease (HD); non-Hodgkin's lymphoma (NHL), including follicular lymphoma and mantle cell lymphoma; B-cell lymphoma; T-cell lymphoma; multiple myeloma (MM); Waldenstrom's macroglobulinemia;
myelodysplastic syndromes (MDS), including refractory anemia (RA), refractory anemia with ringed siderblasts (RARS), (refractory anemia with excess blasts (RAEB), and RAEB in transformation (RAEB-T); and myeloproliferative syndromes.
[073] In some embodiments, the examples of the cancer to be treated include, but are not limited to, lung cancer, head and neck cancer, colorectal cancer, pancreatic cancer, colon cancer, breast cancer, ovarian cancer, prostate cancer, stomach cancer, kidney cancer, liver cancer, brain cancer, bone cancer, and leukemia.
[074] In some embodiments, the at least one compound and/or at least one pharmaceutically acceptable salt described herein, is administered in conjunction with another therapeutic agent. In some embodiments, the other therapeutic agent is one that is normally administered to patients with the disease or condition being treated. The at least one compound and/or at least one pharmaceutically acceptable salt described herein, may be administered with the other therapeutic agent in a single dosage form or as a separate dosage form. When administered as a separate dosage form, the other therapeutic agent may be administered prior to, at the same time as, or following administration of the at least one compound and/or at least one
pharmaceutically acceptable salt described herein. [075] In some embodiments, at least one compound and/or at least one pharmaceutically acceptable salt described herein, is administered in conjunction with an anti-neoplastic agent. As used herein, the term "anti-neoplastic agent" refers to any agent that is administered to a subject with cancer for purposes of treating the cancer. Nonlimiting examples of anti-neoplastic agents include: radiotherapy; immunotherapy; DNA damaging chemotherapeutic agents; and chemotherapeutic agents that disrupt cell replication.
[076] Non-limiting examples of DNA damaging chemotherapeutic agents include topoisomerase I inhibitors (e.g., irinotecan, topotecan, camptothecin and analogs or metabolites thereof, and doxorubicin); topoisomerase II inhibitors (e.g., etoposide, teniposide, and daunorubicin); alkylating agents (e.g., melphalan, chlorambucil, busulfan, thiotepa, ifosfamide, carmustine, lomustine, semustine, streptozocin, decarbazine, methotrexate, mitomycin C, and cyclophosphamide); DNA intercalators (e.g., cisplatin, oxaliplatin, and carboplatin); DNA intercalators and free radical generators such as bleomycin; and nucleoside mimetics (e.g., 5-fluorouracil, capecitibine, gemcitabine, fludarabine, cytarabine, mercaptopurine, thioguanine, pentostatin, and hydroxyurea).
[077] Chemotherapeutic agents that disrupt cell replication include: paclitaxel, docetaxel, and related analogs; vincristine, vinblastin, and related analogs;
thalidomide and related analogs (e.g., CC-5013 and CC-4047); protein tyrosine kinase inhibitors (e.g., imatinib mesylate and gefitinib); proteasome inhibitors (e.g., bortezomib); NF-kappa B inhibitors, including inhibitors of I kappa B kinase; antibodies which bind to proteins overexpressed in cancers and thereby downregulate cell replication (e.g., trastuzumab, rituximab, cetuximab, and bevacizumab); and other inhibitors of proteins or enzymes known to be upregulated, over-expressed or activated in cancers, the inhibition of which downregulates cell replication. [078] The examples below are intended to be purely exemplary and should not be considered to be limiting in any way. Efforts have been made to ensure accuracy with respect to numbers used (for example, amounts, temperature, etc.) but some experimental errors and deviations should be accounted for. Unless indicated otherwise, parts are parts by weight, temperature is in degrees Centigrade, and pressure is at or near atmospheric. All MS data were checked by agilent 6120 and /or agilent 1 100. 1H NMR spectra were recorded on Varian 400 MHz NMR spectrometer using CDCI3 or DMSO-d6 as the solvent and tetramethylsilane (TMS) as the internal standard. Chemical shifts (δ) were expressed in ppm downfield from internal TMS, and J values were given in Hz. All reagents, except intermediates, used in this disclosure are commercially available. All compound names except the reagents were generated by Chemdraw 10.
[079] In the following examples, the abbreviations below are used:
AcOH acetic acid
DCM dichloromethane
DMF N,N-dimethylformamide
DMF-DMA 1 ,1 -dimethoxy-N,N-dimethylmethanamine
DMSO dimethyl sulfoxide
DTT dithiothreitol
EtOAc ethyl acetate
h hour
ISCO Flash chromatography
ml_ milliliter(s)
min minute(s)
Py pyridine
THF tetrahydrofuran HATU 2-(1 H-7-azabenzotriazol-1 -yl)-1 ,1 ,3,3-tetrannethyl uronium
hexafluorophosphate Methanaminium
DIPEA N,N-diisopropylethylannine
DCM dichloromethane
EA ethyl acetate
PE petroleum ether
Pd(dppf)Cl2 1 ,1 '-Bis(diphenylphosphino)ferrocene-palladium(ll)dichloride
dichloromethane complex
PTLC preparative thin-layer chromatography
HEPES 2-[4-(2-hydroxyethyl)piperazin-1 -yl]ethanesulfonic acid
EGTA ethylene glycol tetraacetic acid)
CHAPS 3-[(3-Cholamidopropyl)-dimethylammonio]-1 -propanesulfonate TEA triethylamine
TLC thin-layer chromatography
Intermediate 1
2- 5-aminopyridin-2-yl)-2-methylpropanenitrile
Figure imgf000031_0001
1-1 a 1-1 b 1-1 c 1-1 d 1-1
[080] To a solution of 2-chloro-5-nitropyridine (10 g, 63 mmol) in THF (150 mL) at room temperature was added K2CO3 (17.4 g, 126 mmol) and tert-butyl 2-cyanoacetate (10.7 g, 76 mmol). The reaction mixture was heated to reflux and stirred overnight. Then the solid was filtered off, and the filtrate was concentrated to give the
intermediate 1-1 b (16.6 g). m/z 208 (M+H)+. [081] The crude product tert-butyl 2-cyano-2-(5-nitropyridin-2-yl)acetate (1-1 b, 12 g, 47 mmol) was dissolved in 100 mL of HCI/EtOH (v/v 1 :5) and stirred at 80°C for 4 h. Then the mixture was concentrated, diluted with H2O, and extracted with EtOAc (4 x 40 mL). The combined extracts were concentrated, and the residue was purified by chromatography on silica gel (PE : EtOAc = 3 : 1 ) to afford 2-(5-nitropyridin-2- yl)acetonitrile (1-1 c) as a solid (7.7 g, 42.0% yield).
[082] To the mixture of 2-(5-nitropyridin-2-yl)acetonitrile (1-1 c, 8 g, 46 mmol) and K2CO3 (18 g, 1 10 mmol) in CH3CN (200 mL) was added iodomethane (7.5 mL, 120 mmol) dropwise at room temperature. The reaction mixture was stirred at 40°C overnight. Then the mixture was filtered, the filtrate was concentrated, and the residue was purified by chromatography on silica gel (PE : EtOAc = 5:1 ) to afford 2- methyl-2-(5-nitropyridin-2-yl)propanenitrile (1-1 d) (4.2 g, 48% yield), m/z 192 (M+H)+. [083] The mixture of 2-methyl-2-(5-nitropyridin-2-yl)propanenitrile (1-1 d, 2 g, 10.4 mmol) and SnCI2-2H2O (9.3 g, 41 .6 mmol) in EtOAc (10 mL) was stirred at reflux for 4 h. After cooling to room temperature, aqueous 2 M NaOH (80 mL) was added to adjust the pH to 8-9, the solid was filtered off, and the filtrate was extracted with ethyl acetate (3 x 40 mL). The combined organic layers were dried over anhydrous Na2SO4, and concentrated to afford 2-(5-aminopyridin-2-yl)-2-methylpropanenitrile (1-1 ) (1 .7 g, 65.3% yield). Intermediate 2
1 ,1 ,1 -trif luoro-N-(2-methoxy-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- yl)pyridin-3-yl)methanesulfonamide
Figure imgf000032_0001
[084] The mixture of 2-methoxy-3-nitro-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- yl) pyridine (300mg, 1 .1 mmol) and Raney-Ni(I Omg) in MeOH (10mL) was subject to H2 and stirred for 2h. After filtration, the filtrate was concentrated to give the title compound as a white solid (261 mg). Yield: 95.0%.
[085] To a solution of 2-methoxy-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl) pyridin-3-amine (100mg, 0.4mmol) and 2,6-di-tert-butyl-4-methylpyridine (1 15mg, 0.56mmol) in dichloromethane (5ml_) was added trifluoromethanesulfonic anhydride (147mg, 0.52mmol) drop wise at -20°C, and the mixture was stirred at this
temperature for 2h. Solvent was removed in vacuo and the residue was used in the next step without further purification (141 mg). Yield: 92.0%.
Intermediate 3
6-chloro-3-nitro-1 ,5-naphthyridin-4-ol
Figure imgf000033_0001
l-3a l-3b l-3c I-3
[086] The mixture of 6-chloropyridin-3-amine (5.0 g, 38.8 mmol) and 5- (methoxymethylene)-2,2-dimethyl-1 ,3-dioxane-4,6-dione (7.2 g, 38.8 mmol) in i-PrOH (60 ml_) was stirred at reflux for 2 h, and the solvent was removed to afford 5-((6- chloropyridin-3-ylamino)methylene)-2,2-dimethyl-1 ,3-dioxane-4,6-dione as a solid in 91 % yield (10.0 g). m/z 283 (M+H)+
[087] To the heated Dowtherm A (200 ml_) at 200 °C was added 5-((6-chloropyridin- 3-ylamino)methylene)-2,2-dimethyl-1 ,3-dioxane-4,6-dione(3.5 g, 12.4 mmol) and then stirred for additional 5 min. After cooling to r.t., PE was added. The precipitate was collected, and dried in vacuo to afford 6-chloro-1 ,5-naphthyridin-4-ol as off-white solid in 42% yield (0.94 g). m/z 183 (M+H)+
[088] 6-chloro-1 ,5-naphthyridin-4-ol(1 .45 g, 8 mmol) was added to ice-cooled cone. H2SO4 (15 ml_) and followed by the addition of KNO3 (1 .62 g, 16 mmol) slowly at 0°C. The mixture was heated to 100°C for 1 h, and then was poured into ice-water. The precipitate was collected and dried in vacuo to afford 6-chloro-3-nitro-1 ,5-naphthyridin- 4-ol as a solid in 90% yield (1 .97 g).
Intermediate 4
2,4-difluoro-N-(2-methoxy-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborol
an-2-yl)pyridin-3-yl)benzenesulfonamide
Figure imgf000034_0001
[089] The mixture of 5-bromo-2-methoxy-3-nitropyridine (5 g, 21 .5 mmol),
4,4,4',4',5,5,5',5' -octamethyl-2,2'-bi(1 ,3,2-dioxaborolane) (6.6 g, 25.8 mmol) ,
PdCl2(dppf)-CH2Cl2 (500 mg) and potassium acetate (6.3 g, 64.5 mmol) in anhydrous 1 ,4-dioxane (200 ml_) was refluxed for 2h. Then the solvents were removed. The crude product was purified by chromatography on silica gel using petroleum
ether:EtOAc =10:1 as eluent to afford 2-methoxy-3-nitro-5-(4,4,5,5-tetramethyl-1 ,3,2- dioxaborolan-2-yl)pyridine in 81 % yield (5 g). m/z 281 (M+H)+.
[090] To the solution of 2-methoxy-3-nitro-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan- 2-yl) pyridine (500 mg, 1 .79 mmol) in MeOH (50 ml_) was added Raney-Ni (50 mg). The reaction mixture was stirred at room temperature under H2 for 2h. Then the solid was filtered off, and the solvent was removed to afford 2-methoxy-5-(4,4,5,5- tetramethyl-1 ,3,2-dioxaborolan-2-yl)pyridin-3-amine in 89% yield (400 mg). m/z 251 (M+H)+.
[091 ] To the solution of 2-methoxy-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- yl)pyhdin-3-amine (400 mg, 1 .6 mmol) in pyridine (5 mL) was added 2,4- difluorobenzenesulfonyl chloride (407 mg, 1 .9 mmol) slowly, the reaction mixture was stirred at room temperature overnight, the solvent was evaporated in vacuo, and the residue was treated with brine (5 mL) and extracted with EtOAc (3 x 10 mL). The combined organic layer was evaporated in vacuo, and the residue was purified by column chromatography using petroleum ether:EtOAc=5:1 as eluent to afford the desired product 2,4-difluoro-N-(2-methoxy-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan- 2-yl)pyridin-3-yl)benzenesulfonamide as white solid in 59% yield (400 mg). m/z 427(M+H)+.
Intermediate 5
8-bromo-[1 ,2,4]triazolo[4,3-a]quinoxaline
Figure imgf000035_0001
l-5a l-5b l-5c l-5d 1-5
[092] Bromine (0.895 mL, 15.5 mmol) was added to the solution of quinoxalin-2-ol (1 .5 g, 10.3 mmol) in HOAc (15 mL), the mixture was stirred at r.t. for 6 h, and the precipitate was collected and washed with ethyl ether and dried to afford 7- bromoquinoxalin-2-ol as a solid in 90% yield (2 g).
[093] To the suspension of 7-bromoquinoxalin-2-ol (2 g, 8.88 mmol) in neat phosphorus oxychloride (7 mL) was added DMF (2drops). The mixture was heated to 100°C for 3h. Then it was cooled to room temperature. Phosphorus oxychloride was removed under vacuum, and the residue was dissolved into EtOAc and dropped into ice water with stirring. The mixture was extracted with EtOAc for three times, the combined organic layer was washed with saturated NaHCO3 solution. Then the organic layer was concentrated to afford 7-bromo-2-chloroquinoxaline as a solid in 93% yield (2 g).
[094] To the solution of 7-bromo-2-chloroquinoxaline (2 g, 8.2 mmol) in ethanol (20 ml_) was added hydrazine hydrate (85%, 4.5 ml_, 32.8 mmol). The mixture was heated to 78°C for 2h. After cooling to r.t., the precipitate was collected by filtration to afford 7-bromo-2-hydrazinylquinoxaline as white solid in 87% yield (1 .7 g).
[095] The solution of 7-bromo-2-hydrazinylquinoxaline (200 mg, 0.83 mmol) in triethyl orthoformate (3 ml_) was heated to 100°C for 4h; after cooling to r.t., the mixture was diluted with ethyl ether, and the precipitate was collected by filtration and dried in vacuo to afford the title product 8-bromo-[1 ,2,4]triazolo[4,3-a]quinoxaline as yellow solid in 87% yield (180 mg). m/z 251 (M+H)+.
Intermediate 6
8-bromo-1 -cyclopropyl-[1 ,2,4]triazolo[4,3-a]quinoxaline
Figure imgf000036_0001
[096] To the solution of 7-bromo-2-hydrazinylquinoxaline (200 mg, 0.83 mmol) in DMF (3 ml_) was added HATU (380 mg, 1 mmol), DIPEA (0.205 ml_, 1 mmol) and cyclopropanecarboxylic acid (71 mg, 0.83 mmol). After stirring at r.t. for 5 h, the mixture was diluted with EtOAc. The organic layer was washed with water for three times, then concentrated to afford N'-(7-bromoquinoxalin-2-yl)cyclopropane- carbohydrazide as yellow solid which was used directly in the next step, m/z 309 (M+H)+. [097] The crude product N'-(7-bromoquinoxalin-2-yl)cyclopropanecarbohydrazide was dissolved in HOAc (5 mL) and then heated to 100°C overnight. The solvent was removed under vacuum and the residue was washed with water and dried in vacuo to afford the title product 8-bromo-1 -cyclopropyl-[1 ,2,4]triazolo[4,3-a]quinoxaline as yellow solid in 42% overall yield (100 mg). m/z 291 (M+H)+.
Intermediate 7
8-bromo-[1 ,2,4]triazolo[4,3-a]quinoxalin-1(2H)-one
Figure imgf000037_0001
[098] To the solution of 7-bromo-2-hydrazinylquinoxaline (200 mg, 0.83 mmol) in dichloromethane (5 mL) was added triethylamine (0.175 mL, 1 .2 mmol), then the solution of diphosgene (0.055 mL, 0.46 mmol) in dichloromethane was added dropwise at 0°C with stirring under nitrogen atmosphere. After stirring at r.t. for 5 h, the solvent was removed under vacuum, the residue was washed with water, and dried in vacuo to afford the title product 8-bromo-[1 ,2,4]triazolo[4,3-a]quinoxalin-1 (2H)- one as yellow solid in 82% yield (180 mg). m/z 265 (M+H)+.
Intermediate 8
5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-3-(trifluoromethyl)pyridin-2-amine
Figure imgf000037_0002
[099] An orange suspension of 5-bromo-3-(trifluoromethyl)pyridin-2-amine (4 g, 16.60 mmol), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(1 ,3,2-dioxaborolane)(5.90 g, 23.24 mmol), KOAc (4.07 g, 41 .5 mmol) and PdCI2(dppf).CH2CI2 (0.678 g, 0.830 mmol) in Dioxane (60 mL) was heated to 1 10°C for 10 h under N2. After concentration under vacuum to remove the solvent, the crude product was purified using a silica gel column, with PE/EtOAc as eluant to give pure product as pale yellow solid (4.5g, yield 94%). MS (m/z): 289 (M+H)+.
Example 1 : Synthesis of Compounds 1-20
Compound 1
(S)-1 -(4-(8-(6-amino-5-(trifluoromethyl)pyridin-3-yl)-1 H-[1 ,2,3]triazolo[4,5- c][1 ,5]naphthyridin-1 -yl)piperidin-1 -yl)-2-hydroxypropan-1 -one
Figure imgf000038_0001
[0100] To a suspension of 6-chloro-3-nitro-1 ,5-naphthyridin-4-ol (5 g, 22.1 mmol) in 15 mL of DMF, was added a solution of phosphorous oxychloride (2.7 mL, 28.8 mmol) in anhydrous DMF(10 mL) over 3min. The mixture was stirred at room temperature overnight. The mixture was thenpoured onto crushed ice. The resulting precipitate was collected by filtration, washed with H2O, and dried in vacuo to afford 2,8-dichloro-7- nitro-1 ,5-naphthyridine as yellow solid (4 g, yield 74.0%). MS (m/z): 244(M+H)+. [0101] A mixture of 2,8-dichloro-7-nitro-1 ,5-naphthyridine (4 g, 16.4 mmol), 4-amino- piperidine-1 -carboxylic acid tert-butyl ester (4 g, 19.7 mmol) and triethylamine (3.5 mL) in DMF (15 mL) was stirred at room temperature overnight. The reaction mixture was poured into water (250 mL). The precipitate was collected by filtration, washed with H2O, and dried in vacuo to afford tert-butyl 4-(6-chloro-3-nitro-1 ,5 -naphthyridin-4- ylamino) piperidine-1 -carboxylate as yellow solid(6.54 g, yield 97.8%). The crude product was used in the next step without further purification. MS (m/z): 408(M+H)+.
[0102] To the solution of tert-butyl 4-(6-chloro-3-nitro-1 ,5-naphthyridin-4-ylamino) piperidine-1 -carboxylate (6.43 g, 15.7 mmol) in EtOAc (250 mL) was added
SnCl2-2H2O(18 g, 78.8 mmol), and the mixture was stirred at room temperature for 3h. Then saturated NaHCO3 was added to adjust the pH= 8. The solid was filtered off. The filtrate was concentrated and the crude product was purified by silica gel chromatography using EtOAcPE as eluant to afford tert-butyl 4-(3-amino-6-chloro-1 ,5- naphthyridin-4-ylamino) piperidine-1 -carboxylate as yellow solid in 85.6% yield (5.1 g). MS (m/z): 378 (M+H)+.
[0103] To a solution of tert-butyl 4-(3-amino-6-chloro-1 ,5-naphthyridin-4-ylamino) piperidine-1 -carboxylate(2.5 g, 6.6 mmol) in acetic acid(8 mL) was added NaNO2 (460 mg, 6.6 mmol) at 0°C. After the mixture was stirred at room temperature for 2 hours, saturated sodium bicarbonate and ice water were added. The resulting mixture was extracted with CH2CI2. The organic layer was dried over anhydrous Na2SO4, and concentrated. The crude product was purified by silica gel chromatography using EtOAcPE as eluant to afford tert-butyl 4-(8-chloro-1 H-[1 ,2,3]triazolo[4,5- c][1 ,5]naphthyridin-1 -yl) piperidine-1 -carboxylate as yellow solid in 73.8% yield (1 .9 g). MS (m/z): 388.8(M+H)+.
[0104] A solution of tert-butyl 4-(8-chloro-1 H-[1 ,2,3]triazolo[4,5-c][1 ,5]naphthyridin-1 - yl) piperidine-1 -carboxylate (4 g) in CH2CI2/ MeOH was treated with 3 mL cone. HCI. The resulting solution was then concentrated to give 8-chloro-1 -(piperidin-4-yl)-1 H- [1 ,2,3]triazolo[4,5-c][1 ,5] naphthyridine hydrochloride as yellow solid. (3.81 g, yield 100%). MS (m/z): 289 (M+H)+. [0105] To a solution of 8-chloro-1 -(pipehdin-4-yl)-1 H-[1 ,2,3]thazolo[4,5-c][1 ,5] naphthyhdine hydrochloride (1 g, 3.08 mmol) in 10ml_ of CH2CI2 was added (S)-1 - chloro-1 -oxopropan-2-yl acetate(1 .4 g, 9.23 mmol) and Et3N (2.2 mL ). After the mixture was stirred at room temperature for 2h, it was quenched with water (10 mL). The resulting mixture was extracted with CH2CI2 (2 x 20 mL). The combined organic layer was dried over anhydrous Na2SO4, and then was concentrated in vacuo to afford the (S)-1 -(4-(8-chloro-1 H-[1 ,2,3]triazolo[4,5-c][1 ,5]naphthyridin-1 -yl) piperidin-1 -yl) -1 - oxopropan-2-yl acetate (1 .2 g). MS (m/z): 403 (M+H)+.
[0106] To a solution of (S)-1 -(4-(8-chloro-1 H-[1 ,2,3]triazolo[4,5-c][1 ,5]naphthyridin-1 - yl) piperidin-1 -yl)-1 -oxopropan-2-yl acetate (1 .2 g, 2.97 mmol) in a mixture of THF (30 mL) and MeOH (30 mL), was added LiOH (650 mg, 14.9 mmol) drop-wise. The mixture was stirred at room temperature for 3h. After concentration in vacuo, the residue was diluted with water and the pH was adjusted to 7 with 2N HCI. The resulting mixture was concentrated and the precipitate was collected by filtration, washed with H2O, and dried in vacuo to afford (S)-1 -(4-(8-chloro-1 H-
[1 ,2,3]triazolo[4,5-c] [1 ,5]naphthyridin-1 -yl) piperidin-1 -yl)-2-hydroxypropan-1 -one as yellow solid. The crude product was used in next step without further purification. (918 mg, yield 85.4%). MS (m/z): 361 (M+H)+.
[0107] To a solution of (S)-1 -(4-(8-chloro-1 H-[1 ,2,3]triazolo[4,5-c][1 ,5]naphthyridin-1 - yl) piperidin-1 -yl)-2-hydroxypropan-1 -one (150 mg, 0.42 mmol) in a mixture of 20 mL dixoane and 2 mL H2O was added 5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-3- (trifluoromethyl)pyridin-2-amine (120 mg, 0.42 mmol), Pd(dppf)CI2 (20 mg, 0.02 mmol), and Na2COs(100 mg, 0.84 mmol). The resulting mixture was purged with N2 and stirred at 100°C overnight, then the resulting mixture was purified on silica gel using MeOH/H2O as eluent to afford compound 1 as yellow solid (59.1 mg). 1H NMR (400 MHz, DMSO-de) δ 9.60 (s, 1 H), 9.20 (s, 1 H), 8.67 (s, 1 H), 8.64 (d, J = 8.8 Hz, 1 H), 8.54 (d, J = 8.8Hz, 1 H), 7.09 (s, 2H), 6.23 - 6.02 (m, 1 H), 5.17 - 5.01 (m, 1 H), 4.75 - 4.50 (m, 2H), 4.46 - 4.27 (m, 1 H), 3.00 - 2.79 (m, 1 H), 2.39 - 1 .90 (m, 3H), 1 .36 - 1 .16 (m, 6H). MS (m/z): 487(M+H)+.
[0108] The following compounds 2-20 were prepared according to the procedures for Compound 1 by using the corresponding intermediates and boronic acids or esters under appropriate conditions that could be recognized by one skilled in the art.
Figure imgf000041_0001
Figure imgf000042_0001
Figure imgf000043_0001
Figure imgf000044_0001
Example 2: Synthesis of Compounds 21-29 Compound 21
(R)-1-(4-(8-(6-amino-5-(trifluoromethyl)pyridin-3-yl)-1 H-[1 ,2,3]triazolo[4,5- c][1 ,5]naphthyridin-1 -yl)piperidin-1 -yl)-2-hydroxypropan-1 -one
Figure imgf000044_0002
[0109] To a solution of 8-chloro-1 -(pipehdin-4-yl)-1 H-[1 ,2,3]thazolo[4,5-c][1 ,5]naphthy -hdine hydrochloride (600 mg, 1 .85 mmol) in DMF(15 mL) was added (R)-2- hydroxypropanoic acid (540 mg, 1 .85 mmol), HATU (850 mg, 2.21 mmol) and DIEA (0.8 mL, 3.70 mmol). The mixture was stirred at room temperature overnight. Another 3 eq of HATU and 2 eq of DIEA were added because the reaction did not go to completion as shown by HPLC-MS. The mixture was stirred for another 30 min, and was then diluted with water. The resulting mixture was extracted with ethyl acetate twice. The combined organic layers were dried over anhydrous Na2SO4, and concentrated to give the crude product, which was subsequently purified by chromatography on silica gel using EtOAc : PE as eluent to afford (R)-1 -(4-(8-chloro- 1 H-[1 ,2,3]triazolo[4,5-c][1 ,5]naphthyridin-1 -yl)piperidin-1 -yl)-2- hydroxypropan-1 -one as a yellow solid (136 mg, yield 20.4%). MS (m/z): 361 (M+H)+.
[0110] The next step for the synthesis of compound 21 was similar to the
corresponding step used for the synthesis of Compound 1.
[0111] Compound 21 : a solid, 31.0 mg. 1H NMR (400 MHz, dmso) δ 9.59 (s, 1 H), 9.19 (s, 1 H), 8.66 (s, 1 H), 8.63 (d, J = 8.9 Hz, 1 H), 8.53 (d, J = 8.9 Hz, 1 H), 7.07 (s, 2H), 6.09 (m, 1 H), 5.02 (m, 1 H), 4.74 - 4.27 (m, 2H), 2.87 (m, 1 H), 2.44 - 1.90 (m, 3H), 1.27 - 1.21 (m, 4H). MS (m/z): 487(M+H)+.
[0112] The following compounds 22-29 were prepared according to the procedures for Compound 21 by using the corresponding intermediates and boronic acid or ester under appropriate conditions that could be recognized by one skilled in the art.
Figure imgf000045_0001
Figure imgf000046_0001
Example 3: Synthesis of Compounds 30-42
Compound 30
1-(4-(8-(6-amino-5-(trifluoromethyl)pyridin-3-yl)-1 H-[1 ,2,3]triazolo[4,5- c][1 ,5]naphthyridin-1-yl)piperidin-1-yl)-2-methylpropan-1-one:
Figure imgf000047_0001
[0113] A mixture of 8-chloro-1 -(piperidin-4-yl)-1 H-[1 ,2,3]triazolo[4,5-c][1 ,5]naphthyrid -ine (200 mg, 0.69 mmol), isobutyric acid ( 67 mg, 0.76 mmol), HATU (315 mg, 0.83 mmol) and DIEA (133 mg, 1 .04 mmol) in DMF (5 ml_) was stirred at r.t. overnight.
Then water (10 ml_) was added; the precipitate was collected and dried in vacuo to afford 1 -(4-(8-chloro-1 H-[1 ,2,3]triazolo[4,5-c][1 ,5]naphthyridin-1 -yl)piperidin -1 -yl) -2- methylpropan-1 -one as a solid (200 mg).
[0114] A mixture of 1 -(4-(8-chloro-1 H-[1 ,2,3]triazolo[4,5-c][1 ,5]naphthyridin-1 -yl)piperi -din-1 -yl)-2-methylpropan-1 -one (60 mg, 0.17 mmol), 5-(4,4,5,5-tetramethyl-1 ,3,2- dioxaborolan-2-yl)-3-(trifluoromethyl)pyridin-2-amine (53 mg, 0.18 mmol), K2CO3 (70 mg, 0.51 mmol) and Pd(dppf)Cl2 (6 mg) in dioxane/H2O (3 : 1 , 4 ml_) was stirred and microwaved at 160°C for 0.5 h. The solvent was removed, and the residue was purified by ISCO (MeOH/H2O=20%-80%) to afford 1 -(4-(8-(6-amino-5-
(trifluoromethyl)pyridin-3-yl)-1 H-[1 ,2,3]triazolo[4,5-c][1 ,5]naphthyridin-1 -yl)piperidin-1 - yl)-2-methylpropan-1 -one as white solid (46 mg). 1HNMR (400 MHz, dmso) δ 9.61 (s, 1 H), 9.21 (d, J = 2.1 Hz, 1 H), 8.66 (dd, J = 12.5, 5.5 Hz, 2H), 8.55 (d, J = 8.9 Hz, 1 H), 7.10 (s, 2H), 6.17 - 6.01 (m, 1 H), 4.78 - 4.59 (m, 1 H), 4.35-4.26 (m, 1 H), 3.40-3.37 (m, 2H), 3.05-2.97 (m, 1 H), 2.93 - 2.82 (m, 1 H), 2.64 - 2.52 (m, 1 H), 2.39 - 2.21 (m, 1 H), 2.16 - 1 .96 (m, 1 H), 1 .07 (s, 6H). MS (m/z): 485(M+H)+.
[0115] The following compounds 31 -42 were prepared according to the procedures for Compound 30 by using the corresponding intermediates and boronic acid or ester under appropriate conditions that could be recognized by one skilled in the art.
Compound Structure LC/MS NMR
Figure imgf000049_0001
Example 4: Synthesis of Compounds 43-47
Compound 43
2-(4-(8-(6-amino-5-(trifluoromethyl)pyridin-3-yl)-1 H-[1 ,2,3]triazolo[4,5- c][1 ,5]naphthyridin-1 -yl)piperidin-1 -yl)-N,N-dimethylacetamide
Figure imgf000049_0002
[0116] A mixture of 8-chloro-1 -(pi perid i n -4-y I )- 1 H-[1 ,2,3]triazolo[4,5-c][1 ,5]naphthyri- dine (100 mg, 0.35 mmol), 2-chloro-N,N-dimethylacetamide (46 mg, 0.38 mmol), and K2CO3 (97 mg, 1 .04 mmol) in DMF (5 ml_) was stirred at r.t. overnight. The solvent was removed, and the residue was extracted with EtOAc (3 x10 ml_). The combined organic layers were dried over Na2SO4, and concentrated to afford crude 2-(4-(8- chloro-1 H- [1 ,2,3]triazolo[4,5-c] [1 ,5]naphthyridin-1 -yl) piperidin -1 -yl) -N,N- dimethylacetamide, which was used in the next step without further purification.
[0117] A mixture of the above product, 5-(4,4,5,5-tetramethyl-1 ,3,2- dioxaborolan-2-yl) -3-(trifluoromethyl)pyridin-2-amine (109 mg, 0.38 mmol), K2CO3 (145 mg, 1 .05 mmol) and Pd(dppf)Cl2 (10 mg) in dioxane/ H2O (3 : 1 , 4 ml_) was stirred and microwaved at 160 °C for 0.5h. The solvent was removed, and the residue was purified by
chromatography to afford compound 43 as a pale yellow solid (50 mg). 1HNMR (400 MHz, dmso-d6) δ 9.60 (s, 1 H), 9.20 (d, J = 2.0 Hz, 1 H), 8.66 (dd, J = 16.5, 5.4 Hz, 2H), 8.54 (d, J = 8.9 Hz, 1 H), 7.12 (s, 2H), 5.97 - 5.77 (m, 1 H), 3.40 (s, 2H), 3.29 - 3.14 (m, 4H), 3.10 (s, 3H), 2.86 (s, 3H), 2.44-2.35 (m, 4H). MS (m/z): 500(M+H)+.
[0118] The following compounds 44-47 were prepared according to the procedures for Compound 43 by using the corresponding intermediates, and boronic acid or ester under appropriate conditions that could be recognized by one skilled in the art:
Figure imgf000050_0001
Figure imgf000051_0001
Example 5: Synthesis of Compound 48
Compound 48
5-(1 -(1 -((tetrahydro-2H-pyran-4-yl)methyl)piperidin-4-yl)-1 H-[1 ,2,3]triazolo
[4,5-c][1 ,5]naphthyridin-8-yl)-3-(trifluoromethyl)pyridin-2-amine
Figure imgf000051_0002
[0119] Under N2, a white suspension of 8-chloro-1 -(piperidin-4-yl)-1 H-
[1 ,2,3]triazolo[4,5-c][1 ,5] naphthyridine (130 mg, 0.450 mmol), 4-(bromomethyl) tetrahydro-2H-pyran (97 mg, 0.540 mmol) and K2CO3 (124 mg, 0.900 mmol) in acetonitrile (15 ml_) was heated to reflux for 4h. After cooling to room temperature, the mixture was diluted with EtOAc 20 ml_. The mixture was then filtered through a Buchner funnel, and the organic phase was collected and concentrated. The crude product was used in the next step without further purification (35 mg). MS (m/z): 387 (M+H)+. [0120] Under N2, an orange suspension of 8-chloro-1 -(1 -((tetrahydro-2H-pyran-4-yl) methyl) piperidin-4-yl) - H-[1 ,2,3] triazolo [4,5-c][1 ,5]naphthyridine(35 mg, 0.090 mmol), 5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-3-(trifluoromethyl)pyridin-2-amine (26.1 mg, 0.090 mmol), Na2CO3 (19.18 mg, 0.181 mmol) and PdCI2(dppf).CH2CI2 (3.69 mg, 4.52 μηηοΙ) in a mixture of dioxane (20 mL) and H2O(2 mL) was stirred for 10 minutes, before the resulting mixture was heated to 120°C for 2 h. After concentration in vacuo, the resulting residue was purified by ISCO with 12 g silica gel (PE/EtOAc) to give the product as pale yellow powder (10 mg). 1H NMR (400 MHz, dmso) δ 9.53 (s, 1 H), 9.15 (d, J = 2.1 , 1 H), 8.65 (s, 1 H), 8.61 (d, J = 8.9, 1 H), 8.51 (d, J = 8.9, 1 H), 7.07 (s, 2H), 5.99 - 5.80 (m, 1 H), 3.91 - 3.85 (m, 4H), 3.22 - 3.08 (m, 4H), 2.41 - 2.15 (m, 10H), 1 .73-1 .58(m,2H). MS (m/z): 513 (M+H)+.
Example 6: Synthesis of Compounds 49-78
Compound 49
2-(4-(8-(6-amino-5-(trifluoromethyl)pyridin-3-yl)-[1 ,2,4]triazolo[4,3-a]quinoxalin-1 - yl)phenyl)-2-methylpropanenitrile
Figure imgf000052_0001
[0121 ] To a solution of 7-bromo-2-hydrazinylquinoxaline (1 .5 g, 0.063 mol) and 4-(2- cyanopropan-2-yl) benzoic acid (1 .1 g, 0.063 mol) in DMF(5 mL) was added HATU (2.4 g, 0.063 mol) and DIEA (1 .2 g, 0.095 mol). The reaction mixture was then stirred at r.t.overnight. The solution was diluted with water (5 mL), and the solid was collected on a filter to give N'-(7-bromoquinoxalin-2-yl)-4-(2-cyanopropan-2-yl)benzohydrazide as a yellow solid (2.2 g, yield 85.0%). MS (m/z): 412 (M+H)+. [0122] A solution of N'-(7-bromoquinoxalin-2-yl)-4-(2-cyanopropan-2- yl)benzohydrazide (2.2 g, 0.054 mol) in 3 mL of AcOH was stirred at 100°C overnight. After cooling to room temperature, the reaction mixture was diluted with water (5 mL). The solid was collected on a filter, and washed with Sat. NaHCO3 (5 mL) to give 2-(4- (8-bromo- [1 ,2,4] triazolo[4,3-a]quinoxalin-1 -yl) phenyl)-2-methylpropanenitrile as a yellow solid (1 .8 g, yield 85.0%). MS (m/z): 392 (M+H)+.
[0123] To a mixture of 2-(4-(8-bromo-[1 ,2,4]triazolo[4,3-a]quinoxalin-1 -yl) phenyl) -2- methylpropanenitrile (80 mg, 0.21 mmol), 5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2- yl)-3-(trifluoromethyl)pyridin-2-amine (59 mg, 0.21 mmol) and K2CO3(87 mg, 0.63 mmol) in dioxane (3 mL) and H2O(1 mL) was added Pd(dppf)Cl2 (3 mg). The reaction mixture was microwaved at 150°C for 30min. After cooling to room temperature, the mixture was concentrated and purified by chromatography to give compound 49 as a yellow solid (52 mg). 1H NMR (400 MHz, dmso) δ 9.38 (s, 1 H), 8.26 (s, 1 H), 8.1 1 (d, J = 8.5, 1 H), 7.99 - 7.95 (m, 1 H), 7.92 (d, J = 8.3, 2H), 7.83 (d, J = 8.4, 2H), 7.63 (m, 1 H), 7.52 (d, J = 1 .5, 1 H), 6.77 (s, 2H), 1 .77 (s, 6H). MS (m/z): 474 (M+H)+.
[0124] The following compounds 50-78 were prepared according to the procedures for Compound 49 by using the corresponding intermediates and boronic acid or ester under appropriate conditions that could be recognized by one skilled in the art.
Figure imgf000053_0001
Figure imgf000054_0001
Figure imgf000055_0001
Figure imgf000056_0001
Figure imgf000057_0001
Example 7: Synthesis of Compounds 79-87
Compound 79
(S)-1 -(4-(8-(6-amino-5-(trif luoromethyl)pyridin-3-yl)-[1 ,2,4]triazolo[4,3- a]quinoxalin-1 -yl)piperidin-1 -yl)-2-hydroxypropan-1 -one
Figure imgf000057_0002
[0125] A mixture of 1 -tert-butyl 4-methyl piperidine-1 ,4-dicarboxylate (4.84 g 20 mmol), and LiOH(2.52 g,60 mmol) in THF(90 mL) /MeOH (90 mL) /H2O(30 mL) was stirred at r.t overnight. Then the solvents were removed, and the pH of the residue was adjusted to 2 by using 2N HCI. The resulting mixture was extracted with EtOAc (3 x 20 mL). The combined organic layers were dried over Na2SO4, and concentrated to give 1 -(tert-butoxycarbonyl)piperidine -4-carboxylic acid (4.6 g, yield 100.0%). [0126] A mixture of 7-bromo-2-hydrazinylquinoxaline (3 g, 12.55 mmol), 1 -(tert- butoxycarbonyl)piperidine-4-carboxylic acid (3.16 g, 13.81 mmol), EDCI (2.89 g, 15.06 mmol), HOBt (2.03 g, 15.06 mmol), and TEA (1 .9 g, 18.83 mmol) in DMF (100 ml_) was stirred at r.t. overnight. The mixture was diluted with water (10 ml_), and extracted with EtOAc (3 x 100 ml_). The combined layers were dried over Na2SO4, and concentrated in vacuo to afford tert-butyl 4-(2-(7-bromoquinoxalin-2- yl)hydrazinecarbonyl) piperidine-1 -carboxylate as pale yellow solid (3.5g, yield 62 %).
[0127] A mixture of tert-butyl 4-(2-(7-bromoquinoxalin-2-yl)hydrazinecarbonyl) piperidine-1 -carboxylate (900 mg, 2.0 mmol) in AcOH(10 ml_) was refluxed overnight. Then the solvent was removed, and the residue was purified by ISCO (MeOH / H2O = 20%-90%) to afford 8-bromo-1 -(piperidin-4-yl)-[1 ,2,4] triazolo[4,3-a] quinoxaline as a pale yellow solid (550 mg, yield 83.0%). MS (m/z): 332 (M+H)+.
[0128] A mixture of 8-bromo-1 -(piperidin-4-yl)-[1 ,2,4]triazolo[4,3-a]quinoxaline (250 mg, 0.75 mmol), (S)-2-hydroxypropanoic acid (75 mg, 0.83 mmol), HATU (346 mg, 0.90 mmol), and DIEA (1 16 mg, 0.90 mmol) in DMF (5 ml_) was stirred at r.t. for 6h. Then the solvents was removed, and the residue was purified by ISCO
(MeOH/H2O=20%-90%) to afford (S)-1 -(4-(8-bromo-[1 ,2,4]triazolo[4,3-a]quinoxalin-1 - yl) piperidin-1 -yl) -2-hydroxypropan-1 -one as pale yellow solid (200 mg, yield 66.0%). MS (m/z): 404 (M+H)+.
[0129] A mixture of (S)-1 -(4-(8-bromo-[1 ,2,4]triazolo[4,3-a]quinoxalin-1 -yl) piperidin-1 - yl)-2-hydroxypropan-1 -one (65 mg, 0.16 mmol), 5-(4,4,5,5- tetramethyl-1 ,3,2- dioxaborolan-2-yl)-3-(trifluoromethyl)pyridin-2-amine (51 mg, 0.18 mmol), K2CO3 (67 mg, 0.48 mmol) and Pd(dppf)Cl2 (5 mg) in dioxane/H2O(3 : 1 , 4 ml_) was microwaved at 150°C for 0.5 h. Then the solvents were removed, and the residue was purified by ISCO (MeOH/H2O=20%-80%) to afford compound 72 as yellow solid (30 mg). 1HNMR (400 MHz, dmso) δ 9.28 (s, 1 H), 8.72 (d, J = 2.0 Hz, 1 H), 8.32 (s, 1 H), 8.16-8.12 (m, 2H), 8.05 (dd, J = 8.5, 1 .5 Hz, 1 H), 6.82 (s, 2H), 4.91 (dd, J = 7.2, 6.5 Hz, 1 H), 4.47 (dd, J = 17.8, 1 1 .8 Hz, 2H), 4.29 (t, J = 10.6 Hz, 1 H), 4.15 (t, J = 1 1 .2 Hz, 1 H), 3.09 - 2.97 (m, 1 H), 2.27 (dd, J = 15.6, 14.5 Hz, 2H), 1 .97 (dd, J = 12.0, 4.5 Hz, 1 H), 1 .78 (dd, J = 7.1 , 5.0 Hz, 1 H), 1 .25-1 .15 (m, 3H). MS (m/z): 486 (M+H)+.
[0130] The following compounds 80-87 were prepared according to the procedures for Compound 79 by using the corresponding intermediates and boronic acid or ester under appropriate conditions that could be recognized by one skilled in the art:
Figure imgf000059_0001
Figure imgf000060_0001
Example 8: Synthesis of Compounds 88-118
Compound 88
(4-(8-(6-amino-5-(trifluoromethyl)pyridine-3yl)-1 H-imidazo[4,5-c][1 ,5]
naphthyridin-1 -yl)piperidin-1-yl)(cyclopropyl)methanone
Figure imgf000060_0002
[0131 ] A mixture of 2,8-dichloro-7-nitro-1 ,5-naphthyridine (3.55 g, 14.55 mmol) and K2CO3 (6.02 g, 43.65 mmol) in DMF (8 mL) was stirred at r.t. overnight, and then poured into ice-water (-20 mL). The precipitate was collected, washed with water three times, and dried in vacuo to afford tert-butyl 4-(6-chloro-3-nitro-1 ,5-naphthyridin 4-ylamino)piperidine-1 -carboxlate as yellow solid (5.22 g, yield 88%) which was used in next step without further purification. MS (m/z): 409 (M+H)+.
[0132] A mixture of tert-butyl 4-(6-chloro-3-nitro-1 ,5-naphthyridin-4-ylamino)
piperidine-1 -carboxlate (600 mg, 1 .47 mmol) and SnCl2.H2O (996 mg, 4.41 mmol) in ethyl acetate (20 mL) was stirred at r.t. for 2h, and was then alkalized with 5% NaOH solution. The mixture was filtered through a pad of celite. The filtrate was extracted with ethyl acetate (3 x 15 mL). The organic layers were combined and washed with brine (10 mL) , dried over anhydrous Na2SO4, filtered, and concentrated to give tert- butyl 4-(3-amino-6-chloro-1 ,5-naphthyridin-4-ylamino) piperidine-1 -carboxlate as a yellow solid (499 mg, yield 90%) which was used in the next step without further purification. MS (m/z): 378(M+H)+.
[0133] A mixture of tert-butyl 4-(3-amino-6-chloro-1 ,5-naphthyridin-4-ylamino) piperidine-1 -carboxlate (200 mg, 0.53 mmol), triethyl orthofornnate (94 mg, 0.64 mmol), and PyHCI (6 mg, 0.053 mmol) in Toluene (5 mL) was refluxed for 3.5h. The solvent was removed under vacuum and the residue was added to a solution of HCI in MeOH (6N, 3 mL). The reaction mixture was stirred at r.t. for 3h, and was then concentrated under vacuum. The residue was dissolved in dichloromethane(20 mL). the resulting solution was washed with saturated NaHCO3 (10 mL) and brine (10 mL), dried over anhydrous Na2SO4, filtered, and concentrated to to give 8-chloro-1 -(piperidin-4-yl)-1 H- imidazo[4,5-c][1 ,5]naphthayridine as a yellow solid (1 10 mg, yield 72%) which was used in next step without further purification.
[0134] To a solution of 8-chloro-1 -(piperidin-4-yl)-1 /-/-imidazo[4,5- c][1 ,5]naphthayridine (1 10 mg, 0.382 mmol) and Et3N (106 μί, 0.764 mmol) in THF (15 mL) was added cyclopropanecarbonyl chloride (38 μί, 0.420 mmol) while cooling with an ice-water bath. The reaction mixture was stirred at r.t. for 3h, and was then concentrated under vacuum. The residue was dissolved in ethyl acetate(20 mL). The resulting solution was washed with saturated NaHCO3 (10 mL) and brine (10 mL), dried over anhydrous Na2SO4, filtered, and concentrated to give (4-(8-chloro-1 H- imidazo[4,5-c][1 ,5]naphthyridin-1 -yl)piperidin- 1 -yl)(cyclopropyl)methanone as a yellow solid (100 mg, yield 73%) which was used in the next step without further purification. MS (m/z): 356 (M+H)+.
[0135] A mixture of (4-(8-chloro-1 /-/-imidazo[4,5-c][1 ,5]naphthyridin-1 -yl)piperidin- 1 - yl)(cyclopropyl)methanone (100 mg, 0.281 mmol), PdCI2(dppf)2(12 mg, 0.014 mmol), 5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-3-(trifluoromethyl)pyridine -2-amine(97 mg, 0.337 mmol), and 2 N K2CO3 solution (1 ml_) in dioxane (4 ml_) was microwaved at 150°C for 30min. The solvent was removed and the residue was purified by ISCO (MeOH/H2O 0%~100%) to give compound 88 as a yellowish solid (70 mg). 1H NMR (400 MHz, dmso) δ 9.20 (s, 1 H), 9.13 (d, J = 1 .9, 1 H), 8.71 (s, 1 H), 8.59 (d, J = 2.0, 1 H), 8.49 (d, J = 8.9, 1 H), 8.32 (d, J = 8.9, 1 H), 6.99 (s, 2H), 5.98 - 5.92 (m, 1 H), 4.66 (s, 1 H), 4.58 (s, 1 H), 3.25 (s, 2H), 2.35 (s, 2H), 2.09 - 2.01 (m, 2H), 2.01 - 1 .90 (m, 1 H), 0.80 - 0.71 (m, 4H). MS (m/z): 482 (M+H)+.
[0136] The following compounds 89-1 18 were prepared according to the procedures for Compound 88 by using the corresponding intermediates and boronic acid or ester under appropriate conditions that could be recognized by one skilled in the art.
Figure imgf000062_0001
Figure imgf000063_0001
Figure imgf000064_0001
Figure imgf000065_0001
Figure imgf000066_0001
Figure imgf000067_0001
Example 9: Synthesis of Compound 119-146
Compound 119
(4-(8-(6-amino-5-(trifluoromethyl)pyridine-3yl) -2-methyl-1 «-imidazo[4,5-c][1 ,5] naphthyridin-1-yl)piperidin-1-yl)(cyclopropyl)methanone
Figure imgf000068_0001
[0137] A mixture of tert-butyl 4-(3amino-6-chloro-1 ,5-naphthyridin-4-ylamino) piperidine-1 -carboxlate (200 mg, 0.53 mmol) in acetic acid(3 mL) was stirred at 100 °C overnight. The solvent was removed under vacuum and the residue was added to a solution of HCI in MeOH (6N, 3 mL). The reaction mixture was stirred at r.t. for 2h, and was then concentrated under vacuum. The residue was dissolved in dichloromethane (20 mL). The resulting solution was washed with saturated NaHCO3 (10mL) and brine (10 mL), dried over anhydrous Na2SO4, filtered, and concentrated to give crude 8- chloro-2-methyl-1 -(piperidin-4-yl)-1 /-/-imidazo[4,5-c][1 ,5] naphthyridine as a yellow solid (1 10 mg, yield 68%) which was used in the next step without further purification.
[0138] To a solution of 8-chloro-2-methyl-1 -(piperidin-4-yl)-1 /-/-imidazo[4,5-c][1 ,5] naphthyridine (1 10 mg, 0.364 mmol) and Et3N (101 μΙ_, 0.728 mmol) in THF (15 mL) was added cyclopropanecarbonyl chloride (36 μί, 0.401 mmol) while cooling with an ice-water bath. The reaction mixture was stirred at r.t. for 3h, and was then
concentrated under vacuum. The residue was dissolved in a mixture of CH2CI2 (10 mL) and H2O (10 mL). The resulting solution was then extracted with EA (2 x 10 mL). The combined organic layers were concentrated to give crude (4-(8-chloro-2-methyl- 1 H-imidazo[4,5-c][1 ,5]naphthyridin-1 -yl) piperidin- 1 -yl)(cyclopropyl)methanone as a yellow solid (105 mg, yield 80%) which was used in the next step without further purification. MS (m/z): 370 (M+H)+.
[0139] A mixture of crude (4-(8-chloro-2-methyl-1 /-/-imidazo[4,5-c][1 ,5]naphthyridin-1 - yl) piperidin- 1 -yl)(cyclopropyl)methanone (105 mg, 0.284 mmol), PdCl2(dppf)2(12 mg, 0.014mmol), 5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl)-3-(trifluoromethyl) pyridine-2-amine(98 mg, 0.340 mmol), and 2 N K2CO3 solution (1 mL) in dioxane(4 mL) was microwaved at 150°C for 30min. The solvent was removed and the residue was purified by ISCO(MeOH/H2O 0%~100%) to obtain compound 1 19 as a yellowish solid (57 mg). 1H NMR (400 MHz, dmso) δ 9.1 1 (d, J = 6.8, 2H), 8.58 (s, 1 H), 8.48 (d, J = 8.8, 1 H), 8.29 (d, J = 8.3, 1 H), 6.97 (s, 2H), 4.67 - 4.58 (m, 2H), 4.06 (s, 1 H), 2.75 (m, 5H), 2.1 1 - 2.04 (m, 4H), 0.81 (s, 1 H), 0.71 (m, 4H). MS (m/z): 496 (M+H)+.
[0140] The following compounds 120-146 were prepared according to the procedures for Compound 1 19 by using the corresponding intermediates and boronic acid or ester under appropriate conditions that could be recognized by one skilled in the art.
Figure imgf000069_0001
Figure imgf000070_0001
Figure imgf000071_0001
Figure imgf000072_0001
Figure imgf000073_0001
Example 10: Synthesis of Compounds 147-178
Compound 147 2,4-difluoro-N-(2-methoxy-5-(1-methyl-[1 ,2,4]triazolo[4,3-a]quinoxalin-8- yl)pyridin-3-yl)benzenesulfonamide
Figure imgf000074_0001
[0141] A mixture of 7-bromo-2-hydrazinylquinoxaline (200 mg, 0.84 mmol) and AcOH (5 ml_) was refluxed overnight. After cooling to r.t., the mixture was treated with water (10 ml_); the solid was collected on a filter and dried by vacuum to give 8-bromo-1 - methyl-[1 ,2,4]triazolo [4,3-a]quinoxaline as gray solid (200 mg, yield: 91 .0%).
[0142] A mixture of 8-bromo-1 -methyl-[1 ,2,4]triazolo[4,3-a]quinoxaline (60 mg, 0.23 mmol), 2,4-difluoro-N-(2-methoxy-5-(4,4,5,5-tetramethyl-1 ,3,2-dioxaborolan-2-yl) pyridine-3-yl)benzenesulfonamide (107 mg, 0.25 mmol), K2CO3 (95 mg, 0.69 mmol) and Pd(dppf)Cl2 (6 mg) in dioxane/H2O (3 : 1 , 4 ml_) was microwaved at 160°C for 0.5h. Then the solvent was removed, and the residue was purified by ISCO
(MeOH/H2O = 20%-80%) to give compound 147 as white solid (60 mg). 1HNMR (400 MHz, dmso) δ 10.44 (s, 1 H), 9.26 (s, 1 H), 8.47 (d, J = 1 .7 Hz, 1 H), 8.34 (d, J = 1 .5 Hz, 1 H), 8.13 (t, J = 10.3 Hz, 1 H), 8.02 (d, J = 2.1 Hz, 1 H), 7.92 (dd, J = 8.4, 1 .5 Hz, 1 H), 7.76 (dd, J = 14.9, 8.5 Hz, 1 H), 7.51 (dd, J = 14.2, 5.4 Hz, 1 H), 7.18 (t, J = 7.5 Hz, 1 H), 3.68 (s, 3H), 3.17 (s, 3H). MS (m/z): 483 (M+H)+.
[0143] The following compounds 148-178 were prepared according to the procedures for Compound 147 by using the corresponding intermediates and boronic acid or ester under appropriate conditions that could be recognized by one skilled in the art.
Compounds Structure LC/MS NMR
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
Figure imgf000078_0001
Figure imgf000079_0001
Figure imgf000080_0001
Example 11 : Synthesis of Compounds 179-182
Compound 179
N-(2-methoxy-5-(1 -methyl-[1 ,2,4]triazolo[4,3-a]quinoxalin-8-yl)pyrid
methyl benzenesulfonamide
Figure imgf000080_0002
[0114] A mixture of 7-bromo-2-hydrazinylquinoxaline (1 g, 4.18 mmol) in acetic acid (10 mL) was refluxed for 18h. Half of the acetic acid was removed under vacuum and the residue was poured into ice-water. The precipitate was collected on a filter, washed with water, and dried under vacuo to afford crude 8-bromo-1 -methyl- [1 ,2,4]triazolo[4,3-a]quinoxaline as a dark red solid (1 g, yield 90%) which was used in the next step without further purification. MS (m/z): 263(M+H)+.
[0115] A mixture of crude 8-bromo-1 -methyl-[1 ,2,4]triazolo[4,3- a]quinoxaline(809 mg, 3.07 mmol), PdCI2(dppf)2(132.6 mg, 0.153 mmol), 2-methoxy-5- (4,4,5,5-tetramethyl -1 ,3,2-dioxaborolan-2-yl)pyridin-3-amine (1 g, 4.3 mmol) and K2CO3(1 .7 g, 12.28 mmol) in DMF(40 ml_) and water (15 mL) was stirred at 100°C overnight. Half of solvent was removed. After cooling to room temperature, the resulting mixture was poured into ice-water. The precipitate was collected, washed with water three times, and dried in vacuo to afford crude 2-methoxy-5-(1 -methyl - [1 ,2,4]triazolo [4,3-a] quinoxalin-8-yl) pyridin-3-amine as grey solid (730 mg, yield 77% ) which was used in next step without further purification. MS (m/z): 307 (M+H)+.
[0116] To a mixture of crude 2-methoxy-5-(1 -methyl-[1 ,2,4]triazolo[4,3- a]quinoxalin-8-yl) pyridin-3-amine (50 mg, 0.163 mmol) in pyridine(2 mL) was added 4-methylbenzene -1 -sulfonyl chloride (31 .3 mg, 0.164 mmol). The mixture was stirred at room temperature overnight, and was then heated at 50°C for 5h. The solvent was removed, and the residue was purified by PTLC to afford compound 179 as a grey solid (22 mg). 1H NMR (400 MHz, dmso) δ 10.00 (s, 1 H), 9.27 (s, 1 H), 8.44 (d, J = 2.3, 1 H), 8.32 (d, J = 1 .8, 1 H), 8.15 (d, J = 8.4, 1 H), 7.98 (d, J = 2.3, 1 H), 7.89 (dd, J = 8.4, 1 .8, 1 H), 7.68 (dd, J = 8.4, 1 .8, 2H), 7.35 (d, J = 8.0, 2H), 3.71 (s, 3H), 3.17 (s, 3H), 2.33 (s, 3H). MS (m/z): 461 (M+H)+.
[0117] The following compounds 180-182 were prepared according to the procedures for Compound 179 by using the corresponding intermediates and boronic acid or ester under appropriate conditions that could be recognized by one skilled in the art.
Compound Structure LC/MS NMR
Figure imgf000082_0001
Example 12: Synthesis of Compounds 183 and184
Compound 183
2-(4-(8-(6-amino-5-(trifluoromethyl)pyridin-3-yl)-1 H-imidazo[4,5-c]cinnolin-1 - yl)phenyl)-2-methylpropanenitrile
Figure imgf000082_0002
[0118] To a white suspension of 1 -(2-aminophenyl)ethanone (25 g, 185 mmol) and Et3N (33.4 mL, 240 mmol) in DCM at 0°C was added dropwise AcCI (15.70 mL, 222 mmol) in 25 mL DCM. The reaction mixture was then stirred at r.t. for 2 h. After the reaction went to completion as monitored by LC-MS, the reaction mixture was cooled to 0°C, and was then quenched with H2O (100 mL). The organic phase was isolated; the aqueous layer was extracted with DCM. The organic phases were combined, and washed with H2O and brine. The resulting organic phase was dried over anhydrous MgSO4, filtered and concentrated to dryness to give crude N-(2- acetylphenyl)acetamide, which was used in the next step without further purification (30g, yield 92%). MS (m/z): 136 (M+H)+.
[0119] Under N2, a pale yellow suspension of crude N-(2-acetylphenyl)acetamide(28 g, 158 mmol) in AcOH (300 mL) was stirred for about 5 min, and Br2 (12.95 mL, 253 mmol) was added within 1 h at room temperature. The resulting mixture was then stirred at room temperature for 75 min. After the reaction went to completion as monitored by LC-MS, the reaction mixture was quenched with H2O (200 mL). The mixture was filtered through Buchner funnel, and the solid was collected as N-(2- acetyl-4-bromophenyl)acetamide (35g, yield 86%). MS (m/z): 216 (M+H)+.
[0120] Under N2, a mixture of N-(2-acetyl-4-bromophenyl)acetamide (35 g, 137 mmol) and HCI (100 mL, 600 mmol) in THF (400 mL) was heated to reflux for 1 h. After being concentrated under vacuum to remove the solvent, the mixture was treated with EtOAc( 100 mL). The aqueous layer was concentrated to remove THF, and 6N HCI (100 mL, 600 mmol) was added at room temperature. After cooling to 0°C, the resulting mixture was treated dropwise with NaNO2 (9.43 g, 137 mmol) in 20 mL H2O. The reaction mixture was then stirred at r.t. for 15 h. Then the resulting mixture was heated to reflux for another 6 h. The mixture was cooled to r.t. The solid was collected and dried in vacuo, to afford the desired product as a white solid. The crude product was used directly in next step without further purification. (19.5g, yield 63.4%) MS (m/z): 227 (M+H)+. [0121] Under N2, an orange solution of 6-bromocinnolin-4-ol (18.5 g, 82 mmol) in HNO3 (90 ml_, 82 mmol) was cooled to 0°C, and H2SO4 (30 ml_) was added. The resulting mixture was then heated at 60°C for 3h. After the reaction went to completion as monitored by LC-MS, the reaction mixture was cooled to 0°C, and was then quenched with H2O (20 ml_). The mixture was diluted with EtOAc (25 ml_). Following general workup procedures, the crude residue was added to silica gel, and eluted with PE/EtOAc to give 6-bromo-3-nitrocinnolin-4-ol as a pale yellow solid (13 g, yield
58.6%).
[0122] Under N2, a brown solution of 6-bromo-3-nitrocinnolin-4-ol (2 g, 7.41 mmol) in DMF (10 ml_) was cooled to 0°C, and POCI3 (0.897 ml_, 9.63 mmol) was added dropwise. The reaction mixture was stirred at r.t. for 5 h. After the reaction went to completion as monitored by LC-MS, the reaction mixture was cooled to 0°C, and then quenched with H2O (50 ml_). The mixture was filtered through a Buchner funnel, and the solid was collected. The crude 6-bromo-4-chloro-3-nitrocinnoline was used in the next step without further purification (1 .75 g, yield 82%). MS (m/z): 290 (M+H)+.
[0123] A yellow suspension of 6-bromo-4-chloro-3-nitrocinnoline (1 .75 g, 6.07 mmol), 2-(4-aminophenyl)-2-methylpropanenitrile (1 .069 g, 6.67 mmol) and K2CO3 (1 .677 g, 12.13 mmol) in MeCN (2 mL) was heated to reflux for 5 min. The workup followed general workup procedures, and the crude was purified on silica gel with PE/EtOAc as eluant to give 2-(4-(6-bromo-3- nitrocinnolin-4-ylamino)phenyl)-2-methylpropanenitrile as yellow solid (2.5g, yield 100%). MS (m/z): 414 (M+H)+.
[0124] Under N2, an orange solution of 2-(4-(6-bromo-3-nitrocinnolin-4- ylamino)phenyl)-2-methylpropanenitrile (2.5 g, 6.06 mmol) and SnCI2.2H2O (5.21 g, 24.26 mmol) in EtOAc(50 mL) was heated at 45°C for 3 h. After cooling to r.t., the pH of the mixture was adjusted to 8 with saturated Na2CO3. The mixture was filtered through a Buchner funnel, and the filtrate was collected and concentrated to give 2-(4- (3-amino-6-bromocinnolin-4-ylamino)phenyl)-2-methylpropaneni (1 .6 g, yield 69%). MS (m/z): 384 (M+H)+.
[0125] Under N2, a brown solution of 2-(4-(3-amino-6-bromocinnolin-4- ylamino)phenyl)-2-methyl-propanenitrile (250 mg, 0.654 mmol) in HCO2H (3 mL) was heated to reflux for 4 h. The reaction mixture was quenched with H2O, and was then concentrated to remove the solvent. The crude product was used in thenext step directly without further purification (250 mg, yield 97%). MS (m/z): 394 (M+H)+.
[0126] Under N2, an orange suspension of 2-(4-(8-bromo-1 H-imidazo[4,5-c]cinnolin- 1 -yl)phenyl)-2-methylpropanenitrile (100mg, 0.255 mmol), 5-(4,4,5,5-tetramethyl-1 ,3,2- dioxaborolan-2-yl)-3-(trifluoromethyl)pyridin-2-amine (95 mg, 0.331 mmol), Na2CO3 (54.0 mg, 0.510 mmol) and PdCI2(dppf).CH2CI2 (10.41 mg, 0.013 mmol) in dioxane (20 mL) and H2O (2 mL) was stirred for 10 minutes at r.t. The resulting mixture was then heated at 120°C for 2h. The mixture was concentrated in vacuo, and the residue was then purified by chromatography to give compound 183 as a pale yellow powder (50 mg). 1H NMR (400 MHz, dmso) δ 8.87 (s, 1 H), 8.63 (d, J = 8.9, 1 H), 8.48 (d, J = 2.0, 1 H), 8.22 (dd, J = 9.0, 1 .9, 1 H), 7.95 (d, J = 8.6, 2H), 7.88 (d, J = 8.6, 2H), 7.76 (d, J = 2.1 , 1 H), 7.52 (d, J = 1 .8, 1 H), 6.84 (s, 2H), 1 .78 (s, 6H). MS (m/z): 474 (M+H)+.
[0127] The following compound 184 was prepared according to the procedures of Compound 183 by using the corresponding intermediates and boronic acid or ester under appropriate conditions that could be recognized by one skilled in the art.
Figure imgf000085_0001
Example 13 PI3Ka Transcreener ADP Assay
[0128] Fluorescence polarization was used in this assay. The final conditions for kinase assay are 10 μΜ of ATP, 0.2 ng/ L of PI3Ka kinase, 30 μηηοΙ/L of lipid substrate and assay buffer (50 mmol/L of HEPES (pH 7.5),100 mmol/L of NaCI, 1 mmol/L of EGTA, 3 mmol/L of MgCI2, 1 mmol/L of DTT and 0.03% CHAPS and 2% DMSO).
[0129] 5 μί of test compounds in 10% DMSO and10 pL of 0.5 ng/ L PI3K kinase (Invitrogen, PV4788) in assay buffer are put into a 96-well plate (Greiner, Cat.
675076), and then the reaction is started by the addition of 10 μί of 75 μηηοΙ/L PIP2, (PS Lipid Substrate Invitorgen, PV5100), and 25 μηηοΙ/L ATP mixture. After the mixture is incubated for 60 minutes at room temperature, 25 μί of transcreener Kinase Kit reagent - ADP Detection Mix - (Bellbrook Labs) is added, and the reaction is continued for an additional 1 .5 hours. At the end, the plate is read in a Tecan Infinite F500 at excitation of 610 nm and emission of 670 nm.
[0130] A standard curve for ADP is obtained in a parallel way by replacing compound and PI3Ka kinase with DMSO and assay buffer, respectively. Different concentration of ADP, 0 -10 μΜ, and ATP, 10-0 μΜ ( ATP+ADP equal to 10 μΜ ) are applied instead of a fixed concentration of ATP in this standard curve assay. Other conditions are the same as described above. The standard curve is plotted using Origin 8.0 software. The inhibition of test compound on ADP production is calculated based on ADP concentration from standard curve. IC50 is obtained using XLfit 2.0 software.
[0131] Results
Above compounds 1 , 2, 3, 5, 6,7, 8, 9,10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 20, 21 , 22, 23, 24, 25, 26, 28, 29, 30, 31 , 33, 35, 36, 37, 38, 39, 40, 41 , 42, 43, 44, 45, 46, 47, 48, 49, 50, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61 , 62, 63, 64, 65, 66, 67, 68, 69, 70, 71 , 72, 73, 74, 75, 76, 77, 79, 80, 81 , 82, 83, 84, 85, 87, 88, 89, 90, 92, 94, 95, 96, 97, 98, 99, 100, 101 , 102, 103, 104, 105, 106, 107, 108, 109, 1 10, 1 1 1 , 1 12, 1 13, 1 19,120, 121 , 122, 123, 124, 125, 126, 127, 128, 129, 130, 131 , 132, 133, 134, 135, 136, 137, 138, 139, 140, 142, 143, 144, 145, 147, 148, 149, 150, 151 , 152, 153, 154, 155, 156, 157, 158, 159,160, 161 , 162, 163, 164, 165,166, 167, 168, 169, 170, 171 , 172, 173, 174, 175, 176, 177, 178, 179, 180, 181 , 182, 183, 184 inhibited the PI3Ka kinase receptor with IC50 < 100 nM.
Example 14
P-Akt Acumen Assay
[0132] Compounds are tested using p-Akt Acumen cell-based assay. The human prostate cancer cell line PC3 (ATCC) is cultured in F-12 medium with 10% fetal bovine serum. For p-Akt Acumen assay, PC3 cells are seeded at a density of 5000 cells/90 μΙ_ in Poly-D-Lysine 96-well plate (BD, 356692). After incubation for 24 hours, different concentrations of test compounds (10 μΙ_) is added and cells are incubated for another 2 hours. 100 μΙ_ of 4% prewarmed paraformaldehyde is added, and cells are fixed for 45min at room temperature. After the removal of paraformaldehyde, 100 μΙ_ of 0.1 % Triton X-100 is added, and cells are incubated for extra 30 min at room temperature. After the cells are washed twice with 160 μΙ_ of PBS, 100 μΙ_ of blocking buffer (1 % BSA, in PBS) is added, and the cells are continued to incubate for 2-3 hours. Cells are washed with 160 μΙ_ of PBS again, treated with 30 μΙ_ of Ser473-p-Akt (Cell signaling, CAT: 4060) which is diluted in 0.1 % BSA at 1 :250, and incubated at 4°C overnight. Cells are then washed twice with 160 μΙ_ of PBS. 35 μΙ_ of Alexa Fluor 488 goat anti-rabbit IgG (Invitrogen, A1 1034), in a 1 :1000 dilution buffer (0.1 % BSA in PBS), is added, and the reaction mixture is incubated for 1 .5 hours in the dark. It is washed twice with 160 μΙ_ of PBS, and then 35 μΙ_ of 1 .5 μΜ propidium iodide (Sigma, P4170) is added to each well, and reaction plate is incubated at 37 °C, 5% CO2 for 30min. Finally, the plate is loaded into the Acumen eX3 (TTP LabTech) and scanned with the appropriate instrument settings. [0133] The inhibition of test compound is calculated based on the ratio of compound treated and untreated cells. IC50 is generated using XLfit 2.0 software.
Each compound specifically exemplified in the invention inhibited the PI3Ka kinase receptor with IC50 < 1 -0 μΜ.
Example 15
mTOR TR-FRET Assay
[0134] Compounds are tested using LanthaScreen TR-FRET Assay. The kinase reaction is completed in 384-well black plate (Corning, Cat. 3676). The final
conditions for kinase assay are: 10 μηηοΙ/L of ATP, 0.2 ng/ L of mTOR kinase, 0.4 μηηοΙ/L of GFP-4EBP1 substrate and assay buffer (50 mmol/L of HEPES,pH 7.5, 0.01 % of Tween 20, 1 mmol/L of EGTA, 10 mmol/L of MnCI2, 2 mmol/L of DTT and 1 % DMSO).
[0135] To each well, 2.5 μί of test compounds in 4% DMSO and 2.5 μί of 0.8 ng/μί mTOR kinase (Invitrogen, PV4753) diluted in assay buffer are added. The reaction is initiated by the addition of 5 μί mixture of 0.8 μηηοΙ/L GFP-4EBP1 Substrate
(Invitorgen, PV4759) and 20 μηηοΙ/L ATP mixtures. The mixture is incubated at room temperature for 60 minutes. 10 μΙ_ of 20 mmol/L EDTA and 4 nmol/L Tb-anti-p4EBP1 [pThr46] antibody (Invitrogen, PV4755) diluted in TR-FRET dilution buffer are added and incubated for an additional 1 hour. The plate is then read in a BioTek Synergy2 Reader at excitation of 340nm and emission of 490 nm and 528 nm.
[0136] The inhibition of test compound is calculated based on the ratio of 528 nm/490 nm. IC50 of test compound is obtained using XLfit 2.0 software.
[0137] Results: Above compounds 1 , 6, 7, 9, 10, 12, 14, 16, 17, 21 , 25, 26, 30, 33, 35, 42, 43, 44, 45, 46, 49, 50, 52, 53, 55, 56, 58, 63, 66, 72, 75, 88, 96, 98, 102, 103, 105, 106, 107, 1 19, 120, 121 , 122, 123, 129, 131 , 147,148,149,151 , 152, 153, 154, 155, 156, 157, 158, 159, 160, 165, 166, 178, 180, 181 inhibited mTOR with IC50 < 100 nM.
Example 16 P-S6 Acumen Assay
[0138] Compounds are tested using p-Akt Acumen cell-based assay. The human prostate cancer cell line PC3 (ATCC) is cultured in F-12 medium with 10% fetal bovine serum. For p-S6 Acumen assay, PC3 cells are seeded at density of 5000 cells/90 μΙ_ in Poly-D-Lysine 96-well plate (BD, 356692). After incubation for 24 hours, 10 μΙ_ of different concentration of test compounds is added and cells are incubated for 2 hours, followed by the addition of 100 μΙ_ of 4% pre-warmed paraformaldehyde. The cells are fixed at room temperature for 45min. After removal of the paraformaldehyde solution, 100 μΙ_ of 0.1 % Triton X-100 is added, and cells are incubated at room temperature for 30 min. After the cells are washed twice with 160 μΙ_ of PBS, 100 μΙ_ of blocking buffer (1 % BSA, in PBS) is added, and the cells are incubated for an additional 2-3 hours. Again, cells are washed with 160 μΙ_ of PBS, treated with 30 μΙ_ of p-S6 antibody (Cell signaling, CAT: 4858) which is diluted in 0.1 % BSA at 1 :250, and incubated at 4°C overnight. After the cells are washed twice with 160 μΙ_ of PBS, 35 μΙ_ of Alexa Fluor 488 goat anti-rabbit IgG (Invitrogen, A1 1034), in a 1 :1000 dilution buffer (0.1 % BSA in PBS) is added and the reaction mixture is incubated in the dark for 1 .5 hours. After washing twice with 160 μΙ_ of PBS, 35μΙ_ of 1 .5μΜ propidium iodide (Sigma, P4170) is added to each well and reaction plate is incubated at 37 °C, 5% CO2 for 30min. Finally, the plate is loaded into the Acumen eX3 (TTP LabTech) and scanned with the appropriate instrument settings.
[0139] The inhibition of the test compound is calculated based on the ratio of compound treated and untreated cells. IC50 is generated using XLfit 2.0 software.
Each compound specifically exemplified in the invention inhibited mTOR with IC50 < 10.0 μΜ.

Claims

CLAIMS What is claimed is:
1 . A compound of formula 1 :
Figure imgf000090_0001
(1 )
and/or at least one pharmaceutically acceptable salt thereof wherein
A1 is N or CH;
A4 and A5 are independently N or CR2;
A2 and A3, together with B ring, are a 5-membered heteroaryl or heterocycle containing 1 to 4 heteroatoms selected from N, O and S, and said 5-membered heteroaryl or heterocycle is optionally substituted by one or more groups
independently chosen from alkyl, alkenyl, alkynyl, aryl, cycloalkyi, oxo, -C(O)Ra, - C(O)ORb, -CN, -C(O)NRcRd, -NRcRd, -NRcC(O)Ra, -NRcS(O)nRe, -NRcS(O)nNRfRg, - NRcC(O)ORb, -NRcC(O)NRdRe, -NO2, -ORb, -S(O)nRe, -S(O)nNRcRd, halo, haloalkyl, heteroaryl, and heterocycle;
provided that A and A , together with the B ring, are not
Figure imgf000090_0002
;
is a single bond or a double bond;
R1 is heteroaryl, optionally substituted by one or more groups independently chosen from alkyl, alkenyl, alkynyl, aryl, cycloalkyi, oxo, -C(O)Ra, -C(O)ORb, -CN, - C(O)NRcRd, -NRcRd, -NRcC(O)Ra, -NRcS(O)nRe, -NRcS(O)nNRfRg, -NRcC(O)ORb, - NRcC(O)NRdRe, -NO2, -ORb, -S(O)nRe, -S(O)nNRcRd, halo, haloalkyl, heteroaryl, and heterocycle; R and R2 are independently chosen from H, alkyl, alkenyl, alkynyl, aryl, cycloalkyl, -C(O)Ra, -C(O)ORb, -CN, -C(O)NRcRd, -NRcRd, -NRcC(O)Ra, -NRcS(O)nRe, -NRcS(O)nNRfRg, -NRcC(O)ORb, -NRcC(O)NRdRe, -NO2, -ORb, -OC(O)Ra, - OC(O)NRcRd, -S(O)nRe, -S(O)nNRcRd, halo, haloalkyl, heteroaryl, and heterocycle; and each of said above alkyl, alkenyl, alkynyl, aryl, cycloalkyl, haloalkyl, heteroaryl and heterocycle can be optionally substituted by one or more groups independently chosen from optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted cycloalkyl, -OH, oxo, -C(O)Ra, -C(O)ORb, -CN, -C(O)NRcRd, -NRcRd, -NRcC(O)Ra, - NRcS(O)nRe, -NRcS(O)nNRfRg, -NRcC(O)ORb, -NRcC(O)NRdRe, -NO2, ORb, -S(O)nRe, - S(O)nNRcRd, halo, optionally substituted haloalkyl, optionally substituted heteroaryl, and optionally substituted heterocycle;
Ra, Rb, Rc, Rd, Re, Rf and Rg are each independently chosen from H, optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl optionally substituted aryl, optionally substituted cycloalkyl, optionally substituted haloalkyl, optionally substituted heteroaryl, and optionally substituted heterocycle,
or Ra and Rc, and/or Rc and Rd, and/or Rc and Re, and/or Rc and Rf, and/or Rd and Re, and /or Rg and Rf together with the atom(s) to which they are attached, form a 3-10 membered optionally substituted heterocycle ring; and
for each occurrence, n is independently 0, 1 , or 2.
2. The compound according to Claim 1 , wherein A2 and A3, together with B ring, are selected from structures (2)-(6)
Figure imgf000091_0001
(2) (3) (4) (5) wherein,
t is 1 , 2 or 3; and
R3 is independently chosen from H, Ci-C6 alkyl,C2-C8 alkenyl, C2-C8 alkynyl, C6-Ci4aryl, C3-C9 membered cydoalkyi, -C(O)ORb, -CN, -C(O)NRcRd, -NRcRd, - NRcC(O)Ra, -NRcS(O)nRe, -NRcS(O)nNRfRg, -NRcC(O)ORb, -NRcC(O)NRdRe, -NO2, - ORb, -OC(O)Ra, -OC(O)NRcRd, -S(O)nRe, -S(O)nNRcRd, halo, haloalkyi, heteroaryl, and heterocyde;
and each of said above alkyl, alkenyl, alkynyl, aryl, cydoalkyi, heteroaryl and heterocyde can be optionally substituted by one or more groups independently chosen from optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted cydoalkyi, -OH, oxo, -C(O)Ra, -C(O)ORb, -CN, -C(O)NRcRd, -NRcRd, -NRcC(O)Ra, -NRcS(O)nRe, - NRcS(O)nNRfRg, -NRcC(O)ORb, -NRcC(O)NRdRe, -NO2, ORb, -S(O)nRe, -S(O)nNRcRd, halo, optionally substituted haloalkyi, optionally substituted heteroaryl, and optionally substituted heterocyde;
provided that, when A2 and A3, together with B ring, are structure (4), A4 is not
CR2.
3. The compound according to Claim 2, wherein said R3 is chosen from H, OH, CN, NO2, halo, C1-C6 alkyl, C2-C8 alkenyl, C2-C8 alkynyl, aryl, C3-C9 cydoalkyi, heteroaryl, and heterocyde, wherein each of the alkyl, alkenyl, alkynyl, aryl, cydoalkyi, heteroaryl, and heterocyde is optionally substituted by one or more groups
independently chosen from optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted cydoalkyi, -OH, oxo, -C(O)Ra, -C(O)ORb, -CN, -C(O)NRcRd, -NRcRd, -NRcC(O)Ra, - NRcS(O)nRe, -NRcS(O)nNRfRg, -NRcC(O)ORb, -NRcC(O)NRdRe, -NO2, ORb, -S(O)nRe, - S(O)nNRcRd, halo, optionally substituted haloalkyl, optionally substituted heteroaryl, and optionally substituted heterocycle.
4. The compound according to any one of Claims 2 and 3, wherein A2 and A3, together with B ring, are chosen from the following structures (2)-(5)
Figure imgf000093_0001
(2) (3) (4) (5)
5. The compound according to any one of Claims 2-4, wherein A2 and A3, together with the B ring, are chosen from structures (3)-(4)
Figure imgf000093_0002
(3) (4)
6. The compound according to any one of Claims 1 -5, wherein A4 is N or CH.
7. The compound according to any one of Claims 1 -6, wherein A5 is N or CH.
8. The compound according to any one of Claims 1 -7, wherein A1, A4, and A5 are CH.
9. The compound according to any one of Claims 1 -7, wherein A1 and A5 are CH, and A4 is N.
10. The compound according to any one of Claims 1 -9, wherein R1 is a heteroaryl chosen from the following structures
Figure imgf000094_0001
wherein each of which is optionally substituted by one or more groups independently chosen from alkyl, alkenyl, alkynyl, aryl, cydoalkyi, haloalkyi, heteroaryl, heterocyde, oxo, -C(O)Ra, -C(O)ORb, -CN, -C(O)NRcRd, -NRcRd, - NRcC(O)Ra, -NRcS(O)nRe, -NRcS(O)nNRfRg, -NRcC(O)ORb, -NRcC(O)NRdRe, -NO2, - ORb, -OC(O)Ra, -OC(O)NRcRd, -S(O)nRe, -S(O)nNRcRd and halo, and wherein each of the alkyl, alkenyl, alkynyl, aryl, cydoalkyi, haloalkyi, heteroaryl and heterocyde is optionally substituted by one or more groups independently chosen from optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted cydoalkyi, -OH, oxo, -C(O)Ra, - C(O)ORb, -CN, -C(O)NRcRd, -NRcRd, -NRcC(O)Ra, -NRcS(O)nRe, -NRcS(O)nNRfRg, - NRcC(O)ORb, -NRcC(O)NRdRe, -NO2, ORb, -S(O)nRe, -S(O)nNRcRd, halo, optionally substituted haloalkyi, optionally substituted heteroaryl, and optionally substituted heterocyde;
wherein Ra, Rb, Rc, Rd, Re, Rf and Rg are each independently chosen from H, alkyl, aryl, cydoalkyi, haloalkyi, heteroaryl, and heterocyde, further wherein each of the alkyl, aryl, cydoalkyi, heteroaryl, and heterocyde in Ra, Rb, Rc, Rd, Re, Rf and Rg is optionally substituted by one or more substitutents independently selected from halo and alkyl.
1 1 . The compound according to Claim 10, wherein R1 is a heteroaryl chosen from
Figure imgf000095_0001
Figure imgf000095_0002
wherein each of which is optionally substituted by one or more groups independently chosen from alkyl, alkenyl, alkynyl, aryl, cydoalkyi, haloalkyi, heteroaryl, heterocyde, oxo, -C(O)Ra, -C(O)ORb, -CN, -C(O)NRcRd, -NRcRd, - NRcC(O)Ra, -NRcS(O)nRe, -NRcS(O)nNRfRg, -NRcC(O)ORb, -NRcC(O)NRdRe, -NO2, - ORb, -OC(O)Ra, -OC(O)NRcRd, -S(O)nRe, -S(O)nNRcRd and halo, and wherein each of the alkyl, alkenyl, alkynyl, aryl, cydoalkyi, haloalkyi, heteroaryl and heterocyde is optionally substituted by one or more groups independently chosen from optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted cycloalkyi, -OH, oxo, -C(O)Ra, - C(O)ORb, -CN, -C(O)NRcRd, -NRcRd, -NRcC(O)Ra, -NRcS(O)nRe, -NRcS(O)nNRfRg, - NRcC(O)ORb, -NRcC(O)NRdRe, -NO2, ORb, -S(O)nRe, -S(O)nNRcRd, halo, optionally substituted haloalkyi, optionally substituted heteroaryl, and optionally substituted heterocycle;
wherein Ra, Rb, Rc, Rd, Re, Rf and Rg are each independently chosen from H, alkyl, aryl, cycloalkyi, haloalkyi, heteroaryl, and heterocycle, further wherein each of the alkyl, aryl, cycloalkyi, heteroaryl, and heterocycle in Ra, Rb, Rc, Rd, Re, Rf and Rg is optionally substituted by one or more substitutents independently selected from halo and alkyl.
12. Thecompound according to any one of Claims 1 -1 1 , wherein R1 is
Figure imgf000096_0001
which is optionally substituted with one or more groups independently chosen from:
- alkyl, alkenyl, alkynyl, wherein each of which is optionally substituted by one or more groups independently chosen from optionally substituted alkyl, optionally substituted alkenyl, optionally substituted alkynyl, optionally substituted aryl, optionally substituted cycloalkyi, -OH, oxo, -C(O)Ra, -C(O)ORb, -CN, -C(O)NRcRd, -
NRcRd, -NRcC(O)Ra, -NRcS(O)nRe, -NRcS(O)nNRfRg, -NRcC(O)ORb, -NRcC(O)NRdRe,
-NO2, ORb, -S(O)nRe, -S(O)nNRcRd, halo, optionally substituted haloalkyi, optionally substituted heteroaryl, and optionally substituted heterocycle;
-C(O)NRcRd;
-NRcRd;
-ORb;
-halo;
-cyano;
-NRcS(O)nRe, wherein Ra, Rb, Rc, Rd, Re, Rf and Rg are each independently chosen from H, alkyi, aryl, cycloalkyi, haloalkyi, heteroaryl, and heterocycle, wherein each of the alkyi, aryl, cycloalkyi, heteroaryl, and heterocycle in Ra, Rb, Rc, Rd, Re, Rf and Rg is optionally substituted by one or more substitutents independently selected from halo and alkyi.
13. A compound selected from compounds 1 to 184 and/or at least one pharmaceutically acceptable salt.
14. A pharmaceutical composition comprising at least one compound and/or at least one pharmaceutically acceptable salt thereof according to any one of Claims 1 - 13 and at least one pharmaceutically acceptable carrier.
15. A method of inhibiting the activity of PI3K and/or mTOR comprising contacting the enzyme with an effective amount of at least one compound and/or at least one pharmaceutically acceptable salt thereof according to any one of Claims 1 -13.
16. A method of treating cancer responsive to inhibition of PI3K and/or mTOR comprising administering to a subject in need thereof an effective amount of at least one compound and/or at least one pharmaceutically acceptable salt thereof according to any one of Claims 1 -13.
17. A use of at least one compound and/or at least one pharmaceutically acceptable salt thereof according to any one of Claims 1 -13 in the manufacture of a medicament for inhibiting the activity of PI3K and/or mTOR.
18. A use of at least one compound and/or at least one pharmaceutically acceptable salt thereof according to any one of Claims 1 -13 in the manufacture of a medicament for treating cancer.
PCT/CN2011/079684 2010-09-16 2011-09-15 Fused heteroaryls and their uses WO2012034526A1 (en)

Priority Applications (13)

Application Number Priority Date Filing Date Title
EP11824596.8A EP2616468A4 (en) 2010-09-16 2011-09-15 Fused heteroaryls and their uses
MX2013002784A MX2013002784A (en) 2010-09-16 2011-09-15 Fused heteroaryls and their uses.
AU2011301518A AU2011301518B2 (en) 2010-09-16 2011-09-15 Fused heteroaryls and their uses
NZ60752711A NZ607527A (en) 2010-09-16 2011-09-15 Fused heteroaryls and their uses
US13/823,604 US9181264B2 (en) 2010-09-16 2011-09-15 Fused heteroaryls and their uses
KR1020137009341A KR20130087020A (en) 2010-09-16 2011-09-15 Fused heteroaryls and their uses
SG2013014162A SG188302A1 (en) 2010-09-16 2011-09-15 Fused heteroaryls and their uses
JP2013528507A JP5634609B2 (en) 2010-09-16 2011-09-15 Fused heteroaryls and uses thereof
RU2013108348/04A RU2552114C2 (en) 2010-09-16 2011-09-15 Condensed heteroaryls and their application
CN201180044818.8A CN103124731B (en) 2010-09-16 2011-09-15 The heteroaryl compound condensed and application thereof
BR112013005855A BR112013005855A2 (en) 2010-09-16 2011-09-15 fused heteroaryls and their uses
CA2810708A CA2810708A1 (en) 2010-09-16 2011-09-15 Fused heteroaryls and their uses
TW101107297A TW201311682A (en) 2011-09-15 2012-03-05 Fused heteroaryls and their uses

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2010076996 2010-09-16
CNPCT/CN2010/076996 2010-09-16

Publications (1)

Publication Number Publication Date
WO2012034526A1 true WO2012034526A1 (en) 2012-03-22

Family

ID=45831015

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2011/079684 WO2012034526A1 (en) 2010-09-16 2011-09-15 Fused heteroaryls and their uses

Country Status (12)

Country Link
US (1) US9181264B2 (en)
EP (1) EP2616468A4 (en)
JP (1) JP5634609B2 (en)
KR (1) KR20130087020A (en)
AU (1) AU2011301518B2 (en)
BR (1) BR112013005855A2 (en)
CA (1) CA2810708A1 (en)
MX (1) MX2013002784A (en)
NZ (1) NZ607527A (en)
RU (1) RU2552114C2 (en)
SG (1) SG188302A1 (en)
WO (1) WO2012034526A1 (en)

Cited By (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2648733A1 (en) * 2010-12-06 2013-10-16 Piramal Enterprises Limited Substituted imidazoquinoline derivatives
US9227969B2 (en) 2013-08-14 2016-01-05 Novartis Ag Compounds and compositions as inhibitors of MEK
JP2016508484A (en) * 2013-01-23 2016-03-22 アストラゼネカ アクチボラグ Compound
WO2017046675A1 (en) * 2015-09-14 2017-03-23 Pfizer Inc. Novel imidazo [4,5-c] quinoline and imidazo [4,5-c][1,5] naphthyridine derivatives as lrrk2 inhibitors
US9642855B2 (en) 2012-06-29 2017-05-09 Pfizer Inc. Substituted pyrrolo[2,3-d]pyrimidines as LRRK2 inhibitors
US9695171B2 (en) 2013-12-17 2017-07-04 Pfizer Inc. 3,4-disubstituted-1 H-pyrrolo[2,3-b]pyridines and 4,5-disubstituted-7H-pyrrolo[2,3-c]pyridazines as LRRK2 inhibitors
WO2018127195A1 (en) 2017-01-09 2018-07-12 上海瑛派药业有限公司 Substituted fused heteroaryl compound serving as a kinase inhibitor, and applications thereof
CN109810112A (en) * 2019-02-01 2019-05-28 华侨大学 A kind of preparation method of indoles simultaneously [2,1-a] phthalazine derivatives
CN109912606A (en) * 2019-04-16 2019-06-21 新乡医学院 A kind of synthetic method of pyrimido indazole compounds

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA2954999C (en) * 2014-07-14 2020-01-07 Advenchen Pharmaceuticals, Nanjing Ltd. Fused quinoline compounds as pi3k, mtor inhibitors
CN110156783B (en) * 2015-07-11 2021-09-07 南京爱德程医药科技有限公司 Pyrimidinyl substituted fused quinoline compounds as PI3K/MTOR inhibitors
KR20180095595A (en) * 2015-12-18 2018-08-27 노파르티스 아게 Tricyclic compounds and compositions as kinase inhibitors
KR102143320B1 (en) 2018-08-28 2020-08-12 주식회사 스템랩 METHODS FOR DIRECT CONVERTION OF HUMAN URINE CELLS INTO NEURAL STEM CELLS USING A SYNTHETIC mRNA

Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101330916A (en) * 2005-12-16 2008-12-24 科勒制药集团公司 Substituted imidazoquinolines, imidazonaphthyridines, and imidazopyridines, compositions, and methods
WO2009155527A2 (en) * 2008-06-19 2009-12-23 Progenics Pharmaceuticals, Inc. Phosphatidylinositol 3 kinase inhibitors
WO2010038165A1 (en) * 2008-09-30 2010-04-08 Pfizer Inc. Imidazo[1,5]naphthyridine compounds, their pharmaceutical use and compositions
US20100120763A1 (en) * 2008-11-07 2010-05-13 Wyeth Imidazo[5,1-c][1,2,4]benzotriazine derivatives as inhibitors of phosphodiesterases
US20100120762A1 (en) * 2008-11-07 2010-05-13 Wyeth Triazine derivatives as inhibitors of phosphodiesterases
CN101784547A (en) * 2007-08-14 2010-07-21 诺瓦提斯公司 Pyrazoloquinoline and pyrazolonaphthyridine derivatives as gaba alpha ligands
WO2011044157A1 (en) * 2009-10-06 2011-04-14 Biogen Idec Ma Inc. Heterocyclic compounds useful as pdk1 inhibitors

Family Cites Families (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
IN160956B (en) * 1982-10-18 1987-08-22 Pfizer
NZ292107A (en) * 1994-09-16 1999-02-25 Novo Nordisk As [1,2,4]triazolo[4,3-a]quinoxalinone derivatives and medicaments
PL1687305T3 (en) * 2003-11-21 2008-12-31 Novartis Ag 1h-imidazoquinoline derivatives as protein kinase inhibitors
AR046845A1 (en) * 2003-11-21 2005-12-28 Novartis Ag DERIVATIVES OF 1H-IMIDAZO [4,5-C] QUINOLINE FOR THE TREATMENT OF PROTEIN-KINASE DEPENDENT DISEASES
US20100078444A1 (en) 2008-09-30 2010-04-01 Thomas Gerard Shannon Compressed tissue product
TW201204727A (en) * 2010-03-10 2012-02-01 Kalypsys Inc Heterocyclic inhibitors of histamine receptors for the treatment of disease

Patent Citations (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101330916A (en) * 2005-12-16 2008-12-24 科勒制药集团公司 Substituted imidazoquinolines, imidazonaphthyridines, and imidazopyridines, compositions, and methods
CN101784547A (en) * 2007-08-14 2010-07-21 诺瓦提斯公司 Pyrazoloquinoline and pyrazolonaphthyridine derivatives as gaba alpha ligands
WO2009155527A2 (en) * 2008-06-19 2009-12-23 Progenics Pharmaceuticals, Inc. Phosphatidylinositol 3 kinase inhibitors
WO2010038165A1 (en) * 2008-09-30 2010-04-08 Pfizer Inc. Imidazo[1,5]naphthyridine compounds, their pharmaceutical use and compositions
US20100120763A1 (en) * 2008-11-07 2010-05-13 Wyeth Imidazo[5,1-c][1,2,4]benzotriazine derivatives as inhibitors of phosphodiesterases
US20100120762A1 (en) * 2008-11-07 2010-05-13 Wyeth Triazine derivatives as inhibitors of phosphodiesterases
WO2011044157A1 (en) * 2009-10-06 2011-04-14 Biogen Idec Ma Inc. Heterocyclic compounds useful as pdk1 inhibitors

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of EP2616468A4 *

Cited By (17)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2648733A1 (en) * 2010-12-06 2013-10-16 Piramal Enterprises Limited Substituted imidazoquinoline derivatives
US9642855B2 (en) 2012-06-29 2017-05-09 Pfizer Inc. Substituted pyrrolo[2,3-d]pyrimidines as LRRK2 inhibitors
JP2016508484A (en) * 2013-01-23 2016-03-22 アストラゼネカ アクチボラグ Compound
US10011599B2 (en) 2013-08-14 2018-07-03 Novartis Ag Compounds and compositions as inhibitors of MEK
US9227969B2 (en) 2013-08-14 2016-01-05 Novartis Ag Compounds and compositions as inhibitors of MEK
US9629836B2 (en) 2013-08-14 2017-04-25 Novartis Ag Compounds and compositions as inhibitors of MEK
US9695171B2 (en) 2013-12-17 2017-07-04 Pfizer Inc. 3,4-disubstituted-1 H-pyrrolo[2,3-b]pyridines and 4,5-disubstituted-7H-pyrrolo[2,3-c]pyridazines as LRRK2 inhibitors
US10039753B2 (en) 2015-09-14 2018-08-07 Pfizer Inc. Imidazo[4,5-c]quinoline and imidazo[4,5-c][1,5]naphthyridine derivatives as LRRK2 inhibitors
CN108137586A (en) * 2015-09-14 2018-06-08 辉瑞大药厂 As novel imidazo [4,5-c] quinoline of LRRK2 inhibitor and imidazo [4,5-c] [1,5] 7-naphthyridine derivatives
WO2017046675A1 (en) * 2015-09-14 2017-03-23 Pfizer Inc. Novel imidazo [4,5-c] quinoline and imidazo [4,5-c][1,5] naphthyridine derivatives as lrrk2 inhibitors
WO2018127195A1 (en) 2017-01-09 2018-07-12 上海瑛派药业有限公司 Substituted fused heteroaryl compound serving as a kinase inhibitor, and applications thereof
US10874670B2 (en) 2017-01-09 2020-12-29 Impact Therapeutics, Inc. Substituted fused heteroaromatic compounds as kinase inhibitors and the use thereof
AU2018206111B2 (en) * 2017-01-09 2022-07-21 Impact Therapeutics (Shanghai), Inc Substituted fused heteroaryl compound serving as a kinase inhibitor, and applications thereof
CN109810112A (en) * 2019-02-01 2019-05-28 华侨大学 A kind of preparation method of indoles simultaneously [2,1-a] phthalazine derivatives
CN109810112B (en) * 2019-02-01 2021-03-23 华侨大学 Preparation method of indolo [2,1-a ] phthalazine derivative
CN109912606A (en) * 2019-04-16 2019-06-21 新乡医学院 A kind of synthetic method of pyrimido indazole compounds
CN109912606B (en) * 2019-04-16 2021-05-04 新乡医学院 Synthesis method of pyrimido indazole compound

Also Published As

Publication number Publication date
US9181264B2 (en) 2015-11-10
EP2616468A4 (en) 2014-02-26
JP2013537204A (en) 2013-09-30
AU2011301518A1 (en) 2013-03-14
RU2013108348A (en) 2014-10-27
AU2011301518B2 (en) 2014-07-03
EP2616468A1 (en) 2013-07-24
BR112013005855A2 (en) 2016-05-17
RU2552114C2 (en) 2015-06-10
NZ607527A (en) 2015-04-24
SG188302A1 (en) 2013-04-30
KR20130087020A (en) 2013-08-05
JP5634609B2 (en) 2014-12-03
CA2810708A1 (en) 2012-03-22
MX2013002784A (en) 2013-04-24
US20130190307A1 (en) 2013-07-25

Similar Documents

Publication Publication Date Title
JP5634609B2 (en) Fused heteroaryls and uses thereof
JP6486999B2 (en) Specific triazolopyridines and triazolopyrazines, their compositions and methods of using them
AU2011291185C1 (en) Pyrrolopyrimidine compounds and uses thereof
WO2012022045A1 (en) Pyrrolopyrimidine compounds and uses thereof
WO2012167423A1 (en) Substituted pyridopyrazines as novel syk inhibitors
JP2021506979A (en) Aryl-bipyridineamine derivative as a phosphatidylinositol phosphate kinase inhibitor
JP2021506977A (en) Chromenopyridine derivative as a phosphatidylinositol phosphate kinase inhibitor
KR20140014104A (en) Substituted imidazoquinoline derivatives
CN103124731A (en) Fused heteroaryls and their uses
TW201311682A (en) Fused heteroaryls and their uses

Legal Events

Date Code Title Description
WWE Wipo information: entry into national phase

Ref document number: 201180044818.8

Country of ref document: CN

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

Ref document number: 11824596

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2810708

Country of ref document: CA

WWE Wipo information: entry into national phase

Ref document number: MX/A/2013/002784

Country of ref document: MX

ENP Entry into the national phase

Ref document number: 2013528507

Country of ref document: JP

Kind code of ref document: A

Ref document number: 2011301518

Country of ref document: AU

Date of ref document: 20110915

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 13823604

Country of ref document: US

WWE Wipo information: entry into national phase

Ref document number: 12013500500

Country of ref document: PH

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 20137009341

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2013108348

Country of ref document: RU

Kind code of ref document: A

WWE Wipo information: entry into national phase

Ref document number: 2011824596

Country of ref document: EP

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112013005855

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 112013005855

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20130312