WO2011163527A1 - Pyrrolo- pyrazine derivatives useful as inhibitors of atr kinase - Google Patents

Pyrrolo- pyrazine derivatives useful as inhibitors of atr kinase Download PDF

Info

Publication number
WO2011163527A1
WO2011163527A1 PCT/US2011/041705 US2011041705W WO2011163527A1 WO 2011163527 A1 WO2011163527 A1 WO 2011163527A1 US 2011041705 W US2011041705 W US 2011041705W WO 2011163527 A1 WO2011163527 A1 WO 2011163527A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
nitrogen
group
carcinoma
membered
Prior art date
Application number
PCT/US2011/041705
Other languages
French (fr)
Inventor
Jean-Damien Charrier
Joanne Pinder
Ronald Marcellus Alphonsus Knegtel
Steven John Durrant
Damien Fraysse
Somhairle Maccormick
Aniza Nizarali Virani
Philip Michael Reaper
Original Assignee
Vertex Pharmaceuticals Incorporated
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 Vertex Pharmaceuticals Incorporated filed Critical Vertex Pharmaceuticals Incorporated
Priority to NZ605627A priority Critical patent/NZ605627A/en
Priority to EP11729535.2A priority patent/EP2585468A1/en
Priority to CA2803802A priority patent/CA2803802A1/en
Priority to JP2013516779A priority patent/JP2013529643A/en
Priority to MX2013000103A priority patent/MX2013000103A/en
Priority to AU2011270807A priority patent/AU2011270807A1/en
Publication of WO2011163527A1 publication Critical patent/WO2011163527A1/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
    • C07D487/02Heterocyclic 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
    • 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
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/4985Pyrazines or piperazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N5/00Radiation therapy
    • A61N5/10X-ray therapy; Gamma-ray therapy; Particle-irradiation therapy
    • 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
    • C07D495/00Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms
    • C07D495/02Heterocyclic compounds containing in the condensed system at least one hetero ring having sulfur atoms as the only ring hetero atoms in which the condensed system contains two hetero rings
    • C07D495/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N5/00Radiation therapy
    • A61N5/10X-ray therapy; Gamma-ray therapy; Particle-irradiation therapy
    • A61N2005/1092Details
    • A61N2005/1098Enhancing the effect of the particle by an injected agent or implanted device

Definitions

  • ATR ATM and Rad3 related
  • ATM and Rad3 related kinase is a protein kinase involved in cellular responses to DNA damage.
  • ATR kinase acts with ATM ("ataxia telangiectasia mutated") kinase and many other proteins to regulate a cell's response to DNA damage, commonly referred to as the DNA Damage Response ("DDR").
  • DDR DNA Damage Response
  • the DDR stimulates DNA repair, promotes survival and stalls cell cycle progression by activating cell cycle checkpoints, which provide time for repair. Without the DDR, cells are much more sensitive to DNA damage and readily die from DNA lesions induced by endogenous cellular processes such as DNA replication or exogenous DNA damaging agents commonly used in cancer therapy.
  • Healthy cells can rely on a host of different proteins for DNA repair including the DDR kinase ATR. In some cases these proteins can compensate for one another by activating functionally redundant DNA repair processes. On the contrary, many cancer cells harbour defects in some of their DNA repair processes, such as ATM signaling, and therefore display a greater reliance on their remaining intact DNA repair proteins which include ATR.
  • ATR inhibitors may be useful for cancer treatment, either used alone or in combination with DNA damaging agents, because they shut down a DNA repair mechanism that is more important for cellular survival in many cancer cells than in healthy normal cells.
  • ATR peptide can be expressed and isolated using a variety of methods known in the literature (see e.g., Unsal-Kacmaz et al, PNAS 99: 10, pp6673-6678, May 14, 2002; see also Kumagai et al. Cell 124, pp943-955, March 10, 2006; Unsal-Kacmaz et al. Molecular and Cellular Biology. Feb 2004, pi 292- 1300; and Hall-Jackson et al. Oncogene 1999, 18, 6707-6713).
  • the present invention relates to pyrrolopyrazine compounds useful as inhibitors of ATR protein kinase.
  • the invention also relates to pharmaceutically acceptable compositions comprising the compounds of this invention; methods of treating of various diseases, disorders, and conditions using the compounds of this invention; processes for preparing the compounds of this invention; intermediates for the preparation of the compounds of this invention; and methods of using the compounds in in vitro applications, such as the study of kinases in biological and pathological phenomena; the study of intracellular signal transduction pathways mediated by such kinases; and the comparative evaluation of new kinase inhibitors.
  • These compounds have an unexpected ability to treat cancer as single agents. These compounds also show surprising synergy with other cancer agents, such as cisplatin, in combination therapies.
  • R 1 is hydrogen, Ci- 6 alkyl, or a 3-7 membered monocyclic fully saturated, partially
  • a 1 is a 5 -membered heteroaryl wherein X is carbon, nitrogen, oxygen, or sulfur; when X is nitrogen or carbon;
  • a 2 is phenyl or a 6-membered heteroaryl having 1-3 nitrogen atoms; A 2 is independently and optionally substituted with up to 2 occurrences of halo or CN;
  • a 3 is an 8-10 membered bicyclic heteroaromatic ring having 1-3 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur;
  • Z 1 is H, a Ci_ 6 aliphatic; wherein 0-2 methylene units of said Ci_ioaliphatic are optionally replaced with -NR.'-, -0-, -S-, C(O); or a 5-6 membered monocyclic aromatic ring having 0-3 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or an 8-10 membered bicyclic aromatic ring having 0-6 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or a Ci-ioaliphatic; wherein 0-4 methylene units of said Ci-ioaliphatic are optionally replaced with -NR'-, -0-, -S-, C(O); a C3_ 6 cycloalkyl, or a 3-6 membered heterocyclic ring having 1-2 heteroatoms selected from O, NR', or S; Z 1 is optionally substituted with 1-5 J 1 groups;
  • Z 2 is H, a 5-6 membered monocyclic aromatic ring having 0-3 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or an 8-10 membered bicyclic aromatic ring having 0-6 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or a Ci-ioaliphatic; wherein 0-4 methylene units of said Ci-ioaliphatic are optionally replaced with -NR'-, -0-, -S-, C(O), a C3_ 6 cycloalkyl, or a 3-6 membered heterocyclic ring having 1-2 heteroatoms selected from O, NR', or S; Z 2 is optionally substituted with 1-5 J 2 groups;
  • Z 3 is H, C3_ 6 cycloalkyl, halo, CN, NO 2 , or a Ci-ioaliphatic; wherein 0-4 methylene units of said Ci-ioaliphatic are optionally replaced with -NR'-, -0-, -S-, or C(O); Z 3 is optionally substituted with 1-5 J 3 groups;
  • Z 4 is a 5-6 membered monocyclic aromatic ring having 0-3 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or an 8-10 membered bicyclic aromatic ring having 0-6 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; Z 4 is optionally substituted with 1-5 J 4 groups; J is halo, CN, or V, or (V) t -R 2 ;
  • V is a Ci-ioaliphatic group wherein up to 3 methylene units are optionally replaced with O, NR", C(O), S, S(O), or S(0)2; wherein said Ci-ioaliphatic group is optionally substituted with 1-3 occcurences of halo or CN;
  • R 2 is 3-7 membered aromatic or nonaromatic monocyclic ring having 0-3 heteroatoms
  • R 2 is optionally substituted with 1-3 occurences of halo, CN, C3_ 6 cycloalkyl, or Ci-ioaliphatic; wherein up to 3 methylene units of said Ci-ioaliphatic are optionally replaced with NR', O, S, or CO; each J 1 , J 2 , and J 4 is independently halo, CN, NO2, or X 1 ;
  • J A is X A or X A -Q A ;
  • X A is a Ci- 6 aliphatic; wherein 0-4 methylene units of said Ci-ioaliphatic are optionally
  • Ci-6aliphatic is optionally substituted with halo or Ci_ 3 alkyl
  • Q A is phenyl
  • X 1 is a Ci_ 6 aliphatic; wherein 0-4 methylene units of said Ci-ioaliphatic are optionally
  • J X1 is halo or a 3-6 membered monocyclic ring having 0-2 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur;
  • J 3 is halo, CN, phenyl, a 4-6 membered heterocyclic ring having 1-2 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or a Ci- 6 aliphatic optionally substituted with 1-3 occurrences of halo;
  • each R' and R" is independently hydrogen or Ci_ 6 alkyl
  • t 0 or 1.
  • R 1 is not optionally substituted phenyl or optionally substituted pyrazinyl
  • R 1 is not optionally substituted cyclohexyl
  • R 1 is not an optionally substituted group selected from pyridinyl, morpholinyl, or piperazinyl; when A 2 is phenyl and R 1 is phenyl; R 1 is substituted with 4-S0 2 (Ci_ 6 alkyl) as shown in formula ii-a;
  • R 1 is hydrogen, Ci_ 6 alkyl, or a 3-7 membered monocyclic fully saturated, partially
  • R 1 is optionally substituted with 0-4 occurrences of J;
  • a 1 is a 5 -membered heteroaryl wherein X is carbon, nitrogen, oxygen, or sulfur;
  • a 2 is a 6-membered heteroaryl having 1-3 nitrogen atoms; A 2 is independently and optionally substituted with up to 2 occurrences of halo or CN;
  • a 3 is an 8-10 membered bicyclic heteroaromatic ring having 1-3 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur;
  • Z 1 is a 5-6 membered monocyclic aromatic ring having 0-3 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or an 8-10 membered bicyclic aromatic ring having 0-6 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or a Ci-ioaliphatic; wherein 0-4 methylene units of said Ci-ioaliphatic are optionally replaced with -NR'-, -0-, -S-, C(O); a C3_ 6 cycloalkyl, or a 3-6 membered heterocyclic ring having 1-2 heteroatoms selected from O, NR.', or S; Z 1 is optionally substituted with 1-5 J 1 groups;
  • Z 2 is hydrogen, a 5-6 membered monocyclic aromatic ring having 0-3 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or an 8-10 membered bicyclic aromatic ring having 0-6 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or a Ci_ioaliphatic; wherein 0-4 methylene units of said Ci-ioaliphatic are optionally replaced with -NR.'-, -0-, -S-, C(O), a C3_ 6 cycloalkyl, or a 3-6 membered heterocyclic ring having 1-2 heteroatoms selected from O, NR', or S; Z 2 is optionally substituted with 1-5 J 2 groups;
  • Z 3 is H, C 3 _ 6 cycloalkyl, halo, CN, NO 2 , or a Ci-ioaliphatic; wherein 0-4 methylene units of said Ci-ioaliphatic are optionally replaced with -NR'-, -0-, -S-, or C(O); Z 3 is optionally substituted with 1-5 J 3 groups;
  • Z 4 is a 5-6 membered monocyclic aromatic ring having 0-3 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or an 8-10 membered bicyclic aromatic ring having 0-6 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; Z 4 is optionally substituted with 1-5 J 4 groups;
  • J is halo, CN, or V, or (V) t -R 2 ;
  • V is a Ci-ioaliphatic group wherein up to 3 methylene units are optionally replaced with O, NR", C(O), S, S(O), or S(0) 2 ; wherein said Ci-ioaliphatic group is optionally substituted with 1-3 occcurences of halo or CN;
  • R 2 is 3-7 membered aromatic or nonaromatic monocyclic ring having 0-3 heteroatoms
  • R 2 is optionally substituted with 1-3 occurences of halo, CN, C 3 _ 6 cycloalkyl, or Ci-ioaliphatic; wherein up to 3 methylene units of said Ci-ioaliphatic are optionally replaced with NR', O, S, or CO; each J 1 , J 2 , and J 4 is independently halo, CN, NO 2 , or X 1 ;
  • X 1 is a Ci_ 6 aliphatic; wherein 0-4 methylene units of said Ci-ioaliphatic are optionally
  • J X1 is halo or a 3-6 membered monocyclic ring having 0-2 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur;
  • J 3 is halo, CN, a 4-6 membered heterocyclic ring having 1-2 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or a Ci_ 6 aliphatic optionally substituted with 1-3 occurrences of halo;
  • each R' and R" is independently hydrogen or Ci- 6 alkyl; t is 0 or 1.
  • a 2 is a 6-membered heteroaryl having 1-3 nitrogen atoms
  • Z 1 is a 5-6 membered monocyclic aromatic ring having 0-3 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or an 8-10 membered bicyclic aromatic ring having 0-6 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or a Ci-ioaliphatic; wherein 0-4 methylene units of said Ci-ioaliphatic are optionally replaced with -NR.'-, -0-, -S-, C(O); a C3_ 6 cycloalkyl, or a 3-6 membered heterocyclic ring having 1-2 heteroatoms selected from O, NR', or S; Z 1 is optionally substituted with 1-5 J 1 groups; and
  • J 3 is halo, CN, phenyl, a 4-6 membered heterocyclic ring having 1-2 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or a Ci_ 6 aliphatic optionally substituted with 1-3 occurrences of halo.
  • A is N-(0012] According to one embodiment, A is N-(0012]
  • a 1 is selected from the following: wherein X 1 is O, NR, or S; R is H or C 1-6 alkyl.
  • a 1 is selected from the following: or 3 ⁇ 4 X ' ⁇ some embodiments, X is O, NR, or S; wherein R is H or Ci- 6 alkyl. In other embodiments, A is . In yet other embodiments, A 1 is x ' ⁇ ⁇ i . i n some embodiments, X 1 is S. In other embodiments, X 1 is O.
  • a 1 groups can be bonded to the pyrrolopyrazine and Z 1 in at least two different ways.
  • X 1 is S or O. In other embodiments, X 1 is O.
  • Z 1 is a 5-6 membered aromatic ring.
  • Z 1 is phenyl.
  • Z 1 is optionally substituted with 1-2 J 1 groups.
  • J 1 is -CH 2 NHR' or CHCi_ 6 alkyl)NHR'.
  • R' is H (J 1 is -CH 2 NH 2 or CHCi_ 6 alkyl)NH 2 ).
  • Z 1 is COOH.
  • Z 1 is Ci- 6 alkyl.
  • a ome embodiments A 2 is a
  • a 2 is pyridinyl or pyrimidinyl.
  • a 2 is substituted with one occurrence of Z 2 .
  • the one occurrence of Z 2 is bonded at the 3 -position of the 6-membered ring.
  • a 2 is selected from the following:
  • a 2 is phenyl and Z 2 is CN, CH 2 OH, CH 2 OCH 3 , CONH 2 , CON(CH 3 ) 2 , OCH 3 , or tretrazolyl.
  • A In some embodiment,
  • a 3 is an 8-10 membered bicyclic heteroaromatic ring having 1-2 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur.
  • said heteroaromatic ring is benzoxazolyl, benzisoxazolyl, benzothiazolyl, benzimidazolyl, azaindolyl, indolyl, indazolyl, benzothienyl, benzofuranyl, dihydrothienodioxinyl, quinolinyl, or isoquinolinyl.
  • said heteroaromatic ring is benzoxazolyl, benzisoxazolyl, benzothiazolyl, benzimidazolyl, indolyl, indazolyl, benzothienyl, or benzofuranyl.
  • A is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
  • A is O, NR, or S; wherein R is H or Ci- 4 alkyl.
  • A is selected from the following:
  • Z 1 and Z 2 can be a Ci-ioaliphatic wherein 0-4 methylene units are optionally replaced with -NR.'-, -0-, -S-, C(O); a C3_ 6 cycloalkyl, or a 3-6 membered heterocyclic ring having 1-2 heteroatoms selected from O, N, or S. It shall be understood that when a methylene unit is replaced by a cyclic ring, such as a C3_ 6 cycloalkyl or a 3-6 membered heterocyclic ring, the cyclic ring can be attached to the methylene units of the aliphatic chain via any two points of attachment on the ring.
  • piperidine is replacing the first methylene unit of the C 4 aliphatic.
  • the piperidine ring is bonded to its surrounding atoms via two points of attachment on ring: a carbon atom and a nitrogen atom.
  • a C5 aliphatic wherein one methylene unit (the second one, in this case) is replaced with a 5-membered heterocyclyl having one nitrogen atom could look like this: .
  • the heterocyclic ring pyrrolidine
  • a C3 aliphatic wherein one methylene unit is replaced with a cyclohexyl could look like this:
  • the cyclohexyl ring has replaced the second methylene unit of the aliphatic.
  • the cyclohexyl ring is bonded to the surrounding methylene units of the aliphatic via two different carbon atoms of the cyclohexyl ring.
  • Z 4 is a 5-6 membered aromatic ring. In other embodiments, Z 4 is phenyl.
  • R 1 is Ci- 6 alkyl.
  • R 1 is a monocyclic ring.
  • R 1 is a 3-7 membered cycloaliphatic or heterocyclyl.
  • R 1 is a 5-6 membered aromatic ring having 0-4 heteroatoms independently selected from the group consisting of nitrogen, oxygen, and sulfur.
  • R 1 is phenyl, pyridinyl, or pyrimidinyl.
  • R 1 is phenyl.
  • R 1 is substituted by one to two occurrences of J. In some embodiments, R 1 is substituted in the para position with J. In certain embodiments, J is V. In some embodiments, V is a Ci_ 6 aliphatic group wherein one methylene unitis optionally replaced with S(0)2. In some embodiments, V is -S(0)2(Ci_4alkyl). In other embodiments, V is S(0) 2 CH(CH 3 ) 2 .
  • J is CN and substituted in the ortho position.
  • J is -(V) t -R 2 .
  • V is -S(0)2.
  • R 2 is a C3-7 cycloalkyl or a 3-7 membered heterocyclyl having 1-3 heteroatoms selected from the group consisting of oxygen, nitrogen, and sulfur.
  • R 1 is phenyl substituted in the para position with J, wherein J is -S(0)2(Ci_4alkyl).
  • Another embodiment provides a compound from the following Table:
  • the variables are as depicted in the compounds of the disclosure including compounds in the tables above.
  • a specified number range of atoms includes any integer therein.
  • a group having from 1-4 atoms could have 1, 2, 3, or 4 atoms.
  • compounds of the invention may optionally be substituted with one or more substituents, such as are illustrated generally herein, or as exemplified by particular classes, subclasses, and species of the invention. It will be appreciated that the phrase “optionally substituted” is used interchangeably with the phrase “substituted or unsubstituted.” In general, the term “substituted”, whether preceded by the term “optionally” or not, refers to the replacement of hydrogen radicals in a given structure with the radical of a specified substituent.
  • an optionally substituted group may have a substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position.
  • Combinations of substituents envisioned by this invention are preferably those that result in the formation of stable or chemically feasible compounds.
  • a substituent connected by a bond drawn from the center of a ring means that the substituent can be bonded to any position in the ring.
  • J 1 can be bonded to any position on the pyridyl ring.
  • a bond drawn through both rings indicates that the substituent can be bonded from any position of the bicyclic ring.
  • J 1 can be bonded to the 5-membered ring (on the nitrogen atom, for instance), and to the 6-membered ring.
  • stable refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, recovery, purification, and use for one or more of the purposes disclosed herein.
  • a stable compound or chemically feasible compound is one that is not substantially altered when kept at a temperature of 40°C or less, in the absence of moisture or other chemically reactive conditions, for at least a week.
  • aliphatic or "aliphatic group”, as used herein, means a straight-chain (i.e., unbranched), branched, or cyclic, substituted or unsubstituted hydrocarbon chain that is completely saturated or that contains one or more units of unsaturation that has a single point of attachment to the rest of the molecule.
  • aliphatic groups contain 1-20 aliphatic carbon atoms. In some embodiments, aliphatic groups contain 1-10 aliphatic carbon atoms. In other embodiments, aliphatic groups contain 1-8 aliphatic carbon atoms. In still other
  • aliphatic groups contain 1-6 aliphatic carbon atoms, and in yet other embodiments aliphatic groups contain 1-4 aliphatic carbon atoms.
  • Aliphatic groups may be linear or branched, substituted or unsubstituted alkyl, alkenyl, or alkynyl groups. Specific examples include, but are not limited to, methyl, ethyl, isopropyl, n-propyl, sec -butyl, vinyl, n-butenyl, ethynyl, and tert-butyl.
  • Aliphatic groups may also be cyclic, or have a combination of linear or branched and cyclic groups.
  • Examples of such types of aliphatic groups include, but are not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, -CH 2 - cyclopropyl, CH 2 CH 2 CH(CH 3 )-cyclohexyl.
  • cycloaliphatic refers to a monocyclic C3-C8 hydrocarbon or bicyclic C 8 -C 12 hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic, that has a single point of attachment to the rest of the molecule wherein any individual ring in said bicyclic ring system has 3-7 members.
  • cycloaliphatic groups include, but are not limited to, cycloalkyl and cycloalkenyl groups. Specific examples include, but are not limited to, cyclohexyl, cyclopropenyl, and cyclobutyl.
  • heterocycle means non-aromatic, monocyclic, bicyclic, or tricyclic ring systems in which one or more ring members are an independently selected heteroatom.
  • heterocycle has three to fourteen ring members in which one or more ring members is a heteroatom independently selected from oxygen, sulfur, nitrogen, or phosphorus, and each ring in the system contains 3 to 7 ring members.
  • heterocycles include, but are not limited to, 3-lH-benzimidazol-2- one, 3-(l-alkyl)-benzimidazol-2-one, 2-tetrahydrofuranyl, 3-tetrahydrofuranyl, 2- tetrahydrothiophenyl, 3-tetrahydrothiophenyl, 2-morpholino, 3-morpholino, 4-morpholino, 2- thiomorpholino, 3-thiomorpholino, 4-thiomorpholino, 1 -pyrrolidinyl, 2-pyrrolidinyl, 3- pyrrolidinyl, 1 -tetrahydropiperazinyl, 2-tetrahydropiperazinyl, 3-tetrahydropiperazinyl, 1- piperidinyl, 2-piperidinyl, 3-piperidinyl, 1-pyrazolinyl, 3-pyrazolinyl, 4-pyrazolinyl, 5- pyrazolinyl, 1 -piperidin
  • Cyclic groups (e.g. cycloaliphatic and heterocycles), can be linearly fused, bridged, or spirocyclic.
  • heteroatom means one or more of oxygen, sulfur, nitrogen, phosphorus, or silicon (including, any oxidized form of nitrogen, sulfur, phosphorus, or silicon; the quaternized form of any basic nitrogen or; a substitutable nitrogen of a heterocyclic ring, for example N (as in 3,4-dihydro-2H-pyrrolyl), ⁇ (as in pyrrolidinyl) or NR + (as in N-substituted pyrrolidinyl)).
  • unsaturated as used herein, means that a moiety has one or more units of unsaturation.
  • unsaturated groups can be partially unsaturated or fully unsaturated.
  • partially unsaturated groups include, but are not limited to, butene, cyclohexene, and tetrahydropyridine.
  • Fully unsaturated groups can be aromatic, anti-aromatic, or non-aromatic. Examples of fully unsaturated groups include, but are not limited to, phenyl, cyclooctatetraene, pyridyl, thienyl, and 1- methylpyridin-2(lH)-one.
  • alkoxy refers to an alkyl group, as previously defined, attached through an oxygen (“alkoxy”) or sulfur (“thioalkyl”) atom.
  • haloalkyl mean alkyl, alkenyl or alkoxy, as the case may be, substituted with one or more halogen atoms.
  • This term includes perfluorinated alkyl groups, such as -CF 3 and -CF 2 CF 3 .
  • halogen means F, CI, Br, or I.
  • aralkoxy refers to monocyclic, bicyclic, and tricyclic ring systems having a total of five to fourteen ring members, wherein at least one ring in the system is aromatic and wherein each ring in the system contains 3 to 7 ring members.
  • aryl may be used interchangeably with the term “aryl ring”.
  • heteroaryl used alone or as part of a larger moiety as in
  • heteroarylkyl refers to monocyclic, bicyclic, and tricyclic ring systems having a total of five to fourteen ring members, wherein at least one ring in the system is aromatic, at least one ring in the system contains one or more heteroatoms, and wherein each ring in the system contains 3 to 7 ring members.
  • heteroaryl may be used interchangeably with the term “heteroaryl ring” or the term “heteroaromatic".
  • heteroaryl rings include, but are not limited to, 2-furanyl, 3-furanyl, N-imidazolyl, 2- imidazolyl, 4-imidazolyl, 5-imidazolyl, benzimidazolyl, 3-isoxazolyl, 4-isoxazolyl, 5- isoxazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, N-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, pyridazinyl (e.g., 3- pyridazinyl), 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, tetrazolyl (e.g., 5-tetrazolyl), triazolyl (e.g., 2-triazolyl and 5-triazolyl), 2-thien
  • heteroaryl includes certain types of heteroaryl rings that exist in equilibrium between two different forms. More specifically, for example, species such hydropyridine and pyridinone (and likewise hydroxypyrimidine and pyrimidinone) are meant to be encompassed within the definition of "heteroaryl.”
  • a protecting group and “protective group” as used herein, are interchangeable and refer to an agent used to temporarily block one or more desired functional groups in a compound with multiple reactive sites.
  • a protecting group has one or more, or preferably all, of the following characteristics: a) is added selectively to a functional group in good yield to give a protected substrate that is b) stable to reactions occurring at one or more of the other reactive sites; and c) is selectively removable in good yield by reagents that do not attack the regenerated, deprotected functional group. As would be understood by one skilled in the art, in some cases, the reagents do not attack other reactive groups in the compound.
  • the reagents may also react with other reactive groups in the compound.
  • protecting groups are detailed in Greene, T.W., Wuts, P. G in "Protective Groups in Organic Synthesis", Third Edition, John Wiley & Sons, New York: 1999 (and other editions of the book), the entire contents of which are hereby incorporated by reference.
  • nitrogen protecting group refers to an agent used to temporarily block one or more desired nitrogen reactive sites in a multifunctional compound.
  • Preferred nitrogen protecting groups also possess the characteristics exemplified for a protecting group above, and certain exemplary nitrogen protecting groups are also detailed in Chapter 7 in Greene, T.W., Wuts, P. G in "Protective Groups in Organic Synthesis", Third Edition, John Wiley & Sons, New York: 1999, the entire contents of which are hereby incorporated by reference.
  • a methylene unit of an alkyl or aliphatic chain is optionally replaced with another atom or group.
  • R is, for example, H or Ci_ 6 aliphatic.
  • R is, for example, H or Ci_ 6 aliphatic.
  • R is, for example, H or Ci_ 6 aliphatic.
  • R is, for example, H or Ci_ 6 aliphatic.
  • R is, for example, H or Ci_ 6 aliphatic.
  • R is, for example, H or Ci_ 6 aliphatic.
  • R is, for example, H or Ci_ 6 aliphatic.
  • methylene unit can also refer to branched or substituted methylene units.
  • a nitrogen atom e.g. NR
  • dimethylamine e.g. N(CH 3 ) 2 .
  • the optional replacements form a chemically stable compound.
  • Optional replacements can occur both within the chain and/or at either end of the chain; i.e. both at the point of attachment and/or also at the terminal end.
  • Two optional replacements can also be adjacent to each other within a chain so long as it results in a chemically stable compound.
  • the optional replacements can also completely replace all of the carbon atoms in a chain.
  • a C 3 aliphatic can be optionally replaced by -NR-, -C(O)-, and -NR- to form -NRC(0)NR- (a urea).
  • the replacement atom is bound to a hydrogen atom on the terminal end.
  • the resulting compound could be -OCH 2 CH 3 , -CH 2 OCH 3 , or -CH 2 CH 2 OH.
  • structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, geometric, conformational, and rotational) forms of the structure.
  • isomeric e.g., enantiomeric, diastereomeric, geometric, conformational, and rotational
  • the R and S configurations for each asymmetric center, (Z) and (E) double bond isomers, and (Z) and (E) conformational isomers are included in this invention.
  • a substituent can freely rotate around any rotatable bonds.
  • structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms.
  • compounds having the present structures except for the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13 C- or 14 C-enriched carbon are within the scope of this invention.
  • Such compounds are useful, for example, as analytical tools or probes in biological assays.
  • the compounds of this invention can exist in free form for treatment, or where appropriate, as a pharmaceutically acceptable salt.
  • a "pharmaceutically acceptable salt” means any non-toxic salt of a compound of this invention that, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this invention or an inhibitorily active metabolite or residue thereof.
  • inhibitorily active metabolite or residue thereof means that a metabolite or residue thereof is also an inhibitor of the ATR protein kinase.
  • Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge et ah, describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 1977, 66, 1-19, incorporated herein by reference.
  • Pharmaceutically acceptable salts of the compounds of this invention include those derived from suitable inorganic and organic acids and bases. These salts can be prepared in situ during the final isolation and purification of the compounds. Acid addition salts can be prepared by 1) reacting the purified compound in its free-based form with a suitable organic or inorganic acid and 2) isolating the salt thus formed.
  • Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange.
  • inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid
  • organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange.
  • salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, glycolate, gluconate, glycolate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, ox
  • Base addition salts can be prepared by 1) reacting the purified compound in its acid form with a suitable organic or inorganic base and 2) isolating the salt thus formed.
  • Salts derived from appropriate bases include alkali metal (e.g., sodium, lithium, and potassium), alkaline earth metal (e.g., magnesium and calcium), ammonium and + (Ci_ 4 alkyl) 4 salts.
  • alkali metal e.g., sodium, lithium, and potassium
  • alkaline earth metal e.g., magnesium and calcium
  • ammonium and + (Ci_ 4 alkyl) 4 salts e.g., sodium, lithium, and potassium
  • alkaline earth metal e.g., magnesium and calcium
  • ammonium and + (Ci_ 4 alkyl) 4 salts e.g., sodium, lithium, and potassium
  • ammonium and + (Ci_ 4 alkyl) 4 salts e.g., sodium, lithium, and potassium
  • Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, loweralkyl sulfonate and aryl sulfonate.
  • Other acids and bases while not in themselves pharmaceutically acceptable, may be employed in the preparation of salts useful as intermediates in obtaining the compounds of the invention and their pharmaceutically acceptable acid or base addition salts.
  • One aspect of this invention provides compounds that are inhibitors of ATR kinase, and thus are useful for treating or lessening the severity of a disease, condition, or disorder where ATR is implicated in the disease, condition, or disorder.
  • Another aspect of this invention provides compounds that are useful for the treatment of diseases, disorders, and conditions characterized by excessive or abnormal cell proliferation.
  • diseases include, a proliferative or hyperproliferative disease.
  • proliferative and hyperproliferative diseases include, without limitation, cancer and myeloproliferative disorders.
  • said compounds are selected from the group consisting of a compound of formula I.
  • cancer includes, but is not limited to the following cancers. Oral: buccal cavity, lip, tongue, mouth, pharynx; Cardiac: sarcoma (angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma), myxoma, rhabdomyoma, fibroma, lipoma and teratoma; Lung: bronchogenic carcinoma (squamous cell or epidermoid, undifferentiated small cell, undifferentiated large cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hamartoma, mesothelioma;
  • Gastrointestinal esophagus (squamous cell carcinoma, larynx, adenocarcinoma,
  • leiomyosarcoma lymphoma
  • stomach carcinoma, lymphoma, leiomyosarcoma
  • pancreas ductal adenocarcinoma, insulinoma, glucagonoma, gastrinoma, carcinoid tumors, vipoma), small bowel or small intestines (adenocarcinoma, lymphoma, carcinoid tumors, Karposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, fibroma), large bowel or large intestines (adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, leiomyoma), colon, colon-rectum, colorectal; rectum, Genitourinary tract: kidney (adenocarcinoma, Wilm's tumor [nephroblastoma], lymphoma, leukemia), bladder
  • uterus endometrial carcinoma
  • cervix cervical carcinoma, pre-tumor cervical dysplasia
  • ovaries ovarian carcinoma [serous cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma], granulosa-thecal cell tumors, Sertoli-Leydig cell tumors, dysgerminoma, malignant teratoma), vulva (squamous cell carcinoma, intraepithelial carcinoma,
  • adenocarcinoma, fibrosarcoma, melanoma), vagina clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma (embryonal rhabdomyosarcoma), fallopian tubes (carcinoma), breast; Hematologic: blood (myeloid leukemia [acute and chronic], acute lymphoblastic leukemia, chronic lymphocytic leukemia, myeloproliferative diseases, multiple myeloma, myelodysplastic syndrome), Hodgkin's disease, non-Hodgkin's lymphoma [malignant lymphoma] hairy cell; lymphoid disorders; Skin: malignant melanoma, basal cell carcinoma, squamous cell carcinoma, Karposi's sarcoma, keratoacanthoma, moles dysplastic nevi, lipoma, angioma, dermatofibroma, keloids, psorias
  • cancer includes a cell afflicted by any one of the above-identified conditions.
  • the cancer is selected from colorectal, thyroid, or breast cancer.
  • myeloproliferative disorders includes disorders such as polycythemia vera, thrombocythemia, myeloid metaplasia with myelofibrosis, hypereosinophilic syndrome, juvenile myelomonocytic leukemia, systemic mast cell disease, and hematopoietic disorders, in particular, acute-myelogenous leukemia (AML), chronic-myelogenous leukemia (CML), acute-promyelocytic leukemia (APL), and acute lymphocytic leukemia (ALL).
  • AML acute-myelogenous leukemia
  • CML chronic-myelogenous leukemia
  • APL acute-promyelocytic leukemia
  • ALL acute lymphocytic leukemia
  • compositions to treat or prevent the herein identified disorders.
  • the compounds of this invention can also exist as pharmaceutically acceptable derivatives.
  • a "pharmaceutically acceptable derivative” is an adduct or derivative which, upon administration to a patient in need, is capable of providing, directly or indirectly, a compound as otherwise described herein, or a metabolite or residue thereof.
  • pharmaceutically acceptable derivatives include, but are not limited to, esters and salts of such esters.
  • a "pharmaceutically acceptable derivative or prodrug” means any one of
  • ester, salt of an ester or other derivative or salt thereof of a compound, of this invention which, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this invention or an inhibitorily active metabolite or residue thereof.
  • Particularly favoured derivatives or prodrugs are those that increase the bioavailability of the compounds of this invention when such compounds are administered to a patient (e.g., by allowing an orally administered compound to be more readily absorbed into the blood) or which enhance delivery of the parent compound to a biological compartment (e.g., the brain or lymphatic system) relative to the parent species.
  • compositions of this invention include, without limitation, esters, amino acid esters, phosphate esters, metal salts and sulfonate esters.
  • the present invention also provides compounds and compositions that are useful as inhibitors of ATR kinase.
  • compositions that comprise any of the compounds as described herein, and optionally comprise a pharmaceutically acceptable carrier, adjuvant or vehicle.
  • the pharmaceutically acceptable carrier, adjuvant, or vehicle includes any and all solvents, diluents, or other liquid vehicle, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired.
  • Remington's Pharmaceutical Sciences, Sixteenth Edition, E. W. Martin (Mack Publishing Co., Easton, Pa., 1980) discloses various carriers used in formulating pharmaceutically acceptable
  • compositions and known techniques for the preparation thereof are contemplated to be within the scope of this invention.
  • any conventional carrier medium is incompatible with the compounds of the invention, such as by producing any undesirable biological effect or otherwise interacting in a deleterious manner with any other component(s) of the pharmaceutically acceptable composition, its use is contemplated to be within the scope of this invention.
  • Some examples of materials which can serve as pharmaceutically acceptable carriers include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, or potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, polyacrylates, waxes, polyethylene-polyoxypropylene- block polymers, wool fat, sugars such as lactose, glucose and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin
  • Another aspect of this invention is directed towards a method of treating cancer in a subject in need thereof, comprising administration of a compound of this invention or a pharmaceutically acceptable salt thereof, and an additional therapeutic agent.
  • said method comprises the sequential or co-administration of the compound or a pharmaceutically acceptable salt thereof, and the additional therapeutic agent.
  • said additional therapeutic agent is an anti-cancer agent.
  • said additional therapeutic agent is a DNA-damaging agent.
  • said additional therapeutic agent is selected from radiation therapy, chemotherapy, or other agents typically used in combination with radiation therapy or chemotherapy, such as radios ens itizers and chemosensitizers.
  • radiosensitizers are agents that can be used in combination with radiation therapy. Radiosensitizers work in various different ways, including, but not limited to, making cancer cells more sensitive to radiation therapy, working in synergy with radiation therapy to provide an improved synergistic effect, acting additively with radiation therapy, or protecting surrounding healthy cells from damage caused by radiation therapy. Likewise chemosensitizers are agents that can be used in combination with chemotherapy. Similarly, chemosensitizers work in various different ways, including, but not limited to, making cancer cells more sensitive to chemotherapy, working in synergy with chemotherapy to provide an improved synergistic effect, acting additively to
  • DNA-damaging agents that may be used in combination with compounds of this invention include, but are not limited to Platinating agents, such as Carboplatin, Nedaplatin, Satraplatin and other derivatives; Topo I inhibitors, such as
  • Topotecan irinotecan/SN38, rubitecan and other derivatives
  • Antimetabolites such as Folic family (Methotrexate, Pemetrexed and relatives); Purine antagonists and Pyrimidine antagonists (Thioguanine, Fludarabine, Cladribine, Cytarabine, Gemcitabine,
  • Alkylating agents such as Nitrogen mustards (Cyclophosphamide, Melphalan, Chlorambucil, mechlorethamine, Ifosfamide and relatives); nitrosoureas (eg Carmustine); Triazenes (Dacarbazine, temozolomide); Alkyl s
  • therapies or anticancer agents that may be used in combination with the inventive agents of the present invention include surgery, radiotherapy (in but a few examples, gamma-radiation, neutron beam radiotherapy, electron beam radiotherapy, proton therapy, brachytherapy, and systemic radioactive isotopes, to name a few), endocrine therapy, biologic response modifiers (interferons, interleukins, and tumor necrosis factor (TNF) to name a few), hyperthermia and cryotherapy, agents to attenuate any adverse effects (e.g., antiemetics), and other approved chemotherapeutic drugs, including, but not limited to, the DNA damaging agents listed herein, spindle poisons (Vinblastine, Vincristine, Vinorelbine, Paclitaxel), podophyllotoxins (Etoposide, Irinotecan, Topotecan), nitrosoureas (Carmustine, Lomustine), inorganic ions (Cisplatin, Carbop
  • a compound of the instant invention may also be useful for treating cancer in combination with any of the following therapeutic agents: abarelix (Plenaxis depot®);
  • aldesleukin Prokine®
  • Aldesleukin Proleukin
  • Alemtuzumabb Pierath®
  • alitretinoin Panretin®
  • allopurinol Zayloprim®
  • altretamine Hexalen®
  • amifostine Ethyol®
  • anastrozole (Arimidex®); arsenic trioxide (Trisenox®); asparaginase (Elspar®); azacitidine (Vidaza®); bevacuzimab (Avastin®); bexarotene capsules (Targretin®); bexarotene gel (Targretin®); bleomycin (Blenoxane®); bortezomib (Velcade®); busulfan intravenous (Busulfex®); busulfan oral (Myleran®); calusterone (Methosarb®); capecitabine (Xeloda®); carboplatin (Paraplatin®); carmustine (BCNU®, BiCNU®); carmustine (Gliadel®);
  • cetuximab (Erbitux®); chlorambucil (Leukeran®); cisplatin (Platinol®); cladribine
  • dactinomycin actinomycin D (Cosmegen®); Darbepoetin alfa (Aranesp®); daunorubicin liposomal (DanuoXome®); daunorubicin, daunomycin (Daunorubicin®); daunorubicin, daunomycin (Cerubidine®); Denileukin diftitox (Ontak®); dexrazoxane (Zinecard®);
  • docetaxel Taxotere®
  • doxorubicin Adriamycin PFS®
  • doxorubicin doxorubicin
  • Rubex® doxorubicin
  • doxorubicin liposomal Doxil®
  • dromostanolone propionate dromostanolone®
  • dromostanolone propionate masterone injection®
  • Elliott's B Solution Elliott's B Solution®
  • epirubicin Ellence®
  • Epoetin alfa epogen®
  • erlotinib Tarceva®
  • estramustine Emcyt®
  • Nepogen® floxuridine (intraarterial) (FUDR®); fludarabine (Fludara®); fluorouracil, 5- FU (Adrucil®); fulvestrant (Faslodex®); gefitinib (Iressa®); gemcitabine (Gemzar®); gemtuzumab ozogamicin (Mylotarg®); goserelin acetate (Zoladex Implant®); goserelin acetate (Zoladex®); histrelin acetate (Histrelin implant®); hydroxyurea (Hydrea®);
  • Ibritumomab Tiuxetan Zevalin®; idarubicin (Idamycin®); ifosfamide (IFEX®); imatinib mesylate (Gleevec®); interferon alfa 2a (Roferon A®); Interferon alfa-2b (Intron A®); irinotecan (Camptosar®); lenalidomide (Revlimid®); letrozole (Femara®); leucovorin (Wellcovorin®, Leucovorin®); Leuprolide Acetate (Eligard®); levamisole (Ergamisol®); lomustine, CCNU (CeeBU®); meclorethamine, nitrogen mustard (Mustargen®); megestrol acetate (Megace®); melphalan, L-PAM (Alkeran®); mercaptopurine, 6-MP (Purinethol®); mesna (Mesnex
  • Nofetumomab (Verluma®); Oprelvekin (Neumega®); oxaliplatin (Eloxatin®); paclitaxel (Paxene®); paclitaxel (Taxol®); paclitaxel protein-bound particles (Abraxane®); palifermin (Kepivance®); pamidronate (Aredia®); pegademase (Adagen (Pegademase Bovine)®); pegaspargase (Oncaspar®); Pegfilgrastim (Neulasta®); pemetrexed disodium (Alimta®); pentostatin (Nipent®); pipobroman (Vercyte®); plicamycin, mithramycin (Mithracin®); porfimer sodium (Photofrin®); procarbazine (Matulane®); quinacrine (Atabrine®);
  • Rasburicase Elitek®; Rituximab (Rituxan®); sargramostim (Leukine®); Sargramostim (Prokine®); sorafenib (Nexavar®); streptozocin (Zanosar®); sunitinib maleate (Sutent®); talc (Sclerosol®); tamoxifen (Nolvadex®); temozolomide (Temodar®); teniposide, VM-26 (Vumon®); testolactone (Teslac®); thioguanine, 6-TG (Thioguanine®); thiotepa
  • Thioplex® topotecan (Hycamtin®); toremifene (Fareston®); Tositumomab (Bexxar®); Tositumomab/I-131 tositumomab (Bexxar®); Trastuzumab (Herceptin®); tretinoin, ATRA (Vesanoid®); Uracil Mustard (Uracil Mustard Capsules®); valrubicin (Valstar®); vinblastine (Velban®); vincristine (Oncovin®); vinorelbine (Navelbine®); zoledronate (Zometa®) and vorinostat (Zolinza®).
  • the ATR kinase inhibitors or pharmaceutical salts thereof may be formulated into pharmaceutical compositions for administration to animals or humans.
  • These pharmaceutical compositions which comprise an amount of the ATR inhibitor effective to treat or prevent the diseases or conditions described herein and a pharmaceutically acceptable carrier, are another embodiment of the present invention.
  • the exact amount of compound required for treatment will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the infection, the particular agent, its mode of administration, and the like.
  • the compounds of the invention are preferably formulated in dosage unit form for ease of administration and uniformity of dosage.
  • dosage unit form refers to a physically discrete unit of agent appropriate for the patient to be treated. It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment.
  • the specific effective dose level for any particular patient or organism will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed, and like factors well known in the medical arts.
  • patient means an animal, preferably a mammal, and most preferably a human.
  • these compositions optionally further comprise one or more additional therapeutic agents.
  • additional therapeutic agents chemotherapeutic agents or other antiproliferative agents may be combined with the compounds of this invention to treat proliferative diseases and cancer. Examples of known agents with which these compositions can be combined are listed above under the "Combination Therapies" section and also throughout the specification. Some embodiments provide a simultaneous, separate or sequential use of a combined preparation.
  • compositions of this invention can be administered to humans and other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), bucally, as an oral or nasal spray, or the like, depending on the severity of the infection being treated.
  • the compounds of the invention may be administered orally or parenterally at dosage levels of about 0.01 mg/kg to about 50 mg/kg and preferably from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect.
  • Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs.
  • the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.
  • the oral compositions can also include adj
  • Injectable preparations for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol.
  • the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S. P. and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil can be employed including synthetic mono- or diglycerides.
  • fatty acids such as oleic acid are used in the preparation of injectables.
  • the injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
  • a compound of the present invention In order to prolong the effect of a compound of the present invention, it is often desirable to slow the absorption of the compound from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the compound then depends upon its rate of dissolution that, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered compound form is accomplished by dissolving or suspending the compound in an oil vehicle. Injectable depot forms are made by forming microencapsule matrices of the compound in biodegradable polymers such as polylactide-polyglycolide.
  • the rate of compound release can be controlled.
  • biodegradable polymers include poly(orthoesters) and poly(anhydrides).
  • Depot injectable formulations are also prepared by entrapping the compound in liposomes or microemulsions that are compatible with body tissues.
  • compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non- irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
  • suitable non- irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
  • Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules.
  • the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar— agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl
  • Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polethylene glycols and the like.
  • the active compounds can also be in microencapsulated form with one or more excipients as noted above.
  • the solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art.
  • the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch.
  • Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose.
  • the dosage forms may also comprise buffering agents. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner.
  • buffering agents include polymeric substances and waxes.
  • Dosage forms for topical or transdermal administration of a compound of this invention include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches.
  • the active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required.
  • Ophthalmic formulation, eardrops, and eye drops are also contemplated as being within the scope of this invention.
  • the present invention contemplates the use of transdermal patches, which have the added advantage of providing controlled delivery of a compound to the body.
  • Such dosage forms can be made by dissolving or dispensing the compound in the proper medium.
  • Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.
  • compositions of the present invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir.
  • parenteral as used herein includes, but is not limited to, subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques.
  • Sterile injectable forms of the compositions of this invention may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents.
  • the sterile injectable preparation may also be a sterile injectable solution or suspension in a nontoxic parenterally-acceptable diluent or solvent, for example as a solution in 1,3-butanediol.
  • the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution.
  • sterile, fixed oils are conventionally employed as a solvent or suspending medium.
  • any bland fixed oil may be employed including synthetic mono- or di-glycerides.
  • Fatty acids such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions.
  • These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersing agents which are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions.
  • Other commonly used surfactants such as Tweens, Spans and other emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation.
  • compositions of this invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions.
  • carriers commonly used include, but are not limited to, lactose and corn starch.
  • Lubricating agents such as magnesium stearate, are also typically added.
  • useful diluents include lactose and dried cornstarch.
  • aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added.
  • compositions of this invention may be administered in the form of suppositories for rectal administration.
  • suppositories for rectal administration.
  • suppositories can be prepared by mixing the agent with a suitable non-irritating excipient that is solid at room temperature but liquid at rectal temperature and therefore will melt in the rectum to release the drug.
  • suitable non-irritating excipient include, but are not limited to, cocoa butter, beeswax and polyethylene glycols.
  • compositions of this invention may also be administered topically, especially when the target of treatment includes areas or organs readily accessible by topical application, including diseases of the eye, the skin, or the lower intestinal tract. Suitable topical formulations are readily prepared for each of these areas or organs.
  • Topical application for the lower intestinal tract can be effected in a rectal suppository formulation (see above) or in a suitable enema formulation. Topically- transdermal patches may also be used.
  • the pharmaceutical compositions may be formulated in a suitable ointment containing the active component suspended or dissolved in one or more carriers.
  • Carriers for topical administration of the compounds of this invention include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water.
  • the pharmaceutical compositions can be formulated in a suitable lotion or cream containing the active components suspended or dissolved in one or more pharmaceutically acceptable carriers.
  • Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
  • the pharmaceutical compositions may be formulated as micronized suspensions in isotonic, pH adjusted sterile saline, or, preferably, as solutions in isotonic, pH adjusted sterile saline, either with or without a preservative such as
  • compositions may be formulated in an ointment such as petrolatum.
  • compositions of this invention may also be administered by nasal aerosol or inhalation.
  • Such compositions are prepared according to techniques well- known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other conventional solubilizing or dispersing agents.
  • compositions should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of the inhibitor can be administered to a patient receiving these compositions.
  • a specific dosage and treatment regimen for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, and the judgment of the treating physician and the severity of the particular disease being treated.
  • the amount of inhibitor will also depend upon the particular compound in the composition.
  • those additional agents may be administered separately, as part of a multiple dosage regimen, from the protein kinase inhibitor-containing compound or composition.
  • those agents may be part of a single dosage form, mixed together with the protein kinase inhibitor in a single composition.
  • Another aspect of this invention is directed towards a method of treating cancer in a subject in need thereof, comprising the sequential or co-administration of a compound of this invention or a pharmaceutically acceptable salt thereof, and an anti-cancer agent.
  • said anti-cancer agent is selected from Platinating agents, such as Cisplatin, Oxaliplatin, Carboplatin, Nedaplatin, or Satraplatin and other derivatives; Topo I inhibitors, such as Camptothecin, Topotecan, irinotecan/SN38, rubitecan and other derivatives; Antimetabolites, such as Folic family (Methotrexate, Pemetrexed and relatives); Purine family (Thioguanine, Fludarabine, Cladribine, 6-Mercaptopurine and relatives); Pyrimidine family (Cytarabine, Gemcitabine, 5-Fluorouracil and relatives); Alkylating agents, such as Nitrogen mustards (Cyclophosphamide, Melphalan, Ch
  • Aziridines Antibiotics, such as Hydroxyurea; Anthracyclines (doxorubicin, daunorubicin, epirubicin and other derivatives); Anthracenediones (Mitoxantrone and relatives);
  • Streptomyces family (Bleomycin, Mitomycin C, actinomycin) and Ultraviolet light.
  • the compounds and compositions of this invention are also useful in biological samples.
  • One aspect of the invention relates to inhibiting ATR kinase activity in a biological sample, which method comprises contacting said biological sample with a compound described herein or a composition comprising said compound.
  • biological sample means an in vitro or an ex vivo sample, including, without limitation, cell cultures or extracts thereof; biopsied material obtained from a mammal or extracts thereof; and blood, saliva, urine, feces, semen, tears, or other body fluids or extracts thereof.
  • compounds described herein includes compounds of formula I.
  • Inhibition of ATR kinase activity in a biological sample is useful for a variety of purposes that are known to one of skill in the art. Examples of such purposes include, but are not limited to, blood transfusion, organ-transplantation, and biological specimen storage.
  • Another aspect of this invention relates to the study of protein kinases in biological and pathological phenomena; the study of intracellular signal transduction pathways mediated by such protein kinases; and the comparative evaluation of new protein kinase inhibitors.
  • uses include, but are not limited to, biological assays such as enzyme assays and cell-based assays.
  • the activity of the compounds as protein kinase inhibitors may be assayed in vitro, in vivo or in a cell line.
  • In vitro assays include assays that determine inhibition of either the kinase activity or ATPase activity of the activated kinase. Alternate in vitro assays quantitate the ability of the inhibitor to bind to the protein kinase and may be measured either by radiolabelling the inhibitor prior to binding, isolating the inhibitor/kinase complex and determining the amount of radiolabel bound, or by running a competition experiment where new inhibitors are incubated with the kinase bound to known radioligands.
  • Detailed conditions for assaying a compound utilized in this invention as an inhibitor of ATR is set forth in the Examples below.
  • Another aspect of the invention provides a method for modulating enzyme activity by contacting a compound described herein with ATR kinase.
  • the present invention provides a method for treating or lessening the severity of a disease, condition, or disorder where ATR kinase is implicated in the disease state. In another aspect, the present invention provides a method for treating or lessening the severity of an ATR kinase disease, condition, or disorder where inhibition of enzymatic activity is implicated in the treatment of the disease. In another aspect, this invention provides a method for treating or lessening the severity of a disease, condition, or disorder with compounds that inhibit enzymatic activity by binding to the ATR kinase.
  • Another aspect provides a method for treating or lessening the severity of a kinase disease, condition, or disorder by inhibiting enzymatic activity of ATR kinase with an ATR kinase inhibitor.
  • One aspect of the invention relates to a method of inhibiting ATR kinase activity in a patient, which method comprises administering to the patient a compound described herein, or a composition comprising said compound. In some embodiments, said method is used to treat or prevent a condition selected from proliferative and hyperproliferative diseases, such as cancer.
  • Another aspect of this invention provides a method for treating, preventing, or lessening the severity of proliferative or hyperproliferative diseases comprising administering an effective amount of a compound, or a pharmaceutically acceptable composition comprising a compound, to a subject in need thereof.
  • said subject is a patient.
  • patient means an animal, preferably a human.
  • said method is used to treat or prevent cancer.
  • said method is used to treat or prevent a type of cancer with solid tumors.
  • said cancer is selected from the following cancers: Oral: buccal cavity, lip, tongue, mouth, pharynx; Cardiac: sarcoma (angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma), myxoma, rhabdomyoma, fibroma, lipoma and teratoma; Lung: bronchogenic carcinoma (squamous cell or epidermoid, undifferentiated small cell, undifferentiated large cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hamartoma, mesothelioma; Gastrointestinal: esophagus (
  • adenocarcinoma tubular adenoma, villous adenoma, hamartoma, leiomyoma), colon, colon- rectum, colorectal; rectum, Genitourinary tract: kidney (adenocarcinoma, Wilm's tumor [nephroblastoma], lymphoma), bladder and urethra (squamous cell carcinoma, transitional cell carcinoma, adenocarcinoma), prostate (adenocarcinoma, sarcoma), testis (seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma); Liver: hepatoma
  • osteogenic sarcoma hepatocellular carcinoma
  • cholangiocarcinoma hepatoblastoma
  • angiosarcoma hepatocellular adenoma
  • hemangioma hemangioma
  • biliary passages Bone: osteogenic sarcoma
  • nerveous system skull (osteoma, hemangioma, granuloma, xanthoma, osteitis deformans), meninges (meningioma, meningiosarcoma, gliomatosis), brain (astrocytoma, meduUoblastoma, glioma, ependymoma, germinoma [pinealoma], glioblasto
  • cystadenocarcinoma mucinous cystadenocarcinoma, unclassified carcinoma] granulosa- thecal cell tumors, Sertoli-Leydig cell tumors, dysgerminoma, malignant teratoma), vulva (squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, melanoma), vagina (clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma (embryonal rhabdomyosarcoma), fallopian tubes (carcinoma), breast; Skin: malignant melanoma, basal cell carcinoma, squamous cell carcinoma, Karposi's sarcoma,
  • keratoacanthoma moles dysplastic nevi, lipoma, angioma, dermatofibroma, keloids, psoriasis, Thyroid gland: papillary thyroid carcinoma, follicular thyroid carcinoma;
  • medullary thyroid carcinoma multiple endocrine neoplasia type 2A, multiple endocrine neoplasia type 2B, familial medullary thyroid cancer, pheochromocytoma, paraganglioma; and Adrenal glands: neuroblastoma.
  • the cancer is selected from the cancers described herein.
  • said cancer is lung cancer, head and neck cancer, pancreatic cancer, gastric cancer, or brain cancer.
  • composition is that amount effective in order to treat said disease.
  • the compounds and compositions, according to the method of the present invention may be administered using any amount and any route of administration effective for treating or lessening the severity of said disease.
  • One aspect provides a method for inhibiting ATR in a patient comprising administering a compound described herein as described herein.
  • Another embodiment provides a method of treating cancer comprising administering to a patient a compound described herein, wherein the variables are as defined herein.
  • Some embodiments comprising administering to said patient an additional therapeutic agent selected from a DNA-damaging agent; wherein said additional therapeutic agent is appropriate for the disease being treated; and said additional therapeutic agent is administered together with said compound as a single dosage form or separately from said compound as part of a multiple dosage form.
  • said DNA-damaging agent is selected from ionizing radiation, radiomimetic neocarzinostatin, a platinating agent, a Topo I inhibitor, a Topo II inhibitor, an antimetabolite, an alkylating agent, an alkyl sulphonates, an antimetabolite, or an antibiotic.
  • said DNA-damaging agent is selected from ionizing radiation, a platinating agent, a Topo I inhibitor, a Topo II inhibitor, or an antibiotic.
  • Platinating agents include Cisplatin, Oxaliplatin, Carboplatin, Nedaplatin, Satraplatin and other derivatives.
  • Other platinating agents include Lobaplatin, and Triplatin.
  • Other platinating agents include Tetranitrate, Picoplatin, Satraplatin,
  • Topo I inhibitor examples include Camptothecin, Topotecan, irinotecan/SN38, rubitecan and other derivatives.
  • Other Topo I inhibitors include Belotecan.
  • Topo II inhibitors examples include Etoposide, Daunorubicin, Doxorubicin,
  • Antimetabolites include members of the Folic family, Purine family (purine antagonists), or Pyrimidine family (pyrimidine antagonists).
  • Examples of the Folic family include methotrexate, pemetrexed and relatives; examples of the Purine family include Thioguanine, Fludarabine, Cladribine, 6-Mercaptopurine, and relatives; examples of the Pyrimidine family include Cytarabine, gemcitabine, 5-Fluorouracil (5FU) and relatives.
  • antimetabolites include Aminopterin,
  • Methotrexate Pemetrexed, Raltitrexed, Pentostatin, Cladribine, Clofarabine, Fludarabine, Thioguanine, Mercaptopurine, Fluorouracil, Capecitabine, Tegafur, Carmofur, Floxuridine, Cytarabine, Gemcitabine, Azacitidine and Hydroxyurea.
  • alkylating agents include Nitrogen mustards, Triazenes, alkyl sulphonates, Procarbazine and Aziridines.
  • Nitrogen mustards include Nitrogen mustards, Triazenes, alkyl sulphonates, Procarbazine and Aziridines.
  • Nitrogen mustards include Nitrogen mustards, Triazenes, alkyl sulphonates, Procarbazine and Aziridines.
  • alkylating agents include Mechlorethamine,
  • Cyclophosphamide Ifosfamide, Trofosfamide, Chlorambucil, Melphalan, Prednimustine, Bendamustine, Uramustine, Estramustine, Carmustine, Lomustine, Semustine, Fotemustine, Nimustine, Ranimustine, Streptozocin, Busulfan, Mannosulfan, Treosulfan, Carboquone, ThioTEPA, Triaziquone, Triethylenemelamine, Procarbazine, dacarbazine, Temozolomide, Altretamine, Mitobronitol, Actinomycin, Bleomycin, Mitomycin and Plicamycin.
  • antibiotics include Mitomycin, Hydroxyurea; Anthracyclines, Anthracenediones, Streptomyces family.
  • Anthracyclines include doxorubicin, daunorubicin, epirubicin and other derivatives; examples of Anthracenediones include Mitoxantrone and relatives; examples of Streptomyces family includee Bleomycin, Mitomycin C, and actinomycin.
  • said platinating agent is Cisplatin or Oxaliplatin; said Topo I inhibitor is Camptothecin; said Topo II inhibitor is Etoposide; and said antibiotic is Mitomycin.
  • said platinating agent is selected from Cisplatin, Oxaliplatin, Carboplatin, Nedaplatin, or Satraplatin; said Topo I inhibitor is selected from Camptothecin, Topotecan, irinotecan/SN38, rubitecan; said Topo II inhibitor is selected from Etoposide; said antimetabolite is selected from a member of the Folic Family, the Purine Family, or the Pyrimidine Family; said alkylating agent is selected from nitrogen mustards, nitrosoureas, triazenes, alkyl sulfonates, Procarbazine, or aziridines; and said antibiotic is selected from Hydroxyurea, Anthracyclines, Anthracenediones, or Streptomyces family.
  • Another embodiment provides a method of promoting cell death in cancer cells comprising administering to a patient a compound described herein, , or a composition comprising said compound.
  • Another embodiment provides a method of sensitizing cells to DNA damaging agents comprising administering to a patient a compound described herein, or a composition comprising said compound.
  • the method is used on a cancer cell having defects in the ATM signaling cascade.
  • said defect is altered expression or activity of one or more of the following: ATM, p53, CHK2, MRE1 1, RAD50, NBS 1, 53BP1, MDC1 or H2AX.
  • the cell is a cancer cell expressing DNA damaging oncogenes.
  • said cancer cell has altered expression or activity of one or more of the following: K-Ras, N-Ras, H-Ras, Raf, Myc, Mos, E2F, Cdc25A, CDC4, CDK2, Cyclin E, Cyclin A and Rb.
  • Yet another embodiment provides use of a compound described herein as a radio- sensitizer or a chemo-sensitizer.
  • a compound of formula I as a single agent (monotherapy) for treating cancer.
  • the compounds of formula I are used for treating patients having cancer with a DNA-damage response (DDR) defect.
  • said defect is a mutation or loss of ATM, p53, CHK2, MREl 1, RAD50, NBS 1 , 53BP 1 , MDC1 , or H2AX.
  • the compounds of the disclosure may be prepared in light of the specification using steps generally known to those of ordinary skill in the art. Those compounds may be analyzed by known methods, including but not limited to LCMS (liquid chromatography mass spectrometry) and NMR (nuclear magnetic resonance). Below are a set of generic schemes that illustrate generally how to prepare the compounds of the present disclosure. Scheme A
  • Scheme A depicts a general method for making compounds of Formula I.
  • Compound A is functionalised via either an SNAr reaction or a range of known metal mediated reactions such as, but not limited to, Suzuki reactions, Stille couplings, Buchwald- Hartwig reactions and carbonylation reactions to give compounds of Formula A-i.
  • J substituents on Rl can undergo further functionalisation by known reactions such as, but not limited to, Mitsunobu reactions and acylation.
  • These compounds are then brominated under standard conditions known to those skilled in the art such as, but not limited to, treatment with N-bromosuccinimide to give compounds of Formula A-ii. Reaction of these compounds under Sonagashira conditions with a suitable protected alkyne gives rise to compounds of the Formula A-iii.
  • Suitable alkyne protecting groups PG include, but are not limited to, TMS, TES or TIPS.
  • the protecting group PG can be removed under standard conditions known to those skilled in the art such as, but not limited to, treatment with aqueous base or TBAF to give compounds of Formula A-iv.
  • Compound A-iv can be cyclised to the corresponding pyrrolo[2,3-b]pyrazine of Formula A-v using methods known to those skilled in the art such as, but not limited to heating with KO l Bu in ⁇ .
  • the protecting group PG of Compound A-iii can be removed concurrently with cyclisation to pyrrolo[2,3-b]pyrazine of Formula A-v, without need to isolate compounds of Formula A-iv.
  • Compounds of Formula A-v can be iodinated under standard conditions known to those skilled in the art such as, but not limited to, treatment with IC1 to give compounds of Formula A-vi.
  • Compounds of Formula A-vi are then protected with a suitable protecting group PG' such as, but not limited to Tosyl (p-toluenesulfonyl), to give compounds of Formula A-vii.
  • Scheme B depicts an alternative general method for making compounds of Formula A-iv and A-v where protection of the aminopyrazine motif allows for functional group transformation on the J substituents on Ri.
  • Compounds of Formula A-iii are protected with a suitable amine protecting group PG" such as, but not limited to BOC (tert- butoxycarbonyl), to give compounds of Formula B-i.
  • PG amine protecting group
  • J substituents on Ri can undergo further functionalisation by known reactions such as, but not limited to, Mitsunobu reactions and acylation.
  • Scheme C depicts an alternative method for the synthesis of compounds of Formula I.
  • Compounds of Formula A-vii are transformed into the corresponding boronic acid (or ester) C-i under standard conditions known to those skilled in the art such as, but not limited to treatment with bis(pinacolato)diboron under Palladium catalysis. These compounds are functionalised under Suzuki conditions to give compounds of Formula A-viii.
  • substituents on A Z 1 , Z 2 , Z 3 or Z 4
  • the protecting group PG' can be removed under standard conditions known to those skilled in the art such as, but not limited to, treatment with aqueous base or TBAF to give compounds of Formula I.
  • Scheme D depicts an alternative method for the synthesis of compounds of Formula I where parameter A can derive from an alkyne functional group.
  • Compounds of Formula A-vii are reacted under Sonagashira conditions with a suitable protected alkyne to give rise to compounds of the Formula D-i.
  • Suitable alkyne protecting groups PG'" include, but are not limited to, TMS, TES or TIPS.
  • the protecting groups PG' and PG' " can be removed under standard conditions known to those skilled in the art such as, but not limited to, treatment with aqueous base or TBAF to give compounds of Formula D-ii. These compounds are further elaborated under reaction conditions known in the art to give compounds of Formula I.
  • Scheme E depicts an alternative method for the synthesis of compounds of Formula I where parameter A can derive from an alkyne functional group where protection of the pyrrolo[2,3-b]pyrazine allows for further elaboration of the substituents on A.
  • Compounds are Formula D-ii are protected with a suitable protecting group PG' such as, but not limited to Tosyl (p-toluenesulfonyl), to give compounds of Formula E-i. These compounds are further elaborated under reaction conditions known in the art to give compounds of Formula E-ii. Where appropriate, substituents on A (Zl , Z2, Z3 opr Z4) can undergo further functionalisation by reactions known to those skilled in the art such as, but not limited to, Mitsunobu reactions, nucleophilic displacement and acylation. The protecting group PG' can then be removed under standard conditions known to those skilled in the art such as, but not limited to, treatment with aqueous base or TBAF to give compounds of Formula I.
  • a suitable protecting group PG' such as, but not limited to Tosyl (p-toluenesulfonyl), to give compounds of Formula E-i.
  • substituents on A Zl , Z2, Z3 opr Z4
  • the protecting group PG'
  • Triethylamine (47.58 g, 65.54 mL, 470.2 mmol) was added to a stirred suspension of 3-bromo-5-(4-isopropylsulfonylphenyl)pyrazin-2-amine (33.5 g, 94.04 mmol) in anhydrous DMF (234.5 mL). Copper(I) iodide (2.148 g, 11.28 mmol) was then added followed by Pd(PPh3) 4 (5.433 g, 4.702 mmol).
  • Step 4 tert-Butyl N-teri-butoxycarbonyl-N-[5-(4-isopropylsulfonylphi
  • Triethylamine (18.42 g, 25.37 niL, 182.0 mmol) was added to a stirred solution of 5-(4-isopropylsulfonylphenyl)-3-(24rimethylsilylethynyl)pyrazin-2-amine (34 g, 91.02 mmol) in DCM (1.020 L).
  • DMAP 1.112 g, 9.102 mmol
  • tert- butoxycarbonyl tert-butyl carbonate 59.60 g, 62.74 mL, 273.1 mmol
  • Step 5 teri-Butyl N-teri-butoxycarbonyl-N-[3-ethynyl-5-(4- isopropylsulfonylphenyl)pyrazin-2-yl]carbamate
  • tert-Butyl N-ter?-butoxycarbonyl-N-[5-(4-isopropylsulfonylphenyl)-3-(2- trimethylsilylethynyl)pyrazin-2-yl]carbamate (42.6 g, 74.25 mmol) was dissolved/suspended in methanol (510 mL) and 2M aqueous sodium carbonate (425.8 mL, 851.6 mmol) was added to the rapidly stirred mixture. An oil separated out and more methanol (100 mL) was added to dissolve. The mixture was stirred at ambient temperature for 1 hour.
  • Step 6 3-Ethynyl-5-(4-isopropylsulfonylphenyl)pyrazin-2-amine
  • TFA 34.45 g, 23.28 mL, 302.1 mmol
  • tert-butyl N-tert-butoxycarbonyl-N-[3-ethynyl-5-(4-isopropylsulfonylphenyl)pyrazin-2-yl]carbamate 5 g, 9.968 mmol
  • DCM 200 mL
  • the solvent was removed in vacuo and the residue re-dissolved in DCM and stirred with saturated aqueous aHC03 for 30 minutes.
  • the layers were separated and the organic layer washed with saturated aqueous aHC03 (x 3).
  • Step 8 7-Iodo-2-(4-isopropylsulfonylphenyl)-5H-pyrrolo [2,3-b]pyrazine
  • Step 9 7-Iodo-2-(4-isopropylsulfonylphenyl)-5-(p-tolylsulfonyl)pyrrolo[2,3-b]pyrazine
  • Step 10 2-(4-Isopropylsulfonylphenyl)-5-(p-tolylsulfonyl)-7-(3-pyridyl)pyrrolo[2,3- b]pyrazine
  • Step 11 2-(4-Isopropylsulfonylphenyl)-7-(3-pyridyl)-5H-pyrrolo[2,3-b]pyrazine
  • Step 1 2-(4-isopropylsulfonylphenyl)-7-(2-phenylethynyl)-5-(p- tolylsulfonyl)pyrrolo[2,3-b]pyrazine
  • Step 1 2-[2-(4-Isopropylsulfonylphenyl)-5-(p-tolylsulfonyl)pyrrolo[2,3-b]pyrazin-7- yl] ethynyl-trimethyl-silane
  • Step 2 7-Ethynyl-2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazine
  • Step 3 7-Ethynyl-2-(4-isopropylsulfonylphenyl)-5-(p-tolylsulfonyl)pyrrolo [2,3- b]pyrazine
  • Step 4 5-[2-(4-Isopropylsulfonylphenyl)-5-(p-tolylsulfonyl)pyrrolo[2,3-b]pyrazin-7-yl]- 3-phenyl-isoxazole
  • Triethylamine (16.45 mg, 22.66 ⁇ ,, 0.1626 mmol) was added to a stirred solution of 7-ethynyl-2-(4-isopropylsulfonylphenyl)-5-(p-tolylsulfonyl)pyrrolo[2,3-b]pyrazine (65 mg, 0.1355 mmol) and N-hydroxybenzimidoyl chloride (21.08 mg, 0.1355 mmol) in THF (2 mL). The reaction mixture was stirred at ambient temperature for 45 minutes then heated at 65 °C for 3 hours. The reaction mixture was concentrated in vacuo and the residue partitioned between DCM and water.
  • Step 5 5-[2-(4-Isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazin-7-yl]-3-phenyl- isoxazole
  • Step 4 teri-Butyl /V-[l-[3-(hydroxymethyl)phenyl] ethyljcarbamate
  • Step 5 teri-Butyl iV-[l-(3-formylphenyl)ethyl]carbamate
  • Step 6 tert-Butyl l-(3-((hydroxyimino)methyl)phenyl)ethylcarbamate
  • Step 7 tert-Butyl l-(3-(chloro(hydroxyimino)methyl)phenyl) ethylcarbamate
  • Step 8 tert-Butyl N-[l-[3-[5-[2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazin-7- yl]isoxazol-3-yl]phenyl]et
  • Step 9 l-[3-[5-[2-(4-Isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazin-7-yl]isoxazol-3- yl]phenyl]ethanamine
  • Compounds can be screened for their ability to inhibit intracellular ATR using an immunofluorescence microscopy assay to detect phosphorylation of the ATR substrate histone H2AX in hydroxyurea treated cells.
  • HT29 cells are plated at 14,000 cells per well in 96-well black imaging plates (BD 353219) in McCoy's 5A media (Sigma M8403) supplemented with 10% foetal bovine serum (JRH Biosciences 12003),
  • the cells are washed in PBS, fixed for lOmin in 4% formaldehyde diluted in PBS (Polysciences Inc 18814), washed in 0.2% Tween-20 in PBS (wash buffer), and permeabilised for lOmin in 0.5% Triton X-100 in PBS, all at room temperature. The cells are then washed once in wash buffer and blocked for 30min at room temperature in 10% goat serum (Sigma G9023) diluted in wash buffer (block buffer).
  • the cells are then incubated for lh at room temperature in primary antibody (mouse monoclonal anti-phosphorylated histone H2AX Serl39 antibody; Upstate 05-636) diluted 1 :250 in block buffer. The cells are then washed five times in wash buffer before incubation for lh at room temperature in the dark in a mixture of secondary antibody (goat anti-mouse Alexa Fluor 488 conjugated antibody;
  • Invitrogen A11029 Invitrogen A11029
  • Hoechst stain Invitrogen H3570
  • diluted 1 :500 and 1 :5000, respectively, in wash buffer The cells are then washed five times in wash buffer and finally lOOul PBS is added to each well before imaging.
  • Pathway 855 Bioimager and Attovision software (BD Biosciences, Version 1.6/855) to quantify phosphorylated H2AX Serl39 and DNA staining, respectively.
  • the percentage of phosphorylated H2AX -positive nuclei in a montage of 9 images at 20x magnification is then calculated for each well using BD Image Data Explorer software (BD Biosciences Version 2.2.15).
  • Phosphorylated H2AX-positive nuclei are defined as Hoechst-positive regions of interest containing Alexa Fluor 488 intensity at 1.75-fold the average Alexa Fluor 488 intensity in cells not treated with hydroxyurea.
  • the percentage of H2AX positive nuclei is finally plotted against concentration for each compound and IC50s for intracellular ATR inhibition are determined using Prism software(GraphPad Prism version 3.0cx for Macintosh, GraphPad Software, San Diego California, USA).
  • Assays were carried out at 25°C in the presence of 5 nM full-length ATR.
  • An assay stock buffer solution was prepared containing all of the reagents listed above, with the exception of ATP and the test compound of interest. 13.5 ⁇ ., of the stock solution was placed in a 96 well plate followed by addition of 2 ⁇ ⁇ of DMSO stock containing serial dilutions of the test compound (typically starting from a final concentration of 15 ⁇ with 3- fold serial dilutions) in duplicate (final DMSO concentration 7%). The plate was pre- incubated for 10 minutes at 25°C and the reaction initiated by addition of 15 ⁇ ⁇ [ ⁇ -33 ⁇ ] ⁇ (final concentration 10 ⁇ ).
  • the reaction was stopped after 24 hours by the addition of 30 ⁇ , 0.1M phosphoric acid containing 2mM ATP.
  • a multiscreen phosphocellulose filter 96-well plate (Millipore, Cat no. MAPHN0B50) was pretreated with ⁇ . 0.2M phosphoric acid prior to the addition of 45 ⁇ , of the stopped assay mixture.
  • the plate was washed with 5 x 200 ⁇ , 0.2M phosphoric acid. After drying, 100 ⁇ ., Optiphase 'SuperMix' liquid scintillation cocktail (Perkin Elmer) was added to the well prior to scintillation counting (1450 Microbeta Liquid Scintillation Counter, Wallac).
  • Ki(app) data were calculated from non-linear regression analysis of the initial rate data using the Prism software package (GraphPad Prism version 3.0cx for Macintosh, GraphPad Software, San Diego California, USA).
  • HCT116 cells which possess a defect in ATM signaling to Cisplatin (see, Kim et al.; Oncogene 21 :3864 (2002); see also, Takemura et al; JBC 281 :30814 (2006)) are plated at 470 cells per well in 96-well polystyrene plates (Costar 3596) in 150 ⁇ 1 of McCoy's 5A media (Sigma M8403) supplemented with 10% foetal bovine serum (JRH Biosciences 12003),
  • Compounds and Cisplatin are then both added simultaneously to the cell media in 2-fold serial dilutions from a top final concentration of 10 ⁇ as a full matrix of concentrations in a final cell volume of 200 ⁇ 1, and the cells are then incubated at 37°C in 5% C0 2 .
  • 40 ⁇ 1 of MTS reagent (Promega G358a) is added to each well and the cells are incubated for lh at 37°C in 5% CO 2 .

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Biomedical Technology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pathology (AREA)
  • Radiology & Medical Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)

Abstract

The present invention relates to pyrrolopyrazines compounds useful as inhibitors of ATR protein kinase. The invention also relates to pharmaceutically acceptable compositions comprising the compounds of this invention; methods of treating of various diseases, disorders, and conditions using the compounds of this invention; processes for preparing the compounds of this invention; intermediates for the preparation of the compounds of this invention; and methods of using the compounds in in vitro applications, such as the study of kinases in biological and pathological phenomena; the study of intracellular signal transduction pathways mediated by such kinases; and the comparative evaluation of new kinase inhibitors. The compounds of this invention have formula I: wherein the variables are as defined herein.

Description

PYRROLO- PYRAZINE DERIVATIVES USEFUL AS
INHIBITORS OF ATR KINASE
BACKGROUND OF THE INVENTION
[0001] ATR ("ATM and Rad3 related") kinase is a protein kinase involved in cellular responses to DNA damage. ATR kinase acts with ATM ("ataxia telangiectasia mutated") kinase and many other proteins to regulate a cell's response to DNA damage, commonly referred to as the DNA Damage Response ("DDR"). The DDR stimulates DNA repair, promotes survival and stalls cell cycle progression by activating cell cycle checkpoints, which provide time for repair. Without the DDR, cells are much more sensitive to DNA damage and readily die from DNA lesions induced by endogenous cellular processes such as DNA replication or exogenous DNA damaging agents commonly used in cancer therapy.
[0002] Healthy cells can rely on a host of different proteins for DNA repair including the DDR kinase ATR. In some cases these proteins can compensate for one another by activating functionally redundant DNA repair processes. On the contrary, many cancer cells harbour defects in some of their DNA repair processes, such as ATM signaling, and therefore display a greater reliance on their remaining intact DNA repair proteins which include ATR.
[0003] In addition, many cancer cells express activated oncogenes or lack key tumour suppressors, and this can make these cancer cells prone to dysregulated phases of DNA replication which in turn cause DNA damage. ATR has been implicated as a critical component of the DDR in response to disrupted DNA replication. As a result, these cancer cells are more dependent on ATR activity for survival than healthy cells. Accordingly, ATR inhibitors may be useful for cancer treatment, either used alone or in combination with DNA damaging agents, because they shut down a DNA repair mechanism that is more important for cellular survival in many cancer cells than in healthy normal cells.
[0004] In fact, disruption of ATR function (e.g. by gene deletion) has been shown to promote cancer cell death both in the absence and presence of DNA damaging agents. This suggests that ATR inhibitors may be effective both as single agents and as potent sensitizers to radiotherapy or genotoxic chemotherapy. [0005] ATR peptide can be expressed and isolated using a variety of methods known in the literature (see e.g., Unsal-Kacmaz et al, PNAS 99: 10, pp6673-6678, May 14, 2002; see also Kumagai et al. Cell 124, pp943-955, March 10, 2006; Unsal-Kacmaz et al. Molecular and Cellular Biology. Feb 2004, pi 292- 1300; and Hall-Jackson et al. Oncogene 1999, 18, 6707-6713).
[0006] For all of these reasons, there is a need for the development of potent and selective ATR inhibitors for the treatment of cancer, either as single agents or as combination therapies with radiotherapy or genotoxic chemotherapy.
SUMMARY OF THE INVENTION
[0007] The present invention relates to pyrrolopyrazine compounds useful as inhibitors of ATR protein kinase. The invention also relates to pharmaceutically acceptable compositions comprising the compounds of this invention; methods of treating of various diseases, disorders, and conditions using the compounds of this invention; processes for preparing the compounds of this invention; intermediates for the preparation of the compounds of this invention; and methods of using the compounds in in vitro applications, such as the study of kinases in biological and pathological phenomena; the study of intracellular signal transduction pathways mediated by such kinases; and the comparative evaluation of new kinase inhibitors. These compounds have an unexpected ability to treat cancer as single agents. These compounds also show surprising synergy with other cancer agents, such as cisplatin, in combination therapies.
DETAILED DESCRIPTION OF THE INVENTION
[0008] One aspect of this invention pro und of Formula I:
Figure imgf000003_0001
I
or a pharmaceutically acceptable salt thereof, wherein
R1 is hydrogen, Ci-6alkyl, or a 3-7 membered monocyclic fully saturated, partially
unsaturated, or aromatic ring having 0-4 heteroatoms independently selected from the group consisting of nitrogen, oxygen, and sulfur; or an 8-10 membered bicyclic aromatic ring having 0-6 heteroatoms selected from the group consisting of nitrogen, oxygen, and 1 is optionally substituted with 0-4 occurrences of J;
Figure imgf000004_0001
A1 is a 5 -membered heteroaryl wherein X is carbon, nitrogen, oxygen, or sulfur; when X is nitrogen or carbon;
A2 is phenyl or a 6-membered heteroaryl having 1-3 nitrogen atoms; A2 is independently and optionally substituted with up to 2 occurrences of halo or CN;
A3 is an 8-10 membered bicyclic heteroaromatic ring having 1-3 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur;
Z1 is H, a Ci_6aliphatic; wherein 0-2 methylene units of said Ci_ioaliphatic are optionally replaced with -NR.'-, -0-, -S-, C(O); or a 5-6 membered monocyclic aromatic ring having 0-3 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or an 8-10 membered bicyclic aromatic ring having 0-6 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or a Ci-ioaliphatic; wherein 0-4 methylene units of said Ci-ioaliphatic are optionally replaced with -NR'-, -0-, -S-, C(O); a C3_6cycloalkyl, or a 3-6 membered heterocyclic ring having 1-2 heteroatoms selected from O, NR', or S; Z1 is optionally substituted with 1-5 J1 groups;
Z2 is H, a 5-6 membered monocyclic aromatic ring having 0-3 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or an 8-10 membered bicyclic aromatic ring having 0-6 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or a Ci-ioaliphatic; wherein 0-4 methylene units of said Ci-ioaliphatic are optionally replaced with -NR'-, -0-, -S-, C(O), a C3_6cycloalkyl, or a 3-6 membered heterocyclic ring having 1-2 heteroatoms selected from O, NR', or S; Z2 is optionally substituted with 1-5 J2 groups;
Z3 is H, C3_6cycloalkyl, halo, CN, NO2, or a Ci-ioaliphatic; wherein 0-4 methylene units of said Ci-ioaliphatic are optionally replaced with -NR'-, -0-, -S-, or C(O); Z3 is optionally substituted with 1-5 J3 groups;
Z4 is a 5-6 membered monocyclic aromatic ring having 0-3 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or an 8-10 membered bicyclic aromatic ring having 0-6 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; Z4 is optionally substituted with 1-5 J4 groups; J is halo, CN, or V, or (V)t-R2;
V is a Ci-ioaliphatic group wherein up to 3 methylene units are optionally replaced with O, NR", C(O), S, S(O), or S(0)2; wherein said Ci-ioaliphatic group is optionally substituted with 1-3 occcurences of halo or CN;
R2 is 3-7 membered aromatic or nonaromatic monocyclic ring having 0-3 heteroatoms
selected from the group consisting of oxygen, nitrogen, and sulfur; R2 is optionally substituted with 1-3 occurences of halo, CN, C3_6cycloalkyl, or Ci-ioaliphatic; wherein up to 3 methylene units of said Ci-ioaliphatic are optionally replaced with NR', O, S, or CO; each J1, J2, and J4 is independently halo, CN, NO2, or X1;
JA is XA or XA-QA;
XA is a Ci-6aliphatic; wherein 0-4 methylene units of said Ci-ioaliphatic are optionally
replaced with -NR'-, -0-, -S-, or C(O); wherein said Ci-6aliphatic is optionally substituted with halo or Ci_3alkyl;
QA is phenyl;
X1 is a Ci_6aliphatic; wherein 0-4 methylene units of said Ci-ioaliphatic are optionally
replaced with -NR'-, -0-, -S-, or C(O), wherein X1 is optionally and independently substituted with 1-4 occurrences of JX1;
JX1 is halo or a 3-6 membered monocyclic ring having 0-2 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur;
J3 is halo, CN, phenyl, a 4-6 membered heterocyclic ring having 1-2 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or a Ci-6aliphatic optionally substituted with 1-3 occurrences of halo;
each R' and R" is independently hydrogen or Ci_6alkyl;
t is 0 or 1.
In some embodiments,
when A
Figure imgf000005_0001
is not optionally substituted phenyl;
when A2 is pyridinyl, then R1 is not optionally substituted phenyl or optionally substituted pyrazinyl;
when A2 is pyrrolyl, then R1 is not optionally substituted cyclohexyl;
when A2 is phenyl, then R1 is not an optionally substituted group selected from pyridinyl, morpholinyl, or piperazinyl; when A2 is phenyl and R1 is phenyl; R1 is substituted with 4-S02(Ci_6alkyl) as shown in formula ii-a;
Figure imgf000006_0001
lkyl
[0009] Another embodiment provides a mpound of formula I:
Figure imgf000006_0002
I
or a pharmaceutically acceptable salt thereof, wherein
R1 is hydrogen, Ci_6alkyl, or a 3-7 membered monocyclic fully saturated, partially
unsaturated, or aromatic ring having 0-4 heteroatoms independently selected from the group consisting of nitrogen, oxygen, and sulfur; or an 8-10 membered bicyclic aromatic ring having 0-6 heteroatoms selected from the group consisting of nitrogen, oxygen, and lfur; R1 is optionally substituted with 0-4 occurrences of J;
Figure imgf000006_0003
A1 is a 5 -membered heteroaryl wherein X is carbon, nitrogen, oxygen, or sulfur;
A2 is a 6-membered heteroaryl having 1-3 nitrogen atoms; A2 is independently and optionally substituted with up to 2 occurrences of halo or CN;
A3 is an 8-10 membered bicyclic heteroaromatic ring having 1-3 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur;
Z1 is a 5-6 membered monocyclic aromatic ring having 0-3 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or an 8-10 membered bicyclic aromatic ring having 0-6 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or a Ci-ioaliphatic; wherein 0-4 methylene units of said Ci-ioaliphatic are optionally replaced with -NR'-, -0-, -S-, C(O); a C3_6cycloalkyl, or a 3-6 membered heterocyclic ring having 1-2 heteroatoms selected from O, NR.', or S; Z1 is optionally substituted with 1-5 J1 groups;
Z2 is hydrogen, a 5-6 membered monocyclic aromatic ring having 0-3 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or an 8-10 membered bicyclic aromatic ring having 0-6 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or a Ci_ioaliphatic; wherein 0-4 methylene units of said Ci-ioaliphatic are optionally replaced with -NR.'-, -0-, -S-, C(O), a C3_6cycloalkyl, or a 3-6 membered heterocyclic ring having 1-2 heteroatoms selected from O, NR', or S; Z2 is optionally substituted with 1-5 J2 groups;
Z3 is H, C3_6cycloalkyl, halo, CN, NO2, or a Ci-ioaliphatic; wherein 0-4 methylene units of said Ci-ioaliphatic are optionally replaced with -NR'-, -0-, -S-, or C(O); Z3 is optionally substituted with 1-5 J3 groups;
Z4 is a 5-6 membered monocyclic aromatic ring having 0-3 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or an 8-10 membered bicyclic aromatic ring having 0-6 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; Z4 is optionally substituted with 1-5 J4 groups;
J is halo, CN, or V, or (V)t-R2;
V is a Ci-ioaliphatic group wherein up to 3 methylene units are optionally replaced with O, NR", C(O), S, S(O), or S(0)2; wherein said Ci-ioaliphatic group is optionally substituted with 1-3 occcurences of halo or CN;
R2 is 3-7 membered aromatic or nonaromatic monocyclic ring having 0-3 heteroatoms
selected from the group consisting of oxygen, nitrogen, and sulfur; R2 is optionally substituted with 1-3 occurences of halo, CN, C3_6cycloalkyl, or Ci-ioaliphatic; wherein up to 3 methylene units of said Ci-ioaliphatic are optionally replaced with NR', O, S, or CO; each J1, J2, and J4 is independently halo, CN, NO2, or X1;
X1 is a Ci_6aliphatic; wherein 0-4 methylene units of said Ci-ioaliphatic are optionally
replaced with -NR'-, -0-, -S-, or C(O), wherein X1 is optionally and independently substituted with 1-4 occurrences of JX1;
JX1 is halo or a 3-6 membered monocyclic ring having 0-2 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur;
J3 is halo, CN, a 4-6 membered heterocyclic ring having 1-2 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or a Ci_6aliphatic optionally substituted with 1-3 occurrences of halo;
each R' and R" is independently hydrogen or Ci-6alkyl; t is 0 or 1.
[0010] In some embodiments,
Figure imgf000008_0001
A2 is a 6-membered heteroaryl having 1-3 nitrogen atoms;
Z1 is a 5-6 membered monocyclic aromatic ring having 0-3 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or an 8-10 membered bicyclic aromatic ring having 0-6 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or a Ci-ioaliphatic; wherein 0-4 methylene units of said Ci-ioaliphatic are optionally replaced with -NR.'-, -0-, -S-, C(O); a C3_6cycloalkyl, or a 3-6 membered heterocyclic ring having 1-2 heteroatoms selected from O, NR', or S; Z1 is optionally substituted with 1-5 J1 groups; and
[0011] J3 is halo, CN, phenyl, a 4-6 membered heterocyclic ring having 1-2 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or a Ci_6aliphatic optionally substituted with 1-3 occurrences of halo.
[0012] According to one embodiment, A is
[0013] According to another embodiment,
Figure imgf000008_0002
. In some embodiments, A1 is selected from the following:
Figure imgf000008_0003
Figure imgf000008_0004
wherein X1 is O, NR, or S; R is H or C1-6alkyl.
[0014] In other embodiments, A1 is selected from the following:
Figure imgf000009_0001
or ¾ X ' <Μη some embodiments, X is O, NR, or S; wherein R is H or Ci-6alkyl. In other embodiments, A is
Figure imgf000009_0002
. In yet other embodiments, A1 is x '^ ^i . in some embodiments, X1 is S. In other embodiments, X1 is O.
[0015] It should be understood that the A1 groups can be bonded to the pyrrolopyrazine and Z1 in at least two different ways. For example,
Figure imgf000009_0003
can be bonded in the two ways shown below:
Figure imgf000009_0004
(as drawn) (reverse)
[0016] In some embodiments, X1 is S or O. In other embodiments, X1 is O.
[0017] According to another embodiment, Z1 is a 5-6 membered aromatic ring. In some embodiments, Z1 is phenyl. In certain embodiments, Z1 is optionally substituted with 1-2 J1 groups. In some embodiments, J1 is -CH2NHR' or CHCi_6alkyl)NHR'. In other
embodiments, R' is H (J1 is -CH2NH2 or CHCi_6alkyl)NH2). According to another embodiment, Z1 is COOH. According to yet another embodiment, Z1 is Ci-6alkyl.
According to another embodiment, A
Figure imgf000009_0005
ome embodiments, A2 is a
6-membered heteroaryl having 1-2 nitrogen atoms. In other embodiments, A2 is pyridinyl or pyrimidinyl. In some embodiments, A2 is substituted with one occurrence of Z2. In some embodiments, the one occurrence of Z2 is bonded at the 3 -position of the 6-membered ring. In t other embodiments A2 is selected from the following:
Figure imgf000010_0001
[0018] In some embodiments, A2 is phenyl and Z2 is CN, CH2OH, CH2OCH3, CONH2, CON(CH3)2, OCH3, or tretrazolyl.
[0019] According to another embodiment, A
Figure imgf000010_0002
In some
embodiments, A3 is an 8-10 membered bicyclic heteroaromatic ring having 1-2 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur. In some embodiments, said heteroaromatic ring is benzoxazolyl, benzisoxazolyl, benzothiazolyl, benzimidazolyl, azaindolyl, indolyl, indazolyl, benzothienyl, benzofuranyl, dihydrothienodioxinyl, quinolinyl, or isoquinolinyl. In other embodiments, said heteroaromatic ring is benzoxazolyl, benzisoxazolyl, benzothiazolyl, benzimidazolyl, indolyl, indazolyl, benzothienyl, or benzofuranyl.
In some embodiments, A is
Figure imgf000010_0003
[0020] In other embodiments, A
Figure imgf000010_0004
. In some embodiments, Y is O, NR, or S; wherein R is H or Ci-4alkyl. In other embodiments, A is selected from the following:
Figure imgf000011_0001
[0021] According to another embodiment, A
Figure imgf000011_0002
[0022] In some embodiments, Z1 and Z2 can be a Ci-ioaliphatic wherein 0-4 methylene units are optionally replaced with -NR.'-, -0-, -S-, C(O); a C3_6cycloalkyl, or a 3-6 membered heterocyclic ring having 1-2 heteroatoms selected from O, N, or S. It shall be understood that when a methylene unit is replaced by a cyclic ring, such as a C3_6cycloalkyl or a 3-6 membered heterocyclic ring, the cyclic ring can be attached to the methylene units of the aliphatic chain via any two points of attachment on the ring. These could be from two different atoms on the ring or one atom of the ring (forming a spirocycle). For example, a C4 aliphatic wherein one methylene unit is replaced with a 6-membered heterocyclyl having one nitrogen atom could look like this: \— / . Here, the heterocyclic ring
(piperidine) is replacing the first methylene unit of the C4 aliphatic. The piperidine ring is bonded to its surrounding atoms via two points of attachment on ring: a carbon atom and a nitrogen atom.
[0023] A C5 aliphatic wherein one methylene unit (the second one, in this case) is replaced with a 5-membered heterocyclyl having one nitrogen atom could look like this:
Figure imgf000011_0003
. In this case, the heterocyclic ring (pyrrolidine) is bonded to the methylene units of the aliphatic via two points of attachment of the same carbon atom. Finally, a C3 aliphatic wherein one methylene unit is replaced with a cyclohexyl could look like this:
Figure imgf000011_0004
Here, the cyclohexyl ring has replaced the second methylene unit of the aliphatic. The cyclohexyl ring is bonded to the surrounding methylene units of the aliphatic via two different carbon atoms of the cyclohexyl ring.
[0024] In some embodiments, Z4 is a 5-6 membered aromatic ring. In other embodiments, Z4 is phenyl.
[0025] According to another embodiment, R1 is Ci-6alkyl. In some embodiments, R1 is a monocyclic ring. In other embodiments, R1 is a 3-7 membered cycloaliphatic or heterocyclyl. In yet other embodiments, R1 is a 5-6 membered aromatic ring having 0-4 heteroatoms independently selected from the group consisting of nitrogen, oxygen, and sulfur. In some embodiments, R1 is phenyl, pyridinyl, or pyrimidinyl. In some embodiments, R1 is phenyl.
[0026] According to another embodiment, R1 is substituted by one to two occurrences of J. In some embodiments, R1 is substituted in the para position with J. In certain embodiments, J is V. In some embodiments, V is a Ci_6aliphatic group wherein one methylene unitis optionally replaced with S(0)2. In some embodiments, V is -S(0)2(Ci_4alkyl). In other embodiments, V is S(0)2CH(CH3)2.
[0027] According to another embodiment, J is CN and substituted in the ortho position. In some embodiments, J is -(V)t-R2. In certain embodiments, V is -S(0)2.
[0028] According to another embodiment, R2 is a C3-7 cycloalkyl or a 3-7 membered heterocyclyl having 1-3 heteroatoms selected from the group consisting of oxygen, nitrogen, and sulfur.
[0029] According to another embodiment, R1 is phenyl substituted in the para position with J, wherein J is -S(0)2(Ci_4alkyl).
[0030] Another embodiment provides a compound from the following Table:
Figure imgf000012_0001
Figure imgf000013_0001
Figure imgf000014_0001
Figure imgf000015_0001
Figure imgf000016_0001
Figure imgf000017_0001
1-67 1-68 1-69
Figure imgf000017_0002
1-70
[0031] In some embodiments, the variables are as depicted in the compounds of the disclosure including compounds in the tables above.
[0032] Compounds of this invention include those described generally herein, and are further illustrated by the classes, subclasses, and species disclosed herein. As used herein, the following definitions shall apply unless otherwise indicated. For purposes of this invention, the chemical elements are identified in accordance with the Periodic Table of the Elements, CAS version, Handbook of Chemistry and Physics, 75th Ed. Additionally, general principles of organic chemistry are described in "Organic Chemistry", Thomas Sorrell, University Science Books, Sausalito: 1999, and "March's Advanced Organic Chemistry", 5th Ed., Ed.: Smith, M.B. and March, J., John Wiley & Sons, New York: 2001, the entire contents of which are hereby incorporated by reference.
[0033] As described herein, a specified number range of atoms includes any integer therein. For example, a group having from 1-4 atoms could have 1, 2, 3, or 4 atoms.
[0034] As described herein, compounds of the invention may optionally be substituted with one or more substituents, such as are illustrated generally herein, or as exemplified by particular classes, subclasses, and species of the invention. It will be appreciated that the phrase "optionally substituted" is used interchangeably with the phrase "substituted or unsubstituted." In general, the term "substituted", whether preceded by the term "optionally" or not, refers to the replacement of hydrogen radicals in a given structure with the radical of a specified substituent. Unless otherwise indicated, an optionally substituted group may have a substituent at each substitutable position of the group, and when more than one position in any given structure may be substituted with more than one substituent selected from a specified group, the substituent may be either the same or different at every position.
Combinations of substituents envisioned by this invention are preferably those that result in the formation of stable or chemically feasible compounds.
[0035] Unless otherwise indicated, a substituent connected by a bond drawn from the center of a ring means that the substituent can be bonded to any position in the ring. In example i below, for instance, J1 can be bonded to any position on the pyridyl ring. For bicyclic rings, a bond drawn through both rings indicates that the substituent can be bonded from any position of the bicyclic ring. In example ii below, for instance, J1 can be bonded to the 5-membered ring (on the nitrogen atom, for instance), and to the 6-membered ring.
Figure imgf000018_0001
1 11
[0036] The term "stable", as used herein, refers to compounds that are not substantially altered when subjected to conditions to allow for their production, detection, recovery, purification, and use for one or more of the purposes disclosed herein. In some embodiments, a stable compound or chemically feasible compound is one that is not substantially altered when kept at a temperature of 40°C or less, in the absence of moisture or other chemically reactive conditions, for at least a week.
[0037] The term "aliphatic" or "aliphatic group", as used herein, means a straight-chain (i.e., unbranched), branched, or cyclic, substituted or unsubstituted hydrocarbon chain that is completely saturated or that contains one or more units of unsaturation that has a single point of attachment to the rest of the molecule.
[0038] Unless otherwise specified, aliphatic groups contain 1-20 aliphatic carbon atoms. In some embodiments, aliphatic groups contain 1-10 aliphatic carbon atoms. In other embodiments, aliphatic groups contain 1-8 aliphatic carbon atoms. In still other
embodiments, aliphatic groups contain 1-6 aliphatic carbon atoms, and in yet other embodiments aliphatic groups contain 1-4 aliphatic carbon atoms. Aliphatic groups may be linear or branched, substituted or unsubstituted alkyl, alkenyl, or alkynyl groups. Specific examples include, but are not limited to, methyl, ethyl, isopropyl, n-propyl, sec -butyl, vinyl, n-butenyl, ethynyl, and tert-butyl. Aliphatic groups may also be cyclic, or have a combination of linear or branched and cyclic groups. Examples of such types of aliphatic groups include, but are not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, -CH2- cyclopropyl, CH2CH2CH(CH3)-cyclohexyl.
[0039] The term "cycloaliphatic" (or "carbocycle" or "carbocyclyl") refers to a monocyclic C3-C8 hydrocarbon or bicyclic C8-C12 hydrocarbon that is completely saturated or that contains one or more units of unsaturation, but which is not aromatic, that has a single point of attachment to the rest of the molecule wherein any individual ring in said bicyclic ring system has 3-7 members. Examples of cycloaliphatic groups include, but are not limited to, cycloalkyl and cycloalkenyl groups. Specific examples include, but are not limited to, cyclohexyl, cyclopropenyl, and cyclobutyl.
[0040] The term "heterocycle", "heterocyclyl", or "heterocyclic" as used herein means non-aromatic, monocyclic, bicyclic, or tricyclic ring systems in which one or more ring members are an independently selected heteroatom. In some embodiments, the
"heterocycle", "heterocyclyl", or "heterocyclic" group has three to fourteen ring members in which one or more ring members is a heteroatom independently selected from oxygen, sulfur, nitrogen, or phosphorus, and each ring in the system contains 3 to 7 ring members.
[0041] Examples of heterocycles include, but are not limited to, 3-lH-benzimidazol-2- one, 3-(l-alkyl)-benzimidazol-2-one, 2-tetrahydrofuranyl, 3-tetrahydrofuranyl, 2- tetrahydrothiophenyl, 3-tetrahydrothiophenyl, 2-morpholino, 3-morpholino, 4-morpholino, 2- thiomorpholino, 3-thiomorpholino, 4-thiomorpholino, 1 -pyrrolidinyl, 2-pyrrolidinyl, 3- pyrrolidinyl, 1 -tetrahydropiperazinyl, 2-tetrahydropiperazinyl, 3-tetrahydropiperazinyl, 1- piperidinyl, 2-piperidinyl, 3-piperidinyl, 1-pyrazolinyl, 3-pyrazolinyl, 4-pyrazolinyl, 5- pyrazolinyl, 1 -piperidinyl, 2-piperidinyl, 3-piperidinyl, 4-piperidinyl, 2-thiazolidinyl, 3- thiazolidinyl, 4-thiazolidinyl, 1 -imidazolidinyl, 2-imidazolidinyl, 4-imidazolidinyl, 5- imidazolidinyl, indolinyl, tetrahydroquinolinyl, tetrahydroisoquinolinyl, benzothiolane, benzodithiane, and l,3-dihydro-imidazol-2-one.
[0042] Cyclic groups, (e.g. cycloaliphatic and heterocycles), can be linearly fused, bridged, or spirocyclic.
[0043] The term "heteroatom" means one or more of oxygen, sulfur, nitrogen, phosphorus, or silicon (including, any oxidized form of nitrogen, sulfur, phosphorus, or silicon; the quaternized form of any basic nitrogen or; a substitutable nitrogen of a heterocyclic ring, for example N (as in 3,4-dihydro-2H-pyrrolyl), ΝΗ (as in pyrrolidinyl) or NR+ (as in N-substituted pyrrolidinyl)). [0044] The term "unsaturated", as used herein, means that a moiety has one or more units of unsaturation. As would be known by one of skill in the art, unsaturated groups can be partially unsaturated or fully unsaturated. Examples of partially unsaturated groups include, but are not limited to, butene, cyclohexene, and tetrahydropyridine. Fully unsaturated groups can be aromatic, anti-aromatic, or non-aromatic. Examples of fully unsaturated groups include, but are not limited to, phenyl, cyclooctatetraene, pyridyl, thienyl, and 1- methylpyridin-2(lH)-one.
[0045] The term "alkoxy", or "thioalkyl", as used herein, refers to an alkyl group, as previously defined, attached through an oxygen ("alkoxy") or sulfur ("thioalkyl") atom.
[0046] The terms "haloalkyl", "haloalkenyl", "haloaliphatic", and "haloalkoxy" mean alkyl, alkenyl or alkoxy, as the case may be, substituted with one or more halogen atoms. This term includes perfluorinated alkyl groups, such as -CF3 and -CF2CF3.
[0047] The terms "halogen", "halo", and "hal" mean F, CI, Br, or I.
[0048] The term "aryl" used alone or as part of a larger moiety as in "aralkyl",
"aralkoxy", or "aryloxyalkyl", refers to monocyclic, bicyclic, and tricyclic ring systems having a total of five to fourteen ring members, wherein at least one ring in the system is aromatic and wherein each ring in the system contains 3 to 7 ring members. The term "aryl" may be used interchangeably with the term "aryl ring".
[0049] The term "heteroaryl", used alone or as part of a larger moiety as in
"heteroaralkyl" or "heteroarylalkoxy", refers to monocyclic, bicyclic, and tricyclic ring systems having a total of five to fourteen ring members, wherein at least one ring in the system is aromatic, at least one ring in the system contains one or more heteroatoms, and wherein each ring in the system contains 3 to 7 ring members. The term "heteroaryl" may be used interchangeably with the term "heteroaryl ring" or the term "heteroaromatic". Examples of heteroaryl rings include, but are not limited to, 2-furanyl, 3-furanyl, N-imidazolyl, 2- imidazolyl, 4-imidazolyl, 5-imidazolyl, benzimidazolyl, 3-isoxazolyl, 4-isoxazolyl, 5- isoxazolyl, 2-oxazolyl, 4-oxazolyl, 5-oxazolyl, N-pyrrolyl, 2-pyrrolyl, 3-pyrrolyl, 2-pyridyl, 3-pyridyl, 4-pyridyl, 2-pyrimidinyl, 4-pyrimidinyl, 5-pyrimidinyl, pyridazinyl (e.g., 3- pyridazinyl), 2-thiazolyl, 4-thiazolyl, 5-thiazolyl, tetrazolyl (e.g., 5-tetrazolyl), triazolyl (e.g., 2-triazolyl and 5-triazolyl), 2-thienyl, 3-thienyl, benzofuryl, benzothiophenyl, indolyl (e.g., 2- indolyl), pyrazolyl (e.g., 2-pyrazolyl), isothiazolyl, 1,2,3-oxadiazolyl, 1,2,5-oxadiazolyl, 1,2,4-oxadiazolyl, 1,2,3 -triazolyl, 1,2,3-thiadiazolyl, 1,3,4-thiadiazolyl, 1,2,5-thiadiazolyl, purinyl, pyrazinyl, 1,3,5-triazinyl, quinolinyl (e.g., 2-quinolinyl, 3-quinolinyl, 4-quinolinyl), and isoquinolinyl (e.g., 1-isoquinolinyl, 3-isoquinolinyl, or 4-isoquinolinyl). [0050] It shall be understood that the term "heteroaryl" includes certain types of heteroaryl rings that exist in equilibrium between two different forms. More specifically, for example, species such hydropyridine and pyridinone (and likewise hydroxypyrimidine and pyrimidinone) are meant to be encompassed within the definition of "heteroaryl."
Figure imgf000021_0001
[0051] The term "protecting group" and "protective group" as used herein, are interchangeable and refer to an agent used to temporarily block one or more desired functional groups in a compound with multiple reactive sites. In certain embodiments, a protecting group has one or more, or preferably all, of the following characteristics: a) is added selectively to a functional group in good yield to give a protected substrate that is b) stable to reactions occurring at one or more of the other reactive sites; and c) is selectively removable in good yield by reagents that do not attack the regenerated, deprotected functional group. As would be understood by one skilled in the art, in some cases, the reagents do not attack other reactive groups in the compound. In other cases, the reagents may also react with other reactive groups in the compound. Examples of protecting groups are detailed in Greene, T.W., Wuts, P. G in "Protective Groups in Organic Synthesis", Third Edition, John Wiley & Sons, New York: 1999 (and other editions of the book), the entire contents of which are hereby incorporated by reference. The term "nitrogen protecting group", as used herein, refers to an agent used to temporarily block one or more desired nitrogen reactive sites in a multifunctional compound. Preferred nitrogen protecting groups also possess the characteristics exemplified for a protecting group above, and certain exemplary nitrogen protecting groups are also detailed in Chapter 7 in Greene, T.W., Wuts, P. G in "Protective Groups in Organic Synthesis", Third Edition, John Wiley & Sons, New York: 1999, the entire contents of which are hereby incorporated by reference.
[0052] In some embodiments, a methylene unit of an alkyl or aliphatic chain is optionally replaced with another atom or group. Examples of such atoms or groups include, but are not limited to, nitrogen, oxygen, sulfur, -C(O)-, -C(=N-CN)-, -C(=NR)-, -C(=NOR)-, -SO-, and -S02-. These atoms or groups can be combined to form larger groups. Examples of such larger groups include, but are not limited to, -OC(O)-, -C(0)CO-, -C02-, -C(0)NR-, -C(=N- CN), -NRCO-, -NRC(0)0-, -S02NR-, -NRS02-, -NRC(0)NR-, -OC(0)NR-, and
-NRS02NR-, wherein R is, for example, H or Ci_6aliphatic. It should be understood that these groups can be bonded to the methylene units of the aliphatic chain via single, double, or triple bonds. An example of an optional replacement (nitrogen atom in this case) that is bonded to the aliphatic chain via a double bond would be -CH2CH=N-CH3. In some cases, especially on the terminal end, an optional replacement can be bonded to the aliphatic group via a triple bond. One example of this would be CH2CH2CH2C=N. It should be understood that in this situation, the terminal nitrogen is not bonded to another atom.
[0053] It should also be understood that, the term "methylene unit" can also refer to branched or substituted methylene units. For example, in an isopropyl moiety [-CH(CH3)2], a nitrogen atom (e.g. NR) replacing the first recited "methylene unit" would result in dimethylamine [-N(CH3)2]. In instances such as these, one of skill in the art would understand that the nitrogen atom will not have any additional atoms bonded to it, and the "R" from "NR" would be absent in this case.
[0054] Unless otherwise indicated, the optional replacements form a chemically stable compound. Optional replacements can occur both within the chain and/or at either end of the chain; i.e. both at the point of attachment and/or also at the terminal end. Two optional replacements can also be adjacent to each other within a chain so long as it results in a chemically stable compound. For example, a C3 aliphatic can be optionally replaced by 2 nitrogen atoms to form -C-N=N. The optional replacements can also completely replace all of the carbon atoms in a chain. For example, a C3 aliphatic can be optionally replaced by -NR-, -C(O)-, and -NR- to form -NRC(0)NR- (a urea).
[0055] Unless otherwise indicated, if the replacement occurs at the terminal end, the replacement atom is bound to a hydrogen atom on the terminal end. For example, if a methylene unit of -CH2CH2CH3 were optionally replaced with -0-, the resulting compound could be -OCH2CH3, -CH2OCH3, or -CH2CH2OH. It should be understood that if the terminal atom does not contain any free valence electrons, then a hydrogen atom is not required at the terminal end (e.g., -CH2CH2CH=0 or -CH2CH2C≡N).
[0056] Unless otherwise indicated, structures depicted herein are also meant to include all isomeric (e.g., enantiomeric, diastereomeric, geometric, conformational, and rotational) forms of the structure. For example, the R and S configurations for each asymmetric center, (Z) and (E) double bond isomers, and (Z) and (E) conformational isomers are included in this invention. As would be understood to one skilled in the art, a substituent can freely rotate around any rotatable bonds. For example, a substituent drawn as
Figure imgf000023_0001
also
Figure imgf000023_0002
represents
[0057] Therefore, single stereochemical isomers as well as enantiomeric, diastereomeric, geometric, conformational, and rotational mixtures of the present compounds are within the scope of the invention.
[0058] Unless otherwise indicated, all tautomeric forms of the compounds of the invention are within the scope of the invention.
[0059] Additionally, unless otherwise indicated, structures depicted herein are also meant to include compounds that differ only in the presence of one or more isotopically enriched atoms. For example, compounds having the present structures except for the replacement of hydrogen by deuterium or tritium, or the replacement of a carbon by a 13C- or 14C-enriched carbon are within the scope of this invention. Such compounds are useful, for example, as analytical tools or probes in biological assays.
Pharmaceutically Acceptable Salts
[0060] The compounds of this invention can exist in free form for treatment, or where appropriate, as a pharmaceutically acceptable salt.
[0061] A "pharmaceutically acceptable salt" means any non-toxic salt of a compound of this invention that, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this invention or an inhibitorily active metabolite or residue thereof. As used herein, the term "inhibitorily active metabolite or residue thereof means that a metabolite or residue thereof is also an inhibitor of the ATR protein kinase.
[0062] Pharmaceutically acceptable salts are well known in the art. For example, S. M. Berge et ah, describe pharmaceutically acceptable salts in detail in J. Pharmaceutical Sciences, 1977, 66, 1-19, incorporated herein by reference. Pharmaceutically acceptable salts of the compounds of this invention include those derived from suitable inorganic and organic acids and bases. These salts can be prepared in situ during the final isolation and purification of the compounds. Acid addition salts can be prepared by 1) reacting the purified compound in its free-based form with a suitable organic or inorganic acid and 2) isolating the salt thus formed. [0063] Examples of pharmaceutically acceptable, nontoxic acid addition salts are salts of an amino group formed with inorganic acids such as hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid and perchloric acid or with organic acids such as acetic acid, oxalic acid, maleic acid, tartaric acid, citric acid, succinic acid or malonic acid or by using other methods used in the art such as ion exchange. Other pharmaceutically acceptable salts include adipate, alginate, ascorbate, aspartate, benzenesulfonate, benzoate, bisulfate, borate, butyrate, camphorate, camphorsulfonate, citrate, cyclopentanepropionate, digluconate, dodecylsulfate, ethanesulfonate, formate, fumarate, glucoheptonate, glycerophosphate, glycolate, gluconate, glycolate, hemisulfate, heptanoate, hexanoate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxy-ethanesulfonate, lactobionate, lactate, laurate, lauryl sulfate, malate, maleate, malonate, methanesulfonate, 2-naphthalenesulfonate, nicotinate, nitrate, oleate, oxalate, palmitate, palmoate, pectinate, persulfate, 3-phenylpropionate, phosphate, picrate, pivalate, propionate, salicylate, stearate, succinate, sulfate, tartrate, thiocyanate, p-toluenesulfonate, undecanoate, valerate salts, and the like.
[0064] Base addition salts can be prepared by 1) reacting the purified compound in its acid form with a suitable organic or inorganic base and 2) isolating the salt thus formed. Salts derived from appropriate bases include alkali metal (e.g., sodium, lithium, and potassium), alkaline earth metal (e.g., magnesium and calcium), ammonium and +(Ci_ 4alkyl)4 salts. This invention also envisions the quaternization of any basic nitrogen- containing groups of the compounds disclosed herein. Water or oil-soluble or dispersible products may be obtained by such quaternization.
[0065] Further pharmaceutically acceptable salts include, when appropriate, nontoxic ammonium, quaternary ammonium, and amine cations formed using counterions such as halide, hydroxide, carboxylate, sulfate, phosphate, nitrate, loweralkyl sulfonate and aryl sulfonate. Other acids and bases, while not in themselves pharmaceutically acceptable, may be employed in the preparation of salts useful as intermediates in obtaining the compounds of the invention and their pharmaceutically acceptable acid or base addition salts.
Abbreviations
[0066] The following abbreviations are used:
DMSO dimethyl sulfoxide
ATP adenosine triphosphate
1HNMR proton nuclear magnetic resonance
HPLC high performance liquid chromatography
LCMS liquid chromatography-mass spectrometry TLC thin layer chromatography
Rt retention time
Compound Uses
[0067] One aspect of this invention provides compounds that are inhibitors of ATR kinase, and thus are useful for treating or lessening the severity of a disease, condition, or disorder where ATR is implicated in the disease, condition, or disorder.
[0068] Another aspect of this invention provides compounds that are useful for the treatment of diseases, disorders, and conditions characterized by excessive or abnormal cell proliferation. Such diseases include, a proliferative or hyperproliferative disease. Examples of proliferative and hyperproliferative diseases include, without limitation, cancer and myeloproliferative disorders.
[0069] In some embodiments, said compounds are selected from the group consisting of a compound of formula I. Te term "cancer" includes, but is not limited to the following cancers. Oral: buccal cavity, lip, tongue, mouth, pharynx; Cardiac: sarcoma (angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma), myxoma, rhabdomyoma, fibroma, lipoma and teratoma; Lung: bronchogenic carcinoma (squamous cell or epidermoid, undifferentiated small cell, undifferentiated large cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hamartoma, mesothelioma;
Gastrointestinal: esophagus (squamous cell carcinoma, larynx, adenocarcinoma,
leiomyosarcoma, lymphoma), stomach (carcinoma, lymphoma, leiomyosarcoma), pancreas (ductal adenocarcinoma, insulinoma, glucagonoma, gastrinoma, carcinoid tumors, vipoma), small bowel or small intestines (adenocarcinoma, lymphoma, carcinoid tumors, Karposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, fibroma), large bowel or large intestines (adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, leiomyoma), colon, colon-rectum, colorectal; rectum, Genitourinary tract: kidney (adenocarcinoma, Wilm's tumor [nephroblastoma], lymphoma, leukemia), bladder and urethra (squamous cell carcinoma, transitional cell carcinoma, adenocarcinoma), prostate (adenocarcinoma, sarcoma), testis (seminoma, teratoma, embryonal carcinoma, teratocarcinoma,
choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma); Liver: hepatoma (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, hemangioma, biliary passages; Bone: osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Ewing's sarcoma, malignant lymphoma (reticulum cell sarcoma), multiple myeloma, malignant giant cell tumor chordoma, osteochronfroma (osteocartilaginous exostoses), benign chondroma, chondroblastoma, chondromyxofibroma, osteoid osteoma and giant cell tumors; Nervous system: skull (osteoma, hemangioma, granuloma, xanthoma, osteitis deformans), meninges (meningioma, meningiosarcoma, gliomatosis), brain
(astrocytoma, medulloblastoma, glioma, ependymoma, germinoma [pinealoma], glioblastoma multiform, oligodendroglioma, schwannoma, retinoblastoma, congenital tumors), spinal cord neurofibroma, meningioma, glioma, sarcoma); Gynecological: uterus (endometrial carcinoma), cervix (cervical carcinoma, pre-tumor cervical dysplasia), ovaries (ovarian carcinoma [serous cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma], granulosa-thecal cell tumors, Sertoli-Leydig cell tumors, dysgerminoma, malignant teratoma), vulva (squamous cell carcinoma, intraepithelial carcinoma,
adenocarcinoma, fibrosarcoma, melanoma), vagina (clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma (embryonal rhabdomyosarcoma), fallopian tubes (carcinoma), breast; Hematologic: blood (myeloid leukemia [acute and chronic], acute lymphoblastic leukemia, chronic lymphocytic leukemia, myeloproliferative diseases, multiple myeloma, myelodysplastic syndrome), Hodgkin's disease, non-Hodgkin's lymphoma [malignant lymphoma] hairy cell; lymphoid disorders; Skin: malignant melanoma, basal cell carcinoma, squamous cell carcinoma, Karposi's sarcoma, keratoacanthoma, moles dysplastic nevi, lipoma, angioma, dermatofibroma, keloids, psoriasis, Thyroid gland: papillary thyroid carcinoma, follicular thyroid carcinoma; undifferentiated thyroid cancer, medullary thyroid carcinoma, multiple endocrine neoplasia type 2A, multiple endocrine neoplasia type 2B, familial medullary thyroid cancer, pheochromocytoma, paraganglioma; and Adrenal glands: neuroblastoma.
[0070] Thus, the term "cancerous cell" as provided herein, includes a cell afflicted by any one of the above-identified conditions. In some embodiments, the cancer is selected from colorectal, thyroid, or breast cancer.
[0071] The term "myeloproliferative disorders", includes disorders such as polycythemia vera, thrombocythemia, myeloid metaplasia with myelofibrosis, hypereosinophilic syndrome, juvenile myelomonocytic leukemia, systemic mast cell disease, and hematopoietic disorders, in particular, acute-myelogenous leukemia (AML), chronic-myelogenous leukemia (CML), acute-promyelocytic leukemia (APL), and acute lymphocytic leukemia (ALL). Pharmaceutically Acceptable Derivatives or Prodrugs
[0072] In addition to the compounds of this invention, pharmaceutically acceptable derivatives or prodrugs of the compounds of this invention may also be employed in compositions to treat or prevent the herein identified disorders.
[0073] The compounds of this invention can also exist as pharmaceutically acceptable derivatives.
[0074] A "pharmaceutically acceptable derivative" is an adduct or derivative which, upon administration to a patient in need, is capable of providing, directly or indirectly, a compound as otherwise described herein, or a metabolite or residue thereof. Examples of
pharmaceutically acceptable derivatives include, but are not limited to, esters and salts of such esters.
[0075] A "pharmaceutically acceptable derivative or prodrug" means any
pharmaceutically acceptable ester, salt of an ester or other derivative or salt thereof of a compound, of this invention which, upon administration to a recipient, is capable of providing, either directly or indirectly, a compound of this invention or an inhibitorily active metabolite or residue thereof. Particularly favoured derivatives or prodrugs are those that increase the bioavailability of the compounds of this invention when such compounds are administered to a patient (e.g., by allowing an orally administered compound to be more readily absorbed into the blood) or which enhance delivery of the parent compound to a biological compartment (e.g., the brain or lymphatic system) relative to the parent species.
[0076] Pharmaceutically acceptable prodrugs of the compounds of this invention include, without limitation, esters, amino acid esters, phosphate esters, metal salts and sulfonate esters.
Pharmaceutical Compositions
[0077] The present invention also provides compounds and compositions that are useful as inhibitors of ATR kinase.
[0078] One aspect of this invention provides pharmaceutically acceptable compositions that comprise any of the compounds as described herein, and optionally comprise a pharmaceutically acceptable carrier, adjuvant or vehicle.
[0079] The pharmaceutically acceptable carrier, adjuvant, or vehicle, as used herein, includes any and all solvents, diluents, or other liquid vehicle, dispersion or suspension aids, surface active agents, isotonic agents, thickening or emulsifying agents, preservatives, solid binders, lubricants and the like, as suited to the particular dosage form desired. Remington's Pharmaceutical Sciences, Sixteenth Edition, E. W. Martin (Mack Publishing Co., Easton, Pa., 1980) discloses various carriers used in formulating pharmaceutically acceptable
compositions and known techniques for the preparation thereof. Except insofar as any conventional carrier medium is incompatible with the compounds of the invention, such as by producing any undesirable biological effect or otherwise interacting in a deleterious manner with any other component(s) of the pharmaceutically acceptable composition, its use is contemplated to be within the scope of this invention.
[0080] Some examples of materials which can serve as pharmaceutically acceptable carriers include, but are not limited to, ion exchangers, alumina, aluminum stearate, lecithin, serum proteins, such as human serum albumin, buffer substances such as phosphates, glycine, sorbic acid, or potassium sorbate, partial glyceride mixtures of saturated vegetable fatty acids, water, salts or electrolytes, such as protamine sulfate, disodium hydrogen phosphate, potassium hydrogen phosphate, sodium chloride, zinc salts, colloidal silica, magnesium trisilicate, polyvinyl pyrrolidone, polyacrylates, waxes, polyethylene-polyoxypropylene- block polymers, wool fat, sugars such as lactose, glucose and sucrose; starches such as corn starch and potato starch; cellulose and its derivatives such as sodium carboxymethyl cellulose, ethyl cellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients such as cocoa butter and suppository waxes; oils such as peanut oil, cottonseed oil; safflower oil; sesame oil; olive oil; corn oil and soybean oil; glycols; such a propylene glycol or polyethylene glycol; esters such as ethyl oleate and ethyl laurate; agar; buffering agents such as magnesium hydroxide and aluminum hydroxide; alginic acid; pyrogen-free water; isotonic saline; Ringer's solution; ethyl alcohol, and phosphate buffer solutions, as well as other non-toxic compatible lubricants such as sodium lauryl sulfate and magnesium stearate, as well as coloring agents, releasing agents, coating agents, sweetening, flavoring and perfuming agents, preservatives and antioxidants can also be present in the composition, according to the judgment of the formulator.
Combination Therapies
[0081] Another aspect of this invention is directed towards a method of treating cancer in a subject in need thereof, comprising administration of a compound of this invention or a pharmaceutically acceptable salt thereof, and an additional therapeutic agent. In some embodiments, said method comprises the sequential or co-administration of the compound or a pharmaceutically acceptable salt thereof, and the additional therapeutic agent. [0082] In some embodiments, said additional therapeutic agent is an anti-cancer agent. In other embodiments, said additional therapeutic agent is a DNA-damaging agent. In yet other embodiments, said additional therapeutic agent is selected from radiation therapy, chemotherapy, or other agents typically used in combination with radiation therapy or chemotherapy, such as radios ens itizers and chemosensitizers.
[0083] As would be known by one of skill in the art, radiosensitizers are agents that can be used in combination with radiation therapy. Radiosensitizers work in various different ways, including, but not limited to, making cancer cells more sensitive to radiation therapy, working in synergy with radiation therapy to provide an improved synergistic effect, acting additively with radiation therapy, or protecting surrounding healthy cells from damage caused by radiation therapy. Likewise chemosensitizers are agents that can be used in combination with chemotherapy. Similarly, chemosensitizers work in various different ways, including, but not limited to, making cancer cells more sensitive to chemotherapy, working in synergy with chemotherapy to provide an improved synergistic effect, acting additively to
chemotherapy, or protecting surrounding healthy cells from damage caused by chemotherapy.
[0084] Examples of DNA-damaging agents that may be used in combination with compounds of this invention include, but are not limited to Platinating agents, such as Carboplatin, Nedaplatin, Satraplatin and other derivatives; Topo I inhibitors, such as
Topotecan, irinotecan/SN38, rubitecan and other derivatives; Antimetabolites, such as Folic family (Methotrexate, Pemetrexed and relatives); Purine antagonists and Pyrimidine antagonists (Thioguanine, Fludarabine, Cladribine, Cytarabine, Gemcitabine,
6-Mercaptopurine, 5-Fluorouracil (5FU) and relatives); Alkylating agents, such as Nitrogen mustards (Cyclophosphamide, Melphalan, Chlorambucil, mechlorethamine, Ifosfamide and relatives); nitrosoureas (eg Carmustine); Triazenes (Dacarbazine, temozolomide); Alkyl sulphonates (eg Busulfan); Procarbazine and Aziridines; Antibiotics, such as Hydroxyurea, Anthracyclines (doxorubicin, daunorubicin, epirubicin and other derivatives);
Anthracenediones (Mitoxantrone and relatives); Streptomyces family (Bleomycin,
Mitomycin C, actinomycin); and Ultraviolet light.
[0085] Other therapies or anticancer agents that may be used in combination with the inventive agents of the present invention include surgery, radiotherapy (in but a few examples, gamma-radiation, neutron beam radiotherapy, electron beam radiotherapy, proton therapy, brachytherapy, and systemic radioactive isotopes, to name a few), endocrine therapy, biologic response modifiers (interferons, interleukins, and tumor necrosis factor (TNF) to name a few), hyperthermia and cryotherapy, agents to attenuate any adverse effects (e.g., antiemetics), and other approved chemotherapeutic drugs, including, but not limited to, the DNA damaging agents listed herein, spindle poisons (Vinblastine, Vincristine, Vinorelbine, Paclitaxel), podophyllotoxins (Etoposide, Irinotecan, Topotecan), nitrosoureas (Carmustine, Lomustine), inorganic ions (Cisplatin, Carboplatin), enzymes (Asparaginase), and hormones (Tamoxifen, Leuprolide, Flutamide, and Megestrol), Gleevec™, adriamycin, dexamethasone, and cyclophosphamide.
[0086] A compound of the instant invention may also be useful for treating cancer in combination with any of the following therapeutic agents: abarelix (Plenaxis depot®);
aldesleukin (Prokine®); Aldesleukin (Proleukin®); Alemtuzumabb (Campath®); alitretinoin (Panretin®); allopurinol (Zyloprim®); altretamine (Hexalen®); amifostine (Ethyol®);
anastrozole (Arimidex®); arsenic trioxide (Trisenox®); asparaginase (Elspar®); azacitidine (Vidaza®); bevacuzimab (Avastin®); bexarotene capsules (Targretin®); bexarotene gel (Targretin®); bleomycin (Blenoxane®); bortezomib (Velcade®); busulfan intravenous (Busulfex®); busulfan oral (Myleran®); calusterone (Methosarb®); capecitabine (Xeloda®); carboplatin (Paraplatin®); carmustine (BCNU®, BiCNU®); carmustine (Gliadel®);
carmustine with Polifeprosan 20 Implant (Gliadel Wafer®); celecoxib (Celebrex®);
cetuximab (Erbitux®); chlorambucil (Leukeran®); cisplatin (Platinol®); cladribine
(Leustatin®, 2-CdA®); clofarabine (Clolar®); cyclophosphamide (Cytoxan®, Neosar®); cyclophosphamide (Cytoxan Injection®); cyclophosphamide (Cytoxan Tablet®); cytarabine (Cytosar-U®); cytarabine liposomal (DepoCyt®); dacarbazine (DTIC-Dome®);
dactinomycin, actinomycin D (Cosmegen®); Darbepoetin alfa (Aranesp®); daunorubicin liposomal (DanuoXome®); daunorubicin, daunomycin (Daunorubicin®); daunorubicin, daunomycin (Cerubidine®); Denileukin diftitox (Ontak®); dexrazoxane (Zinecard®);
docetaxel (Taxotere®); doxorubicin (Adriamycin PFS®); doxorubicin (Adriamycin®, Rubex®); doxorubicin (Adriamycin PFS Injection®); doxorubicin liposomal (Doxil®); dromostanolone propionate (dromostanolone®); dromostanolone propionate (masterone injection®); Elliott's B Solution (Elliott's B Solution®); epirubicin (Ellence®); Epoetin alfa (epogen®); erlotinib (Tarceva®); estramustine (Emcyt®); etoposide phosphate
(Etopophos®); etoposide, VP- 16 (Vepesid®); exemestane (Aromasin®); Filgrastim
(Neupogen®); floxuridine (intraarterial) (FUDR®); fludarabine (Fludara®); fluorouracil, 5- FU (Adrucil®); fulvestrant (Faslodex®); gefitinib (Iressa®); gemcitabine (Gemzar®); gemtuzumab ozogamicin (Mylotarg®); goserelin acetate (Zoladex Implant®); goserelin acetate (Zoladex®); histrelin acetate (Histrelin implant®); hydroxyurea (Hydrea®);
Ibritumomab Tiuxetan (Zevalin®); idarubicin (Idamycin®); ifosfamide (IFEX®); imatinib mesylate (Gleevec®); interferon alfa 2a (Roferon A®); Interferon alfa-2b (Intron A®); irinotecan (Camptosar®); lenalidomide (Revlimid®); letrozole (Femara®); leucovorin (Wellcovorin®, Leucovorin®); Leuprolide Acetate (Eligard®); levamisole (Ergamisol®); lomustine, CCNU (CeeBU®); meclorethamine, nitrogen mustard (Mustargen®); megestrol acetate (Megace®); melphalan, L-PAM (Alkeran®); mercaptopurine, 6-MP (Purinethol®); mesna (Mesnex®); mesna (Mesnex tabs®); methotrexate (Methotrexate®); methoxsalen (Uvadex®); mitomycin C (Mutamycin®); mitotane (Lysodren®); mitoxantrone
(Novantrone®); nandrolone phenpropionate (Durabolin-50®); nelarabine (Arranon®);
Nofetumomab (Verluma®); Oprelvekin (Neumega®); oxaliplatin (Eloxatin®); paclitaxel (Paxene®); paclitaxel (Taxol®); paclitaxel protein-bound particles (Abraxane®); palifermin (Kepivance®); pamidronate (Aredia®); pegademase (Adagen (Pegademase Bovine)®); pegaspargase (Oncaspar®); Pegfilgrastim (Neulasta®); pemetrexed disodium (Alimta®); pentostatin (Nipent®); pipobroman (Vercyte®); plicamycin, mithramycin (Mithracin®); porfimer sodium (Photofrin®); procarbazine (Matulane®); quinacrine (Atabrine®);
Rasburicase (Elitek®); Rituximab (Rituxan®); sargramostim (Leukine®); Sargramostim (Prokine®); sorafenib (Nexavar®); streptozocin (Zanosar®); sunitinib maleate (Sutent®); talc (Sclerosol®); tamoxifen (Nolvadex®); temozolomide (Temodar®); teniposide, VM-26 (Vumon®); testolactone (Teslac®); thioguanine, 6-TG (Thioguanine®); thiotepa
(Thioplex®); topotecan (Hycamtin®); toremifene (Fareston®); Tositumomab (Bexxar®); Tositumomab/I-131 tositumomab (Bexxar®); Trastuzumab (Herceptin®); tretinoin, ATRA (Vesanoid®); Uracil Mustard (Uracil Mustard Capsules®); valrubicin (Valstar®); vinblastine (Velban®); vincristine (Oncovin®); vinorelbine (Navelbine®); zoledronate (Zometa®) and vorinostat (Zolinza®).
[0087] For a comprehensive discussion of updated cancer therapies see,
http://www.nci.nih.gov/, a list of the FDA approved oncology drugs at
http://www.fda.gov/cder/cancer/druglistframe.htm, and The Merck Manual, Seventeenth Ed. 1999, the entire contents of which are hereby incorporated by reference. Compositions for Administration into a Subject
[0088] The ATR kinase inhibitors or pharmaceutical salts thereof may be formulated into pharmaceutical compositions for administration to animals or humans. These pharmaceutical compositions, which comprise an amount of the ATR inhibitor effective to treat or prevent the diseases or conditions described herein and a pharmaceutically acceptable carrier, are another embodiment of the present invention.
[0089] The exact amount of compound required for treatment will vary from subject to subject, depending on the species, age, and general condition of the subject, the severity of the infection, the particular agent, its mode of administration, and the like. The compounds of the invention are preferably formulated in dosage unit form for ease of administration and uniformity of dosage. The expression "dosage unit form" as used herein refers to a physically discrete unit of agent appropriate for the patient to be treated. It will be understood, however, that the total daily usage of the compounds and compositions of the present invention will be decided by the attending physician within the scope of sound medical judgment. The specific effective dose level for any particular patient or organism will depend upon a variety of factors including the disorder being treated and the severity of the disorder; the activity of the specific compound employed; the specific composition employed; the age, body weight, general health, sex and diet of the patient; the time of administration, route of administration, and rate of excretion of the specific compound employed; the duration of the treatment; drugs used in combination or coincidental with the specific compound employed, and like factors well known in the medical arts. The term "patient", as used herein, means an animal, preferably a mammal, and most preferably a human.
[0090] In some embodiments, these compositions optionally further comprise one or more additional therapeutic agents. For example, chemotherapeutic agents or other antiproliferative agents may be combined with the compounds of this invention to treat proliferative diseases and cancer. Examples of known agents with which these compositions can be combined are listed above under the "Combination Therapies" section and also throughout the specification. Some embodiments provide a simultaneous, separate or sequential use of a combined preparation.
Modes of Administration and Dosage Forms
[0091] The pharmaceutically acceptable compositions of this invention can be administered to humans and other animals orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, or drops), bucally, as an oral or nasal spray, or the like, depending on the severity of the infection being treated. In certain embodiments, the compounds of the invention may be administered orally or parenterally at dosage levels of about 0.01 mg/kg to about 50 mg/kg and preferably from about 1 mg/kg to about 25 mg/kg, of subject body weight per day, one or more times a day, to obtain the desired therapeutic effect.
[0092] Liquid dosage forms for oral administration include, but are not limited to, pharmaceutically acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active compounds, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, dimethylformamide, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor, and sesame oils), glycerol, tetrahydrofurfuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof. Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
[0093] Injectable preparations, for example, sterile injectable aqueous or oleaginous suspensions may be formulated according to the known art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution, suspension or emulsion in a nontoxic parenterally acceptable diluent or solvent, for example, as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution, U.S. P. and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose any bland fixed oil can be employed including synthetic mono- or diglycerides. In addition, fatty acids such as oleic acid are used in the preparation of injectables.
[0094] The injectable formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions which can be dissolved or dispersed in sterile water or other sterile injectable medium prior to use.
[0095] In order to prolong the effect of a compound of the present invention, it is often desirable to slow the absorption of the compound from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. The rate of absorption of the compound then depends upon its rate of dissolution that, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally administered compound form is accomplished by dissolving or suspending the compound in an oil vehicle. Injectable depot forms are made by forming microencapsule matrices of the compound in biodegradable polymers such as polylactide-polyglycolide. Depending upon the ratio of compound to polymer and the nature of the particular polymer employed, the rate of compound release can be controlled. Examples of other biodegradable polymers include poly(orthoesters) and poly(anhydrides). Depot injectable formulations are also prepared by entrapping the compound in liposomes or microemulsions that are compatible with body tissues.
[0096] Compositions for rectal or vaginal administration are preferably suppositories which can be prepared by mixing the compounds of this invention with suitable non- irritating excipients or carriers such as cocoa butter, polyethylene glycol or a suppository wax which are solid at ambient temperature but liquid at body temperature and therefore melt in the rectum or vaginal cavity and release the active compound.
[0097] Solid dosage forms for oral administration include capsules, tablets, pills, powders, and granules. In such solid dosage forms, the active compound is mixed with at least one inert, pharmaceutically acceptable excipient or carrier such as sodium citrate or dicalcium phosphate and/or a) fillers or extenders such as starches, lactose, sucrose, glucose, mannitol, and silicic acid, b) binders such as, for example, carboxymethylcellulose, alginates, gelatin, polyvinylpyrrolidinone, sucrose, and acacia, c) humectants such as glycerol, d) disintegrating agents such as agar— agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate, e) solution retarding agents such as paraffin, f) absorption accelerators such as quaternary ammonium compounds, g) wetting agents such as, for example, cetyl alcohol and glycerol monostearate, h) absorbents such as kaolin and bentonite clay, and i) lubricants such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof. In the case of capsules, tablets and pills, the dosage form may also comprise buffering agents.
[0098] Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polyethylene glycols and the like. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings and other coatings well known in the pharmaceutical formulating art. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes. Solid compositions of a similar type may also be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugar as well as high molecular weight polethylene glycols and the like.
[0099] The active compounds can also be in microencapsulated form with one or more excipients as noted above. The solid dosage forms of tablets, dragees, capsules, pills, and granules can be prepared with coatings and shells such as enteric coatings, release controlling coatings and other coatings well known in the pharmaceutical formulating art. In such solid dosage forms the active compound may be admixed with at least one inert diluent such as sucrose, lactose or starch. Such dosage forms may also comprise, as is normal practice, additional substances other than inert diluents, e.g., tableting lubricants and other tableting aids such a magnesium stearate and microcrystalline cellulose. In the case of capsules, tablets and pills, the dosage forms may also comprise buffering agents. They may optionally contain opacifying agents and can also be of a composition that they release the active ingredient(s) only, or preferentially, in a certain part of the intestinal tract, optionally, in a delayed manner. Examples of embedding compositions that can be used include polymeric substances and waxes.
[00100] Dosage forms for topical or transdermal administration of a compound of this invention include ointments, pastes, creams, lotions, gels, powders, solutions, sprays, inhalants or patches. The active component is admixed under sterile conditions with a pharmaceutically acceptable carrier and any needed preservatives or buffers as may be required. Ophthalmic formulation, eardrops, and eye drops are also contemplated as being within the scope of this invention. Additionally, the present invention contemplates the use of transdermal patches, which have the added advantage of providing controlled delivery of a compound to the body. Such dosage forms can be made by dissolving or dispensing the compound in the proper medium. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate can be controlled by either providing a rate controlling membrane or by dispersing the compound in a polymer matrix or gel.
[00101] The compositions of the present invention may be administered orally, parenterally, by inhalation spray, topically, rectally, nasally, buccally, vaginally or via an implanted reservoir. The term "parenteral" as used herein includes, but is not limited to, subcutaneous, intravenous, intramuscular, intra-articular, intra-synovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques.
Preferably, the compositions are administered orally, intraperitoneally or intravenously. [00102] Sterile injectable forms of the compositions of this invention may be aqueous or oleaginous suspension. These suspensions may be formulated according to techniques known in the art using suitable dispersing or wetting agents and suspending agents. The sterile injectable preparation may also be a sterile injectable solution or suspension in a nontoxic parenterally-acceptable diluent or solvent, for example as a solution in 1,3-butanediol. Among the acceptable vehicles and solvents that may be employed are water, Ringer's solution and isotonic sodium chloride solution. In addition, sterile, fixed oils are conventionally employed as a solvent or suspending medium. For this purpose, any bland fixed oil may be employed including synthetic mono- or di-glycerides. Fatty acids, such as oleic acid and its glyceride derivatives are useful in the preparation of injectables, as are natural pharmaceutically-acceptable oils, such as olive oil or castor oil, especially in their polyoxyethylated versions. These oil solutions or suspensions may also contain a long-chain alcohol diluent or dispersant, such as carboxymethyl cellulose or similar dispersing agents which are commonly used in the formulation of pharmaceutically acceptable dosage forms including emulsions and suspensions. Other commonly used surfactants, such as Tweens, Spans and other emulsifying agents or bioavailability enhancers which are commonly used in the manufacture of pharmaceutically acceptable solid, liquid, or other dosage forms may also be used for the purposes of formulation.
[00103] The pharmaceutical compositions of this invention may be orally administered in any orally acceptable dosage form including, but not limited to, capsules, tablets, aqueous suspensions or solutions. In the case of tablets for oral use, carriers commonly used include, but are not limited to, lactose and corn starch. Lubricating agents, such as magnesium stearate, are also typically added. For oral administration in a capsule form, useful diluents include lactose and dried cornstarch. When aqueous suspensions are required for oral use, the active ingredient is combined with emulsifying and suspending agents. If desired, certain sweetening, flavoring or coloring agents may also be added.
[00104] Alternatively, the pharmaceutical compositions of this invention may be administered in the form of suppositories for rectal administration. These can be prepared by mixing the agent with a suitable non-irritating excipient that is solid at room temperature but liquid at rectal temperature and therefore will melt in the rectum to release the drug. Such materials include, but are not limited to, cocoa butter, beeswax and polyethylene glycols.
[00105] The pharmaceutical compositions of this invention may also be administered topically, especially when the target of treatment includes areas or organs readily accessible by topical application, including diseases of the eye, the skin, or the lower intestinal tract. Suitable topical formulations are readily prepared for each of these areas or organs.
[00106] Topical application for the lower intestinal tract can be effected in a rectal suppository formulation (see above) or in a suitable enema formulation. Topically- transdermal patches may also be used.
[00107] For topical applications, the pharmaceutical compositions may be formulated in a suitable ointment containing the active component suspended or dissolved in one or more carriers. Carriers for topical administration of the compounds of this invention include, but are not limited to, mineral oil, liquid petrolatum, white petrolatum, propylene glycol, polyoxyethylene, polyoxypropylene compound, emulsifying wax and water. Alternatively, the pharmaceutical compositions can be formulated in a suitable lotion or cream containing the active components suspended or dissolved in one or more pharmaceutically acceptable carriers. Suitable carriers include, but are not limited to, mineral oil, sorbitan monostearate, polysorbate 60, cetyl esters wax, cetearyl alcohol, 2-octyldodecanol, benzyl alcohol and water.
[00108] For ophthalmic use, the pharmaceutical compositions may be formulated as micronized suspensions in isotonic, pH adjusted sterile saline, or, preferably, as solutions in isotonic, pH adjusted sterile saline, either with or without a preservative such as
benzylalkonium chloride. Alternatively, for ophthalmic uses, the pharmaceutical compositions may be formulated in an ointment such as petrolatum.
[00109] The pharmaceutical compositions of this invention may also be administered by nasal aerosol or inhalation. Such compositions are prepared according to techniques well- known in the art of pharmaceutical formulation and may be prepared as solutions in saline, employing benzyl alcohol or other suitable preservatives, absorption promoters to enhance bioavailability, fluorocarbons, and/or other conventional solubilizing or dispersing agents.
[00110] The amount of protein kinase inhibitor that may be combined with the carrier materials to produce a single dosage form will vary depending upon the host treated, the particular mode of administration. Preferably, the compositions should be formulated so that a dosage of between 0.01 - 100 mg/kg body weight/day of the inhibitor can be administered to a patient receiving these compositions.
[00111] It should also be understood that a specific dosage and treatment regimen for any particular patient will depend upon a variety of factors, including the activity of the specific compound employed, the age, body weight, general health, sex, diet, time of administration, rate of excretion, drug combination, and the judgment of the treating physician and the severity of the particular disease being treated. The amount of inhibitor will also depend upon the particular compound in the composition.
Administering with another Agent
[00112] Depending upon the particular protein kinase-mediated conditions to be treated or prevented, additional drugs, which are normally administered to treat or prevent that condition, may be administered together with the compounds of this invention.
[00113] Those additional agents may be administered separately, as part of a multiple dosage regimen, from the protein kinase inhibitor-containing compound or composition. Alternatively, those agents may be part of a single dosage form, mixed together with the protein kinase inhibitor in a single composition.
[00114] Another aspect of this invention is directed towards a method of treating cancer in a subject in need thereof, comprising the sequential or co-administration of a compound of this invention or a pharmaceutically acceptable salt thereof, and an anti-cancer agent. In some embodiments, said anti-cancer agent is selected from Platinating agents, such as Cisplatin, Oxaliplatin, Carboplatin, Nedaplatin, or Satraplatin and other derivatives; Topo I inhibitors, such as Camptothecin, Topotecan, irinotecan/SN38, rubitecan and other derivatives; Antimetabolites, such as Folic family (Methotrexate, Pemetrexed and relatives); Purine family (Thioguanine, Fludarabine, Cladribine, 6-Mercaptopurine and relatives); Pyrimidine family (Cytarabine, Gemcitabine, 5-Fluorouracil and relatives); Alkylating agents, such as Nitrogen mustards (Cyclophosphamide, Melphalan, Chlorambucil, mechlorethamine, Ifosfamide, and relatives); nitrosoureas (e.g. Carmustine); Triazenes (Dacarbazine, temozolomide); Alkyl sulphonates (e.g. Busulfan); Procarbazine and
Aziridines; Antibiotics, such as Hydroxyurea; Anthracyclines (doxorubicin, daunorubicin, epirubicin and other derivatives); Anthracenediones (Mitoxantrone and relatives);
Streptomyces family (Bleomycin, Mitomycin C, actinomycin) and Ultraviolet light.
Biological Samples
[00115] As inhibitors of ATR kinase, the compounds and compositions of this invention are also useful in biological samples. One aspect of the invention relates to inhibiting ATR kinase activity in a biological sample, which method comprises contacting said biological sample with a compound described herein or a composition comprising said compound. The term "biological sample", as used herein, means an in vitro or an ex vivo sample, including, without limitation, cell cultures or extracts thereof; biopsied material obtained from a mammal or extracts thereof; and blood, saliva, urine, feces, semen, tears, or other body fluids or extracts thereof. The term "compounds described herein" includes compounds of formula I.
[00116] Inhibition of ATR kinase activity in a biological sample is useful for a variety of purposes that are known to one of skill in the art. Examples of such purposes include, but are not limited to, blood transfusion, organ-transplantation, and biological specimen storage.
Study of Protein Kinases
[00117] Another aspect of this invention relates to the study of protein kinases in biological and pathological phenomena; the study of intracellular signal transduction pathways mediated by such protein kinases; and the comparative evaluation of new protein kinase inhibitors. Examples of such uses include, but are not limited to, biological assays such as enzyme assays and cell-based assays.
[00118] The activity of the compounds as protein kinase inhibitors may be assayed in vitro, in vivo or in a cell line. In vitro assays include assays that determine inhibition of either the kinase activity or ATPase activity of the activated kinase. Alternate in vitro assays quantitate the ability of the inhibitor to bind to the protein kinase and may be measured either by radiolabelling the inhibitor prior to binding, isolating the inhibitor/kinase complex and determining the amount of radiolabel bound, or by running a competition experiment where new inhibitors are incubated with the kinase bound to known radioligands. Detailed conditions for assaying a compound utilized in this invention as an inhibitor of ATR is set forth in the Examples below.
[00119] Another aspect of the invention provides a method for modulating enzyme activity by contacting a compound described herein with ATR kinase.
Methods of Treatment
[00120] In one aspect, the present invention provides a method for treating or lessening the severity of a disease, condition, or disorder where ATR kinase is implicated in the disease state. In another aspect, the present invention provides a method for treating or lessening the severity of an ATR kinase disease, condition, or disorder where inhibition of enzymatic activity is implicated in the treatment of the disease. In another aspect, this invention provides a method for treating or lessening the severity of a disease, condition, or disorder with compounds that inhibit enzymatic activity by binding to the ATR kinase. Another aspect provides a method for treating or lessening the severity of a kinase disease, condition, or disorder by inhibiting enzymatic activity of ATR kinase with an ATR kinase inhibitor. [00121] One aspect of the invention relates to a method of inhibiting ATR kinase activity in a patient, which method comprises administering to the patient a compound described herein, or a composition comprising said compound. In some embodiments, said method is used to treat or prevent a condition selected from proliferative and hyperproliferative diseases, such as cancer.
[00122] Another aspect of this invention provides a method for treating, preventing, or lessening the severity of proliferative or hyperproliferative diseases comprising administering an effective amount of a compound, or a pharmaceutically acceptable composition comprising a compound, to a subject in need thereof. In some embodiments, said subject is a patient. The term "patient", as used herein, means an animal, preferably a human.
[00123] In some embodiments, said method is used to treat or prevent cancer. In some embodiments, said method is used to treat or prevent a type of cancer with solid tumors. In yet another embodiment, said cancer is selected from the following cancers: Oral: buccal cavity, lip, tongue, mouth, pharynx; Cardiac: sarcoma (angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma), myxoma, rhabdomyoma, fibroma, lipoma and teratoma; Lung: bronchogenic carcinoma (squamous cell or epidermoid, undifferentiated small cell, undifferentiated large cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hamartoma, mesothelioma; Gastrointestinal: esophagus (squamous cell carcinoma, larynx, adenocarcinoma, leiomyosarcoma, lymphoma), stomach (carcinoma, lymphoma, leiomyosarcoma), pancreas (ductal adenocarcinoma, insulinoma, glucagonoma, gastrinoma, carcinoid tumors, vipoma), small bowel or small intestines (adenocarcinoma, lymphoma, carcinoid tumors, Karposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, fibroma), large bowel or large intestines
(adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, leiomyoma), colon, colon- rectum, colorectal; rectum, Genitourinary tract: kidney (adenocarcinoma, Wilm's tumor [nephroblastoma], lymphoma), bladder and urethra (squamous cell carcinoma, transitional cell carcinoma, adenocarcinoma), prostate (adenocarcinoma, sarcoma), testis (seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma); Liver: hepatoma
(hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, hemangioma, biliary passages; Bone: osteogenic sarcoma
(osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Ewing's sarcoma, malignant lymphoma (reticulum cell sarcoma), multiple myeloma, malignant giant cell tumor chordoma, osteochronfroma (osteocartilaginous exostoses), benign chondroma, chondroblastoma, chondromyxofibroma, osteoid osteoma and giant cell tumors; Nervous system: skull (osteoma, hemangioma, granuloma, xanthoma, osteitis deformans), meninges (meningioma, meningiosarcoma, gliomatosis), brain (astrocytoma, meduUoblastoma, glioma, ependymoma, germinoma [pinealoma], glioblastoma multiform, oligodendroglioma, schwannoma, retinoblastoma, congenital tumors), spinal cord neurofibroma, meningioma, glioma, sarcoma); Gynecological: uterus (endometrial carcinoma), cervix (cervical carcinoma, pre-tumor cervical dysplasia), ovaries (ovarian carcinoma [serous
cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma], granulosa- thecal cell tumors, Sertoli-Leydig cell tumors, dysgerminoma, malignant teratoma), vulva (squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, melanoma), vagina (clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma (embryonal rhabdomyosarcoma), fallopian tubes (carcinoma), breast; Skin: malignant melanoma, basal cell carcinoma, squamous cell carcinoma, Karposi's sarcoma,
keratoacanthoma, moles dysplastic nevi, lipoma, angioma, dermatofibroma, keloids, psoriasis, Thyroid gland: papillary thyroid carcinoma, follicular thyroid carcinoma;
medullary thyroid carcinoma, multiple endocrine neoplasia type 2A, multiple endocrine neoplasia type 2B, familial medullary thyroid cancer, pheochromocytoma, paraganglioma; and Adrenal glands: neuroblastoma.
[00124] In some embodiments, the cancer is selected from the cancers described herein. In some embodiments, said cancer is lung cancer, head and neck cancer, pancreatic cancer, gastric cancer, or brain cancer.
[00125] In certain embodiments, an "effective amount" of the compound or
pharmaceutically acceptable composition is that amount effective in order to treat said disease. The compounds and compositions, according to the method of the present invention, may be administered using any amount and any route of administration effective for treating or lessening the severity of said disease.
[00126] One aspect provides a method for inhibiting ATR in a patient comprising administering a compound described herein as described herein. Another embodiment provides a method of treating cancer comprising administering to a patient a compound described herein, wherein the variables are as defined herein.
[00127] Some embodiments comprising administering to said patient an additional therapeutic agent selected from a DNA-damaging agent; wherein said additional therapeutic agent is appropriate for the disease being treated; and said additional therapeutic agent is administered together with said compound as a single dosage form or separately from said compound as part of a multiple dosage form.
[00128] In some embodiments, said DNA-damaging agent is selected from ionizing radiation, radiomimetic neocarzinostatin, a platinating agent, a Topo I inhibitor, a Topo II inhibitor, an antimetabolite, an alkylating agent, an alkyl sulphonates, an antimetabolite, or an antibiotic. In other embodiments, said DNA-damaging agent is selected from ionizing radiation, a platinating agent, a Topo I inhibitor, a Topo II inhibitor, or an antibiotic.
[00129] Examples of Platinating agents include Cisplatin, Oxaliplatin, Carboplatin, Nedaplatin, Satraplatin and other derivatives. Other platinating agents include Lobaplatin, and Triplatin. Other platinating agents include Tetranitrate, Picoplatin, Satraplatin,
ProLindac and Aroplatin.
[00130] Examples of Topo I inhibitor include Camptothecin, Topotecan, irinotecan/SN38, rubitecan and other derivatives. Other Topo I inhibitors include Belotecan.
[00131] Examples of Topo II inhibitors include Etoposide, Daunorubicin, Doxorubicin,
Aclarubicin, Epirubicin, Idarubicin, Amrubicin, Pirarubicin, Valrubicin, Zorubicin and
Teniposide.
[00132] Examples of Antimetabolites include members of the Folic family, Purine family (purine antagonists), or Pyrimidine family (pyrimidine antagonists). Examples of the Folic family include methotrexate, pemetrexed and relatives; examples of the Purine family include Thioguanine, Fludarabine, Cladribine, 6-Mercaptopurine, and relatives; examples of the Pyrimidine family include Cytarabine, gemcitabine, 5-Fluorouracil (5FU) and relatives.
[00133] Some other specific examples of antimetabolites include Aminopterin,
Methotrexate, Pemetrexed, Raltitrexed, Pentostatin, Cladribine, Clofarabine, Fludarabine, Thioguanine, Mercaptopurine, Fluorouracil, Capecitabine, Tegafur, Carmofur, Floxuridine, Cytarabine, Gemcitabine, Azacitidine and Hydroxyurea.
[00134] Examples of alkylating agents include Nitrogen mustards, Triazenes, alkyl sulphonates, Procarbazine and Aziridines. Examples of Nitrogen mustards include
Cyclophosphamide, Melphalan, Chlorambucil and relatives; examples of nitrosoureas include Carmustine; examples of triazenes include Dacarbazine and temozolomide; examples of alkyl sulphonates include Busulfan.
[00135] Other specific examples of alkylating agents include Mechlorethamine,
Cyclophosphamide, Ifosfamide, Trofosfamide, Chlorambucil, Melphalan, Prednimustine, Bendamustine, Uramustine, Estramustine, Carmustine, Lomustine, Semustine, Fotemustine, Nimustine, Ranimustine, Streptozocin, Busulfan, Mannosulfan, Treosulfan, Carboquone, ThioTEPA, Triaziquone, Triethylenemelamine, Procarbazine, Dacarbazine, Temozolomide, Altretamine, Mitobronitol, Actinomycin, Bleomycin, Mitomycin and Plicamycin.
[00136] Examples of antibiotics include Mitomycin, Hydroxyurea; Anthracyclines, Anthracenediones, Streptomyces family. Examples of Anthracyclines include doxorubicin, daunorubicin, epirubicin and other derivatives; examples of Anthracenediones include Mitoxantrone and relatives; examples of Streptomyces family inclue Bleomycin, Mitomycin C, and actinomycin.
[00137] In certain embodiments, said platinating agent is Cisplatin or Oxaliplatin; said Topo I inhibitor is Camptothecin; said Topo II inhibitor is Etoposide; and said antibiotic is Mitomycin. In other embodiments, said platinating agent is selected from Cisplatin, Oxaliplatin, Carboplatin, Nedaplatin, or Satraplatin; said Topo I inhibitor is selected from Camptothecin, Topotecan, irinotecan/SN38, rubitecan; said Topo II inhibitor is selected from Etoposide; said antimetabolite is selected from a member of the Folic Family, the Purine Family, or the Pyrimidine Family; said alkylating agent is selected from nitrogen mustards, nitrosoureas, triazenes, alkyl sulfonates, Procarbazine, or aziridines; and said antibiotic is selected from Hydroxyurea, Anthracyclines, Anthracenediones, or Streptomyces family.
[00138] Another embodiment provides a method of promoting cell death in cancer cells comprising administering to a patient a compound described herein, , or a composition comprising said compound.
[00139] Yet another embodiment provides a method of preventing cell repair of DNA damage in cancer cells comprising administering to a patient a compound described herein, or a composition comprising said compound. Yet another embodiment provides a method of preventing cell repair caused by of DNA damage in cancer cells comprising administering to a patient a compound of formula I, or composition comprising said compound.
[00140] Another embodiment provides a method of sensitizing cells to DNA damaging agents comprising administering to a patient a compound described herein, or a composition comprising said compound.
[00141] In some embodiments, the method is used on a cancer cell having defects in the ATM signaling cascade. In some embodiments, said defect is altered expression or activity of one or more of the following: ATM, p53, CHK2, MRE1 1, RAD50, NBS 1, 53BP1, MDC1 or H2AX. In another embodiment, the cell is a cancer cell expressing DNA damaging oncogenes. In some embodiments, said cancer cell has altered expression or activity of one or more of the following: K-Ras, N-Ras, H-Ras, Raf, Myc, Mos, E2F, Cdc25A, CDC4, CDK2, Cyclin E, Cyclin A and Rb. [00142] Yet another embodiment provides use of a compound described herein as a radio- sensitizer or a chemo-sensitizer.
[00143] Yet other embodiment provides use of a compound of formula I as a single agent (monotherapy) for treating cancer. In some embodiments, the compounds of formula I are used for treating patients having cancer with a DNA-damage response (DDR) defect. In other embodiments, said defect is a mutation or loss of ATM, p53, CHK2, MREl 1, RAD50, NBS 1 , 53BP 1 , MDC1 , or H2AX.
SCHEMES
[00144] The compounds of the disclosure may be prepared in light of the specification using steps generally known to those of ordinary skill in the art. Those compounds may be analyzed by known methods, including but not limited to LCMS (liquid chromatography mass spectrometry) and NMR (nuclear magnetic resonance). Below are a set of generic schemes that illustrate generally how to prepare the compounds of the present disclosure. Scheme A
Sonagashira
Figure imgf000044_0001
A A-i A-ii
lodination
Figure imgf000044_0002
A-i A-iv A-v
Figure imgf000044_0003
A-vi A-vii A-viii |
[00145] Scheme A depicts a general method for making compounds of Formula I.
Compound A is functionalised via either an SNAr reaction or a range of known metal mediated reactions such as, but not limited to, Suzuki reactions, Stille couplings, Buchwald- Hartwig reactions and carbonylation reactions to give compounds of Formula A-i. Where appropriate, J substituents on Rl can undergo further functionalisation by known reactions such as, but not limited to, Mitsunobu reactions and acylation. These compounds are then brominated under standard conditions known to those skilled in the art such as, but not limited to, treatment with N-bromosuccinimide to give compounds of Formula A-ii. Reaction of these compounds under Sonagashira conditions with a suitable protected alkyne gives rise to compounds of the Formula A-iii. Suitable alkyne protecting groups PG include, but are not limited to, TMS, TES or TIPS. The protecting group PG can be removed under standard conditions known to those skilled in the art such as, but not limited to, treatment with aqueous base or TBAF to give compounds of Formula A-iv. Compound A-iv can be cyclised to the corresponding pyrrolo[2,3-b]pyrazine of Formula A-v using methods known to those skilled in the art such as, but not limited to heating with KOlBu in ΝΜΡ. Under certain circumstances, the protecting group PG of Compound A-iii can be removed concurrently with cyclisation to pyrrolo[2,3-b]pyrazine of Formula A-v, without need to isolate compounds of Formula A-iv. Compounds of Formula A-v can be iodinated under standard conditions known to those skilled in the art such as, but not limited to, treatment with IC1 to give compounds of Formula A-vi. Compounds of Formula A-vi are then protected with a suitable protecting group PG' such as, but not limited to Tosyl (p-toluenesulfonyl), to give compounds of Formula A-vii. These compounds are functionalised using a range of known metal mediated reactions such as, but not limited to, Sonagashira couplings, Negishi couplings, Suzuki reactions and Stille couplings to give compounds of Formula A-vii. Where appropriate, substituents on A (Z1, Z2, Z3 or Z4) can undergo further functionalisation by reactions known to those skilled in the art such as, but not limited to, Mitsunobu reactions, nucleophilic displacement and acylation. The protecting group PG' can be removed under standard conditions known to those skilled in the art such as, but not limited to, treatment with aqueous base or TBAF to give compounds of Formula I.
Scheme B
Figure imgf000046_0001
Functional Group
Interconversion
A-iii B-i B-ii
Deprotection
Figure imgf000046_0002
A-iv A-v
[00146] Scheme B depicts an alternative general method for making compounds of Formula A-iv and A-v where protection of the aminopyrazine motif allows for functional group transformation on the J substituents on Ri. Compounds of Formula A-iii are protected with a suitable amine protecting group PG" such as, but not limited to BOC (tert- butoxycarbonyl), to give compounds of Formula B-i. J substituents on Ri can undergo further functionalisation by known reactions such as, but not limited to, Mitsunobu reactions and acylation. Compounds of this example are then selectively deprotected under standard conditions known to those skilled in the art such as, but not limited to, treatment with aqueous base or TBAF to remove the alkyne protecting group PG' yielding compounds of Formula B-ii. Removal of the nitrogen protecting group PG" from compounds of Formula B- ii under standard conditions known to those skilled in the art such as, but not limited to, treatment with HCl or TFA gives rise to compounds of Formula A-iv. Compound A-iv can be cyclised to the corresponding pyrrolo[2,3-b]pyrazine of Formula A-vas described in Scheme A. Scheme C
P
Figure imgf000047_0001
A-vii C-i
Fu nctionalisation
Figure imgf000047_0002
A-vii i
[00147] Scheme C depicts an alternative method for the synthesis of compounds of Formula I. Compounds of Formula A-vii are transformed into the corresponding boronic acid (or ester) C-i under standard conditions known to those skilled in the art such as, but not limited to treatment with bis(pinacolato)diboron under Palladium catalysis. These compounds are functionalised under Suzuki conditions to give compounds of Formula A-viii. Where appropriate, substituents on A (Z1, Z2, Z3 or Z4) can undergo further functionalisation by reactions known to those skilled in the art such as, but not limited to, Mitsunobu reactions, nucleophilic displacement and acylation. The protecting group PG' can be removed under standard conditions known to those skilled in the art such as, but not limited to, treatment with aqueous base or TBAF to give compounds of Formula I.
Scheme D
P
Figure imgf000048_0001
A-vii i
Deprotection
Figure imgf000048_0002
D-ii
[00148] Scheme D depicts an alternative method for the synthesis of compounds of Formula I where parameter A can derive from an alkyne functional group. Compounds of Formula A-vii are reacted under Sonagashira conditions with a suitable protected alkyne to give rise to compounds of the Formula D-i. Suitable alkyne protecting groups PG'" include, but are not limited to, TMS, TES or TIPS. The protecting groups PG' and PG' " can be removed under standard conditions known to those skilled in the art such as, but not limited to, treatment with aqueous base or TBAF to give compounds of Formula D-ii. These compounds are further elaborated under reaction conditions known in the art to give compounds of Formula I.
Scheme E
Figure imgf000049_0001
D-ii E-i
Function alisation
Figure imgf000049_0002
E-ii
[00149] Scheme E depicts an alternative method for the synthesis of compounds of Formula I where parameter A can derive from an alkyne functional group where protection of the pyrrolo[2,3-b]pyrazine allows for further elaboration of the substituents on A.
Compounds are Formula D-ii are protected with a suitable protecting group PG' such as, but not limited to Tosyl (p-toluenesulfonyl), to give compounds of Formula E-i. These compounds are further elaborated under reaction conditions known in the art to give compounds of Formula E-ii. Where appropriate, substituents on A (Zl , Z2, Z3 opr Z4) can undergo further functionalisation by reactions known to those skilled in the art such as, but not limited to, Mitsunobu reactions, nucleophilic displacement and acylation. The protecting group PG' can then be removed under standard conditions known to those skilled in the art such as, but not limited to, treatment with aqueous base or TBAF to give compounds of Formula I.
EXAMPLES
[00150] In order that this invention be more fully understood, the following preparative and testing examples are set forth. These examples are for the purpose of illustration only and are not to be construed as limiting the scope of the invention in any way. H-NMR spectra were recorded at 400 MHz using a Bruker DPX 400 instrument. Mass spec, samples were analyzed on a MicroMass Quattro Micro mass spectrometer operated in single MS mode with electrospray ionization. Example 1; 2-(4-Isopropylsulfonylphenyl)-7-(3-pyridyl)-5H-pyrrolo [2,3-b] pyrazine (Compound 1-1)
SCHEME I
Figure imgf000050_0001
METHOD A:
Step 1: 5-(4-(Isopropylsulfonyl)phenyl)pyrazin-2-amine
Figure imgf000050_0002
[00151] 5-Bromopyrazin-2-amine (5 g, 28.74 mmol), (4-isopropylsulfonylphenyl)boronic acid (7.866 g, 34.49 mmol) and K3P04 (12.20 g, 57.48 mmol) were combined in MeCN (100 mL) / Water (25 mL) and Pd[P(tBu)3]2 (734.4 mg, 1.437 mmol) was added. The reaction was heated at 60 °C for 1 hour. The reaction mixture was cooled to ambient temperature and diluted with EtOAc and water. The layers were separated and the aqueous layer extracted with EtOAc (x 3). The combined organic layers were dried (MgS04), filtered and
concentrated in vacuo. The residue was triturated from DCM and isolated by filtration to give the sub-title compound as an orange solid (6.43 g, 76% Yield). ¾ NMR (400.0MHz, DMSO) δ 1.17 (d, 6H), 3.43 (sept, 1H), 6.86 (s, 2H), 7.87 (d, 2H), 8.00 (s, 1H), 8.20 (d, 2H) and 8.67 (s, 1H) ppm; MS (ES+) 278.2.
Step 2: 3-Bromo-5-(4-(isopropylsulfonyl)phenyl)pyrazin-2-amine
Figure imgf000051_0001
[00152] 5-(4-(Isopropylsulfonyl)phenyl)pyrazin-2-amine (10 g, 36.06 mmol) was dissolved in dry DMF (70 mL) and N-bromosuccinimide (6.418 g, 36.06 mmol) was added portionwise. The mixture was stirred at ambient temperature for 2 hours. The reaction mixture was poured into water (200 mL) and stirred for 5 minutes. The solid was isolated by filtration and washed with water. The wet solid was dissolved in ethyl acetate and any insoluble material removed by filtration. The aqueous layer was separated and the organic phase washed with saturated aqueous a2S203 (x 1) and dried (MgS04). The organic extracts were filtered through a plug of Florisil (200 mesh), eluting with ethyl acetate. The filtrate was concentrated to ca. 50 mL and resultant precipitate isolated by filtration and washed with Petroleum Ether. The solid was further dried under high vacuum to give the sub-title compound as a pale orange solid (8.0 g, 62% Yield). ). XH NMR (400.0MHz, DMSO) δ 1.17
(d, 6H), 3.41 - 3.49 (m, 1H), 7.16 (br s, 2H), 7.89 (d, 2H), 8.17 (d, 2H) and 8.76 (s, 1H) ppm; MS (ES+) 356.1. Step 3: 5-(4-Isopropylsulfonylphenyl)-3-(2-trimethylsilylethynyl)pyrazin-2-amine
Figure imgf000052_0001
[00153] Triethylamine (47.58 g, 65.54 mL, 470.2 mmol) was added to a stirred suspension of 3-bromo-5-(4-isopropylsulfonylphenyl)pyrazin-2-amine (33.5 g, 94.04 mmol) in anhydrous DMF (234.5 mL). Copper(I) iodide (2.148 g, 11.28 mmol) was then added followed by Pd(PPh3)4 (5.433 g, 4.702 mmol). The stirred mixture was cooled to 0 °C in an ice-bath and (trimethylsilyl)acetylene (12.01 g, 17.28 mL, 122.3 mmol) was added dropwise over 10 minutes. The ice bath was then removed and the mixture allowed to warm to ambient temperature over 2 hours. The mixture was poured into EtOAc/water (500 mL/300 mL) and the mixture was filtered through a pad of celite. The organic phase was separated and the aqueous layer extracted with EtOAc. The combined organics were washed with water (x 1) and brine (x 1), dried (MgS04), filtered and concentrated in vacuo. The residue was purified by column chromatography (ISCO Companion™, 750 g column, eluting with 0 to 15% DCM/EtOAc, dry-loaded) to give the sub-title product as a dark yellow solid (28.0 g, 78% Yield). ¾ NMR (400.0 MHz, DMSO) δ 0.15 (s, 9H, 1.03 (d, 6H), 3.29 (sept, 1H), 6.80 (br s, 2H), 7.74 (d, 2H), 8.05 (d, 2H) and 8.60 (s, 1H) ppm; MS (ES+) 375.1.
Step 4: tert-Butyl N-teri-butoxycarbonyl-N-[5-(4-isopropylsulfonylphi
trimethylsilylethynyl)pyrazin-2-yl]carbamate
Figure imgf000053_0001
[00154] Triethylamine (18.42 g, 25.37 niL, 182.0 mmol) was added to a stirred solution of 5-(4-isopropylsulfonylphenyl)-3-(24rimethylsilylethynyl)pyrazin-2-amine (34 g, 91.02 mmol) in DCM (1.020 L). DMAP (1.112 g, 9.102 mmol) was added followed by tert- butoxycarbonyl tert-butyl carbonate (59.60 g, 62.74 mL, 273.1 mmol) in portions. The reaction mixture was stirred at ambient temperature for 16 hours. The reaction mixture was poured into water (500 mL) and the organic layer separated. The organic layer was washed with brine (x 1), dried (MgS04), filtered and concentrated in vacuo. The residue was purified by silica gel chromatography eluting with 20-30% EtOAc/Petroleum Ether to give the subtitle product as an orange foam (42.6 g, 82% Yield). XH NMR (400.0 MHz, CDC13) δ 0.15 (s, 9H), 1.19 (d, 6H), 1.28 (s, 18H), 3.10 (sept, IH), 7.89 (d, 2H), 8.12 (d, 2H) and 8.75 (s, IH) ppm; MS (ES+) 574.1.
Step 5: teri-Butyl N-teri-butoxycarbonyl-N-[3-ethynyl-5-(4- isopropylsulfonylphenyl)pyrazin-2-yl]carbamate
Figure imgf000053_0002
[00155] tert-Butyl N-ter?-butoxycarbonyl-N-[5-(4-isopropylsulfonylphenyl)-3-(2- trimethylsilylethynyl)pyrazin-2-yl]carbamate (42.6 g, 74.25 mmol) was dissolved/suspended in methanol (510 mL) and 2M aqueous sodium carbonate (425.8 mL, 851.6 mmol) was added to the rapidly stirred mixture. An oil separated out and more methanol (100 mL) was added to dissolve. The mixture was stirred at ambient temperature for 1 hour. The slurry was poured into a mixture of EtO Ac/water (800 mL/lL). The organic phase was separated and the aqueous extracted with EtO Ac (2 x 250 mL). The combined organic extracts were washed with brine, dried (MgS04), filtered and concentrated in vacuo. The residue was slurried in 5% EtO Ac/Petroleum Ether and stirred for 90 minutes at ambient temperature. The precipitate was isolated by filtration, washed with Petroleum Ether and dried to give the sub-title product as a white solid (33.1 g, 89% Yield). 'H NMR (400.0 MHz, DMSO) δ 1.18 (d, 6H), 1.37 (s, 18H), 3.50 (sept, 1H), 4.99 (s, 1H), 8.03 (d, 2H), 8.44 (d, 2H) and 9.35 (s, 1H) ppm; MS (ES+) 502.1.
Step 6: 3-Ethynyl-5-(4-isopropylsulfonylphenyl)pyrazin-2-amine
Figure imgf000054_0001
[00156] TFA (34.45 g, 23.28 mL, 302.1 mmol) was added to a stirred solution of tert-butyl N-tert-butoxycarbonyl-N-[3-ethynyl-5-(4-isopropylsulfonylphenyl)pyrazin-2-yl]carbamate (5 g, 9.968 mmol) in DCM (200 mL) and the reaction mixture stirred at ambient temperature for 1.5 hours. The solvent was removed in vacuo and the residue re-dissolved in DCM and stirred with saturated aqueous aHC03 for 30 minutes. The layers were separated and the organic layer washed with saturated aqueous aHC03 (x 3). The combined aqueous layers were extracted with DCM (x 3) and the combined organic extracts dried (MgS04), filtered and concentrated in vacuo. The residue was triturated from DCM and precipitate isolated by filtration and dried under vacuum to give the title product as a beige solid (2.8 g, 93% Yield). XH NMR (400.0 MHz, DMSO) δ 1.18 (d, 6H), 3.44 (t, 1H), 4.76 (s, 1H), 7.01 (s, 2H), 7.89 (d, 2H), 8.20 (d, 2H) and 8.75 (s, 1H) ppm; MS (ES+) 302.12. Step 7: 2-(4-Isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazine
Figure imgf000055_0001
[00157] KO'Bu (2.041 g, 18.19 mmol) in anhydrous NMP (5 mL) was stirred at 80 °C while a solution of 3-ethynyl-5-(4-isopropylsulfonylphenyl)pyrazin-2-amine (2.7685 g, 9.095 mmol) in dry NMP (15 mL) was added slowly over 5 minutes. The resultant solution was stirred at 80°C for 1.5 hours. The reaction mixture was cooled to ambient temperature and the solvent concentrated in vacuo. The residue was diluted with EtOAc and washed with water (x 6). The organic layer was dried (MgSC^), filtered and concentrated in vacuo to give the subtitle compounds as a beige solid (1.88 g, 69% Yield). XH NMR (400.0 MHz, DMSO) δ H NMR (400.0 MHz, DMSO) δ 1.20 (d, 6H), 3.49 (sept, 1H), 6.74 (dd, 1H), 7.96 - 8.00 (m, 3H), 8.43 (d, 2H), 9.00 (s, 1H) and 12.25 (s, 1H) ppm; MS (ES+) 302.1.
Step 8: 7-Iodo-2-(4-isopropylsulfonylphenyl)-5H-pyrrolo [2,3-b]pyrazine
Figure imgf000055_0002
[00158] 2-(4-Isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazine (1 g, 3.318 mmol) was dissolved in anhydrous pyridine (20 mL) and cooled to 0 °C in an ice-bath. 1M ICl in DCM (3.484 mL, 3.484 mmol) was added slowly and the resultant solution stirred at 0 °C. After 5 hours a further portion of 1M ICl in DCM (3.484 mL, 3.484 mmol) was added and the reaction allowed to warm to ambient temperature over 16 hours. A further portion of 1M ICl in DCM (3.484 mL, 3.484 mmol) was added and the reaction stirred at ambient temperature for a further 30 minutes. The reaction mixture was concentrated in vacuo and the residue partitioned between EtOAc and saturated aqueous Na2C03/saturated aqueous a2S203 (1 : 1). The aqueous layer was extracted with EtOAc (100 mL) and the combined organic extracts washed with saturated aqueous Na2C03/saturated aqueous a2S203 (1 : 1) (5 x 50 mL), dried (MgS04), filtered and concentrated in vacuo. The residue was triturated from MeCN to give the sub-title compound as a pale orange solid (1.23 g, 87% Yield). *H NMR (400.0 MHz, DMSO) δ 1.21 (d, 6H), 3.49 (s, IH), 8.01 (d, 2H), 8.22 (s, IH), 8.46 (d, 2H), 9.03 (s, IH) and 12.71 (s, IH) ppm; MS (ES+) 428.0.
Step 9: 7-Iodo-2-(4-isopropylsulfonylphenyl)-5-(p-tolylsulfonyl)pyrrolo[2,3-b]pyrazine
Figure imgf000056_0001
[00159] A 60% dispersion of sodium hydride in mineral oil (130.2 mg, 3.398 mmol) was added slowly to a stirred solution of 7-iodo-2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3- b]pyrazine (1.21 g, 2.832 mmol) in DMF (10 mL) at 0 °C. The reaction mixture was stirred at this temperature for 20 minutes then tosyl chloride (539.9 mg, 2.832 mmol) was added and reaction mixture allowed to warm to ambient temperature over 16 hours. The reaction mixture was quenched by the slow addition of water (30 mL) and the mixture stirred for 20 minutes. The resultant precipitate was isolated by filtration, washed with water and dried under vacuum to give the sub-title product as a yellow solid (1.52 g, 92% Yield). XH NMR (400.0 MHz, DMSO) δ 1.22 - 1.19 (m, 6H), 2.36 (s, 3H), 3.50 - 3.45 (m, IH), 7.48 (d, 2H), 8.05 (q, 4H), 8.42 (d, 2H), 8.64 (s, IH), and 9.18 (s, IH) ppm; MS (ES+) 581.9.
Step 10: 2-(4-Isopropylsulfonylphenyl)-5-(p-tolylsulfonyl)-7-(3-pyridyl)pyrrolo[2,3- b]pyrazine
Figure imgf000057_0001
[00160] A mixture of 7-iodo-2-(4-isopropylsulfonylphenyl)-5-(p-tolylsulfonyl)pyrrolo[2,3- b]pyrazine (100 mg, 0.1720 mmol), pyridylboronic acid (25.37 mg, 0.2064 mmol) and potassium phosphate (46.12 mg, 0.3440 mmol) in acetonitrile (2 mL) / Water (500.0 μΚ) was degassed and placed under an atmosphere of nitrogen. Pd[P(lBu)3]2 (4.395 mg, 0.008600 mmol) was added and the reaction degassed and placed under an atmosphere of nitrogen and heated at 60 °C for 16 hours in a sealed tube. The reaction mixture was cooled to ambient temperature and diluted with EtO Ac/water. The layers were separated and the aqueous layer extracted with EtOAc ( x2). The combined organic extracts were washed with water (x 1), dried (MgS04) and concentrated in vacuo. The residue was purified by column
chromatography (ISCO Companion™, 12 g column, eluting with 2 to 20% EtOAc/DCM, loaded in DCM) and the product contating fractions concentrated in vacuo. The residue was triturated from MeCN and the resultant precipitate isolated by filtration to give the sub-title product as a cream solid (64 mg, 70% Yield). MS (ES+) 533.1.
Step 11: 2-(4-Isopropylsulfonylphenyl)-7-(3-pyridyl)-5H-pyrrolo[2,3-b]pyrazine
Figure imgf000057_0002
[00161] 2-(4-Isopropylsulfonylphenyl)-5-(p-tolylsulfonyl)-7-(3-pyridyl)pyrrolo[2,3- b]pyrazine (64 mg, 0.1202 mmol) was dissolved in THF (1.4 mL) and 1M LiOH (601.0 μί, 0.6010 mmol) was added. The reaction was stirred at ambient temperature for 2 hours. The reaction mixture was partitioned between EtO Ac/brine and the layers separated. The aqueous layer was extracted with EtO Ac (x 2) and the combined organic extracts dried (MgSC^), filtered and concentrated in vacuo. The residue was triturated from MeCN and the resultant precipitate isolated by filtration to give the title compound as a yellow solid (7.5 mg, 17% Yield). ¾ NMR (400.0 MHz, DMSO) δ 1.27 (d, 6H), 3.54 (d, 1H), 7.54 - 7.56 (m, 1H), 8.07 (d, 2H), 8.51 (dd, 1H), 8.56 (d, 2H), 8.71 (d, 2H), 9.11 (s, 1H), 9.58 (d, 1H) and 12.70 (br s, 1H) ppm; MS (ES+) 379.0.
The following compounds were prepared using procedure analogous to that described above in example 1 :
Cmpd
Compound Name
No.
1-5 2-(4-isopropylsulfonylphenyl)-7-phenyl-5H-pyrrolo[2,3-b]pyrazine
2-(4-isopropylsulfonylphenyl)-7-(lH-pyrrolo[2,3-b]pyridin-5-yl)-5H-pyrrolo[2,3-
1-6
b]pyrazine
1-7 7-(l-ethylpyrazol-4-yl)-2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazine
1-8 2-[2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazin-7-yl]benzonitrile
1-9 5-[2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazin-7-yl]pyrazin-2-amine
1-10 2-(4-isopropylsulfonylphenyl)-7-(4-pyridyl)-5H-pyrrolo[2,3-b]pyrazine
3-[[5-[2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazin-7-yl]-2-
1-11
pyridyl]oxy]-N,N-dimethyl-propan-l-amine
1-12 7-(lH-indol-4-yl)-2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazine
5-[2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazin-7-yl]pyrimidin-2-
1-13
amine
1-14 2-[2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazin-7-yl]phenol
1-15 2-(4-isopropylsulfonylphenyl)-7-(2-methoxyphenyl)-5H-pyrrolo[2,3-b]pyrazine
1-16 [2-[2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazin-7-yl]phenyl]methanol
2-(4-isopropylsulfonylphenyl)-7-[2-(methoxymethyl)phenyl]-5H-pyrrolo[2,3-
1-17
b]pyrazine
1-18 2-[2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazin-7-yl]benzamide
2-[2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazin-7-yl]-N,N-dimethyl-
1-19
benzamide
7-(2,3-dihydrothieno[3,4-b][l,4]dioxin-5-yl)-2-(4-isopropylsulfonylphenyl)-5H-
1-20
pyrrolo [2,3 -b]pyrazine
4-[2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazin-7-yl]thiophene-3-
1-21
carbonitrile
1-22 2-(4-isopropylsulfonylphenyl)-7-(3-thienyl)-5H-pyrrolo[2,3-b]pyrazine
1-23 7-(6-fluoro-3-pyridyl)-2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazine -24 7-(2-fluoro-3-pyridyl)-2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazine
4-[2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazin-7-yl]-3,5-dimethyl--25
isoxazole
-26 2-(4-isopropylsulfonylphenyl)-7-(5-methyl-2-thienyl)-5H-pyrrolo[2,3-b]pyrazine-27 2-(4-isopropylsulfonylphenyl)-7-(4-methyl-3-thienyl)-5H-pyrrolo[2,3-b]pyrazine-28 2-(4-isopropylsulfonylphenyl)-7-(3-methyl-2-thienyl)-5H-pyrrolo[2,3-b]pyrazine-29 2-(4-isopropylsulfonylphenyl)-7-(4-methoxyphenyl)-5H-pyrrolo[2,3-b]pyrazine-30 2-(4-isopropylsulfonylphenyl)-7-(3-methoxyphenyl)-5H-pyrrolo[2,3-b]pyrazine-31 2-(4-isopropylsulfonylphenyl)-7-(6-methoxy-3-pyridyl)-5H-pyrrolo[2,3-b]pyrazine-32 2-(4-isopropylsulfonylphenyl)-7-(2-methoxy-3-pyridyl)-5H-pyrrolo[2,3-b]pyrazine-33 2-(4-isopropylsulfonylphenyl)-7-(2-methoxy-4-pyridyl)-5H-pyrrolo[2,3-b]pyrazine
2-(4-isopropylsulfonylphenyl)-7-(2-methoxypyrimidin-5-yl)-5H-pyrrolo[2,3--34
b]pyrazine
-35 7-(lH-indol-5-yl)-2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazine-36 7-(benzofuran-2-yl)-2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazine
5-[2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazin-7-yl]-N,N-dimethyl--37
pyridin-2-amine
2-(4-isopropylsulfonylphenyl)-7-(6-methoxy-5-methyl-3-pyridyl)-5H-pyrrolo[2,3--38
b]pyrazine
7-(5-fluoro-2-methoxy-4-pyridyl)-2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3--39
b]pyrazine
-40 6-[2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazin-7-yl]quinoline-41 5-[2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazin-7-yl]isoquinoline-42 4-[2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazin-7-yl]isoquinoline-43 4-[2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazin-7-yl]quinoline-44 7-(benzothiophen-3-yl)-2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazine-45 7-(benzothiophen-2-yl)-2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazine
7-(2,4-dimethoxypyrimidin-5-yl)-2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3--46
b]pyrazine
-47 2-(4-(isopropylsulfonyl)phenyl)-7-(lH-pyrazol-4-yl)-5H-pyrrolo[2,3-b]pyrazine
2-(4-isopropylsulfonylphenyl)-7-(5-methylsulfonyl-3-pyridyl)-5H-pyrrolo[2,3--48
b]pyrazine
2-(4-isopropylsulfonylphenyl)-7-(l-methylpyrazol-3-yl)-5H-pyrrolo[2,3--49
b]pyrazine
2-(4-isopropylsulfonylphenyl)-7-(l-methylpyrazol-4-yl)-5H-pyrrolo[2,3--50
b]pyrazine
-51 2-(4-isopropylsulfonylphenyl)-7-(6-methyl-3-pyridyl)-5H-pyrrolo[2,3-b]pyrazine
7-(3,5-dimethyl-lH-pyrazol-4-yl)-2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3--52
b]pyrazine
-53 5-[2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazin-7-yl]pyridin-2-amine
7-(5-bromobenzothiophen-2-yl)-2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3--54
b]pyrazine
-55 2-(4-isopropylsulfonylphenyl)-7-(l-methylindol-6-yl)-5H-pyrrolo[2,3-b]pyrazine tert-butyl 5-fluoro-2-[2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazin-7--56
yl] indole- 1-carboxylate
tert-butyl 2-[2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazin-7-yl]-5--57
methoxy-indole- 1 -carboxylate
tert-butyl 4-chloro-2-[2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazin-7--58
yl] indole- 1-carboxylate 7-[l-(benzenesulfonyl)indol-3-yl]-2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-
1-59
b]pyrazine
N-[3-[2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazin-7-yl]-2-pyridyl]-
1-60
2,2-dimethyl-propanamide
tert-butyl 2-[2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazin-7-yl]indole-
1-61
1 -carboxylate
5-[2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazin-7-yl]thiophene-2-
1-62
carboxylic acid
2-[2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazin-7-yl]thiophene-3-
1-63
carboxylic acid
4-[2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazin-7-yl]thiophene-2-
1-64
carboxylic acid
1-65 2-(4-isopropylsulfonylphenyl)-7-(6-methoxy-2-pyridyl)-5H-pyrrolo[2,3-b]pyrazine
2-[5-[2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazin-7-yl]-3-
1-66
pyridyl]propan-2-ol
2-(4-isopropylsulfonylphenyl)-7- [4-( 1 H-tetrazol-5 -yl)phenyl] -5H-pyrrolo [2,3 -
1-67
b]pyrazine
2-(4-isopropylsulfonylphenyl)-7- [3 -( 1 H-tetrazol-5 -yl)phenyl] -5H-pyrrolo [2,3 -
1-68
b]pyrazine
3-[l-(2-chlorophenyl)ethoxy]-5-[2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-
1-69
b]pyrazin-7-yl]thiophene-2-carboxylic acid
3-[(lR)-l-(2-chlorophenyl)ethoxy]-5-[2-(4-isopropylsulfonylphenyl)-5H-
1-70
pyrrolo[2,3-b]pyrazin-7-yl]thiophene-2-carboxylic acid
Example 2; 2-(4-Isopropylsulfonylphenyl)-7-(2-phenylethynyl)-5H-pyrrolo[2,3- b]pyrazine (Compound 1-2)
S
Figure imgf000060_0001
METHOD B:
Step 1: 2-(4-isopropylsulfonylphenyl)-7-(2-phenylethynyl)-5-(p- tolylsulfonyl)pyrrolo[2,3-b]pyrazine
Figure imgf000061_0001
[00162] Ethynylbenzene (34.26 mg, 36.92 μΐ,, 0.3354 mmol) was added dropwise to a stirred solution of 7-iodo-2-(4-isopropylsulfonylphenyl)-5-(p-tolylsulfonyl)pyrrolo[2,3- b]pyrazine (150 mg, 0.2580 mmol), triethylamine (130.5 mg, 179.8 μΐ,, 1.290 mmol) , copper(I) iodide (5.896 mg, 0.03096 mmol) and Pd(PPh3)4 (14.91 mg, 0.01290 mmol) in DMF (1.3 mL) and the resulting solution stirred at ambient temperature for 64 hours. The solution was added dropwise to a mixture of EtO Ac/ice water and the layers separated. The organic layer was washed with brine (x 2), dried (MgS04), filtered and concentrated in vacuo. The residue was purified by column chromatography (ISCO Companion™, 12 g column, eluting with 2 to 20% EtOAc/DCM, loaded in DCM) to give the sub-title product as an off-white solid (96.4 mg, 67% Yield). XH NMR (400.0 MHz, DMSO) δ 1.19 (d, 6H), 2.38 (s, 3H), 3.49 (s, 1H), 7.50 (t, 5H), 7.64 (t, 2H), 8.01 (d, 2H), 8.10 (d, 2H), 8.44 (d, 2H), 8.81 (s, 1H) and 9.24 (s, 1H) ppm; MS (ES+) 556.1.
Step 2: 2-(4-Isopropylsulfonylphenyl)-7-(2-phenylethynyl)-5H-pyrrolo[2,3-b]pyrazine
Figure imgf000061_0002
[00163] 2-(4-Isopropylsulfonylphenyl)-7-(2-phenylethynyl)-5-(p- tolylsulfonyl)pyrrolo[2,3-b]pyrazine (96.4 mg, 0.1735 mmol) was dissolved in THF (2 mL) and 1M aqueous LiOH (867.5 μΐ,, 0.8675 mmol) was added. The reaction was stirred at ambient temperature for 1 hour. The reaction mixture was partitioned between EtO Ac/brine and the layers separated. The aqueous layer was extracted with EtOAc (x 2) and the combined organic extracts dried (MgSC^), filtered and concentrated in vacuo. The residue was triturated from MeCN and the resultant precipitate isolated by filtration to give the title compound as a cream solid (34 mg, 51% Yield). 'H NMR (400.0 MHz, DMSO) δ 1.20 (d, 6H), 3.48 (t, IH), 7.42 - 7.48 (m, 3H), 7.58 - 7.60 (m, 2H), 8.00 (d, 2H), 8.41 (s, IH), 8.47 (d, 2H), 9.08 (s, IH) and 12.75 (br s,lH) ppm; MS (ES+) 402.1.
Example 3; 5-[2-(4-Isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazin-7-yl]-3-phenyl- isoxazole (Compound 1-3)
SCHEME III
Figure imgf000062_0001
METHOD C:
Step 1: 2-[2-(4-Isopropylsulfonylphenyl)-5-(p-tolylsulfonyl)pyrrolo[2,3-b]pyrazin-7- yl] ethynyl-trimethyl-silane
Figure imgf000063_0001
[00164] (Trimethylsilyl)acetylene (168.1 mg, 241.9 μΐ,, 1.71 1 mmol) was added dropwise to a stirred solution of 7-iodo-2-(4-isopropylsulfonylphenyl)-5-(p-tolylsulfonyl)pyrrolo[2,3- b]pyrazine (765 mg, 1.316 mmol), triethylamine (665.8 mg, 917.1 μΐ,, 6.580 mmol), copper(I) iodide (30.07 mg, 0.1579 mmol) and Pd(PPh3)4 (76.04 mg, 0.06580 mmol) in DMF (7 mL) at 0 °C and the resulting solution allowed to warm to ambient temperature over 64 hours. The solution was added dropwise to a mixture of EtO Ac/ice water and the layers separated. The organic layer was washed with brine (x 2), dried (MgS04), filtered and concentrated in vacuo. The residue was purified by column chromatography (ISCO
Companion™, 40 g column, eluting with 2 to 20% EtOAc/DCM, loaded in DCM) to give the sub-title product as a beige solid (410.4 mg, 56% Yield). XH NMR (400.0 MHz, DMSO) δ 0.17 (t, 9H), 1.07 (d, 6H), 2.38 (qn, 3H), 3.45 - 3.50 (m, 1H), 7.36 (d, 2H), 7.90 (d, 2H), 7.96 (d, 2H), 8.27 (d, 2H), 8.60 (s, 1H) and 9.08 (s, 1H) ppm; MS (ES+) 552.0.
Step 2: 7-Ethynyl-2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazine
Figure imgf000063_0002
[00165] 2-[2-(4-Isopropylsulfonylphenyl)-5-(p-tolylsulfonyl)pyrrolo[2,3-b]pyrazin-7- yl]ethynyl-trimethyl-silane (248 mg, 0.4495 mmol) in THF (5 mL) was treated with 1M tetrabutylammonium fluoride trihydrate (674.3 μΐ^, 0.6743 mmol) and the reaction mixture allowed to stir at ambient temperature for 16 hours. A further portion of 1M
tetrabutylammonium fluoride trihydrate (449.5 μϊ^, 0.4495 mmol) was added and the reaction mixture warmed to 40 °C for 6 hours. The reaction was cooled to ambient temperature and added dropwise to a mixture of EtOAc/ice water and the layers separated. The organic layer was washed with 2M NaOH (x 2), brine (xl), dried (MgS04), filtered and concentrated in vacuo. The residue was purified by column silica gel chromatography eluting with 15-20% EtOAc/DCM to give the sub-title compound as colourless crystals (89 mg, 61% Yield). lH NMR (400.0 MHz, DMSO) δ 1.20 (d, 6H), 3.40 - 3.50 (m, 1H), 4.20 (s, 1H), 8.00 (d, 2H), 8.33 (s, 1H), 8.44 (d, 2H), 9.06 (s, 1H) and 12.70 (s, 1H) ppm; MS (ES+) 326.0.
Step 3 : 7-Ethynyl-2-(4-isopropylsulfonylphenyl)-5-(p-tolylsulfonyl)pyrrolo [2,3- b]pyrazine
Figure imgf000064_0001
[00166] A 60% dispersion of sodium hydride in mineral oil (19.79 mg, 0.5164 mmol) was added slowly to a stirred solution of 7-ethynyl-2-(4-isopropylsulfonylphenyl)-5H- pyrrolo[2,3-b]pyrazine (140 mg, 0.4303 mmol) in DMF (1.2 mL) at 0 °C. The reaction mixture was stirred at this temperature for 20 minutes then tosyl chloride (82.04 mg, 0.4303 mmol) was added and reaction mixture allowed to warm to ambient temperature over 16 hours. The reaction mixture was quenched by the slow addition of water (30 mL) and the mixture stirred for 20 minutes. The aqueous layer was extracted with EtOAc (x 3) and the combined organic extracts dried (MgS04), filtered and concentrated in vacuo. The residue was triturated form DCM/diethyl ether and the resultant precipitate isolated by filtration to give the sub-tile compound as an off-white solid (1 12.6 mg, 59% Yield). XH NMR (400.0 MHz, DMSO) δ 1.19 (d, 6H), 2.37 (s, 3H), 3.45 - 3.50 (m, 1H), 4.55 (s, 1H), 7.49 (d, 2H), 8.01 (d, 2H), 8.08 (d, 2H), 8.41 (d, 2H), 8.75 (s, 1H) and 9.22 (s, 1H) ppm; MS (ES+) 480.0.
Step 4: 5-[2-(4-Isopropylsulfonylphenyl)-5-(p-tolylsulfonyl)pyrrolo[2,3-b]pyrazin-7-yl]- 3-phenyl-isoxazole
Figure imgf000065_0001
[00167] Triethylamine (16.45 mg, 22.66 μΐ,, 0.1626 mmol) was added to a stirred solution of 7-ethynyl-2-(4-isopropylsulfonylphenyl)-5-(p-tolylsulfonyl)pyrrolo[2,3-b]pyrazine (65 mg, 0.1355 mmol) and N-hydroxybenzimidoyl chloride (21.08 mg, 0.1355 mmol) in THF (2 mL). The reaction mixture was stirred at ambient temperature for 45 minutes then heated at 65 °C for 3 hours. The reaction mixture was concentrated in vacuo and the residue partitioned between DCM and water. The layers were separated and the aqueous layer extracted with DCM (x 2). The combined organic extracts was dried (MgS04), filtered and concentrated in vacuo. The residue was triturated from MeCN and the resultant precipitate isolated by filtration to give the sub-title compound as an off-white solid (40.7 mg, 50% Yield). XH NMR (400.0 MHz, DMSO) δ 1.21 (d, 6H), 2.33 (s, 3H), 3.53 (t, 1H), 7.51 (d, 2H), 7.57 - 7.60 (m, 3H), 7.73 (s, 1H), 8.03 - 8.06 (m, 4H), 8.16 (d, 2H), 8.60 (d, 2H), 9.00 (s, 1H) and 9.32 (s, 1H) ppm; MS (ES+) 599.08.
Step 5: 5-[2-(4-Isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazin-7-yl]-3-phenyl- isoxazole
Figure imgf000066_0001
[00168] 2M Na2C03 (136 μί, 0.272 mmol) was added to a stirred solution of 5-[2-(4- isopropylsulfonylphenyl)-5-(p-tolylsulfonyl)pyrrolo[2,3-b]pyrazin-7-yl]-3-phenyl-isoxazole (40.7 mg, 0.06798 mmol) in THF (4.6 mL) and the reaction stirred at ambient temperature for 64 hours. The reaction mixture was partitioned between EtO Ac/brine and the layers separated. The aqueous layer was extracted with EtOAc( x2) and the combined organic extracts dried (MgSC^), filtered and concentrated in vacuo. The residue was triturated from MeCN and the precipitate isolated by filtration to give the title compound as a yellow solid (20.3 mg, 67% Yield). 'H NMR (400.0 MHz, DMSO) δ 1.16 (s, 6H), 3.52 (t, 1H), 7.52 - 7.61 (m, 4H), 8.02 - 8.05 (m, 4H), 8.63 (d, 2H), 8.69 (s, 1H), 9.18 (s, 1H) and 13.00 (br s, 1H) ppm; MS (ES+) 445.1.
Example 4; l-[3-[5-[2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazin-7- yl]isoxazol-3-yl]phenyl]ethanamine (Compound 1-4)
SCHE
Figure imgf000066_0002
METHOD C:
Step 1: 3-(l-(Hydroxyimino)ethyl)benzoic acid
Figure imgf000067_0001
[00169] 3-Acetylbenzoic acid (10.0 g, 60.9 mmol), hydroxylamine hydrochloride
(33.9 g, 487.4 mmol) and sodium acetate (45.0 g, 548.3 mmol) were suspended in ethanol (115 mL) and water (1 15 mL) and the mixture heated at reflux for 30 minutes. The solvents were removed under reduced pressure, and the residue was triturated with water. The resulting precipitate was isolated by filtration and dried in vacuo to give the sub-title compound as a white solid (10.34 g, 95% yield). XH NMR (400 MHz, DMSO) δ 2.18 (s, 3H), 7.52 (t, IH), 7.88 (dd, IH), 7.91 - 7.95 (m, IH), 8.22 (s, IH), 1 1.36 (s, IH) and 12.86 (s, IH); MS (ES+) 180.3.
Step 2: 3-(l-Aminoethyl)benzoic acid hydrochloride
Figure imgf000067_0002
[00170] 3-(l-(Hydroxyimino)ethyl)benzoic acid (2.28 g, 12.73 mmol) and
palladium on carbon (10 wt.%, 1.355 g, 12.73 mmol) in ethanol (170 mL) and 12M aqueous HCI (2.1 mL, 24.2 mmol) were stirred under an atmosphere of ¾ at 40 psi for 6 hours. The reaction mixture was purged with nitrogen for 30 minutes then filtered through a pad of Celite washing with methanol. The filtrate was concentrated in vacuo and the residue triturated from Et20 to give the sub-title compound as a white (2.4 g, 94% Yield). ¾ NMR (400 MHz, CD3OD) δ 1.66 (d, 3H), 4.55 (q, IH), 7.58 (t, IH), 7.70 (d, IH), 8.07 (d, IH) and 8.16 (s, IH); MS (ES+) 166.5. Step 3: 3-[l- (teri-Butoxycarbonylamino)ethyl]benzoic acid
Figure imgf000068_0001
[00171] To a solution of 3-(l-aminoethyl)benzoic acid hydrochloride (10.2 g, 50.6 mmol) in water (283 rnL) was added a solution of tert-butyl [(cyano-phenyl- methylene)amino]carbonate (13.1 g, 53. 1 mmol) in acetone (283 mL) followed by triethylamine (21 mL, 151 mmol). After stirring at ambient temperature for 16 hours the reaction mixture was concentrated in vacuo and the residue partitioned between ethyl acetate and water and the layers separated. The organic layer was extracted with saturated sodium bicarbonate solution (x 1). The combined aqueous layers were acidified to pH 2 with 1 M HC1 and extracted with ethyl acetate ( x3). The combined organic extracts were dried ( a2S04), filtered and concentrated in vacuo. The residue was triturated with ethyl acetate/hexanes to give the subtitle compound as a white solid (8.3 g, 62% Yield). ¾ NMR (400 MHz, CDC13) δ 1.37 (s, 9H), 1.48 (d, 3H), 4.88 (s, 1H), 7.43 (t, 1H), 7.51 - 7.60 (m, 1H), 7.94 - 8.02 (m, 1H) and 8.07 (s, 1H) ppm.
Step 4: teri-Butyl /V-[l-[3-(hydroxymethyl)phenyl] ethyljcarbamate
Figure imgf000068_0002
[00172] 3-[l-(/er/-Butoxycarbonylamino)ethyl]benzoic acid (7.57 g, 28.53 mmol) was dissolved in anhydrous THF (45 mL) and 2M borane-dimethylsulfide in THF (42.8 mL, 85.6 mmol) was added dropwise at ambient temperature. The reaction was stirred at this temperature for 3 hours then cooled to 0 °C and quenched with MeOH. The solvents were removed under reduced pressure and the resulting residue partitioned between 1M HC1 and ethyl acetate. The organic layer was washed with saturated sodium bicarbonate (x 1) and brine (x 1), dried (Na2S04), filtered and concentrated in vacuo. The residue was purified by column chromatography (ISCO Companion™ eluting with 0 to 30% EtOAc/petroleum ether) to give the sub-title compound as a white solid (4.91 g, 68% Yield). XH NMR (400 MHz, DMSO) δ 1.28 (d, 3H), 1.36 (s, 9H), 4.47 (d, 2H), 4.54 - 4.64 (m, 1H), 5.17 (t, 1H), 7.14 (d, 2H), 7.23 (d, 2H) and 7.38 (d, 1H) ppm; MS (ES+) 252.3.
Step 5: teri-Butyl iV-[l-(3-formylphenyl)ethyl]carbamate
Figure imgf000069_0001
[00173] To a solution of tert-butyl N-[l-[3-(hydroxymethyl)phenyl]ethyl]carbamate (4.91 g, 19.54 mmol) in DCM (40 mL) was added Mn02 (13.59 g, 156.3 mmol) and the reaction mixture was stirred at ambient temperature for 48 hours. An additional portion of Μη(¾ (3 g 34.5 mmol) was added and the reaction mixture was stirred at ambient temperature for an additional 12 hours. The reaction mixture was filtered through a pad of Celite and the filtrate concentrated in vacuo to give the sub-title compound as a colourless oil (4.0 g, 82% Yield). XH NMR (400 MHz, DMSO) δ 1.30 - 1.41 (m, 12H) 4.64 - 4.75 (m, 1H), 7.55 (dd, 2H), 7.64 (d, 1H), 7.78 (d, 1H), 7.83 (s, 1H) and 10.00 (s, 1H) ppm; MS (ES+) 250.5.
Step 6: tert-Butyl l-(3-((hydroxyimino)methyl)phenyl)ethylcarbamate
Figure imgf000069_0002
[00174] To a solution of tert-butyl N-[l-(3-formylphenyl)ethyl]carbamate (4.0 g, 16.0 mmol) and sodium acetate (3.3 g, 40.1 mmol) in THF (69 mL) at 0 °C was added
hydroxylamine hydrochloride (1.4 g, 20.9 mmol). The reaction was allowed to warm to ambient temperature and stirred for 16 hours. The reaction was concentrated in vacuo and the residue partitioned between EtOAc and water and the layers separated. The organic layer was washed with brine (x 2), dried (Na2S04), filtered and concentrated in vacuo to give the sub- title compound as a colourless gel (4.24 g, quantitative Yield). XH NMR (400 MHz, DMSO) δ 1.30 (d, 3H), 1.33 (d, 9H), 4.61 (dd, 1H), 7.24 - 7.38 (m, 2H), 7.41 (q, 2H), 7.53 (s, 1H), 8.11 (s, lH) and 11.20 (s, lH) ppm; MS (ES+) 265.3.
Step 7: tert-Butyl l-(3-(chloro(hydroxyimino)methyl)phenyl) ethylcarbamate
Figure imgf000070_0001
[00175] To a solution of tert-butyl l-(3- ((hydroxyimino)methyl)phenyl)ethylcarbamate (4.2 g, 16.0 mmol) in DMF (69 mL) at 0 °C was added N-chlorosuccinimide (2.4 g, 17.6 mmol) and the reaction mixture allowed to warm to ambient temperature over 2 hours. The reaction mixture was diluted with EtOAc (300 mL) and washed with brine ( xl). The organic layer was dried (Na2S04), filtered and concentrated in vacuo. The residue was then triturated from Et20/hexanes and the resultant precipitate isolated by filtration to give the sub-title compound as white solid (4.3 g, 90% Yield). ¾ NMR (400 MHz, DMSO) δ 1.25 - 1.45 (m, 12H), 4.64 (dt, 1H), 7.45 (dd, 3H), 7.60 - 7.69 (m, 1H), 7.72 (s, 1H) and 12.37 (s, 1H) ppm; MS (ES+) 299.1.
Step 8: tert-Butyl N-[l-[3-[5-[2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazin-7- yl]isoxazol-3-yl]phenyl]et
Figure imgf000070_0002
[00176] To a solution of 7-ethynyl-2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3- b]pyrazine (72 mg, 0.2213 mmol) and tert-butyl l-(3-(chloro(hydroxyimino)methyl)phenyl) ethylcarbamate (66.12 mg, 0.2213 mmol) in THF (2.5 mL) was added triethylamine (26.88 mg, 37.02 μΐ^, 0.2656 mmol) The mixture was stirred at ambient temperature for 45 minutes then heated at 65 °C for 3.5 hours. The reaction was cooled to ambient temperature and the solvent removed in vacuo. The residues was dissolved in warm DCM (50 mL) and washed with saturated aqueous aHC03 (x 1), dried (MgSC^), filtered and concentrated in vacuo. The residue triturated from hot MeCN and the precipitate obtained on cooling was isolated by filtration and washed with MeCN to give the sub-title compound as a pale yellow powder (83 mg, 64% Yield). XH NMR (400 MHz, DMSO) δ 1.28 (d, 6H), 1.41-1.45 (m, 3H), 1.43 (s, 9H), 3.58 (sept, 1H), 4.79 (br t, 1H), 7.52-7.62 (m, 4H), 7.92 (d, 1H), 8.00 (s, 1H), 8.10 (d, 2H), 8.68 (d, 2H), 8.76 (s, 1H), 9.25 (s, 1H) and 13.08 (br s, 1H) ppm ; MS (ES+) 588.1.
Step 9: l-[3-[5-[2-(4-Isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazin-7-yl]isoxazol-3- yl]phenyl]ethanamine
Figure imgf000071_0001
[00177] tert-Butyl N-[l-[3-[5-[2-(4-isopropylsulfonylphenyl)-5H-pyrrolo[2,3-b]pyrazin-7- yl]isoxazol-3-yl]phenyl]ethyl]carbamate (80 mg, 0.1361 mmol) was suspended in DCM (2 mL) and treated with TFA (1 mL, 12.98 mmol) and the reaction mixture stirred at ambient temperature for 2 hours. The reaction mixture was concentrated in vacuo and the residue azeotroped DCM/MeOH. The residue taken into DCM/MeOH and passed through bicarbonate cartridge. The solvent was removed in vacuo and the residue triturated from MeCN. The resultant precipitate was isolated by filtration and washed with MeCN to give the title compound as a very pale yellow powder (61 mg, 92% Yield). XH NMR (400 MHz, DMSO) δ 1.22 (d, 6H), 1.34 (d, 3H), 3.51 (sept, 1H), 4.15 (q, 1H), 7.47-7.56 (m, 3H), 7.85 (d, 1H), 8.00-8.04 (m, 3H), 8.61 (d, 2H), 8.68 (s, 1H) and 9.15 (s, 1H) ppm ; MS (ES+) 488.0. Analytical Data:
Figure imgf000072_0001
-21 409 2.8-22 384.15 1.71-23 397.18 1.55-24 397.18 1.55-25 397.18 1.44-26 398.12 1.82-27 398.12 1.77-28 398.12 1.75-29 408.19 1.71-30 408.19 1.72-31 409.2 1.57-32 409.2 1.57-33 409.2 1.35-34 410.21 1.4-35 417.19 1.6-36 418.14 1.92-37 422.23 1.02-38 423.17 1.72-39 427.2 1.67-40 429.14 1.2-41 429.14 1.12-42 429.14 1.12-43 429.14 1.08-44 434.11 1.83-45 434.11 1.89-46 440.13 1.49-47 368.12 1.17-48 457.13 1.33-49 382.12 1.29-50 382.12 1.28-51 393.17 1.02-52 396.18 1.12-53 394.15 0.99-54 512.07 2.09-55 431.17 1.74-56 535.23 1.94-57 547.27 1.86-58 551.18 2.05-59 557.2 1.89-60 478.2 1.19-61 517.24 1.9-62
-63
-64
-65 409.24 1.97-66 437.17 1.23
H NMR (400.0 MHz, DMSO) d 12.70 (d, J = 2.4 Hz,
-67 446 0.6 IH), 9.11 (s, IH), 8.72 (d, J = 2.9 Hz, IH), 8.59 (d, J = 8.4 Hz, 2H), 8.55 (d, J = 8.5 Hz, 2H), 8.15 (d, J = 8.4 Hz,
2H), 8.04 (d, J = 8.5 Hz, 2H), 3.51 (q, J = 6.8 Hz, 1H)
and 1.22 (d, J = 6.7 Hz, 6H) ppm
H NMR (400.0 MHz, DMSO) d 12.65 (d, J = 2.5 Hz,
1H), 9.12 (s, 1H), 9.08 (s, 1H), 8.65 (d, J = 2.9 Hz, 1H),
1-68 8.59 (d, J = 8.4 Hz, 3H), 8.02 (d, J = 8.5 Hz, 2H), 7.91 (d, 446.6 0.62 J = 7.6 Hz, 1H), 7.72 (t, J = 7.7 Hz, 1H), 3.52 (q, J = 6.8
Hz, 1H) and 1.22 (d, J = 6.8 Hz, 6H) ppm
H NMR (400.0 MHz, DMSO) d 12.76 (d, J = 2.9 Hz,
1H), 12.53 (s, 1H), 9.09 (s, 1H), 8.54 (d, J = 3.0 Hz, 1H),
8.48 (d, J = 8.5 Hz, 2H), 8.04 (d, J = 8.5 Hz, 2H), 7.70
1-69 (dd, J = 1.6, 7.8 Hz, 1H), 7.64 (s, 1H), 7.45 - 7.37 (m, 582.1 0.67 2H), 7.31 (dd, J = 1.7, 7.7 Hz, 1H), 5.96 (d, J = 6.3 Hz,
1H), 3.54 (t, J = 6.8 Hz, 1H), 1.64 (d, J = 6.3 Hz, 3H) and
1.23 (dd, J = 1.1, 6.7 Hz, 6H) ppm
H NMR (400.0 MHz, DMSO) d 12.76 (d, J = 2.9 Hz,
1H), 12.53 (s, 1H), 9.09 (s, 1H), 8.54 (d, J = 3.0 Hz, 1H),
8.48 (d, J = 8.5 Hz, 2H), 8.04 (d, J = 8.5 Hz, 2H), 7.70
1-70 (dd, J = 1.6, 7.8 Hz, 1H), 7.64 (s, 1H), 7.45 - 7.37 (m, 582 0.67 2H), 7.31 (dd, J = 1.7, 7.7 Hz, 1H), 5.96 (d, J = 6.3 Hz,
1H), 3.54 (t, J = 6.8 Hz, 1H), 1.64 (d, J = 6.3 Hz, 3H) and
1.23 (dd, J = 1.1, 6.7 Hz, 6H)
Example 2: Cellular ATR Inhibition Assay:
[00178] Compounds can be screened for their ability to inhibit intracellular ATR using an immunofluorescence microscopy assay to detect phosphorylation of the ATR substrate histone H2AX in hydroxyurea treated cells. HT29 cells are plated at 14,000 cells per well in 96-well black imaging plates (BD 353219) in McCoy's 5A media (Sigma M8403) supplemented with 10% foetal bovine serum (JRH Biosciences 12003),
Penicillin/Streptomycin solution diluted 1 : 100 (Sigma P7539), and 2mM L-glumtamine (Sigma G7513), and allowed to adhere overnight at 37°C in 5% CO2. Compounds are then added to the cell media from a final concentration of 25μΜ in 3-fold serial dilutions and the cells are incubated at 37°C in 5% CO2. After 15min, hydroxyurea (Sigma H8627) is added to a final concentration of 2mM.
[00179] After 45min of treatment with hydroxyurea, the cells are washed in PBS, fixed for lOmin in 4% formaldehyde diluted in PBS (Polysciences Inc 18814), washed in 0.2% Tween-20 in PBS (wash buffer), and permeabilised for lOmin in 0.5% Triton X-100 in PBS, all at room temperature. The cells are then washed once in wash buffer and blocked for 30min at room temperature in 10% goat serum (Sigma G9023) diluted in wash buffer (block buffer). To detect H2AX phosphorylation levels, the cells are then incubated for lh at room temperature in primary antibody (mouse monoclonal anti-phosphorylated histone H2AX Serl39 antibody; Upstate 05-636) diluted 1 :250 in block buffer. The cells are then washed five times in wash buffer before incubation for lh at room temperature in the dark in a mixture of secondary antibody (goat anti-mouse Alexa Fluor 488 conjugated antibody;
Invitrogen A11029) and Hoechst stain (Invitrogen H3570); diluted 1 :500 and 1 :5000, respectively, in wash buffer. The cells are then washed five times in wash buffer and finally lOOul PBS is added to each well before imaging.
[00180] Cells are imaged for Alexa Fluor 488 and Hoechst intensity using the BD
Pathway 855 Bioimager and Attovision software (BD Biosciences, Version 1.6/855) to quantify phosphorylated H2AX Serl39 and DNA staining, respectively. The percentage of phosphorylated H2AX -positive nuclei in a montage of 9 images at 20x magnification is then calculated for each well using BD Image Data Explorer software (BD Biosciences Version 2.2.15). Phosphorylated H2AX-positive nuclei are defined as Hoechst-positive regions of interest containing Alexa Fluor 488 intensity at 1.75-fold the average Alexa Fluor 488 intensity in cells not treated with hydroxyurea. The percentage of H2AX positive nuclei is finally plotted against concentration for each compound and IC50s for intracellular ATR inhibition are determined using Prism software(GraphPad Prism version 3.0cx for Macintosh, GraphPad Software, San Diego California, USA).
[00181] The compounds described herein can also be tested according to other methods known in the art (see Sarkaria et al, "Inhibition of ATM and ATR Kinase Activities by the Radiosensitizing Agent, Caffeine: Cancer Research 59: 4375-5382 (1999); Hickson et al, "Identification and Characterization of a Novel and Specific Inhibitor of the Ataxia- Telangiectasia Mutated Kinase ATM" Cancer Research 64: 9152-9159 (2004); Kim et al, "Substrate Specificities and Identification of Putative Substrates of ATM Kinase Family Members" The Journal of Biological Chemistry, 274(53): 37538-37543 (1999); and Chiang et al, "Determination of the catalytic activities of mTOR and other members of the phosphoinositide-3-kinase-related kinase family" Methods Mol. Biol. 281 : 125-41 (2004)).
Example 3: ATR Inhibition Assay:
[00182] Compounds were screened for their ability to inhibit ATR kinase using a radioactive-phosphate incorporation assay. Assays were carried out in a mixture of 50mM Tris/HCl (pH 7.5), lOmM MgCl2 and ImM DTT. Final substrate concentrations were 10μΜ [γ-33Ρ]ΑΤΡ (3mCi 33P ATP/mmol ATP, Perkin Elmer) and 800 μΜ target peptide
(ASELPASQPQPFSAKKK). [00183] Assays were carried out at 25°C in the presence of 5 nM full-length ATR. An assay stock buffer solution was prepared containing all of the reagents listed above, with the exception of ATP and the test compound of interest. 13.5 μΐ., of the stock solution was placed in a 96 well plate followed by addition of 2 μϊ^ of DMSO stock containing serial dilutions of the test compound (typically starting from a final concentration of 15 μΜ with 3- fold serial dilutions) in duplicate (final DMSO concentration 7%). The plate was pre- incubated for 10 minutes at 25°C and the reaction initiated by addition of 15 μϊ^ [γ-33Ρ]ΑΤΡ (final concentration 10 μΜ).
[00184] The reaction was stopped after 24 hours by the addition of 30μΙ, 0.1M phosphoric acid containing 2mM ATP. A multiscreen phosphocellulose filter 96-well plate (Millipore, Cat no. MAPHN0B50) was pretreated with ΙΟΟμΙ. 0.2M phosphoric acid prior to the addition of 45μΙ, of the stopped assay mixture. The plate was washed with 5 x 200μΙ, 0.2M phosphoric acid. After drying, 100 μΐ., Optiphase 'SuperMix' liquid scintillation cocktail (Perkin Elmer) was added to the well prior to scintillation counting (1450 Microbeta Liquid Scintillation Counter, Wallac).
[00185] After removing mean background values for all of the data points, Ki(app) data were calculated from non-linear regression analysis of the initial rate data using the Prism software package (GraphPad Prism version 3.0cx for Macintosh, GraphPad Software, San Diego California, USA).
[00186] Below is a chart showing the ATR Inhibition Ki values of compounds of the disclosure. Compounds with a Ki value of < 10 nM are marked with "+++." Compounds with a Ki value > 10 nM but < 50 nM are marked with "++." Compounds with a Ki value > 50 nM are marked with "+."
Figure imgf000076_0001
Figure imgf000076_0002
Figure imgf000076_0003
1-41 + 1-52 + 1-63 ++
1-42 ++ 1-53 +++ 1-64 +++
1-43 + 1-54 + 1-65
1-44 + 1-55 1-66 +++
1-45 ++ 1-56 + 1-67 +
1-46 + 1-57 + 1-68 ++
1-47 ++ 1-58 + 1-69 +++
1-48 ++ 1-59 + 1-70 +++
1-49 ++ 1-60 +
1-50 + 1-61 +
1-51 ++ 1-62 +++
Example 4: Cisplatin Sensitization Assay
[00187] Compounds can be screened for their ability to sensitize HCT116 colorectal cancer cells to Cisplatin using a 96h cell viability (MTS) assay. HCT116 cells, which possess a defect in ATM signaling to Cisplatin (see, Kim et al.; Oncogene 21 :3864 (2002); see also, Takemura et al; JBC 281 :30814 (2006)) are plated at 470 cells per well in 96-well polystyrene plates (Costar 3596) in 150μ1 of McCoy's 5A media (Sigma M8403) supplemented with 10% foetal bovine serum (JRH Biosciences 12003),
Penicillin/Streptomycin solution diluted 1: 100 (Sigma P7539), and 2mM L-glumtamine (Sigma G7513), and allowed to adhere overnight at 37°C in 5% CO2. Compounds and Cisplatin are then both added simultaneously to the cell media in 2-fold serial dilutions from a top final concentration of 10μΜ as a full matrix of concentrations in a final cell volume of 200μ1, and the cells are then incubated at 37°C in 5% C02. After 96h, 40μ1 of MTS reagent (Promega G358a) is added to each well and the cells are incubated for lh at 37°C in 5% CO2. Finally, absorbance is measured at 490nm using a SpectraMax Plus 384 reader (Molecular Devices) and the concentration of compound required to reduce the IC50 of Cisplatin alone by at least 3-fold (to 1 decimal place) can be reported either with an IC50 or Ki value .
[00188] While we have described a number of embodiments of this invention, it is apparent that our basic examples may be altered to provide other embodiments that utilize the compounds, methods, and processes of this invention. Therefore, it will be appreciated that the scope of this invention is to be defined by the appended claims rather than by the specific embodiments that have been represented by way of example herein.

Claims

1. A compound of formula I:
Figure imgf000078_0001
I
or a pharmaceutically acceptable salt thereof, wherein
R1 is hydrogen, Ci-6alkyl, or a 3-7 membered monocyclic fully saturated, partially
unsaturated, or aromatic ring having 0-4 heteroatoms independently selected from the group consisting of nitrogen, oxygen, and sulfur; or an 8-10 membered bicyclic aromatic ring having 0-6 heteroatoms selected from the group consisting of nitrogen, oxygen, and 1 is optionally substituted with 0-4 occurrences of J;
Figure imgf000078_0002
A1 is a 5 -membered heteroaryl wherein X is carbon, nitrogen, oxygen, or sulfur; when X is nitrogen or carbon;
A2 is phenyl or a 6-membered heteroaryl having 1-3 nitrogen atoms; A2 is independently and optionally substituted with up to 2 occurrences of halo or CN;
A3 is an 8-10 membered bicyclic heteroaromatic ring having 1-3 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur;
Z1 is H, a Ci_6aliphatic; wherein 0-2 methylene units of said Ci_ioaliphatic are optionally replaced with -NR'-, -0-, -S-, C(O); or a 5-6 membered monocyclic aromatic ring having 0-3 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or an 8-10 membered bicyclic aromatic ring having 0-6 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or a Ci-ioaliphatic; wherein 0-4 methylene units of said Ci-ioaliphatic are optionally replaced with -NR'-, -0-, -S-, C(O); a C3_6cycloalkyl, or a 3-6 membered heterocyclic ring having 1-2 heteroatoms selected from O, NR', or S; Z1 is optionally substituted with 1-5 J1 groups;
Z2 is H, a 5-6 membered monocyclic aromatic ring having 0-3 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or an 8-10 membered bicyclic aromatic ring having 0-6 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or a Ci-ioaliphatic; wherein 0-4 methylene units of said Ci-ioaliphatic are optionally replaced with -NR.'-, -0-, -S-, C(O), a C3_6cycloalkyl, or a 3-6 membered heterocyclic ring having 1-2 heteroatoms selected from O, NR', or S; Z2 is optionally substituted with 1-5 J2 groups;
Z3 is H, C3_6cycloalkyl, halo, CN, NO2, or a Ci-ioaliphatic; wherein 0-4 methylene units of said Ci-ioaliphatic are optionally replaced with -NR'-, -0-, -S-, or C(O); Z3 is optionally substituted with 1-5 J3 groups;
Z4 is a 5-6 membered monocyclic aromatic ring having 0-3 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or an 8-10 membered bicyclic aromatic ring having 0-6 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; Z4 is optionally substituted with 1-5 J4 groups;
J is halo, CN, or V, or (V)t-R2;
V is a Ci-ioaliphatic group wherein up to 3 methylene units are optionally replaced with O, NR", C(O), S, S(O), or S(0)2; wherein said Ci-ioaliphatic group is optionally substituted with 1-3 occcurences of halo or CN;
R2 is 3-7 membered aromatic or nonaromatic monocyclic ring having 0-3 heteroatoms
selected from the group consisting of oxygen, nitrogen, and sulfur; R2 is optionally substituted with 1-3 occurences of halo, CN, C3_6cycloalkyl, or Ci-ioaliphatic; wherein up to 3 methylene units of said Ci-ioaliphatic are optionally replaced with NR', O, S, or CO; each J1, J2, and J4 is independently halo, CN, NO2, or X1;
JA is XA or XA-QA;
XA is a Ci-6aliphatic; wherein 0-4 methylene units of said Ci-ioaliphatic are optionally
replaced with -NR'-, -0-, -S-, or C(O); wherein said Ci-6aliphatic is optionally substituted with halo or Ci_3alkyl;
QA is phenyl;
X1 is a Ci_6aliphatic; wherein 0-4 methylene units of said Ci-ioaliphatic are optionally
replaced with -NR'-, -0-, -S-, or C(O), wherein X1 is optionally and independently substituted with 1-4 occurrences of JX1;
JX1 is halo or a 3-6 membered monocyclic ring having 0-2 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur;
J3 is halo, CN, phenyl, a 4-6 membered heterocyclic ring having 1-2 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or a Ci_6aliphatic optionally substituted with 1-3 occurrences of halo;
each R' and R" is independently hydrogen or Ci-6alkyl; t is 0 or 1;
provided that
when A3 is
Figure imgf000080_0001
, then R is not optionally substituted phenyl;
when A2 is pyridinyl, then R1 is not optionally substituted phenyl or optionally substituted pyrazinyl;
when A2 is pyrrolyl, then R1 is not optionally substituted cyclohexyl;
when A2 is phenyl, then R1 is not an optionally substituted group selected from pyridinyl, morpholinyl, or piperazinyl;
when A2 is phenyl and R1 is phenyl; R1 is substituted with 4-S02(Ci_6alkyl) as shown in formula ii-a;
Figure imgf000080_0002
lkyl
2. Th compound of claim 1 wherein
Figure imgf000080_0003
A2 is a 6-membered heteroaryl having 1-3 nitrogen atoms; and
Z1 is a 5-6 membered monocyclic aromatic ring having 0-3 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or an 8-10 membered bicyclic aromatic ring having 0-6 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or a Ci_ioaliphatic; wherein 0-4 methylene units of said Ci-ioaliphatic are optionally replaced with -NR'-, -0-, -S-, C(O); a C3_6cycloalkyl, or a 3-6 membered heterocyclic ring having 1-2 heteroatoms selected from O, NR', or S; Z1 is optionally substituted with 1-5 J1 groups; and
J3 is halo, CN, phenyl, a 4-6 membered heterocyclic ring having 1-2 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur; or a Ci_6aliphatic optionally substituted with 1-3 occurrences of halo. The compound of claim 1, wherein A
4. The compound of claim 1, wherein A
Figure imgf000081_0001
5. The compound of claim 3, wherein A1 is selected from the following:
Figure imgf000081_0002
The compound of claim 3, wherein A1 is .
7. The compound of claim 6, wherein X1 is S.
8. The compound of claim 3, wherein A is
Figure imgf000081_0003
9. The compound of claim 8, wherein X1 is S or O.
10. The compound of claim 9, wherein X1 is O.
1 1. The compound of any one of claims 1-10, wherein Z1 is Ci-6alkyl
12. The compound of any one of claims 1-10, wherein Z1 is a 5-6 membered aromatic ring.
13. The compound of claim 6, wherein Z1 is COOH.
14. The compound of any one of claims 12, wherein Z1 is phenyl.
15. The compound of any one of claims 12-14, wherein Z1 is optionally substituted with 1-2 J1 groups.
16. The compound of claim 15, wherein J1 is -CH2NHR' or CHCi_6alkyl)NHR'.
17. The compound of claim 16, wherein R' is H.
18. The compound of claim 1, wherein A is
Figure imgf000082_0001
19. The compound of claim 18, wherein A2 is phenyl and Z2 is CN, CH2OH, CH2OCH3, CONH2, CON(CH3)2, OCH3, or tretrazolyl.
20. The compound of claim 18, wherein A2 is a 6-membered heteroaryl having 1-2 nitrogen atoms.
21. The compound of claim 20, wherein A2 is pyridinyl or pyrimidinyl.
following:
Figure imgf000082_0002
. The compound of claim 1, wherein A is
24. The compound of claim 23, wherein A3 is an 8-10 membered bicyclic heteroaromatic ring having 1-2 heteroatoms selected from the group consisting of nitrogen, oxygen, and sulfur.
25. The compound of claim 24, wherein said heteroaromatic ring is benzoxazolyl,
benzisoxazolyl, benzothiazolyl, benzimidazolyl, azaindolyl, indolyl, indazolyl, benzothienyl, benzofuranyl, dihydrothienodioxinyl, quinolinyl, or isoquinolinyl.
Figure imgf000082_0003
H or Ci-4alkyl.
27. The compound of claim 24, wherein A is selected from the following:
Figure imgf000082_0004
28. The compound of claim 24, wherein A is selected from the following:
Figure imgf000083_0001
The compound of claim 1, wherein A is
30. The compound of claim 29, wherein Z4 is a 5-6 membered aromatic ring.
31. The compound of claim 30, wherein Z4 is phenyl.
32. The compound of any one of claims 1-31, wherein R1 is Ci-6alkyl.
33. The compound of any one of claims 1-31, wherein R1 is a monocyclic ring.
34. The compound of claim 33, wherein R1 is a 3-7 membered cycloaliphatic or heterocyclyl.
35. The compound of claim 33, wherein R1 is a 5-6 membered aromatic ring having 0-4 heteroatoms independently selected from the group consisting of nitrogen, oxygen, and sulfur.
36. The compound of claim 35, wherein R1 is phenyl, pyridinyl, or pyrimidinyl.
37. The compound of claim 36, wherein R1 is phenyl.
38. The compound of any one of claims 1-37, wherein R1 is substituted by one to two
occurrences of J.
39. The compound of claim 38, wherein R1 is substituted in the para position with J.
40. The compound of claim 38 or claim 39, wherein J is V.
41. The compound of claim 40, wherein V is a Ci_6aliphatic group wherein one methylene unitis optionally replaced with S(0)2.
42. The compound of claim 41, wherein V is -S(0)2(Ci_4alkyl).
43. The compound of claim 42, wherein V is S(0)2CH(CH3)2.
44. The compound of claim 38, wherein J is CN and substituted in the ortho position.
45. The compound of claim 38 or claim 39, wherein J is -(V)t-R2.
46. The compound of claim 45, wherein V is -S(0)2.
47. The compound of claim 45 or 46, wherein R2 is a C3-7 cycloalkyl or a 3-7 membered heterocyclyl having 1-3 heteroatoms selected from the group consisting of oxygen, nitrogen, and sulfur.
48. The compound of any one of claims 2, 18, 23, or 29, wherein R1 is phenyl substituted in the para position with J, wherein J is -S(0)2(Ci_4alkyl).
49. A compound selected from the following:
Figure imgf000084_0001
Figure imgf000085_0001
Figure imgf000086_0001
Figure imgf000087_0001
Figure imgf000088_0001
50. A pharmaceutical composition comprising a compound of any one of claims 1-48 and a pharmaceutically acceptable carrier.
51. A method for treating cancer in a patient comprising administering a compound of any one of claims 1-48 or a pharmaceutically acceptable derivative thereof.
52. The method of claim 51, further comprising administering to said patient an additional therapeutic agent selected from a DNA-damaging agent; wherein said additional therapeutic agent is appropriate for the disease being treated; and said additional therapeutic agent is administered together with said compound as a single dosage form or separately from said compound as part of a multiple dosage form.
53. The method of claim 52, wherein said DNA-damaging agent is selected chemotherapy or radiation treatment.
54. The method of claim 52, wherein said DNA-damaging agent is selected from ionizing radiation, radiomimetic neocarzinostatin, a platinating agent, a Topo I inhibitor, a Topo II inhibitor, an antimetabolite, an alkylating agent, an alkyl sulphonates, an antimetabolite, or an antibiotic.
55. The method of claim 54, wherein said DNA-damaging agent is selected from ionizing radiation, a platinating agent, a Topo I inhibitor, a Topo II inhibitor, or an antibiotic.
56. The method of claim 55, wherein said platinating agent is selected from Cisplatin,
Oxaliplatin, Carboplatin, Nedaplatin, Lobaplatin, Triplatin Tetranitrate, Picoplatin, Satraplatin, ProLindac and Aroplatin; said Topo I inhibitor is selected from
Camptothecin, Topotecan, Irinotecan/SN38, Rubitecan and Belotecan; said Topo II inhibitor is selected from Etoposide, Daunorubicin, Doxorubicin, Aclarubicin, Epirubicin, Idarubicin, Amrubicin, Pirarubicin, Valrubicin, Zorubicin and Teniposide; said antimetabolite is selected from Aminopterin, Methotrexate, Pemetrexed, Raltitrexed, Pentostatin, Cladribine, Clofarabine, Fludarabine, Thioguanine, Mercaptopurine, Fluorouracil, Capecitabine, Tegafur, Carmofur, Floxuridine, Cytarabine, Gemcitabine, Azacitidine and Hydroxyurea; said alkylating agent is selected from Mechlorethamine, Cyclophosphamide, Ifosfamide, Trofosfamide, Chlorambucil, Melphalan, Prednimustine, Bendamustine, Uramustine, Estramustine, Carmustine, Lomustine, Semustine,
Fotemustine, Nimustine, Ranimustine, Streptozocin, Busulfan, Mannosulfan, Treosulfan, Carboquone, ThioTEPA, Triaziquone, Triethylenemelamine, Procarbazine, Dacarbazine, Temozolomide, Altretamine, Mitobronitol, Actinomycin, Bleomycin, Mitomycin and Plicamycin.
57. The method of claim 56, wherein said platinating agent is selected from Cisplatin, Oxaliplatin, Carboplatin, Nedaplatin, or Satraplatin; said Topo I inhibitor is selected from Camptothecin, Topotecan, irinotecan/SN38, rubitecan; said Topo II inhibitor is selected from Etoposide; said antimetabolite is selected from methotrexate, pemetrexed,
Thioguanine, Fludarabine, Cladribine, Cytarabine, gemcitabine, 6-Mercaptopurine, or 5-Fluorouracil; said alkylating agent is selected from nitrogen mustards, nitrosoureas, triazenes, alkyl sulfonates, Procarbazine, or aziridines; and said antibiotic is selected from Hydroxyurea, Anthracyclines, Anthracenediones, or Streptomyces family.
58. The method of claim 55 wherein said DNA-damaging agent is a platinating agent or ionizing radiation.
59. The method of any one of claims 51-58, wherein said cancer is a solid tumor selected from the following cancers: Oral: buccal cavity, lip, tongue, mouth, pharynx; Cardiac: sarcoma (angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma), myxoma, rhabdomyoma, fibroma, lipoma and teratoma; Lung: bronchogenic carcinoma (squamous cell or epidermoid, undifferentiated small cell, undifferentiated large cell,
adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hamartoma, mesothelioma; Gastrointestinal: esophagus (squamous cell carcinoma, larynx, adenocarcinoma, leiomyosarcoma, lymphoma), stomach (carcinoma, lymphoma, leiomyosarcoma), pancreas (ductal adenocarcinoma, insulinoma, glucagonoma, gastrinoma, carcinoid tumors, vipoma), small bowel or small intestines (adenocarcinoma, lymphoma, carcinoid tumors, Karposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, fibroma), large bowel or large intestines
(adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, leiomyoma), colon, colon-rectum, colorectal; rectum, Genitourinary tract: kidney (adenocarcinoma, Wilm's tumor [nephroblastoma], lymphoma), bladder and urethra (squamous cell carcinoma, transitional cell carcinoma, adenocarcinoma), prostate (adenocarcinoma, sarcoma), testis (seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma); Liver: hepatoma (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma,
angiosarcoma, hepatocellular adenoma, hemangioma, biliary passages; Bone: osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Ewing's sarcoma, malignant lymphoma (reticulum cell sarcoma), multiple myeloma, malignant giant cell tumor chordoma, osteochronfroma (osteocartilaginous exostoses), benign chondroma, chondroblastoma, chondromyxofibroma, osteoid osteoma and giant cell tumors; Nervous system: skull (osteoma, hemangioma, granuloma, xanthoma, osteitis deformans), meninges (meningioma, meningiosarcoma, gliomatosis), brain (astrocytoma, medulloblastoma, glioma, ependymoma, germinoma [pinealoma], glioblastoma multiform, oligodendroglioma, schwannoma, retinoblastoma, congenital tumors), spinal cord neurofibroma, meningioma, glioma, sarcoma); Gynecological: uterus (endometrial carcinoma), cervix (cervical carcinoma, pre-tumor cervical dysplasia), ovaries (ovarian carcinoma [serous cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma], granulosa-thecal cell tumors, Sertoli-Leydig cell tumors, dysgerminoma, malignant teratoma), vulva (squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, melanoma), vagina (clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma (embryonal rhabdomyosarcoma), fallopian tubes
(carcinoma), breast; Skin: malignant melanoma, basal cell carcinoma, squamous cell carcinoma, Karposi's sarcoma, keratoacanthoma, moles dysplastic nevi, lipoma, angioma, dermatofibroma, keloids, psoriasis, Thyroid gland: papillary thyroid carcinoma, follicular thyroid carcinoma; medullary thyroid carcinoma, multiple endocrine neoplasia type 2A, multiple endocrine neoplasia type 2B, familial medullary thyroid cancer,
pheochromocytoma, paraganglioma; and Adrenal glands: neuroblastoma.
60. The method of claim 59, wherein said cancer is selected from lung cancer, head and neck cancer, pancreatic cancer, gastric cancer, and brain cancer.
61. A method of promoting cell death in cancer cells comprising administering to a patient a compound of any one of claims 1-48.
62. A method of preventing cell repair from DNA damage comprising administering to a patient a compound of any one of claims 1-48.
63. A method of inhibiting ATR in a biological sample comprising the step of contacting a compound of any one of claims 1-48 with said biological sample.
64. The method of claim 63, wherein said biological sample is a cell.
65. A method of sensitizing cells to DNA damaging agents comprising administering to a patient a compound of any one of claims 1-48.
66. The method of any one of claims 51-65, wherein said cell is a cancer cell having defects in the ATM signaling cascade.
67. The method of claim 66, wherein said defect is altered expression or activity of one or more of the following: ATM, p53, CHK2, MRE11, RAD50, NBS 1, 53BP1, MDC1 or H2AX.
68. The method of any one of claims 51-65, wherein said cell is a cancer cell expressing DNA damaging oncogenes.
69. The method of claim 68, wherein said cancer cell has altered expression or activity of one or more of the following: K-Ras, N-Ras, H-Ras, Raf, Myc, Mos, E2F, Cdc25A, CDC4, CDK2, Cyclin E, Cyclin A and Rb.
70. Use of a compound of any one of claims 1-48 as a radio-sensitizer or a chemo-sensitizer.
71. Use of a compound according to any one of claims 1-48 as a single agent (monotherapy) for treating cancer.
72. Use of according to any one of claims 1 -48 for treating patients having cancer with a DNA-damage response (DDR) defect.
73. The use according to claim 72, wherein said defect is a mutation or loss of ATM, p53, CHK2, MRE11, RAD50, NBS1, 53BP1, MDC1, or H2AX.
PCT/US2011/041705 2010-06-23 2011-06-23 Pyrrolo- pyrazine derivatives useful as inhibitors of atr kinase WO2011163527A1 (en)

Priority Applications (6)

Application Number Priority Date Filing Date Title
NZ605627A NZ605627A (en) 2010-06-23 2011-06-23 Pyrrolo-pyrazine derivatives useful as inhibitors of atr kinase
EP11729535.2A EP2585468A1 (en) 2010-06-23 2011-06-23 Pyrrolo- pyrazine derivatives useful as inhibitors of atr kinase
CA2803802A CA2803802A1 (en) 2010-06-23 2011-06-23 Pyrrolo- pyrazine derivatives useful as inhibitors of atr kinase
JP2013516779A JP2013529643A (en) 2010-06-23 2011-06-23 Pyrrolopyrazine derivatives useful as ATR kinase inhibitors
MX2013000103A MX2013000103A (en) 2010-06-23 2011-06-23 Pyrrolo- pyrazine derivatives useful as inhibitors of atr kinase.
AU2011270807A AU2011270807A1 (en) 2010-06-23 2011-06-23 Pyrrolo- pyrazine derivatives useful as inhibitors of ATR kinase

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US35774510P 2010-06-23 2010-06-23
US61/357,745 2010-06-23

Publications (1)

Publication Number Publication Date
WO2011163527A1 true WO2011163527A1 (en) 2011-12-29

Family

ID=44351817

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2011/041705 WO2011163527A1 (en) 2010-06-23 2011-06-23 Pyrrolo- pyrazine derivatives useful as inhibitors of atr kinase

Country Status (8)

Country Link
US (3) US8623869B2 (en)
EP (1) EP2585468A1 (en)
JP (2) JP2013529643A (en)
AU (1) AU2011270807A1 (en)
CA (1) CA2803802A1 (en)
MX (1) MX2013000103A (en)
NZ (1) NZ605627A (en)
WO (1) WO2011163527A1 (en)

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014055756A1 (en) 2012-10-04 2014-04-10 Vertex Pharmaceuticals Incorporated Method for measuring atr inhibition mediated increases in dna damage
JP2015503609A (en) * 2012-01-13 2015-02-02 ノバルティス アーゲー Condensed pyrrole as an IP receptor agonist for the treatment of pulmonary arterial hypertension (PAH) and related disorders
JP2015533151A (en) * 2012-10-16 2015-11-19 エフ・ホフマン−ラ・ロシュ・アクチェンゲゼルシャフト Serine / threonine kinase inhibitor
WO2016020320A1 (en) * 2014-08-04 2016-02-11 Bayer Pharma Aktiengesellschaft 2-(morpholin-4-yl)-l,7-naphthyridines
JP2016510803A (en) * 2013-03-15 2016-04-11 フンダシオン セントロ ナシオナル デ インベスティガシオネス オンコロヒカス カルロス ザ サードFundacion Centro Nacional de Investigaciones Oncologicas Carlos III Chemical substances
WO2017059357A1 (en) 2015-09-30 2017-04-06 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of dna damaging agents and atr inhibitors
WO2018153968A1 (en) 2017-02-24 2018-08-30 Bayer Aktiengesellschaft An inhibitor of atr kinase for use in a method of treating a hyper-proliferative disease
WO2018153973A1 (en) * 2017-02-24 2018-08-30 Bayer Pharma Aktiengesellschaft Combination of atr kinase inhibitors with parp inhibitors
WO2018153972A1 (en) 2017-02-24 2018-08-30 Bayer Pharma Aktiengesellschaft Combination of atr kinase inhibitors and antiandrogens
WO2018153969A1 (en) 2017-02-24 2018-08-30 Bayer Aktiengesellschaft Combination of atr kinase inhibitors with radium-223 salt
WO2018206547A1 (en) 2017-05-12 2018-11-15 Bayer Pharma Aktiengesellschaft Combination of bub1 and atr inhibitors
WO2018210661A1 (en) * 2017-05-15 2018-11-22 Basf Se Heteroaryl compounds as agrochemical fungicides
WO2019025440A1 (en) 2017-08-04 2019-02-07 Bayer Pharma Aktiengesellschaft Combination of atr kinase inhibitors and pd-1/pd-l1 inhibitors
EP3461480A1 (en) 2017-09-27 2019-04-03 Onxeo Combination of a dna damage response cell cycle checkpoint inhibitors and belinostat for treating cancer
WO2019110586A1 (en) 2017-12-08 2019-06-13 Bayer Aktiengesellschaft Predictive markers for atr kinase inhibitors
WO2020064971A1 (en) 2018-09-26 2020-04-02 Merck Patent Gmbh Combination of a pd-1 antagonist, an atr inhibitor and a platinating agent for the treatment of cancer
WO2020078788A1 (en) 2018-10-16 2020-04-23 Bayer Aktiengesellschaft Combination of atr kinase inhibitors with 2,3-dihydroimidazo[1,2-c]quinazoline compounds
WO2020078905A1 (en) 2018-10-15 2020-04-23 Merck Patent Gmbh Combination therapy utilizing dna alkylating agents and atr inhibitors
WO2020259601A1 (en) * 2019-06-28 2020-12-30 Impact Therapeutics, Inc Substituted fused heteroaromatic bicyclic compounds as kinase inhibitors and the use thereof
US11028076B2 (en) 2016-01-11 2021-06-08 Celator Pharmaceuticals, Inc. Inhibiting ataxia telangiectasia and Rad3-related protein (ATR)
WO2022228522A1 (en) * 2021-04-29 2022-11-03 江苏先声药业有限公司 Alkyne compound as hpk1 inhibitor and use thereof
RU2802512C2 (en) * 2018-08-24 2023-08-30 Байер Акциенгезельшафт Method for producing 2-[(3r)-3-methylmorpholin-4-yl]-4-(1-methyl-1h-pyrazol-5-yl)-8-(1h-pyrazol-5-yl)-1,7-nafthyridine

Families Citing this family (45)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CA3013000C (en) * 2008-12-19 2022-12-13 Vertex Pharmaceuticals Incorporated Pyrazine derivatives useful as inhibitors of atr kinase
US9039655B2 (en) 2009-11-06 2015-05-26 Crisi Medical Systems, Inc. Medication injection site and data collection system
US9101534B2 (en) 2010-04-27 2015-08-11 Crisi Medical Systems, Inc. Medication and identification information transfer apparatus
EP2569287B1 (en) 2010-05-12 2014-07-09 Vertex Pharmaceuticals Inc. Compounds useful as inhibitors of atr kinase
WO2011143419A1 (en) 2010-05-12 2011-11-17 Vertex Pharmaceuticals Incorporated Pyrazines useful as inhibitors of atr kinase
EP2569286B1 (en) 2010-05-12 2014-08-20 Vertex Pharmaceuticals Inc. Compounds useful as inhibitors of atr kinase
CA2798760A1 (en) 2010-05-12 2011-11-17 Vertex Pharmaceuticals Incorporated 2-aminopyridine derivatives useful as inhibitors of atr kinase
EP2568984A1 (en) * 2010-05-12 2013-03-20 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
SG185524A1 (en) 2010-05-12 2012-12-28 Vertex Pharma Compounds useful as inhibitors of atr kinase
US9514131B1 (en) 2010-05-30 2016-12-06 Crisi Medical Systems, Inc. Medication container encoding, verification, and identification
US10492991B2 (en) 2010-05-30 2019-12-03 Crisi Medical Systems, Inc. Medication container encoding, verification, and identification
EP2585468A1 (en) * 2010-06-23 2013-05-01 Vertex Pharmaceuticals Incorporated Pyrrolo- pyrazine derivatives useful as inhibitors of atr kinase
JP2014510151A (en) 2011-04-05 2014-04-24 バーテックス ファーマシューティカルズ インコーポレイテッド Aminopyrazine compounds useful as ATR kinase (TRAKINASE) inhibitors
US9078809B2 (en) 2011-06-16 2015-07-14 Crisi Medical Systems, Inc. Medication dose preparation and transfer system
US8822469B2 (en) 2011-06-22 2014-09-02 Vertex Pharmaceuticals Incorporated Pyrrolo[2,3-B]pyrazines useful as inhibitors of ATR kinase
US9744298B2 (en) 2011-06-22 2017-08-29 Crisi Medical Systems, Inc. Selectively controlling fluid flow through a fluid pathway
US10293107B2 (en) 2011-06-22 2019-05-21 Crisi Medical Systems, Inc. Selectively Controlling fluid flow through a fluid pathway
JP2014522818A (en) 2011-06-22 2014-09-08 バーテックス ファーマシューティカルズ インコーポレイテッド Compounds useful as ATR kinase inhibitors
EP2723746A1 (en) 2011-06-22 2014-04-30 Vertex Pharmaceuticals Inc. Compounds useful as inhibitors of atr kinase
WO2013049726A2 (en) 2011-09-30 2013-04-04 Vertex Pharmaceuticals Incorporated Processes for making compounds useful as inhibitors of atr kinase
EP2751088B1 (en) 2011-09-30 2016-04-13 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
CA2850491C (en) 2011-09-30 2020-10-27 Vertex Pharmaceuticals Incorporated Treating pancreatic cancer and non-small cell lung cancer with atr inhibiors
CA2850564A1 (en) 2011-09-30 2013-04-04 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
US8853217B2 (en) 2011-09-30 2014-10-07 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
JP2015502925A (en) 2011-11-09 2015-01-29 バーテックス ファーマシューティカルズ インコーポレイテッドVertex Pharmaceuticals Incorporated Pyrazine compounds useful as inhibitors of ATR kinase
WO2013071088A1 (en) 2011-11-09 2013-05-16 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
EP2776429A1 (en) 2011-11-09 2014-09-17 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
US8841450B2 (en) 2011-11-09 2014-09-23 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
WO2013071090A1 (en) 2011-11-09 2013-05-16 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
MX358818B (en) 2012-04-05 2018-09-05 Vertex Pharma Compounds useful as inhibitors of atr kinase and combination therapies thereof.
US8912198B2 (en) 2012-10-16 2014-12-16 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
ES2946360T3 (en) 2012-12-07 2023-07-17 Vertex Pharma Pyrazolo[1,5-a]pyrimidines useful as ATR kinase inhibitors for the treatment of cancer diseases
US10143830B2 (en) 2013-03-13 2018-12-04 Crisi Medical Systems, Inc. Injection site information cap
WO2014143241A1 (en) 2013-03-15 2014-09-18 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2014143240A1 (en) 2013-03-15 2014-09-18 Vertex Pharmaceuticals Incorporated Fused pyrazolopyrimidine derivatives useful as inhibitors of atr kinase
US8969360B2 (en) 2013-03-15 2015-03-03 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
JP2016535097A (en) * 2013-11-08 2016-11-10 アイティーオス セラペウティクス 4- (Indol-3-yl) -pyrazole derivatives, pharmaceutical compositions and methods for use
RU2687276C2 (en) 2013-12-06 2019-05-13 Вертекс Фармасьютикалз Инкорпорейтед Compounds suitable for use as atr kinase inhibitors
LT3152212T (en) 2014-06-05 2020-05-11 Vertex Pharmaceuticals Inc. Radiolabelled derivatives of a 2-amino-6-fluoro-n-[5-fluoro-pyridin-3-yl]- pyrazolo[1,5-a]pyrimidin-3-carboxamide compound useful as atr kinase inhibitor, the preparation of said compound and different solid forms thereof
SG11201610500WA (en) 2014-06-17 2017-01-27 Vertex Pharma Method for treating cancer using a combination of chk1 and atr inhibitors
RU2708391C2 (en) * 2014-08-01 2019-12-06 Янссен Фармацевтика Нв 6,7-DIHYDROPYRAZOLO[1,5-A]PYRAZINE-4(5H)-ONE COMPOUNDS AND USE THEREOF AS NEGATIVE ALLOSTERIC MODULATORS OF mGluR2 RECEPTORS
ES2856404T3 (en) 2014-10-10 2021-09-27 Becton Dickinson Co Syringe Labeling Device
EP3204303B1 (en) 2014-10-10 2021-09-01 Becton, Dickinson and Company A labelling device with a substrate tensioning control device
AU2016209046A1 (en) * 2015-01-23 2017-07-20 Aclaris Therapeutics, Inc. Heterocyclic ITK inhibitors for treating inflammation and cancer
WO2016164641A1 (en) 2015-04-08 2016-10-13 Plexxikon Inc. Compounds and methods for kinase modulation, and indications therefor

Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006015124A2 (en) * 2004-07-27 2006-02-09 Sgx Pharmaceuticals, Inc. Fused ring heterocycle kinase modulators
WO2006058074A1 (en) * 2004-11-22 2006-06-01 Vertex Pharmaceuticals Incorporated Pyrrolopyrazines and pyrazolopyrazines useful as inhibitors of protein kinases
US20070287711A1 (en) * 2004-07-27 2007-12-13 Sgx Pharmaceuticals, Inc. Fused ring heterocycle kinase modulators
WO2010054398A1 (en) * 2008-11-10 2010-05-14 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2010068483A2 (en) * 2008-11-25 2010-06-17 University Of Rochester Mlk inhibitors and methods of use
WO2011008830A1 (en) * 2009-07-15 2011-01-20 Abbott Laboratories Pyrrolopyrazine inhibitors of kinases

Family Cites Families (123)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4309430A (en) 1980-06-27 1982-01-05 Merck & Co., Inc. Pyrazinyl-1,2,4-oxadiazole-5-ones, for treatment of edema, and processes for preparing same
US5329012A (en) 1987-10-29 1994-07-12 The Research Foundation Of State University Of New York Bis(acyloxmethyl)imidazole compounds
JP2597917B2 (en) 1990-04-26 1997-04-09 富士写真フイルム株式会社 Novel dye-forming coupler and silver halide color photographic material using the same
JP2002241379A (en) 1997-03-21 2002-08-28 Dainippon Pharmaceut Co Ltd 3-oxadiazolylquinoxaline derivative
US6420367B1 (en) 1998-07-16 2002-07-16 Shionogo & Co., Ltd. Pyrimidine derivatives exhibiting antitumor activity
US6660753B2 (en) 1999-08-19 2003-12-09 Nps Pharmaceuticals, Inc. Heteropolycyclic compounds and their use as metabotropic glutamate receptor antagonists
DE60037905T2 (en) 1999-12-17 2009-01-29 Novartis Vaccines and Diagnostics, Inc., Emeryville PYRAZIN-BASED INHIBITORS OF GLYCOGEN SYNTHASE KINASE 3
US6849660B1 (en) 2000-08-01 2005-02-01 Isis Pharmaceuticals, Inc. Antimicrobial biaryl compounds
EP1217000A1 (en) 2000-12-23 2002-06-26 Aventis Pharma Deutschland GmbH Inhibitors of factor Xa and factor VIIa
DE60229059D1 (en) 2001-05-08 2008-11-06 Univ Yale PROTEOMIMETIC COMPOUNDS AND METHOD
SE0102438D0 (en) 2001-07-05 2001-07-05 Astrazeneca Ab New compounds
SE0102439D0 (en) 2001-07-05 2001-07-05 Astrazeneca Ab New compounds
US6992087B2 (en) 2001-11-21 2006-01-31 Pfizer Inc Substituted aryl 1,4-pyrazine derivatives
ATE447561T1 (en) 2001-11-21 2009-11-15 Pharmacia & Upjohn Co Llc SUBSTITUTED ARYL 1,4-PYRAZINE DERIVATIVES
JP4656838B2 (en) 2002-02-06 2011-03-23 バーテックス ファーマシューティカルズ インコーポレイテッド Heteroaryl compounds useful as inhibitors of GSK-3
US7205304B2 (en) 2002-03-13 2007-04-17 Janssen Pharmaceutica N.V. Sulfonyl-Derivatives as novel inhibitors of histone deacetylase
GB0206860D0 (en) 2002-03-22 2002-05-01 Glaxo Group Ltd Compounds
TWI319387B (en) 2002-04-05 2010-01-11 Astrazeneca Ab Benzamide derivatives
GB0209715D0 (en) 2002-04-27 2002-06-05 Astrazeneca Ab Chemical compounds
JP4901102B2 (en) 2002-05-03 2012-03-21 エクセリクシス, インク. Protein kinase modulator and method of use thereof
US7704995B2 (en) 2002-05-03 2010-04-27 Exelixis, Inc. Protein kinase modulators and methods of use
IL164209A0 (en) 2002-05-31 2005-12-18 Eisai Co Ltd Pyrazole derivatives and pharmaceutical compositions containing the same
US7015227B2 (en) 2002-06-21 2006-03-21 Cgi Pharmaceuticals, Inc. Certain amino-substituted monocycles as kinase modulators
WO2004033431A2 (en) 2002-10-04 2004-04-22 Arena Pharmaceuticals, Inc. Hydroxypyrazoles for use against metabolic-related disorders
SE0203754D0 (en) 2002-12-17 2002-12-17 Astrazeneca Ab New compounds
SE0203752D0 (en) 2002-12-17 2002-12-17 Astrazeneca Ab New compounds
CA2517720A1 (en) 2003-03-11 2004-09-23 Pfizer Products Inc. Pyrazine compounds as transforming growth factor (tgf) inhibitors
EP1606266A4 (en) 2003-03-21 2008-06-25 Smithkline Beecham Corp Chemical compounds
WO2004084824A2 (en) 2003-03-24 2004-10-07 Merck & Co., Inc. Biaryl substituted 6-membered heterocyles as sodium channel blockers
GB2400101A (en) 2003-03-28 2004-10-06 Biofocus Discovery Ltd Compounds capable of binding to the active site of protein kinases
CA2524867A1 (en) 2003-05-15 2004-12-02 Merck & Co., Inc. 3-(2-amino-1-azacyclyl)-5-aryl-1,2,4-oxadiazoles as s1p receptor agonists
TWI339206B (en) 2003-09-04 2011-03-21 Vertex Pharma Compositions useful as inhibitors of protein kinases
US7807696B2 (en) 2003-10-07 2010-10-05 The Feinstein Institute For Medical Research Isoxazole and isothiazole compounds useful in the treatment of inflammation
WO2005079802A1 (en) 2004-02-12 2005-09-01 Merck & Co., Inc. Bipyridyl amides as modulators of metabotropic glutamate receptor-5
JP2008502687A (en) 2004-06-14 2008-01-31 タケダ サン ディエゴ インコーポレイテッド Kinase inhibitor
JP5122280B2 (en) * 2004-06-30 2013-01-16 バーテックス ファーマシューティカルズ インコーポレイテッド Azaindoles useful as inhibitors of protein kinases
WO2006050162A2 (en) * 2004-10-28 2006-05-11 Phenomix Corporation Imidazole derivatives
EP1828144A2 (en) 2004-11-12 2007-09-05 OSI Pharmaceuticals, Inc. Integrin antagonists useful as anticancer agents
GB0428235D0 (en) 2004-12-23 2005-01-26 Glaxo Group Ltd Novel compounds
ES2368338T3 (en) 2004-12-27 2011-11-16 Novartis Ag AMINOPIRAZINE ANALOGS FOR THE TREATMENT OF GLAUCOMA AND OTHER DISEASES MEDIATED BY RHO CINASA.
GB0500492D0 (en) 2005-01-11 2005-02-16 Cyclacel Ltd Compound
US7622583B2 (en) 2005-01-14 2009-11-24 Chemocentryx, Inc. Heteroaryl sulfonamides and CCR2
GB0501999D0 (en) 2005-02-01 2005-03-09 Sentinel Oncology Ltd Pharmaceutical compounds
MX2007009945A (en) 2005-02-16 2007-09-26 Schering Corp Heterocyclic substituted piperazines with cxcr3 antagonist activity.
US8859581B2 (en) 2005-04-25 2014-10-14 Merck Patent Gmbh Azaheterocyclic compounds as kinase inhibitors
WO2007015632A1 (en) 2005-08-04 2007-02-08 Cgk Co., Ltd. Atm and atr inhibitor
TW200736260A (en) 2005-11-10 2007-10-01 Smithkline Beecham Corp Inhibitors of Akt activity
AU2006319247B2 (en) 2005-12-01 2010-03-11 F. Hoffmann-La Roche Ag Heteroaryl substituted piperidine derivatives as L-CPT1 inhibitors
EP1970377A4 (en) 2005-12-09 2013-02-27 Meiji Seika Kaisha Lincomycin derivative and antibacterial agent containing the same as active ingredient
ZA200804497B (en) 2005-12-22 2009-10-28 Alcon Res Ltd (Indazol-5-yl)-pyrazines and (1,3-dihydro-indol-2-one)-pyrazines for treating rho kinase-mediated diseases and conditions
ITMI20060311A1 (en) 2006-02-21 2007-08-22 Btsr Int Spa PERFECT DEVICE FOR WIRE OR FILATIO SUPPLY TO A TEXTILE MACHINE AND METHOD TO IMPLEMENT THIS POWER SUPPLY
GB0603684D0 (en) 2006-02-23 2006-04-05 Novartis Ag Organic compounds
WO2007096764A2 (en) 2006-02-27 2007-08-30 Glenmark Pharmaceuticals S.A. Bicyclic heteroaryl derivatives as cannabinoid receptor modulators
TW200800203A (en) 2006-03-08 2008-01-01 Astrazeneca Ab New use
EP2004625B1 (en) 2006-03-22 2009-12-30 Vertex Pharmaceuticals Incorporated C-met protein kinase inhibitors for the treatment of proliferative disorders
WO2007126964A2 (en) 2006-03-31 2007-11-08 Schering Corporation Kinase inhibitors
EP2038261A2 (en) 2006-06-22 2009-03-25 Mallinckrodt Inc. Pyrazine derivatives with extended conjugation and uses thereof
BRPI0713328A2 (en) 2006-06-22 2012-10-30 Biovitrum Ab pyridine and pyrazine derivatives as mnk kinase inhibitors
US20090286779A1 (en) 2006-09-29 2009-11-19 Novartis Ag Pyrazolopyrimidines as lipid kinase inhibitors
GB0619342D0 (en) 2006-09-30 2006-11-08 Vernalis R&D Ltd New chemical compounds
BRPI0717845A2 (en) 2006-10-04 2015-06-16 Hoffmann La Roche Use of compounds, pharmaceutical compositions and methods for the treatment and / or prophylaxis of diseases that can be treated with HDL-cholesterol elevating agents and compounds.
WO2008060907A2 (en) 2006-11-10 2008-05-22 Bristol-Myers Squibb Company Pyrrolo-pyridine kinase inhibitors
EP2125808A2 (en) 2006-12-15 2009-12-02 Bayer Schering Pharma Aktiengesellschaft 3-h-pyrazolopyridines and salts thereof, pharmaceutical compositions comprising same, methods of preparing same and uses of same
WO2008079291A2 (en) 2006-12-20 2008-07-03 Amgen Inc. Substituted heterocycles and methods of use
GB0625659D0 (en) 2006-12-21 2007-01-31 Cancer Rec Tech Ltd Therapeutic compounds and their use
PE20121126A1 (en) 2006-12-21 2012-08-24 Plexxikon Inc PIRROLO [2,3-B] PYRIDINES COMPOUNDS AS KINASE MODULATORS
US8314087B2 (en) 2007-02-16 2012-11-20 Amgen Inc. Nitrogen-containing heterocyclyl ketones and methods of use
EP2132177B1 (en) 2007-03-01 2013-07-17 Novartis AG Pim kinase inhibitors and methods of their use
RU2009141185A (en) 2007-04-10 2011-05-20 Байер КропСайенс АГ (DE) INSECTICIDAL DERIVATIVES OF ARILIZOXOXAZOLINE
JP2010526823A (en) 2007-05-10 2010-08-05 グラクソスミスクライン・リミテッド・ライアビリティ・カンパニー Quinoxaline derivatives as PI3 kinase inhibitors
PE20090717A1 (en) 2007-05-18 2009-07-18 Smithkline Beecham Corp QUINOLINE DERIVATIVES AS PI3 KINASE INHIBITORS
UY31137A1 (en) 2007-06-14 2009-01-05 Smithkline Beecham Corp DERIVATIVES OF QUINAZOLINE AS INHIBITORS OF THE PI3 QUINASA
US20100190980A1 (en) 2007-06-21 2010-07-29 Taisho Pharmaceutical Co., Ltd Pyrazinamide compound
MX2009013990A (en) 2007-06-26 2010-03-30 Lexicon Pharmaceuticals Inc Methods of treating serotonin-mediated diseases and disorders.
GB0713259D0 (en) 2007-07-09 2007-08-15 Astrazeneca Ab Pyrazine derivatives 954
AU2008279776B2 (en) 2007-07-19 2013-05-23 Merck Sharp & Dohme Corp. Heterocyclic amide compounds as protein kinase inhibitors
UA105758C2 (en) 2007-07-19 2014-06-25 Х. Луннбек А/С 5-membered heterocyclic amides and related compounds
BRPI0815042A2 (en) 2007-08-01 2015-02-10 Pfizer PIRAZOL COMPOUNDS
WO2009024825A1 (en) 2007-08-21 2009-02-26 Astrazeneca Ab 2-pyrazinylbenzimidazole derivatives as receptor tyrosine kinase inhibitors
WO2009037247A1 (en) 2007-09-17 2009-03-26 Neurosearch A/S Pyrazine derivatives and their use as potassium channel modulators
CN101910158A (en) 2007-10-25 2010-12-08 阿斯利康(瑞典)有限公司 Pyridine and pyrazine derivatives useful in the treatment of cell proliferative disorders
CA2713710A1 (en) 2008-02-25 2009-09-03 F. Hoffmann-La Roche Ag Pyrrolopyrazine kinase inhibitors
MX2010009023A (en) 2008-02-25 2010-09-07 Hoffmann La Roche Pyrrolopyrazine kinase inhibitors.
CN101945877B (en) 2008-02-25 2013-07-03 霍夫曼-拉罗奇有限公司 Pyrrolopyrazine kinase inhibitors
ATE522536T1 (en) 2008-02-25 2011-09-15 Hoffmann La Roche PYRROLOPYRAZINE KINASE INHIBITORS
TW200940537A (en) 2008-02-26 2009-10-01 Astrazeneca Ab Heterocyclic urea derivatives and methods of use thereof
WO2009152087A1 (en) 2008-06-10 2009-12-17 Plexxikon, Inc. Bicyclic heteroaryl compounds and methods for kinase modulation, and indications therefor
GB0814364D0 (en) 2008-08-05 2008-09-10 Eisai London Res Lab Ltd Diazaindole derivatives and their use in the inhibition of c-Jun N-terminal kinase
AU2009307770A1 (en) 2008-10-21 2010-04-29 Vertex Pharmaceuticals Incorporated C-Met protein kinase inhibitors
AU2009324210A1 (en) 2008-12-05 2010-06-10 F. Hoffmann-La Roche Ag Pyrrolopyrazinyl urea kinase inhibitors
CA3013000C (en) 2008-12-19 2022-12-13 Vertex Pharmaceuticals Incorporated Pyrazine derivatives useful as inhibitors of atr kinase
CN102264737A (en) 2008-12-23 2011-11-30 雅培制药有限公司 Anti-viral compounds
KR20110132564A (en) 2009-02-11 2011-12-08 선오비온 파마슈티컬스 인코포레이티드 Histamine h3 inverse agonists and antagonists and methods of use thereof
US20120065247A1 (en) 2009-03-27 2012-03-15 Discoverybiomed, Inc. Modulating ires-mediated translation
US8518945B2 (en) 2010-03-22 2013-08-27 Hoffmann-La Roche Inc. Pyrrolopyrazine kinase inhibitors
CA2794428A1 (en) 2010-04-08 2011-10-13 Pfizer Inc. Substituted 3,5-diphenyl-isoxazoline derivatives as insecticides and acaricides
EP2558866B1 (en) 2010-04-15 2016-08-17 Tracon Pharmaceuticals, Inc. Potentiation of anti-cancer activity through combination therapy with ber pathway inhibitors
CA2798760A1 (en) 2010-05-12 2011-11-17 Vertex Pharmaceuticals Incorporated 2-aminopyridine derivatives useful as inhibitors of atr kinase
EP2569286B1 (en) 2010-05-12 2014-08-20 Vertex Pharmaceuticals Inc. Compounds useful as inhibitors of atr kinase
EP2568984A1 (en) 2010-05-12 2013-03-20 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2011143419A1 (en) 2010-05-12 2011-11-17 Vertex Pharmaceuticals Incorporated Pyrazines useful as inhibitors of atr kinase
EP2569287B1 (en) 2010-05-12 2014-07-09 Vertex Pharmaceuticals Inc. Compounds useful as inhibitors of atr kinase
SG185524A1 (en) 2010-05-12 2012-12-28 Vertex Pharma Compounds useful as inhibitors of atr kinase
CN102906095A (en) 2010-05-20 2013-01-30 弗·哈夫曼-拉罗切有限公司 Pyrrolopyrazine derivatives as SYK and JAK inhibitors
CA2799904A1 (en) 2010-05-20 2011-11-24 F. Hoffmann-La Roche Ag Pyrrolo [2, 3 - b] pyrazine - 7 - carboxamide derivatives and their use as jak and syk inhibitors
EP2585468A1 (en) 2010-06-23 2013-05-01 Vertex Pharmaceuticals Incorporated Pyrrolo- pyrazine derivatives useful as inhibitors of atr kinase
CN102311396B (en) 2010-07-05 2015-01-07 暨南大学 Pyrazine derivative and preparation method as well as application thereof to pharmacy
EP2407478A1 (en) 2010-07-14 2012-01-18 GENETADI Biotech, S.L. New cyclotetrapeptides with pro-angiogenic properties
JP2014510151A (en) 2011-04-05 2014-04-24 バーテックス ファーマシューティカルズ インコーポレイテッド Aminopyrazine compounds useful as ATR kinase (TRAKINASE) inhibitors
MX2013013331A (en) 2011-05-17 2014-10-17 Principia Biopharma Inc Azaindole derivatives as tyrosine kinase inhibitors.
US8822469B2 (en) 2011-06-22 2014-09-02 Vertex Pharmaceuticals Incorporated Pyrrolo[2,3-B]pyrazines useful as inhibitors of ATR kinase
EP2723746A1 (en) 2011-06-22 2014-04-30 Vertex Pharmaceuticals Inc. Compounds useful as inhibitors of atr kinase
JP2014522818A (en) 2011-06-22 2014-09-08 バーテックス ファーマシューティカルズ インコーポレイテッド Compounds useful as ATR kinase inhibitors
CA2850491C (en) 2011-09-30 2020-10-27 Vertex Pharmaceuticals Incorporated Treating pancreatic cancer and non-small cell lung cancer with atr inhibiors
US8853217B2 (en) 2011-09-30 2014-10-07 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
WO2013049726A2 (en) 2011-09-30 2013-04-04 Vertex Pharmaceuticals Incorporated Processes for making compounds useful as inhibitors of atr kinase
CA2850564A1 (en) 2011-09-30 2013-04-04 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
EP2751088B1 (en) 2011-09-30 2016-04-13 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
EP2776429A1 (en) 2011-11-09 2014-09-17 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
JP2015502925A (en) 2011-11-09 2015-01-29 バーテックス ファーマシューティカルズ インコーポレイテッドVertex Pharmaceuticals Incorporated Pyrazine compounds useful as inhibitors of ATR kinase
WO2013071088A1 (en) 2011-11-09 2013-05-16 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
US8841450B2 (en) 2011-11-09 2014-09-23 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of ATR kinase
WO2013071090A1 (en) 2011-11-09 2013-05-16 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006015124A2 (en) * 2004-07-27 2006-02-09 Sgx Pharmaceuticals, Inc. Fused ring heterocycle kinase modulators
US20070287711A1 (en) * 2004-07-27 2007-12-13 Sgx Pharmaceuticals, Inc. Fused ring heterocycle kinase modulators
WO2006058074A1 (en) * 2004-11-22 2006-06-01 Vertex Pharmaceuticals Incorporated Pyrrolopyrazines and pyrazolopyrazines useful as inhibitors of protein kinases
WO2010054398A1 (en) * 2008-11-10 2010-05-14 Vertex Pharmaceuticals Incorporated Compounds useful as inhibitors of atr kinase
WO2010068483A2 (en) * 2008-11-25 2010-06-17 University Of Rochester Mlk inhibitors and methods of use
WO2011008830A1 (en) * 2009-07-15 2011-01-20 Abbott Laboratories Pyrrolopyrazine inhibitors of kinases

Non-Patent Citations (17)

* Cited by examiner, † Cited by third party
Title
"March's Advanced Organic Chemistry", 2001, JOHN WILEY & SONS
"Remington's Pharmaceutical Sciences", 1980, MACK PUBLISHING CO.
"The Merck Manual", 1999
CHIANG ET AL.: "Determination of the catalytic activities ofmTOR and other members of the phosphoinositide-3-kinase-related kinase family", METHODS MOL. BIOL., vol. 281, 2004, pages 125 - 41
CLARK B A J ET AL: "MASS SPECTROMETRY OF PYRROLOÄ2,3-BÜPYRAZINES AND PYRAZINOÄ2,3-BÜINDOLE", OMS. ORGANIC MASS SPECTROMETRY, WILEY, CHICHESTER, GB, vol. 12, no. 7, 1 July 1997 (1997-07-01), pages 421 - 423, XP001107293, ISSN: 0030-493X, DOI: 10.1002/OMS.1210120703 *
GREENE, T.W., WUTS, P. G: "Protective Groups in Organic Synthesis", 1999, JOHN WILEY & SONS
HALL-JACKSON ET AL., ONCOGENE, vol. 18, 1999, pages 6707 - 6713
HICKSON ET AL.: "Identification and Characterization of a Novel and Specific Inhibitor of the Ataxia-Telangiectasia Mutated Kinase ATM", CANCER RESEARCH, vol. 64, 2004, pages 9152 - 9159
KIM ET AL., ONCOGENE, vol. 21, 2002, pages 3864
KIM ET AL.: "Substrate Specificities and Identification of Putative Substrates of ATM Kinase Family Members", THE JOURNAL OFBIOLOGICAL CHEMISTRY, vol. 274, no. 53, 1999, pages 37538 - 37543
KUMAGAI ET AL., CELL, vol. 124, 10 March 2006 (2006-03-10), pages 943 - 955
S. M. BERGE ET AL., J. PHARMACEUTICAL SCIENCES, vol. 66, 1977, pages 1 - 19
SARKARIA ET AL.: "Inhibition of ATM and ATR Kinase Activities by the Radiosensitizing Agent", CAFFEINE: CANCER RESEARCH, vol. 59, 1999, pages 4375 - 5382
TAKEMURA ET AL., JBC, vol. 281, 2006, pages 30814
THOMAS SORRELL: "Handbook of Chemistry and Physics", 1999, UNIVERSITY SCIENCE BOOKS, article "Organic Chemistry"
UNSAI-KACMAZ ET AL., PNAS, vol. 10, 14 May 2002 (2002-05-14), pages 6673 - 6678
UNSAL-KACMAZ ET AL., MOLECULAR AND CELLULAR BIOLOGY, February 2004 (2004-02-01), pages 1292 - 1300

Cited By (47)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2015503609A (en) * 2012-01-13 2015-02-02 ノバルティス アーゲー Condensed pyrrole as an IP receptor agonist for the treatment of pulmonary arterial hypertension (PAH) and related disorders
WO2014055756A1 (en) 2012-10-04 2014-04-10 Vertex Pharmaceuticals Incorporated Method for measuring atr inhibition mediated increases in dna damage
JP2015533151A (en) * 2012-10-16 2015-11-19 エフ・ホフマン−ラ・ロシュ・アクチェンゲゼルシャフト Serine / threonine kinase inhibitor
JP2016510803A (en) * 2013-03-15 2016-04-11 フンダシオン セントロ ナシオナル デ インベスティガシオネス オンコロヒカス カルロス ザ サードFundacion Centro Nacional de Investigaciones Oncologicas Carlos III Chemical substances
TWI700283B (en) * 2014-08-04 2020-08-01 德商拜耳製藥公司 2-(morpholin-4-yl)-1,7-naphthyridines
KR102180006B1 (en) 2014-08-04 2020-11-18 바이엘 파마 악티엔게젤샤프트 2-(morpholin-4-yl)-1,7-naphthyridines
US11529356B2 (en) 2014-08-04 2022-12-20 Bayer Pharma Aktiengesellschaft 2-(morpholin-4-yl)-1,7-naphthyridines
CN106795156B (en) * 2014-08-04 2019-07-30 拜耳制药股份公司 2- (morpholine -4- base) -1,7- naphthyridines
CN106795156A (en) * 2014-08-04 2017-05-31 拜耳制药股份公司 2 (base of morpholine 4) 1,7 naphthyridines
JP2017523987A (en) * 2014-08-04 2017-08-24 バイエル・ファルマ・アクティエンゲゼルシャフト 2- (Morpholin-4-yl) -1,7-naphthyridine
JP2018062517A (en) * 2014-08-04 2018-04-19 バイエル・ファルマ・アクティエンゲゼルシャフト 2-(morpholin-4-yl)-1,7-naphthyridines
US9993484B2 (en) 2014-08-04 2018-06-12 Bayer Pharma Aktiengesellschaft 2-(morpholin-4-yl)-1,7-naphthyridines
CN110204544B (en) * 2014-08-04 2022-04-12 拜耳制药股份公司 2- (morpholin-4-yl) -1, 7-naphthyridine
CN110256427B (en) * 2014-08-04 2022-02-22 拜耳制药股份公司 2- (morpholin-4-yl) -1, 7-naphthyridine
KR102317169B1 (en) 2014-08-04 2021-10-27 바이엘 파마 악티엔게젤샤프트 2-(morpholin-4-yl)-1,7-naphthyridines
KR20200130755A (en) * 2014-08-04 2020-11-19 바이엘 파마 악티엔게젤샤프트 2-(morpholin-4-yl)-1,7-naphthyridines
EP3395818A1 (en) 2014-08-04 2018-10-31 Bayer Pharma Aktiengesellschaft 2-(morpholin-4-yl)-1,7-naphthyridines
US9549932B2 (en) 2014-08-04 2017-01-24 Bayer Pharma Aktiengesellschaft 2-(morpholin-4-yl)-1,7-naphthyridines
US10772893B2 (en) 2014-08-04 2020-09-15 Bayer Pharma Aktiengesellschaft 2-(morpholin-4-yl)-1,7-naphthyridines
CN110204544A (en) * 2014-08-04 2019-09-06 拜耳制药股份公司 2- (morpholine -4- base) -1,7- naphthyridines
EA031678B1 (en) * 2014-08-04 2019-02-28 Байер Фарма Акциенгезельшафт 2-(morpholin-4-yl)-1,7-naphthyridines
AU2015299173B2 (en) * 2014-08-04 2019-03-14 Bayer Pharma Aktiengesellschaft 2-(morpholin-4-yl)-l,7-naphthyridines
WO2016020320A1 (en) * 2014-08-04 2016-02-11 Bayer Pharma Aktiengesellschaft 2-(morpholin-4-yl)-l,7-naphthyridines
TWI656121B (en) * 2014-08-04 2019-04-11 德商拜耳製藥公司 2-(morpholin-4-yl)-1,7-naphthyridine
AU2019204125B2 (en) * 2014-08-04 2020-06-25 Bayer Pharma Aktiengesellschaft 2-(Morpholin-4-yl)-1,7-naphthyridines
KR20170040300A (en) * 2014-08-04 2017-04-12 바이엘 파마 악티엔게젤샤프트 2-(morpholin-4-yl)-l,7-naphthyridines
EA035039B1 (en) * 2014-08-04 2020-04-21 Байер Фарма Акциенгезельшафт 2-(morpholin-4-yl)-1,7-naphthyridines
CN110256427A (en) * 2014-08-04 2019-09-20 拜耳制药股份公司 2- (morpholine -4- base) -1,7- naphthyridines
WO2017059357A1 (en) 2015-09-30 2017-04-06 Vertex Pharmaceuticals Incorporated Method for treating cancer using a combination of dna damaging agents and atr inhibitors
US11787781B2 (en) 2016-01-11 2023-10-17 Celator Pharmaceuticals, Inc. Inhibiting ataxia telangiectasia and RAD3-related protein (ATR)
US11028076B2 (en) 2016-01-11 2021-06-08 Celator Pharmaceuticals, Inc. Inhibiting ataxia telangiectasia and Rad3-related protein (ATR)
WO2018153973A1 (en) * 2017-02-24 2018-08-30 Bayer Pharma Aktiengesellschaft Combination of atr kinase inhibitors with parp inhibitors
US11660301B2 (en) 2017-02-24 2023-05-30 Bayer Pharma Aktiengesellschaft Combination of ATR kinase inhibitors with PARP inhibitors
WO2018153972A1 (en) 2017-02-24 2018-08-30 Bayer Pharma Aktiengesellschaft Combination of atr kinase inhibitors and antiandrogens
WO2018153968A1 (en) 2017-02-24 2018-08-30 Bayer Aktiengesellschaft An inhibitor of atr kinase for use in a method of treating a hyper-proliferative disease
WO2018153969A1 (en) 2017-02-24 2018-08-30 Bayer Aktiengesellschaft Combination of atr kinase inhibitors with radium-223 salt
WO2018206547A1 (en) 2017-05-12 2018-11-15 Bayer Pharma Aktiengesellschaft Combination of bub1 and atr inhibitors
WO2018210661A1 (en) * 2017-05-15 2018-11-22 Basf Se Heteroaryl compounds as agrochemical fungicides
WO2019025440A1 (en) 2017-08-04 2019-02-07 Bayer Pharma Aktiengesellschaft Combination of atr kinase inhibitors and pd-1/pd-l1 inhibitors
EP3461480A1 (en) 2017-09-27 2019-04-03 Onxeo Combination of a dna damage response cell cycle checkpoint inhibitors and belinostat for treating cancer
WO2019110586A1 (en) 2017-12-08 2019-06-13 Bayer Aktiengesellschaft Predictive markers for atr kinase inhibitors
RU2802512C2 (en) * 2018-08-24 2023-08-30 Байер Акциенгезельшафт Method for producing 2-[(3r)-3-methylmorpholin-4-yl]-4-(1-methyl-1h-pyrazol-5-yl)-8-(1h-pyrazol-5-yl)-1,7-nafthyridine
WO2020064971A1 (en) 2018-09-26 2020-04-02 Merck Patent Gmbh Combination of a pd-1 antagonist, an atr inhibitor and a platinating agent for the treatment of cancer
WO2020078905A1 (en) 2018-10-15 2020-04-23 Merck Patent Gmbh Combination therapy utilizing dna alkylating agents and atr inhibitors
WO2020078788A1 (en) 2018-10-16 2020-04-23 Bayer Aktiengesellschaft Combination of atr kinase inhibitors with 2,3-dihydroimidazo[1,2-c]quinazoline compounds
WO2020259601A1 (en) * 2019-06-28 2020-12-30 Impact Therapeutics, Inc Substituted fused heteroaromatic bicyclic compounds as kinase inhibitors and the use thereof
WO2022228522A1 (en) * 2021-04-29 2022-11-03 江苏先声药业有限公司 Alkyne compound as hpk1 inhibitor and use thereof

Also Published As

Publication number Publication date
AU2011270807A1 (en) 2013-01-31
NZ605627A (en) 2015-06-26
JP2015134834A (en) 2015-07-27
JP2013529643A (en) 2013-07-22
MX2013000103A (en) 2013-06-13
US8623869B2 (en) 2014-01-07
US20120046295A1 (en) 2012-02-23
CA2803802A1 (en) 2011-12-29
US20140288347A1 (en) 2014-09-25
EP2585468A1 (en) 2013-05-01
US20160009723A1 (en) 2016-01-14

Similar Documents

Publication Publication Date Title
US9334244B2 (en) Compounds useful as inhibitors of ATR kinase
US8623869B2 (en) Compounds useful as inhibitors of ATR kinase
EP2569286B1 (en) Compounds useful as inhibitors of atr kinase
EP2569284B1 (en) 2-aminopyridine derivatives useful as inhibitors of atr kinase
US8969356B2 (en) Compounds useful as inhibitors of ATR kinase
US8822469B2 (en) Pyrrolo[2,3-B]pyrazines useful as inhibitors of ATR kinase
JP5836367B2 (en) Compounds useful as ATR kinase inhibitors
US8912198B2 (en) Compounds useful as inhibitors of ATR kinase
EP2723746A1 (en) Compounds useful as inhibitors of atr kinase

Legal Events

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

Ref document number: 11729535

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 2013516779

Country of ref document: JP

Kind code of ref document: A

Ref document number: 2803802

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: MX/A/2013/000103

Country of ref document: MX

WWE Wipo information: entry into national phase

Ref document number: 2011729535

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2011270807

Country of ref document: AU

Date of ref document: 20110623

Kind code of ref document: A