WO2019057757A1 - 1,3-dihydroimidazo[4,5-c]cinnolin-2-one compounds and their use in treating cancer - Google Patents

1,3-dihydroimidazo[4,5-c]cinnolin-2-one compounds and their use in treating cancer Download PDF

Info

Publication number
WO2019057757A1
WO2019057757A1 PCT/EP2018/075327 EP2018075327W WO2019057757A1 WO 2019057757 A1 WO2019057757 A1 WO 2019057757A1 EP 2018075327 W EP2018075327 W EP 2018075327W WO 2019057757 A1 WO2019057757 A1 WO 2019057757A1
Authority
WO
WIPO (PCT)
Prior art keywords
methyl
imidazo
cinnolin
propoxy
pyridyl
Prior art date
Application number
PCT/EP2018/075327
Other languages
French (fr)
Inventor
Kurt Gordon Pike
Bernard Christophe Barlaam
Original Assignee
Astrazeneca Ab
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 Astrazeneca Ab filed Critical Astrazeneca Ab
Priority to CN201880073208.2A priority Critical patent/CN111344293A/en
Publication of WO2019057757A1 publication Critical patent/WO2019057757A1/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
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/17Amides, e.g. hydroxamic acids having the group >N—C(O)—N< or >N—C(S)—N<, e.g. urea, thiourea, carmustine
    • A61K31/175Amides, e.g. hydroxamic acids having the group >N—C(O)—N< or >N—C(S)—N<, e.g. urea, thiourea, carmustine having the group, >N—C(O)—N=N— or, e.g. carbonohydrazides, carbazones, semicarbazides, semicarbazones; Thioanalogues thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • A61K31/196Carboxylic acids, e.g. valproic acid having an amino group the amino group being directly attached to a ring, e.g. anthranilic acid, mefenamic acid, diclofenac, chlorambucil
    • 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
    • A61K31/40Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil
    • A61K31/407Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with one nitrogen as the only ring hetero atom, e.g. sulpiride, succinimide, tolmetin, buflomedil condensed with other heterocyclic ring systems, e.g. ketorolac, physostigmine
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41841,3-Diazoles condensed with carbocyclic rings, e.g. benzimidazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
    • 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
    • A61K31/50Pyridazines; Hydrogenated pyridazines
    • A61K31/5025Pyridazines; Hydrogenated pyridazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7048Compounds having saccharide radicals and heterocyclic rings having oxygen as a ring hetero atom, e.g. leucoglucosan, hesperidin, erythromycin, nystatin, digitoxin or digoxin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Definitions

  • This specification relates to l,3-dihydroimidazo[4,5-c]cinnolin-2-one compounds and pharmaceutically acceptable salts thereof. These compounds and salts selectively modulate ataxia telangiectasia mutated ("ATM") kinase, and the specification therefore also relates to the use of l,3-dihydroimidazo[4,5-c]cinnolin-2-one compounds and salts thereof to treat or prevent ATM mediated disease, including cancer.
  • the specification further relates to pharmaceutical compositions comprising l,3-dihydroimidazo[4,5- c]cinnolin-2-one compounds and pharmaceutically acceptable salts thereof; and kits comprising such compounds and salts.
  • ATM kinase is a serine threonine kinase originally identified as the product of the gene mutated in ataxia telangiectasia.
  • Ataxia telangiectasia is located on human chromosome 1 lq22-23 and codes for a large protein of about 350 kDa, which is characterized by the presence of a phosphatidylinositol ("PI") 3-kinase-like
  • ATM kinase has been identified as a major player of the DNA damage response elicited by double strand breaks. It primarily functions in S/G2/M cell cycle transitions and at collapsed replication forks to initiate cell cycle checkpoints, chromatin modification, HR repair and pro-survival signalling cascades in order to maintain cell integrity after DNA damage (Lavin, M. F.; Rev. Mol. Cell Biol. 2008, 759-769).
  • ATM kinase signalling can be broadly divided into two categories: a canonical pathway, which signals together with the Mrel 1-Rad50-NBS1 complex from double strand breaks and activates the DNA damage checkpoint, and several non-canonical modes of activation, which are activated by other forms of cellular stress (Cremona et al. , Oncogene 2013, 3351-3360).
  • ATM kinase is rapidly and robustly activated in response to double strand breaks and is reportedly able to phosphorylate in excess of 800 substrates (Matsuoka et al., Science 2007, 1160-1166), coordinating multiple stress response pathways (Kurz and Lees Miller, DNA Repair 2004, 889-900).
  • ATM kinase is present predominantly in the nucleus of the cell in an inactive homodimeric form but autophosphorylates itself on Serl981 upon sensing a DNA double strand break (canonical pathway), leading to dissociation to a monomer with full kinase activity (Bakkenist et al., Nature 2003, 499-506). This is a critical activation event, and ATM phospho-Serl981 is therefore both a direct
  • ATM kinase responds to direct double strand breaks caused by common anti-cancer treatments such as ionising radiation and topoisomerase-II inhibitors (doxorubicin, etoposide) and also to topoisomerase-I inhibitors (for example irinotecan and topotecan) via single strand break to double strand break conversion during replication.
  • ionising radiation and topoisomerase-II inhibitors doxorubicin, etoposide
  • topoisomerase-I inhibitors for example irinotecan and topotecan
  • ATM kinase inhibition can potentiate the activity of any these agents.
  • ATM kinase inhibitors are therefore expected to be of use in the treatment of cancer.
  • WO 2015/170081, WO 2017/046216, WO 2017/076895 and WO 2017/076898 describe imidazo[4,5-c]quinolin-2-one compounds that have activity as ATM kinase modulators.
  • WO 2017/170081, WO 2017/046216, WO 2017/076895 and WO 2017/076898 describe imidazo[4,5-c]quinolin-2-one compounds that have activity as ATM kinase modulators.
  • kinase enzymes like ATM kinase, in a highly selective fashion (i.e. by modulating ATM more effectively than other biological targets).
  • the compounds of the present specification generally possess very potent ATM kinase inhibitory activity, but much less potent activity against other tyrosine kinase enzymes, such as PI 3-kinase a, mTOR kinase and ataxia telangiectasia and Rad3-related protein ("ATR") kinase.
  • the compounds of the present specification not only inhibit ATM kinase, but can be considered to be highly selective inhibitors of ATM kinase.
  • the compounds of the present specification are expected to be particularly useful in the treatment of diseases in which ATM kinase is implicated (for example, in the treatment of cancer), but where it is desirable to minimise off-target effects or toxicity that might arise due to the inhibition of other tyrosine kinase enzymes, such as class PI 3-kinase a, mTOR kinase and ATR kinase.
  • other tyrosine kinase enzymes such as class PI 3-kinase a, mTOR kinase and ATR kinase.
  • A is N or CR 4 ;
  • X is -OR 5 or -NR 6 R 7 ;
  • R 1 is (Ci-C6)alkyl, cycloalkyl or heterocycloalkyl;
  • R 2 is H or methyl, wherein methyl is optionally substituted with 1, 2, or 3 halo substituents;
  • R 3 is H, (Ci-C 6 )alkyl or halo
  • R 4 is H, (Ci-C 6 )alkyl or halo
  • R 5 is -(CH 2 ) n -NR 8 R 9 ;
  • R 6 and R 7 together with the nitrogen atom to which they are attached form a 4, 5 or 6 membered non-aromatic monocyclic ring comprising carbon which is substituted with - NR 10 R U ;
  • R 8 and R 9 are independently selected from H and (Ci-Ce)alkyl; or R 8 and R 9 together with the nitrogen atom to which they are attached form a 4, 5 or 6 membered non- aromatic monocyclic ring comprising carbon which can be optionally substituted with halo;
  • R 10 is H or (Ci-C 3 )alkyl
  • R 11 is (Ci-C )alkyl
  • n 2, 3 or 4;
  • alkyl is a linear or branched saturated hydrocarbon
  • alkoxy is a linear or branched O-linked saturated hydrocarbon
  • cycloalkyl is a 3, 4, 5 or 6 membered aliphatic carbocyclic ring, which can be optionally substituted with 1 or 2 substituents selected from (Ci-C 3 )alkyl, (Ci-C 3 )alkoxy and -OH;
  • halo is F, CI or Br
  • heterocycloalkyl is a 4, 5 or 6 membered non-aromatic monocyclic ring comprising carbon and 1 or 2 heteroatoms selected from nitrogen and oxygen;
  • heterocycloalkyl can be optionally substituted with 1 or 2 substituents selected from (Ci- C )alkyl, (Ci-C )alkoxy and -OH.
  • An aspect of this specification includes compounds of formula (I), as defined above, tautomers, isomers, stereoisomers (including enantiomers, diastereoisomers and racemic and scalemic mixtures thereof), pharmaceutically acceptable salts and solvates thereof.
  • the specification provides a prodrug of a compound of formula (I) as herein defined, or a pharmaceutically acceptable salt thereof.
  • the specification provides an N-oxide of a compound of formula (I) as herein defined, or a prodrug or pharmaceutically acceptable salt thereof.
  • composition which comprises a compound of Formula (I), or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient.
  • This specification also describes, in part, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in therapy.
  • This specification also describes, in part, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer.
  • This specification also describes, in part, the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of cancer.
  • This specification also describes, in part, a method for treating cancer in a warm blooded animal in need of such treatment, which comprises administering to said warmblooded animal a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof.
  • A is N or CR 4 ;
  • X is -OR 5 or -NR 6 R 7 ;
  • R 1 is (Ci-C6)alkyl, cycloalkyl or heterocycloalkyl
  • R 2 is H or methyl, wherein methyl is optionally substituted with 1, 2, or 3 halo substituents;
  • R 3 is H, (Ci-C 6 )alkyl or halo
  • R 4 is H, (Ci-C 6 )alkyl or halo
  • R 5 is -(CH 2 ) n -NR 8 R 9 ;
  • R 6 and R 7 together with the nitrogen atom to which they are attached form a 4, 5 or 6 membered non-aromatic monocyclic ring comprising carbon which is substituted with - NR 10 R U ;
  • R 8 and R 9 are independently selected from H and (Ci-Ce)alkyl; or R 8 and R 9 together with the nitrogen atom to which they are attached form a 4, 5 or 6 membered non- aromatic monocyclic ring comprising carbon which can be optionally substituted with halo;
  • R 10 is H or (Ci-C 3 )alkyl
  • R 11 is (Ci-C )alkyl
  • n 2, 3 or 4;
  • alkyl is a linear or branched saturated hydrocarbon
  • alkoxy is a linear or branched O-linked saturated hydrocarbon
  • cycloalkyl is a 3, 4, 5 or 6 membered aliphatic carbocyclic ring, which can be optionally substituted with 1 or 2 substituents selected from (Ci-C 3 )alkyl, (Ci-C 3 )alkoxy and -OH; halo is F, CI or Br;
  • heterocycloalkyl is a 4, 5 or 6 membered non-aromatic monocyclic ring comprising carbon and 1 or 2 heteroatoms selected from nitrogen and oxygen;
  • heterocycloalkyl can be optionally substituted with 1 or 2 substituents selected from (Ci- C 3 )alkyl, (Ci-C )alkoxy and -OH.
  • A is N or CH
  • X is -OR 5 or -NR 6 R 7 ;
  • R 1 is (Ci-C 3 )alkyl, cycloalkyl, heterocycloalkyl;
  • R 2 is H or methyl, wherein methyl is optionally substituted with 1, 2, or 3 fluoro substituents;
  • R 3 is H, methyl, chloro or fluoro
  • R 4 is H, methyl, chloro or fluoro
  • R 5 is -(CH 2 ) n -NR 8 R 9 ;
  • R 6 and R 7 together with the nitrogen atom to which they are attached form a 4, 5 or 6 membered non-aromatic monocyclic ring comprising carbon which is substituted with - NR 10 R U ;
  • R 8 and R 9 are independently selected from H and (Ci-C 3 )alkyl; or R 8 and R 9 together with the nitrogen atom to which they are attached form a 4, 5 or 6 membered non- aromatic monocyclic ring comprising carbon which can be optionally substituted with fluoro;
  • R 10 is H or (Ci-C )alkyl
  • R 11 is (Ci-C )alkyl; n is 2, 3 or 4;
  • alkyl is a linear or branched saturated hydrocarbon
  • cycloalkyl is a 3, 4, 5 or 6 membered aliphatic carbocyclic ring, which can be optionally substituted with 1 or 2 substituents selected from methyl, methoxy and -OH;
  • heterocycloalkyl is a 4, 5 or 6 membered non-aromatic monocyclic ring comprising carbon and a heteroatom selected from oxygen; wherein heterocycloalkyl can be optionally substituted with 1 or 2 substituents selected from methyl, methoxy and -OH.
  • A is N or CH
  • R 1 is (Ci-C3)alkyl, cycloalkyl, heterocycloalkyl;
  • R 2 is H or methyl, wherein methyl is optionally substituted with 1, 2, or 3 fluoro substituents;
  • R 3 is H, methyl or fluoro
  • R 4 is H, methyl or fluoro
  • R 8 and R 9 are independently selected from H and (Ci-C3)alkyl; or R 8 and R 9 together with the nitrogen atom to which they are attached form a 5 or 6 membered non- aromatic monocyclic ring comprising carbon which can be optionally substituted with fluoro;
  • alkyl is a linear or branched saturated hydrocarbon
  • cycloalkyl is a 3, 4, 5 or 6 membered aliphatic carbocyclic ring, which can be optionally substituted with 1 or 2 substituents selected from methyl, methoxy and -OH;
  • heterocycloalkyl is a 5 or 6 membered non-aromatic monocyclic ring comprising carbon and a heteroatom selected from oxygen; wherein heterocycloalkyl can be optionally substituted with 1 or 2 substituents selected from methyl, methoxy and -OH.
  • A is N or CH
  • R 1 is (Ci-C3)alkyl, cycloalkyl, heterocycloalkyl;
  • R 2 is H or methyl, wherein methyl is optionally substituted with 1, 2, or 3 fluoro substituents;
  • R 3 is H, methyl or fluoro
  • R 4 is H, methyl or fluoro
  • R 6 and R 7 together with the nitrogen atom to which they are attached form a 5 or 6 membered non-aromatic monocyclic ring comprising carbon which is substituted with -NR 10 R n ;
  • R 10 is H or (Ci-C 3 )alkyl
  • R 11 is (Ci-C 3 )alkyl
  • alkyl is a linear or branched saturated hydrocarbon
  • cycloalkyl is a 3, 4, 5 or 6 membered aliphatic carbocyclic ring, which can be optionally substituted with 1 or 2 substituents selected from methyl, methoxy and -OH;
  • heterocycloalkyl is a 5 or 6 membered non-aromatic monocyclic ring comprising carbon and a heteroatom selected from oxygen; wherein heterocycloalkyl can be optionally substituted with 1 or 2 substituents selected from methyl, methoxy and -OH.
  • moieties A, X, R ⁇ -R 11 , n and halo may be applied, alone or in combination, to the descriptions of the compounds of Formula (I), (la), (lb) or (Ic) provided above:
  • A is N or CR 4 .
  • A is N or CH.
  • A is N.
  • A is CH.
  • X is -OR 5 or -NR 6 R 7 .
  • X is -OR 5 .
  • X is -NR 6 R 7 .
  • X is selected from the following fragments:
  • X is selected from the following fragments:
  • X is selected from the following fragments:
  • R 1 is (Ci-C6)alkyl, cycloalkyl or heterocycloalkyl.
  • R 1 is (Ci-C3)alkyl, cycloalkyl or heterocycloalkyl.
  • R 1 is methyl, ethyl, n-propyl, iso-propyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, oxetanyl, tetrahydrofuranyl or tertrahydropyranyl; wherein cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, tetrahydrofuranyl or tertrahydropyranyl can be optionally substituted with 1 or 2 substituents selected from (Ci-C3)alkyl, (Ci-C3)alkoxy and -OH.
  • R 1 is n-propyl, iso-propyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, oxetanyl, tetrahydrofuranyl or tertrahydropyranyl; wherein cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, tetrahydrofuranyl or tertrahydropyranyl can be optionally substituted with 1 or 2 substituents selected from methyl, methoxy and -OH.
  • R 1 is iso-propyl, cyclopropyl, cyclobutyl, cyclopentyl, tertrahydropyran-3-yl or tertrahydropyran-4-yl; wherein cyclopropyl, cyclobutyl, cyclopentyl, tertrahydropyran-3-yl or tertrahydropyran-4-yl can be optionally substituted with 1 or 2 substituents selected from methyl and methoxy.
  • R 1 is iso-propyl, cyclopropyl, cyclobutyl or tertrahydropyranyl
  • cyclopropyl, cyclobutyl or tertrahydropyranyl can be optionally substituted with 1 or 2 substituents selected from methyl or methoxy.
  • R 1 is selected from the following fragments:
  • R 1 is selected from the following fragments:
  • R 1 is selected from the followin fragments:
  • R 1 is iso-propyl.
  • R 1 is cyclopropyl, optionally substituted with methyl.
  • R 1 is cyclobutyl, optionally substituted with methoxy.
  • R 1 is tertrahydropyranyl, optionally mono or di substituted with methyl.
  • R 2 is H or methyl, wherein methyl is optionally substituted with 1, 2, or 3 halo substituents.
  • R 2 is H or methyl, wherein methyl is optionally substituted with 1, 2, or 3 fluoro substituents.
  • R 2 is methyl optionally substituted with 1 , 2, or 3 fluoro substituents.
  • R 2 is methyl or CHF 2 .
  • R 2 is methyl
  • R 3 is H, (Ci-C 6 )alkyl or halo.
  • R 3 is H, (Ci-C3)alkyl, chloro or fluro.
  • R 3 is H, methyl, chloro or fluoro.
  • R 3 is H, methyl or fluoro.
  • R 3 is H or fluoro.
  • R 3 is fluoro
  • R 3 is H.
  • R 4 is H, (Ci-C 6 )alkyl or halo.
  • R 4 is H, (Ci-C 3 )alkyl, chloro or fluro.
  • R 4 is H, methyl, chloro or fluoro.
  • R 4 is H, methyl or fluoro.
  • R 4 is H or fluoro.
  • R 4 is H or methyl.
  • R 4 is H.
  • R 5 is -(CH 2 ) 3 -NR3 ⁇ 4 9 .
  • R 6 and R 7 together with the nitrogen atom to which they are attached form a 4, 5 or 6 membered non-aromatic monocyclic ring comprising carbon which is substituted with - NR 10 R n .
  • R 6 and R 7 together with the nitrogen atom to which they are attached form a 5 or 6 membered non-aromatic monocyclic ring comprising carbon which is substituted with - N(CH 3 ) 2 .
  • R 6 and R 7 together with the nitrogen atom to which they are attached form a fragment selected from:
  • ⁇ * designates the point of attachment of the fragment to the aromatic ring containing A.
  • R 6 and R 7 together with the nitrogen atom to which they are attached form a fragment selected from:
  • ⁇ * designates the point of attachment of the fragment to the aromatic ring containing A.
  • R 8 and R 9 are independently selected from H and (Ci-Ce)alkyl; or R 8 and R 9 together with the nitrogen atom to which they are attached form a 4, 5 or 6 membered non- aromatic monocyclic ring comprising carbon which can be optionally substituted with halo.
  • R 8 and R 9 are independently selected from H and (Ci-C3)alkyl; or R 8 and R 9 together with the nitrogen atom to which they are attached form a 5 or 6 membered non- aromatic monocyclic ring comprising carbon which can be optionally substituted with fluoro.
  • R 8 and R 9 are independently selected from H and methyl; or R 8 and R 9 together with the nitrogen atom to which they are attached form a 5 or 6 membered non-aromatic monocyclic ring comprising carbon which can be optionally substituted with fluoro.
  • R 8 and R 9 are methyl; or R 8 and R 9 together with the nitrogen atom to which they are attached form a 5 or 6 membered non-aromatic monocyclic ring comprising carbon which can be optionally substituted with fluoro.
  • R 8 and R 9 together with the nitrogen atom to which they are attached form a 6 membered non-aromatic monocyclic ring comprising carbon which can be optionally substituted with fluoro.
  • R 8 and R 9 together with the nitrogen atom to which they are attached form a 5 membered non-aromatic monocyclic ring comprising carbon which can be optionally substituted with fluoro.
  • R 8 and R 9 together with the nitrogen atom to which they are attached form a fragment selected from:
  • ** designates the point of attachment of the fragment to the remainder of the molecule.
  • R 8 and R 9 together with the nitrogen atom to which they are attached form a fragment selected from:
  • ** designates the point of attachment of the fragment to the remainder of the molecule.
  • R 8 and R 9 together with the nitrogen atom to which they are attached form a fragment selected from:
  • » ⁇ designates the point of attachment of the fragment to the remainder of the molecule.
  • R 8 is H and R 9 is methyl.
  • R 8 and R 9 are methyl.
  • R is H or (Ci-C 3 )alkyl.
  • R 10 is H or methyl.
  • R 10 is methyl
  • R 11 is (Ci-C 3 )alkyl.
  • R 11 is methyl. n is 2, 3 or 4.
  • n 2 or 3.
  • n 3.
  • halo is F, CI or Br.
  • halo is F or CI. halo is F.
  • the compound of Formula (I) is selected from:
  • the compound of Formula (I) is selected from:
  • O-linked such as in "O-linked saturated hydrocarbon" means that the hydrocarbon residue is joined to the remainder of the molecule via an oxygen atom.
  • a suitable pharmaceutically acceptable salt of a compound of Formula (I) is, for example, an acid-addition salt.
  • An acid addition salt of a compound of Formula (I) may be formed by bringing the compound into contact with a suitable inorganic or organic acid under conditions known to the skilled person.
  • An acid addition salt may for example be formed using an inorganic acid selected from the group consisting of hydrochloric acid, hydrobromic acid, sulphuric acid and phosphoric acid.
  • An acid addition salt may also be formed using an organic acid selected from the group consisting of trifluoro acetic acid, citric acid, maleic acid, oxalic acid, acetic acid, formic acid, benzoic acid, fumaric acid, succinic acid, tartaric acid, lactic acid, pyruvic acid, methanesulfonic acid, benzenesulfonic acid and /?ara-toluenesulfonic acid.
  • a compound of Formula (I) or a pharmaceutically acceptable salt thereof where the pharmaceutically acceptable salt is a hydrochloric acid, hydrobromic acid, sulphuric acid, phosphoric acid, trifluoroacetic acid, citric acid, maleic acid, oxalic acid, acetic acid, formic acid, benzoic acid, fumaric acid, succinic acid, tartaric acid, lactic acid, pyruvic acid, methanesulfonic acid, benzenesulfonic acid or /?ara-toluenesulfonic acid salt.
  • the pharmaceutically acceptable salt is a hydrochloric acid, hydrobromic acid, sulphuric acid, phosphoric acid, trifluoroacetic acid, citric acid, maleic acid, oxalic acid, acetic acid, formic acid, benzoic acid, fumaric acid, succinic acid, tartaric acid, lactic acid, pyruvic acid, methanesulfonic acid, benzene
  • a compound of Formula (I) or a pharmaceutically acceptable salt thereof where the pharmaceutically acceptable salt is a methanesulfonic acid salt.
  • a compound of Formula (I) or a pharmaceutically acceptable salt thereof where the pharmaceutically acceptable salt is a mono-methanesulfonic acid salt, i.e. the stoichiometry of the compound of the compound of Formula (I) to methanesulfonic acid is 1 : 1.
  • solvated forms may be a hydrated form, such as a hemi-hydrate, a mono-hydrate, a di-hydrate, a tri-hydrate or an alternative quantity thereof. All such solvated and unsolvated forms of compounds of Formula (I) are encompassed herein, particularly to the extent that such forms possess ATM kinase inhibitory activity, as for example measured using the tests described herein.
  • Atoms of the compounds and salts described in this specification may exist as their isotopes. All compounds of Formula (I) where an atom is replaced by one or more of its isotopes (for example a compound of Formula (I) where one or more carbon atom is an U C or 13 C carbon isotope, or where one or more hydrogen atoms is a 2 H or 3 H isotope) are encompassed herein.
  • Compounds of the application may exist in one or more geometrical, optical, enantiomeric, diastereomeric and tautomeric forms, including, but not limited to, cis- and trans-forms, E- and Z-forms, R-, S- and meso-forms, keto-, and enol-forms. Unless otherwise stated a reference to a particular compound includes all such isomeric forms, including racemic and other mixtures thereof. Where appropriate such isomers can be separated from their mixtures by the application or adaptation of known methods (e.g. chromatographic techniques and recrystallisation techniques). Where appropriate such isomers can be prepared by the application or adaptation of known methods (e.g.
  • fragment includes the cis and trans forms thereof.
  • pharmaceutically acceptable salt thereof which is a single optical isomer being in an enantiomeric excess (%ee) of > 95%, > 98% or > 99%.
  • the single optical isomer is present in an enantiomeric excess (%ee) of > 99%.
  • Compounds and salts described in this specification may be crystalline, and may exhibit one or more crystalline forms. Any crystalline or amorphous form of a compound of Formula (I), or mixture of such forms, which possesses ATM kinase inhibitory activity are encompassed herein.
  • crystalline materials may be characterised using conventional techniques such as X-Ray Powder Diffraction (XRPD), Differential Scanning Calorimetry (DSC), Thermal Gravimetric Analysis (TGA), Diffuse Reflectance Infrared Fourier Transform (DRIFT) spectroscopy, Near Infrared (NIR) spectroscopy, solution and/or solid state nuclear magnetic resonance spectroscopy.
  • XRPD X-Ray Powder Diffraction
  • DSC Differential Scanning Calorimetry
  • TGA Thermal Gravimetric Analysis
  • DRIFT Diffuse Reflectance Infrared Fourier Transform
  • NIR Near Infrared
  • solution and/or solid state nuclear magnetic resonance spectroscopy solution and/or solid state nuclear magnetic resonance spectroscopy.
  • the water content of crystalline materials may be determined by Karl Fischer analysis.
  • the compounds of Formula (I), and pharmaceutically acceptable salts thereof are expected to be useful in therapy, for example in the treatment of diseases or medical conditions mediated at least in part by ATM kinase, including cancer.
  • cancer includes both non-metastatic cancer and also metastatic cancer, such that treating cancer involves treatment of both primary tumours and also tumour metastases.
  • ATM kinase inhibitory activity refers to a decrease in the activity of ATM kinase as a direct or indirect response to the presence of a compound of Formula (I), or pharmaceutically acceptable salt thereof, relative to the activity of ATM kinase in the absence of compound of Formula (I), or pharmaceutically acceptable salt thereof.
  • Such a decrease in activity may be due to the direct interaction of the compound of Formula (I), or pharmaceutically acceptable salt thereof with ATM kinase, or due to the interaction of the compound of Formula (I), or pharmaceutically acceptable salt thereof with one or more other factors that in turn affect ATM kinase activity.
  • the compound of Formula (I), or pharmaceutically acceptable salt thereof may decrease ATM kinase by directly binding to the ATM kinase, by causing (directly or indirectly) another factor to decrease ATM kinase activity, or by (directly or indirectly) decreasing the amount of ATM kinase present in the cell or organism.
  • the term “therapy” is intended to have its normal meaning of dealing with a disease in order to entirely or partially relieve one, some or all of its symptoms, or to correct or compensate for the underlying pathology.
  • the term “therapy” also includes “prophylaxis” unless there are specific indications to the contrary.
  • the terms “therapeutic” and “therapeutically” should be interpreted in a corresponding manner.
  • prophylaxis is intended to have its normal meaning and includes primary prophylaxis to prevent the development of the disease and secondary prophylaxis whereby the disease has already developed and the patient is temporarily or permanently protected against exacerbation or worsening of the disease or the development of new symptoms associated with the disease.
  • treatment is used synonymously with “therapy”.
  • treat can be regarded as “applying therapy” where “therapy” is as defined herein.
  • said disease mediated by ATM kinase is cancer.
  • said cancer is selected from the group consisting of colorectal cancer, glioblastoma, gastric cancer, ovarian cancer, diffuse large B-cell lymphoma, chronic lymphocytic leukaemia, acute myeloid leukaemia, head and neck squamous cell carcinoma, breast cancer, prostate cancer, hepatocellular carcinoma, small cell lung cancer and non-small cell lung cancer.
  • said cancer is selected from the group consisting of colorectal cancer, glioblastoma, gastric cancer, ovarian cancer, diffuse large B-cell lymphoma, chronic lymphocytic leukaemia, head and neck squamous cell carcinoma, breast cancer, prostate cancer and lung cancer.
  • said cancer is colorectal cancer.
  • said cancer is gastric cancer.
  • a “neuroprotective agent” is an agent that aids relative preservation of neuronal structure and/or function.
  • said disease mediated by ATM kinase is cancer.
  • a method for treating a disease in which inhibition of ATM kinase is beneficial in a warm-blooded animal in need of such treatment which comprises administering to said warm-blooded animal a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof.
  • said disease is cancer.
  • a disease in which inhibition of ATM kinase is beneficial may be Huntington' disease.
  • a method of treatment for aiding relative preservation of neuronal structure and/or function in a warm-blooded animal in need of such treatment which comprises administering to said warm-blooded animal a
  • therapeutically effective amount refers to an amount of a compound of Formula (I) as described in any of the embodiments herein which is effective to provide "therapy” in a subject, or to “treat” a disease or disorder in a subject.
  • the therapeutically effective amount may cause any of the changes observable or measurable in a subject as described in the definition of "therapy", “treatment” and “prophylaxis” above.
  • the effective amount can reduce the number of cancer or tumour cells; reduce the overall tumour size; inhibit or stop tumour cell infiltration into peripheral organs including, for example, the soft tissue and bone; inhibit and stop tumour metastasis; inhibit and stop tumour growth; relieve to some extent one or more of the symptoms associated with the cancer; reduce morbidity and mortality; improve quality of life; or a combination of such effects.
  • An effective amount may be an amount sufficient to decrease the symptoms of a disease responsive to inhibition of ATM kinase activity.
  • efficacy in-vivo can, for example, be measured by assessing the duration of survival, time to disease progression (TTP), the response rates (RR), duration of response, and/or quality of life.
  • effective amounts may vary depending on route of administration, excipient usage, and co-usage with other agents.
  • the amount of the compound of formula (I) or pharmaceutcially acceptable salt described in this specification and the amount of the other pharmaceutically active agent(s) are, when combined, jointly effective to treat a targeted disorder in the animal patient.
  • the combined amounts are in a "therapeutically effective amount" if they are, when combined, sufficient to decrease the symptoms of a disease responsive to inhibition of ATM activity as described above.
  • such amounts may be determined by one skilled in the art by, for example, starting with the dosage range described in this specification for the compound of formula (I) or pharmaceutcially acceptable salt thereof and an approved or otherwise published dosage range(s) of the other pharmaceutically active compound(s).
  • Warm-blooded animals include, for example, humans.
  • warm-blooded animal in need of such treatment which comprises administering to said warm-blooded animal a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof.
  • said cancer may be selected from the group consisting of colorectal cancer, glioblastoma, gastric cancer, ovarian cancer, diffuse large B-cell lymphoma, chronic lymphocytic leukaemia, acute myeloid leukaemia, head and neck squamous cell carcinoma, breast cancer, prostate cancer, hepatocellular carcinoma, small cell lung cancer and non-small cell lung cancer.
  • Said cancer may also be selected from the group consisting of colorectal cancer, glioblastoma, gastric cancer, ovarian cancer, diffuse large B-cell lymphoma, chronic lymphocytic leukaemia, head and neck squamous cell carcinoma, breast cancer, prosate cancer and lung cancer.
  • the cancer is colorectal cancer or gastric cancer.
  • the cancer is colorectal cancer.
  • the cancer is glioblastoma.
  • the cancer is gastric cancer.
  • the cancer is oesophageal cancer.
  • the cancer is ovarian cancer.
  • the cancer is endometrial cancer.
  • the cancer is cervical cancer.
  • the cancer is diffuse large B-cell lymphoma.
  • the cancer is chronic lymphocytic leukaemia.
  • the cancer is acute myeloid leukaemia.
  • the cancer is head and neck squamous cell carcinoma.
  • the cancer is prostate cancer.
  • the cancer is breast cancer. In one embodiment the cancer is triple negative breast cancer.
  • Triple negative breast cancer is any breast cancer that does not express the genes for the oestrogen receptor, progesterone receptor and Her2/neu.
  • the cancer is hepatocellular carcinoma.
  • the cancer is lung cancer. In one embodiment the lung cancer is small cell lung cancer. In one embodiment the lung cancer is non-small cell lung cancer.
  • the cancer is metastatic cancer.
  • the metastatic cancer comprises metastases of the central nervous system.
  • the metastases of the central nervous system comprise brain metastases.
  • metastases of the central nervous system comprise leptomeningeal metastases.
  • “Leptomeningeal metastases” occur when cancer spreads to the meninges, the layers of tissue that cover the brain and the spinal cord. Metastases can spread to the meninges through the blood or they can travel from brain metastases, carried by the cerebrospinal fluid (CSF) that flows through the meninges.
  • CSF cerebrospinal fluid
  • the cancer is non-metastatic cancer.
  • the anti-cancer treatment described in this specification may be useful as a sole therapy, or may involve, in addition to administration of the compound of Formula (I), conventional surgery, radiotherapy or chemotherapy; or a combination of such additional therapies.
  • Such conventional surgery, radiotherapy or chemotherapy may be administered simultaneously, sequentially or separately to treatment with the compound of Formula (I).
  • Radiotherapy may include one or more of the following categories of therapy: i. External radiation therapy using electromagnetic radiation, and intraoperative radiation therapy using electromagnetic radiation;
  • iii Systemic radiation therapy, including but not limited to iodine 131 and strontium 89.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof, and radiotherapy for use in the treatment of cancer.
  • the cancer is glioblastoma.
  • the cancer is metastatic cancer.
  • the metastatic cancer comprises metastases of the central nervous system.
  • the metastases of the central nervous system comprise brain metastases.
  • the metastases of the central nervous system comprise leptomeningeal metastases.
  • the cancer is glioblastoma.
  • the cancer is metastatic cancer.
  • the metastatic cancer comprises metastases of the central nervous system.
  • the metastases of the central nervous system comprise brain metastases.
  • the metastases of the central nervous system comprise leptomeningeal metastases.
  • a method of treating cancer in a warmblooded animal who is in need of such treatment which comprises administering to said warm-blooded animal a compound of Formula (I), or a pharmaceutically acceptable salt thereof and radiotherapy.
  • the compound of Formula (I), or a pharmaceutically acceptable salt thereof, and radiotherapy are jointly effective in producing an anti-cancer effect.
  • a method of treating cancer in a warmblooded animal who is in need of such treatment which comprises administering to said warm-blooded animal a compound of Formula (I), or a pharmaceutically acceptable salt thereof and simultaneously, separately or sequentially administering radiotherapy.
  • the compound of Formula (I), or a pharmaceutically acceptable salt thereof, and radiotherapy are jointly effective in producing an anti-cancer effect.
  • the cancer is glioblastoma. In one embodiment, the cancer is metastatic cancer. In one embodiment the metastatic cancer comprises metastases of the central nervous system. In one embodiment the metastases of the central nervous system comprise brain metastases. In one embodiment the metastases of the central nervous system comprise leptomeningeal metastases.
  • said cancer may be selected from the group consisting of glioblastoma, lung cancer (for example small cell lung cancer or non-small cell lung cancer), breast cancer (for example triple negative breast cancer), head and neck squamous cell carcinoma, oesophageal cancer, cervical cancer and endometrial cancer.
  • the cancer is glioblastoma.
  • the cancer is metastatic cancer.
  • the metastatic cancer comprises metastases of the central nervous system.
  • the metastases of the central nervous system comprise brain metastases.
  • metastases of the central nervous system comprise leptomeningeal metastases.
  • the radiotherapy is selected from the group consisting of one or more of the categories of radiotherapy listed under points (i) - (iii) above.
  • Chemotherapy may include one or more of the following categories of anti -tumour substance:
  • Antineoplastic agents and combinations thereof such as DNA alkylating agents (for example cisplatin, oxaliplatin, carboplatin, cyclophosphamide, nitrogen mustards like ifosfamide, bendamustine, melphalan, chlorambucil, busulphan, temozolamide and nitrosoureas like carmustine); antimetabolites (for example gemcitabine and antifolates such as fluoropyrimidines like 5-fluorouracil and tegafur, raltitrexed, methotrexate, cytosine arabinoside, and hydroxyurea); anti-tumour antibiotics (for example anthracyclines like adriamycin, bleomycin, doxorubicin, liposomal doxorubicin, pirarubicin, daunomycin, valrubicin, epirubicin, idarubicin, mitomycin-C, dactinomycin
  • Antiangio genie agents such as those that inhibit the effects of vascular endothelial growth factor, for example the anti-vascular endothelial cell growth factor antibody bevacizumab and for example, a VEGF receptor tyrosine kinase inhibitor such as vandetanib (ZD6474), sorafenib, vatalanib (PTK787), sunitinib (SU11248), axitinib (AG-013736), pazopanib (GW 786034) and cediranib (AZD2171); compounds such as those disclosed in International Patent Applications W097/22596, WO 97/30035, WO 97/32856 and WO 98/13354; and compounds that work by other mechanisms (for example linomide, inhibitors of integrin ⁇ 3 function and angiostatin), or inhibitors of angiopoietins and their receptors (Tie-1 and Tie-2), inhibitors of PLGF, inhibitors of delta
  • Immunotherapy approaches including for example ex-vivo and in-vivo approaches to increase the immunogenicity of patient tumour cells, such as transfection with cytokines such as interleukin 2, interleukin 4 or granulocyte -macrophage colony stimulating factor; approaches to decrease T-cell anergy or regulatory T-cell function; approaches that enhance T-cell responses to tumours, such as blocking antibodies to CTLA4 (for example ipilimumab and tremelimumab), B7H1, PD-1 (for example BMS-936558 or AMP-514), PD-L1 (for example MEDI4736) and agonist antibodies to CD 137; approaches using transfected immune cells such as cytokine -transfected dendritic cells; approaches using cytokine-transfected tumour cell lines, approaches using antibodies to tumour associated antigens, and antibodies that deplete target cell types (e.g., unconjugated anti-CD20 antibodies such as Rituximab, radiolabeled anti-CD20
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer where the compound of Formula (I), or a pharmaceutically acceptable salt thereof is administered in combination with an additional anti-tumour substance.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer where the compound of Formula (I), or a
  • pharmaceutically acceptable salt thereof is administered simultaneously, separately or sequentially with an additional anti-tumour substance.
  • a method of treating cancer in a warmblooded animal who is in need of such treatment which comprises administering to said warm-blooded animal a compound of Formula (I), or a pharmaceutically acceptable salt thereof and at least one additional anti-tumour substance, where the amounts of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, and the additional anti -tumour substance are jointly effective in producing an anti-cancer effect.
  • a method of treating cancer in a warmblooded animal who is in need of such treatment which comprises administering to said warm-blooded animal a compound of Formula (I), or a pharmaceutically acceptable salt thereof, and simultaneously, separately or sequentially administering at least one additional anti -tumour substance to said warm-blooded animal, where the amounts of the compound of Formula (I), or pharmaceutically acceptable salt thereof, and the additional anti-tumour substance are jointly effective in producing an anti-cancer effect.
  • the additional anti-tumour substance is selected from the group consisting of one or more of the anti-tumour substances listed under points (i) - (iv) above.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer where the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered in combination with at least one anti-neoplastic agent.
  • the anti-neoplastic agent is selected from the list of antineoplastic agents in point (i) above.
  • antineoplastic agent for use in the treatment of cancer, where the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered simultaneously, separately or sequentially with at least one anti-neoplastic agent.
  • the antineoplastic agent is selected from the list of antineoplastic agents in point (i) above.
  • cisplatin oxaliplatin, carboplatin, valrubicin, idarubicin, doxorubicin, pirarubicin, irinotecan, topotecan, amrubicin, epirubicin, etoposide, mitomycin, bendamustine, chlorambucil, cyclophosphamide, ifosfamide, carmustine, melphalan, bleomycin, olaparib, MEDI4736, AZD1775 and AZD6738, for use in the treatment of cancer.
  • additional anti -tumour substance selected from the group consisting of cisplatin, oxaliplatin, carboplatin, valrubicin, idarubicin, doxorubicin, pirarubicin, irinotecan, topotecan, amrubicin, epirubicin, etoposide, mitomycin, bendamustine, chlorambucil,
  • anti -tumour substance selected from the group consisting of cisplatin, oxaliplatin, carboplatin, doxorubicin, pirarubicin, irinotecan, topotecan, amrubicin, epirubicin, etoposide, mitomycin, bendamustine, chlorambucil, cyclophosphamide, ifosfamide, carmustine, melphalan, bleomycin, olaparib, AZD1775 and AZD6738, for use in the treatment of cancer.
  • additional anti -tumour substance selected from the group consisting of cisplatin, oxaliplatin, carboplatin, doxorubicin, pirarubicin, irinotecan, topotecan, amrubicin, epirubicin, etoposide, mitomycin, bendamustine, chlorambucil, cyclophosphamide, ifosfamide, car
  • the compound of Formula (I), or a pharmaceutically acceptable salt thereof is administered in combination with at least one additional anti-tumour substance selected from the group consisting of cisplatin, oxaliplatin, carboplatin, valrubicin, idarubicin, doxorubicin, pirarubicin, irinotecan, topotecan, amrubicin, epirubicin, etoposide, mitomycin, bendamustine, chlorambucil, cyclophosphamide, ifosfamide, carmustine, melphalan, bleomycin, olaparib, MEDI4736, AZD1775 and AZD6738.
  • additional anti-tumour substance selected from the group consisting of cisplatin, oxaliplatin, carboplatin, valrubicin, idarubicin, doxorubicin, pirarubicin, irinotecan, topotecan, amrub
  • doxorubicin doxorubicin
  • irinotecan topotecan
  • etoposide mitomycin
  • mitomycin bendamustine
  • chlorambucil cyclophosphamide
  • ifosfamide carmustine
  • melphalan bleomycin
  • olaparib for use in the treatment of cancer.
  • the compound of Formula (I), or a pharmaceutically acceptable salt thereof for use in the treatment of cancer, where the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered in combination with at least one additional anti-tumour substance selected from the group consisting of doxorubicin, irinotecan, topotecan, etoposide, mitomycin, bendamustine, chlorambucil, cyclophosphamide, ifosfamide, carmustine, melphalan, bleomycin and olaparib.
  • at least one additional anti-tumour substance selected from the group consisting of doxorubicin, irinotecan, topotecan, etoposide, mitomycin, bendamustine, chlorambucil, cyclophosphamide, ifosfamide, carmustine, melphalan, bleomycin and olaparib.
  • doxorubicin doxorubicin
  • irinotecan topotecan
  • etoposide mitomycin
  • mitomycin bendamustine
  • chlorambucil cyclophosphamide
  • ifosfamide carmustine
  • melphalan bleomycin
  • pharmaceutically acceptable salt thereof for use in the treatment of cancer, where the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered in combination with at least one additional anti-tumour substance selected from the group consisting of doxorubicin, irinotecan, topotecan, etoposide, mitomycin, bendamustine, chlorambucil, cyclophosphamide, ifosfamide, carmustine, melphalan and bleomycin.
  • additional anti-tumour substance selected from the group consisting of doxorubicin, irinotecan, topotecan, etoposide, mitomycin, bendamustine, chlorambucil, cyclophosphamide, ifosfamide, carmustine, melphalan and bleomycin.
  • the cancer is acute myeloid leukaemia.
  • the cancer is breast cancer (for example triple negative breast cancer).
  • the cancer is hepatocellular carcinoma.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer where the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered in combination with irinotecan.
  • the cancer is colorectal cancer.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer where the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered in combination with FOLFIRI.
  • the cancer is colorectal cancer.
  • FOLFIRI is a dosage regime involving a combination of leucovorin, 5-fluorouracil and irinotecan.
  • the cancer is selected from gastric cancer, triple negative breast cancer, prostate cancer, small cell lung cancer and ovarian cancer.
  • the cancer is gastric cancer.
  • the cancer is triple negative breast cancer.
  • the cancer is prostate cancer.
  • the cancer is small cell lung cancer.
  • the cancer is ovarian cancer.
  • the cancer is small cell lung cancer.
  • a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, where the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered in combination with immunotherapy is one or more of the agents listed under point (iii) above.
  • the immunotherapy is an anti-PD-Ll antibody (for example MEDI4736).
  • Container means for containing said first and further unit dosage forms; and optionally
  • the anti-tumour substance comprises an anti-neoplastic agent.
  • the anti-neoplastic agent is one or more of the agents listed under point (i) above.
  • the compounds of Formula (I), and pharmaceutically acceptable salts thereof, may be administered as pharmaceutical compositions, comprising one or more pharmaceutically acceptable excipients.
  • a pharmaceutical composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient.
  • excipient(s) selected for inclusion in a particular composition will depend on factors such as the mode of administration and the form of the composition provided. Suitable pharmaceutically acceptable excipients are well known to persons skilled in the art and are described, for example, in the Handbook of Pharmaceutical Excipients, Sixth edition, Pharmaceutical Press, edited by Rowe, Ray C; Sheskey, Paul J; Quinn, Marian. Pharmaceutically acceptable excipients may function as, for example, adjuvants, diluents, carriers, stabilisers, flavourings, colorants, fillers, binders, disintegrants, lubricants, glidants, thickening agents and coating agents. As persons skilled in the art will appreciate, certain pharmaceutically acceptable excipients may serve more than one function and may serve alternative functions depending on how much of the excipient is present in the composition and what other excipients are present in the composition.
  • compositions may be in a form suitable for oral use (for example as tablets, lozenges, hard or soft capsules, aqueous or oily suspensions, emulsions, dispersible powders or granules, syrups or elixirs), for topical use (for example as creams, ointments, gels, or aqueous or oily solutions or suspensions), for administration by inhalation (for example as a finely divided powder or a liquid aerosol), for administration by insufflation (for example as a finely divided powder) or for parenteral administration (for example as a sterile aqueous or oily solution for intravenous, subcutaneous, intramuscular or intramuscular dosing), or as a suppository for rectal dosing.
  • the compositions may be obtained by conventional procedures well known in the art.
  • compositions intended for oral use may contain additional components, for example, one or more colouring, sweetening, flavouring and/or preservative agents.
  • the compound of Formula (I) will normally be administered to a warm-blooded animal at a unit dose within the range 2.5-5000 mg/m 2 body area of the animal, or approximately 0.05-100 mg/kg, and this normally provides a therapeutically-effective dose.
  • a unit dose form such as a tablet or capsule will usually contain, for example 0.1-250 mg of active ingredient.
  • the daily dose will necessarily be varied depending upon the host treated, the particular route of administration, any therapies being co-administered, and the severity of the illness being treated. Accordingly the practitioner who is treating any particular patient may determine the optimum dosage.
  • compositions described herein comprise compounds of Formula (I), or a pharmaceutically acceptable salt thereof, and are therefore expected to be useful in therapy.
  • a pharmaceutical composition for use in therapy comprising a compound of Formula (I), or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient.
  • a pharmaceutical composition for use in the treatment of a disease in which inhibition of ATM kinase is beneficial comprising a compound of Formula (I), or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient.
  • a pharmaceutical composition for use in the treatment of cancer comprising a compound of Formula (I), or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient.
  • a pharmaceutical composition for use in the treatment of a cancer in which inhibition of ATM kinase is beneficial comprising a compound of Formula (I), or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient.
  • Examples of such systems include Armen Glider Flash: Spot II Ultimate (Armen Instrument, Saint-Ave, France) or automated Presearch combiflash companions using prepacked Merck normal phase Si60 silica cartridges (granulometry : 15-40 or 40-63 ⁇ ) obtained from Merck, Darmstad, Germany, silicycle silica cartridges or graceresolv silica cartridges;
  • Preparative chromatography was performed on a suitable system.
  • suitable systems include a Waters instrument (600/2700 or 2525) fitted with a ZMD or ZQ ESCi mass spectrometers and a Waters X-Terra or a Waters X-Bridge or a Waters SunFire reverse-phase column (C-18, 5 microns silica, 19 mm or 50 mm diameter, 100 mm length, flow rate of 40 mL / minute) using decreasingly polar mixtures of water (containing 1% ammonia) and acetonitrile or decreasingly polar mixtures of water (containing 0.1% formic acid) and acetonitrile as eluents;
  • LCMS mass spectroscopy following liquid chromatography
  • Methyltertbutylether anhydrous magnesium sulphate
  • Na 2 S0 4 anhydrous sodium sulphate
  • THF tetrahydrofuran
  • sat. saturated aqueous solution
  • SFC supercritical fluid chromatography
  • Example 1 8- [6- [3-(Dimethylamino)propoxy] -3-pyridyl] -3-methyl-l- tetrahydropyran-4-yl-imidazo [4,5-c] cinnolin-2-one
  • Example 2 Isolated as a formic acid salt NMR Spectrum: l H NMR (300MHz, DMSO) ⁇ 1.8-2.0 (2H, m), 2.1-2.2 (6H, m), 2.35-2.45 (2H, m), 2.7-3.1 (4H, m), 3.2 (3H, s), 3.6 (3H, s), 3.8-4.0 (1H, m), 4.3-4.4 (2H,m), 5.0-5.2 (1H, m), 6.9-7.1 (1H, m), 8.0-8.8 (5H, m).
  • Tetrahydro-2H-pyran-4-amine (0.615 mL, 5.94 mmol) was added dropwise to ethyl 6- bromo-4-chloro-3-cinnolinecarboxylate (1.5 g, 4.75 mmol) and triethylamine (1.990 mL, 14.26 mmol) in THF (5 mL) and the reaction stirred at ambient temperature for 30 minutes.
  • Ether (15 mL) was added and the solid was collected by filtration, washed with water (25 mL) and ether (20 mL) and dried under vacuum to afford the desired material (1.78 g) as a beige solid, which was used without further purification.
  • Butyllithium (2.5N, 4.8 mL, 50.96 mmol) was added to a solution of 3-(5-bromopyridin-2- yl)oxy-N,N-dimethylpropan-l -amine (2.07 g, 7.99 mmol) and 4,4,5, 5-tetramethyl-2-
  • reaction mixture was extracted with EtOAc (50 mL), and the organic layer washed twice with saturated brine (20 mL) and evaporated to afford crude product.
  • the crude product was purified by flash silica chromatography, elution gradient 0 to 10% 1% NH 3 in MeOH in DCM, to afford the desired material (0.04 g) as a yellow solid.
  • Dichlorobis(triphenylphosphine)palladium(II) (0.013 g, 0.02 mmol) was added to degassed solution of 8-bromo-l-isopropyl-3-methyl-imidazo[4,5-c]cinnolin-2-one 0.36 mmol) and N,N-dimethyl-3-(4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)phenoxy)propan-l -amine (0.116 g, 0.38 mmol), potassium carbonate (0.542 mL, 1.08 mmol) in 1,4-dioxane (8 mL) and heated in the microwave to 80 °C for 1 hour under nitrogen.
  • the reaction mixture was diluted with EtOAc (25 mL), and washed sequentially with water (10 mL), saturated brine (10 mL) and the organic layer was evaporated.
  • the crude product was purified by flash silica chromatography, elution gradient 0 to 10% (1% NH 3 in MeOH) in DCM. Pure fractions were evaporated to dryness and triturated with ether to afford the desired product (0.055 g, 36 %) as a pale brown solid.
  • N,N-Dimethyl-3-(4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenoxy)propan-l -amine was prepared as described below.
  • the resulting suspension was stirred at 90 °C for 16 hours under an inert atmosphere.
  • the reaction mixture was evaporated to dryness, redissolved in DCM (25 mL), washed with water (20 mL) and the organic layer was dried with a phase separating cartridge, filtered and evaporated.
  • the crude product was purified by flash silica chromatography, elution gradient 0 to 10% MeOH in DCM, to afford the desired material as a brown oil (1.000 g, 42.3 %) which solidified on standing.
  • reaction mixture was diluted with EtOAc (25 mL), and washed sequentially with water (10 mL), saturated brine (10 mL) and the organic layer evaporated to afford crude product as a brown solid residue.
  • the crude product was purified by preparative HPLC to afford the desired material as a dry yellow film. Diethyl ether was added and the mixture evaporated to dryness to afford the desired material (80 mg) as a brown solid.
  • Example 10 can also be isolated as the methane sulfonic acid salt by taking the material as prepared above and subjecting to the following reaction conditions. l-Isopropyl-3-methyl-8-[4-[3-(l-piperidyl)propoxy]phenyl]imidazo[4,5-c]cinnolin-2-one (89 mg, 0.19 mmol) was dissolved in DCM (2 mL) and treated with 1M methanesulfonic acid (0.013 mL, 0.19 mmol) in DCM then the mixture evaporated to dryness. The crude product was purified by ion exchange chromatography, using an SCX column.
  • Propan-2-amine (1.775 mL, 20.66 mmol) was added to 4,6-dichloro-3- cinnolinecarboxamide (5 g, 20.66 mmol) and DIPEA (7.22 mL, 41.31 mmol) in acetonitrile (94 mL) and the resulting suspension stirred at 60 °C for 18 hours. The mixture was allowed to cool and diluted with water. The precipitate was collected by filtration, washed with water (20 mL) followed by acetonitrile (10 mL) and dried under vacuum to afford the desired material (4.06 g) as a brown solid, which was used without further purification.
  • 6-Chloro-4-hydroxy-cinnoline-3-carboxylic acid is available from commercial sources, for example from IChem LP, 5 walnut Hill Park, Suite 13, Woburn, MA, 01801, US; CAS Registry Number: 90272-08-5, or from Vijaya Pharmaceuticals, LLC (V-Pharma), 104 T. W. Alexander Drive, Building 5, PO Box 14547, Research Triangle Park, NC, 27709, US. The preparation is also described in the literature: Schofield, K.; Swain, T., Journal of the Chemical Society, (1949), 2393-9.
  • Butyllithium (139 mL, 347.59 mmol) was added dropwise to 5-bromo-2-(3-(piperidin-l- yl)propoxy)pyridine (80 g, 267.37 mmol) and 2-isopropoxy-4,4,5,5-tetramethyl-l,3,2- dioxaborolane (64.7 g, 347.59 mmol) in THF (400 mL) cooled to -78°C over a period of 10 minutes under an inert atmosphere. The resulting mixture was allowed to warm to ambient temperature and stirred for 12 hours.
  • Potassium acetate (1.036 g, 10.56 mmol) was added to l-(3-(4- bromophenoxy)propyl)pyrrolidine (1 g, 3.52 mmol), 4,4,4',4',5,5,5',5'-octamethyl-2,2'- bi(l,3,2-dioxaborolane) (1.072 g, 4.22 mmol) and [ ⁇ , - bis(diphenylphosphino)ferrocene]dichloropalladium(II) (0.129 g, 0.18 mmol) in 1,4- dioxane (1 mL) at 25°C under nitrogen. The resulting mixture was stirred at 100 °C for 3 hours.
  • the resulting suspension was stirred at 90 °C for 22 hours under an inert atmosphere.
  • the reaction mixture was evaporated to dryness, redissolved in DCM (50 mL), washed with water (40 mL) and the organic layer was dried with a phase separating cartridge, filtered and evaporated.
  • the crude product was purified by flash silica chromatography, elution gradient 0 to 10% MeOH in DCM, to afford the desired material as a brown oil which solidified on standing.
  • Example 12 8-[6-[3-(4-Fluoro-l-piperidyl)propoxy]-3-pyridyl]-l-isopropyl-3-methyl- imidazo 4,5-c] cinnolin-2-one
  • the reaction mixture was diluted with ethyl acetate (40 mL), washed three times with water (3 x 20 mL) and the organic layer was dried over MgS0 4 , filtered and evaporated.
  • the crude product was purified by preparative HPLC to afford the desired material (25 mg) as a yellow solid.
  • Dichlorobis(triphenylphosphine)palladium (II) (31.7 mg, 0.05 mmol) was added to a degassed solution of (6-fluoropyridin-3-yl)boronic acid (134 mg, 0.95 mmol), 8-chloro-l- isopropyl-3-methyl-imidazo[4,5-c]cinnolin-2-one (250 mg, 0.90 mmol) and 2M potassium carbonate solution (1.355 mL, 2.71 mmol) in 1,4-dioxane (3.16 mL) and the mixture heated in a microwave reactor at 80 °C for 1 hour under an inert atmosphere.
  • reaction mixture was diluted with EtOAc (25 mL), and washed sequentially with water (10 mL), saturated brine (10 mL) then the organic layer evaporated to afford the desired material as a brown solid which was used in the subsequent steps without further purification.
  • Example 13 8- [6- [3-[(3R)-3-Fluoropyrrolidin-l-yl] propoxy]-3-pyridyl] -l-isopropyl-3- methyl-imidazo [4,5-c] cinnolin-2-one
  • Dichlorobis(triphenylphosphine)palladium(II) (22.19 mg, 0.03 mmol) was added to a degassed mixture of 8-chloro-l-isopropyl-3-methyl-imidazo[4,5-c]cinnolin-2-one (175 mg, 0.63 mmol), (6-fluoro-2-methyl-3-pyridinyl)boronic acid (108 mg, 0.70 mmol) and 2M aqueous potassium carbonate solution (0.949 mL, 1.90 mmol) in 1,4-dioxane (5.37 mL) and heated to 90 °C for 1 hour in a microwave reactor under an inert atmosphere.
  • reaction mixture was diluted with 10% methanol in DCM (40 mL) and water (15 mL).
  • the aqueous phase was extracted with 10% methanol in DCM (2 x 20 mL), then the combined organic phases dried over a phase separating cartridge and concentrated to afford the desired product, which was used immediately without purification.
  • the reaction mixture was diluted with EtOAc (25 mL), washed sequentially with water (10 mL), saturated brine (10 mL) and the organic layer was evaporated.
  • the crude product was purified by preparative HPLC to afford the desired product (46.2 mg, 29 %) as a brown dry film.
  • Example 17 NMR Spectrum: l H NMR (400 MHz, DMSO) ⁇ 1.62 - 1.78 (2H, m), 1.78 - 1.89 (2H, m), 1.89 - 1.96 (2H, m), 2.29 (IH, d), 2.44 (2H, d), 2.53 - 2.57 (IH, m), 2.63 (4H, td), 3.13 - 3.21 (2H, m), 3.23 (3H, s), 3.61 (3H, s), 4.29 (IH, ddd), 4.38 (2H, t), 4.67 (IH, ddt), 5.58 (IH, ddd), 6.95 - 7.07 (IH, m), 8.06 (IH, dd), 8.2 - 8.31 (2H, m), 8.39 (IH, d), 8.73 (IH, d).
  • Example 21 NMR Spectrum: ! H NMR (400 MHz, DMSO) ⁇ 1.88 (4H, td), 2.17 (6H, s), 2.22 (IH, s), 2.39 (2H, t), 2.54 - 2.66 (IH, m), 3.38 - 3.46 (IH, m), 3.58 (3H, s), 3.94 (IH, d), 4.10 (2H, t), 4.12 - 4.24 (2H, m), 4.90 (IH, tt), 7.1 1 - 7.17 (2H, m), 7.78 - 7.84 (2H, m), 8.05 (IH, dd), 8.23 (OH, s), 8.26 (IH, d), 8.38 (IH, d .
  • Mass Spectrum: m/z (ES+) [M+H]+ 462
  • Example 27 can also be isolated as the methane sulfonic acid salt by taking the material as prepared above and subjecting to the following reaction conditions.
  • Example 29 NMR Spectrum: ! H NMR (400 MHz, DMSO) ⁇ 1.39 (2H, q), 1.50 (4H, p), 1.79 - 1.94 (3H, m), 2.14 - 2.29 (3H, m), 2.33 (4H, d), 2.37 - 2.43 (3H, m), 2.45 - 2.48 (IH, m), 3.28 (3H, s), 3.59 (3H, s), 4.04 - 4.16 (3H, m), 5.47 (IH, p), 7.03 - 7.19 (2H, m), 7.73 - 7.88 (2H, m), 8.03 (IH, dd), 8.22 (IH, d), 8.36 (IH, d . Mass Spectrum: m/z (ES+)
  • Example 30 NMR Spectrum: ! H NMR (400 MHz, DMSO) ⁇ 1.88 (3H, dt), 2.16 (6H, s), 2.17 - 2.46 (7H, m), 3.28 (3H, s), 3.60 (3H, s), 4.05 - 4.19 (3H, m), 5.50 (IH, q), 7.06 - 7.22 (2H, m), 7.75 - 7.86 (2H, m), 8.05 (IH, dd), 8.24 (IH, d), 8.37 (IH, d). Mass
  • Example 31 NMR Spectrum: l H NMR (400 MHz, DMSO) ⁇ 1.4 - 1.48 (2H, m), 1.56 (4H, p), 1.83 - 2.02 (3H, m), 2.21 - 2.51 (11H, m), 3.33 (3H, s), 3.65 (3H, s), 4.20 (IH, p), 4.43 (2H, t), 5.58 (IH, p), 7.03 - 7.08 (IH, m), 8.12 (IH, dd), 8.26 (IH, dd), 8.38 (IH, d),
  • Example 32 NMR Spectrum: l H NMR (400 MHz, DMSO) ⁇ 1.77 - 2.41 (l lH, m), 2.53 - 2.66 (3H, m), 2.77 - 2.88 (2H, m), 3.27 (3H, s), 3.60 (3H, s), 4.1 - 4.18 (IH, m), 4.40 (2H, t), 5.19 (IH, ddd), 5.52 (IH, p), 7.01 (IH, dd), 8.08 (IH, d), 8.21 (IH, dd), 8.33 (IH, d), 8.40 (IH, d), 8.69 - 8.76 (IH, m).
  • reaction mixture was concentrated to dryness, then partitioned between EtOAc (50 mL) and saturated aqueous NH 4 C1 (50 mL). A precipitated crashed out of solution which was collected by filtration, washed with diethyl ether (3 x lOmL) and azeotroped with MeCN (2 x 50 mL) to afford the desired material (1.79 g, 124 %) as a yellow solid.
  • the crude product was purified by flash silica chromatography, elution gradient 0 to 100% EtOAc in heptane followed by 10% MeOH/EtOAc, to afford the desired product (0.110 g, 35.1 %) as a white solid.
  • Cis-3-methoxycyclobutan-l -amine hydrochloride (0.938 g, 6.82 mmol) was added in one portion to 4,6-dichlorocinnoline-3-carboxamide (1.65 g, 6.82 mmol) and DIPEA (3.57 mL, 20.45 mmol) in acetonitrile (13.47 mL) and the resulting suspension was stirred at 90 °C for 18 hours.
  • the crude product was purified by ion exchange chromatography, using an SCX column.
  • the desired product was eluted from the column using 1M NH3/MeOH to afford the desired product (0.300 g, 55.4 %) as a pale yellow oil.
  • Example 37 1:1 mixture of 8-[6-[3-(dimethylamino)propoxy]-3-pyridyl]-l-[(4S)-3,3- dimethyltetrahydropyr an-4-yl] -3-methyl-imidazo [4,5-c] cinnolin-2-one and 8- [6- [3- (dimethylamino)propoxy]-3-pyridyl]-l-[(4R)-3,3-dimethyltetrahydropyran-4-yl]-3- meth l-imidazo [4,5-c] cinnolin-2-one
  • the reaction mixture was diluted with EtOAc (25 mL), washed sequentially with water (10 mL), saturated brine (10 mL) and the organic layer evaporated.
  • the crude product was purified twice by preparative HPLC, using basic then acidic modifiers, to afford the desired material (22.5 mg) as a cream solid.
  • a 1 1 mixture of 8-[6-[3-(dimethylamino)propoxy]-3-pyridyl]-l-[(4S)-3,3- dimethyltetrahydropyran-4-yl] -3 -methyl-imidazo [4,5 -c] cinnolin-2-one and 8-[6- [3 - (dimethylamino)propoxy] -3 -pyridyl] - 1 -[(4R)-3 ,3 -dimethyltetrahydropyran-4-yl] -3 -methyl- imidazo[4,5-c]cinnolin-2-one was separated by preparative chiral-HPLC SFC (Chiralcel OJ-H Technologies IC column, 20 ⁇ silica, 250 mm length, 5 micron), eluting isocratically with 90% supercritical C0 2 in methanol (modified with ammonia) as eluent, to afford the first eluting product, Example
  • Example 39 Isomer 2 NMR Spectrum: l H NMR (400 MHz, CDCb) ⁇ 0.92 (3H, s), 1.33 (3H, s), 1.7 - 1.82 (1H, m), 1.97 - 2.07 (2H, m), 2.27 (6H, s), 2.44 - 2.53 (2H, m), 3.36 (1H, d), 3.57 (1H, dd), 3.65 - 3.72 (2H, m), 3.75 (3H, s), 4.24 - 4.32 (1H, m), 4.44 (2H, t), 4.82 (1H, dd), 6.92 (1H, dd), 7.82 (1H, dd), 7.88 (1H, dd), 8.34 (1H, d), 8.47 - 8.53 (2H, m).
  • reaction mixture was concentrated to dryness, then partitioned between EtOAc (50 mL) and saturated aqueous NH 4 C1 (50 mL). A precipitated crashed out of solution which was collected by filtration, washed with diethyl ether (3 x 10 mL) and azeotroped with MeCN (2 x 50 mL) to afford the desired material (3.60 g) as a beige solid.
  • Example 40 1:1 mixture of l-[(4S)-3,3-dimethyltetrahydropyran-4-yl]-3-methyl-8-[4- [3-(l-piperidyl)propoxy]phenyl]imidazo[4,5-c]cinnolin-2-one and l-[(4R)-3,3- dimethyltetrahydropyran-4-yl]-3-methyl-8-[4-[3-(l- pi eridyl)propoxy] phenyl] imidazo [4,5-c] cinnolin-2-one
  • Example 41 1:1 mixture of 8-[4-[3-(dimethylamino)propoxy]phenyl]-l-[(4S)-3,3- dimethyltetrahydropyr an-4-yl] -3-methyl-imidazo [4,5-c] cinnolin-2-one and 8- [4- [3- (dimethylamino)propoxy]phenyl]-l-[(4R)-3,3-dimethyltetrahydropyran-4-yl]-3- methyl-imidazo [4,5-c] cinnolin-2-one
  • the above material can also be isolated as the methane sulfonic acid salt by taking the material as prepared above and subjecting to the following reaction conditions.
  • a 1 1 mixture of 8- ⁇ 4-[3-(dimethylamino)propoxy]phenyl ⁇ -l-[(4S)-3,3- dimethyltetrahydro-2H-pyran-4-yl] -3 -methyl- 1 ,3 -dihydro-2H-imidazo [4,5 -c] cinnolin-2- one and 8- ⁇ 4-[3-(dimethylamino)propoxy]phenyl ⁇ - 1 -[(4R)-3,3-dimethyltetrahydro-2H- pyran-4-yl]-3-methyl-l,3-dihydro-2H-imidazo[4,5-c]cinnolin-2-one (20 mg, 0.04 mmol) was dissolved in DCM (2 mL) and treated with 1M methanesulfonic acid (0.003 mL, 0.04 mmol) in DCM then the mixture evaporated to dryness.
  • a 1 1 mixture of 8- ⁇ 4-[3-(dimethylamino)propoxy]phenyl ⁇ -l-[(4S)-3,3- dimethyltetrahydro-2H-pyran-4-yl] -3 -methyl- 1 ,3 -dihydro-2H-imidazo [4,5 -c] cinnolin-2- one and 8- ⁇ 4-[3-(dimethylamino)propoxy]phenyl ⁇ - 1 -[(4R)-3,3-dimethyltetrahydro-2H- pyran-4-yl]-3-methyl-l,3-dihydro-2H-imidazo[4,5-c]cinnolin-2-one was separated by preparative chiral-HPLC SFC (Phenomenex LU)
  • the crude product was purified by flash silica chromatography, elution gradient 0 to 10% MeOH in DCM. Pure fractions were evaporated to dryness to afford product as a brown oil.
  • the crude product was purified by preparative HPLC to afford the desired product (50 mg, 35 %>) as a brown solid.
  • the precipitate was collected by filtration, washed with water (100 mL) and dried under vacuum to afford the desired product (0.80 g, 45.1 %) as a red solid.
  • Aluminium trichloride 54 g, 404.98 mmol was added to a solution of 2-[(4-bromo-3- fluorophenyl)hydrazono]-2-cyanoacetamide (39 g, 136.81 mmol) in 1 ,2-dichlorobenzene (500 mL) under an inert atmosphere at ambient temperature.
  • the solution was stirred for 2 days at 120 °C, then cooled and quenched with iced water (3000 mL).
  • the solids formed were collected by filtration, washed with water (3 x 500 mL) and oven dried to afford the desired material (36 g, 92%) as a yellow solid.
  • Example 45 8- [6- [3-(Dimethylamino)propoxy] -3-pyridyl] -7-fluoro-l-isopropyl-3- methyl-imidazo [4,5-c] cinnolin-2-one
  • Example 46 7-Fluoro- l-isopropyl-3-methyl-8- [6- [3-(l -piperidyl)propoxy] -3- p ridyl] imidazo [4,5-c] cinnolin-2-one
  • the crude product was purified by flash silica chromatography, elution gradient 0 to 10% MeOH in DCM. Pure fractions were evaporated to dryness to afford crude product as a yellow oil.
  • the crude product was purified by preparative HPLC to afford the desired product (10 mg, 7 %) as a white solid.
  • Example 48 8- [4- [3-(Dimethylamino)propoxy] phenyl] -l-isopropyl-3,7-dimethyl- imidazo 4,5-c] cinnolin-2-one
  • 6-Bromo-4-chloro-7-methyl-cinnoline-3-carboxamide DMF (0.164 mL, 2.12 mmol) was added to a mixture of 6-bromo-4-hydroxy-7-methyl- cinnoline-3-carboxylic acid (6.00 g, 21.20 mmol) in thionyl chloride (90 mL, 1233.2 mmol) at ambient temperature under an inert atmosphere.
  • the resulting slurry was stirred at 80 °C for 2 hours then allowed to cool, concentrated to dryness and the residue azeotroped with toluene (3 x 100 mL) to afford the crude acid chloride.
  • Potassium hydroxide (62 g, 1.11 mol) was added to a solution of 4-amino-6-bromo-7- methyl-cinnoline-3-carboxamide (31 g, 110.28 mmol) in DMSO/H 2 0 (600 mL / 600 mL) under an inert atmosphere at ambient temperature. The solution was stirred for 2 days at 120 °C, cooled to ambient temperature and quenched with water/ice (3000 mL). The pH value of the solution was adjusted to 7 with 3M hydrochloric acid. The solids formed were collected by filtration, washed with water (2 x 500 mL) and dried. The crude product was purified by re-crystallization from DMSO to afford the desired product (11.5 g, 37%) as a light brown solid.
  • Aluminium trichloride (59.4 g, 445.47 mmol) was added to a solution of (lE)-2-amino-N- (4-bromo-3-methyl-anilino)-2-oxo-acetimidoyl cyanide (42 g, 149.41 mmol) in 1,2- dichlorobenzene (500 mL) under an inert atmosphere at ambient temperature. The solution was stirred for 3 hours at 120 °C, then cooled to 40-50 °C and quenched with water/ice (3000 mL). The solids formed were collected by filtration and washed with water (2 x 500 mL) and oven dried to afford the desired material (31 g, 74%) as a yellow solid.
  • Example 50 8- [6- [3-(Dimethylamino)propoxy] -3-pyridyl] -l-isopropyl-3,7-dimethyl- imidazo [4,5-c] cinnolin-2-one
  • Example 51 l-Isopropyl-3,7-dimethyl-8-[4-[3-(l- piperidyl)propoxy] phenyl] imidazo [4,5-c] cinnolin-2-one
  • Dichlorobis(triphenylphosphine)palladium(II) (0.999 mg, 1.42 ⁇ ) was added to a degassed solution of A ,N-dimethyl-3-[[5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2- pyridyl]oxy]propan-l -amine (9.15 mg, 0.03 mmol) and 8-chloro-3-(difluoromethyl)-l- isopropyl-imidazo[4,5-c]cinnolin-2-one (8.9 mg, 0.03 mmol), 2M potassium carbonate solution (0.043 mL, 0.09 mmol) in 1,4-dioxane (0.527 mL) and heated in a microwave at 80 °C for 1 hour under an inert atmosphere.
  • the reaction mixture was diluted with EtOAc (10 mL), and washed sequentially with water (7 mL), saturated brine (7 mL) and the organic layer was evaporated.
  • the crude product was purified by preparative HPLC to afford the desired product (5.7 mg, 44 %) as a yellow dry film.

Abstract

The specification generally relates to compounds of Formula (I) and pharmaceutically acceptable salts thereof, where R1-R4, A and X have the meanings defined herein. The specification also relates to the use of compounds of Formula (I) and salts thereof to treat or prevent ATM mediated disease, including cancer. The specification further relates to pharmaceutical compositions comprising substituted 1,3-dihydroimidazo[4,5-c]cinnolin-2-one compounds and pharmaceutically acceptable salts thereof; and kits comprising such compounds and salts.

Description

l,3-dihydroimidazo[4,5-c]cinnolin-2-one Compounds and Their Use in Treating
Cancer
FIELD
This specification relates to l,3-dihydroimidazo[4,5-c]cinnolin-2-one compounds and pharmaceutically acceptable salts thereof. These compounds and salts selectively modulate ataxia telangiectasia mutated ("ATM") kinase, and the specification therefore also relates to the use of l,3-dihydroimidazo[4,5-c]cinnolin-2-one compounds and salts thereof to treat or prevent ATM mediated disease, including cancer. The specification further relates to pharmaceutical compositions comprising l,3-dihydroimidazo[4,5- c]cinnolin-2-one compounds and pharmaceutically acceptable salts thereof; and kits comprising such compounds and salts.
BACKGROUND
ATM kinase is a serine threonine kinase originally identified as the product of the gene mutated in ataxia telangiectasia. Ataxia telangiectasia is located on human chromosome 1 lq22-23 and codes for a large protein of about 350 kDa, which is characterized by the presence of a phosphatidylinositol ("PI") 3-kinase-like
serine/threonine kinase domain flanked by FRAP-ATM-TRRAP and FATC domains which modulate ATM kinase activity and function. ATM kinase has been identified as a major player of the DNA damage response elicited by double strand breaks. It primarily functions in S/G2/M cell cycle transitions and at collapsed replication forks to initiate cell cycle checkpoints, chromatin modification, HR repair and pro-survival signalling cascades in order to maintain cell integrity after DNA damage (Lavin, M. F.; Rev. Mol. Cell Biol. 2008, 759-769).
ATM kinase signalling can be broadly divided into two categories: a canonical pathway, which signals together with the Mrel 1-Rad50-NBS1 complex from double strand breaks and activates the DNA damage checkpoint, and several non-canonical modes of activation, which are activated by other forms of cellular stress (Cremona et al. , Oncogene 2013, 3351-3360). ATM kinase is rapidly and robustly activated in response to double strand breaks and is reportedly able to phosphorylate in excess of 800 substrates (Matsuoka et al., Science 2007, 1160-1166), coordinating multiple stress response pathways (Kurz and Lees Miller, DNA Repair 2004, 889-900). ATM kinase is present predominantly in the nucleus of the cell in an inactive homodimeric form but autophosphorylates itself on Serl981 upon sensing a DNA double strand break (canonical pathway), leading to dissociation to a monomer with full kinase activity (Bakkenist et al., Nature 2003, 499-506). This is a critical activation event, and ATM phospho-Serl981 is therefore both a direct
pharmacodynamic and patient selection biomarker for tumour pathway dependency.
The antitumoral effects of ATM inhibitor monotherapy have been reported in a mouse model of acute myeloid leukemia (Fernandez-Capetillo et al., Science Signalling, 2016, 9 (445), ra91). Moreover, ATM kinase responds to direct double strand breaks caused by common anti-cancer treatments such as ionising radiation and topoisomerase-II inhibitors (doxorubicin, etoposide) and also to topoisomerase-I inhibitors (for example irinotecan and topotecan) via single strand break to double strand break conversion during replication. As a result, ATM kinase inhibition can potentiate the activity of any these agents. ATM kinase inhibitors are therefore expected to be of use in the treatment of cancer.
It is desirable for medicinal compounds to have pharmacokinetic properties which allow them to be dosed at tolerable levels to patients. Poor pharmacokinetic properties can be a cause of failure of drug candidates in clinical development. An example of poor pharmacokinetic properties is rapid metabolism which may cause a drug to be cleared rapidly from the body, thus reducing its therapeutic benefit. Although it may be possible to overcome rapid drug clearance by more frequent or higher dosing of the drug, such approaches may decrease patient compliance and/or expose patients to risks of increased side effects. Another approach to addressing problems of rapid metabolism is to substitute one or more atoms in a drug molecule that are suseptible to metabolism with atoms that are less suseptible. The resulting increased stability can impact the pharmacokinetic properties of a drug, for example, by retarding certain pathways of its metabolism.
The compounds described herein are expected to demonstrate pharmacokinetic properties that would be indicative of a profile suitable for administration to patients. WO 2015/170081, WO 2017/046216, WO 2017/076895 and WO 2017/076898 describe imidazo[4,5-c]quinolin-2-one compounds that have activity as ATM kinase modulators. However, there exists a need for new compounds which act against certain kinase enzymes, like ATM kinase, in a highly selective fashion (i.e. by modulating ATM more effectively than other biological targets).
As demonstrated elsewhere in the specification (for example in the cell based assays described in the experimental section), the compounds of the present specification generally possess very potent ATM kinase inhibitory activity, but much less potent activity against other tyrosine kinase enzymes, such as PI 3-kinase a, mTOR kinase and ataxia telangiectasia and Rad3-related protein ("ATR") kinase. As such, the compounds of the present specification not only inhibit ATM kinase, but can be considered to be highly selective inhibitors of ATM kinase.
As a result of their highly selective nature, the compounds of the present specification are expected to be particularly useful in the treatment of diseases in which ATM kinase is implicated (for example, in the treatment of cancer), but where it is desirable to minimise off-target effects or toxicity that might arise due to the inhibition of other tyrosine kinase enzymes, such as class PI 3-kinase a, mTOR kinase and ATR kinase.
SUMMARY
Briefly, this specification describes, in part, a compound of Formula (I):
Figure imgf000004_0001
(I)
or a pharmaceutically acceptable salt thereof, wherein
A is N or CR4;
X is -OR5 or -NR6R7; R1 is (Ci-C6)alkyl, cycloalkyl or heterocycloalkyl;
R2 is H or methyl, wherein methyl is optionally substituted with 1, 2, or 3 halo substituents;
R3 is H, (Ci-C6)alkyl or halo;
R4 is H, (Ci-C6)alkyl or halo;
R5 is -(CH2)n-NR8R9;
R6 and R7 together with the nitrogen atom to which they are attached form a 4, 5 or 6 membered non-aromatic monocyclic ring comprising carbon which is substituted with - NR10RU;
R8 and R9 are independently selected from H and (Ci-Ce)alkyl; or R8 and R9 together with the nitrogen atom to which they are attached form a 4, 5 or 6 membered non- aromatic monocyclic ring comprising carbon which can be optionally substituted with halo;
R10 is H or (Ci-C3)alkyl;
R11 is (Ci-C )alkyl;
n is 2, 3 or 4;
alkyl is a linear or branched saturated hydrocarbon;
alkoxy is a linear or branched O-linked saturated hydrocarbon;
cycloalkyl is a 3, 4, 5 or 6 membered aliphatic carbocyclic ring, which can be optionally substituted with 1 or 2 substituents selected from (Ci-C3)alkyl, (Ci-C3)alkoxy and -OH;
halo is F, CI or Br;
heterocycloalkyl is a 4, 5 or 6 membered non-aromatic monocyclic ring comprising carbon and 1 or 2 heteroatoms selected from nitrogen and oxygen; wherein
heterocycloalkyl can be optionally substituted with 1 or 2 substituents selected from (Ci- C )alkyl, (Ci-C )alkoxy and -OH.
An aspect of this specification includes compounds of formula (I), as defined above, tautomers, isomers, stereoisomers (including enantiomers, diastereoisomers and racemic and scalemic mixtures thereof), pharmaceutically acceptable salts and solvates thereof. In another aspect the specification provides a prodrug of a compound of formula (I) as herein defined, or a pharmaceutically acceptable salt thereof.
In yet another aspect, the specification provides an N-oxide of a compound of formula (I) as herein defined, or a prodrug or pharmaceutically acceptable salt thereof.
It will be understood that certain compounds of formula (I) may exist in solvated, for example hydrated, as well as unsolvated forms. It is to be understood that the specification encompasses all such solvated forms.
This specification also describes, in part, a pharmaceutical composition which comprises a compound of Formula (I), or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient.
This specification also describes, in part, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in therapy.
This specification also describes, in part, a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer.
This specification also describes, in part, the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of cancer.
This specification also describes, in part, a method for treating cancer in a warm blooded animal in need of such treatment, which comprises administering to said warmblooded animal a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof.
DESCRIPTION OF ILLUSTRATIVE EMBODIMENTS
Many embodiments are detailed throughout the specification and will be apparent to a reader skilled in the art. The specification is not to be interpreted as being limited to any particular embodiment(s) described herein.
In an embodiment there is provided a compound of Formula (I):
Figure imgf000007_0001
(I)
or a pharmaceutically acceptable salt thereof, wherein
A is N or CR4;
X is -OR5 or -NR6R7;
R1 is (Ci-C6)alkyl, cycloalkyl or heterocycloalkyl;
R2 is H or methyl, wherein methyl is optionally substituted with 1, 2, or 3 halo substituents;
R3 is H, (Ci-C6)alkyl or halo;
R4 is H, (Ci-C6)alkyl or halo;
R5 is -(CH2)n-NR8R9;
R6 and R7 together with the nitrogen atom to which they are attached form a 4, 5 or 6 membered non-aromatic monocyclic ring comprising carbon which is substituted with - NR10RU;
R8 and R9 are independently selected from H and (Ci-Ce)alkyl; or R8 and R9 together with the nitrogen atom to which they are attached form a 4, 5 or 6 membered non- aromatic monocyclic ring comprising carbon which can be optionally substituted with halo;
R10 is H or (Ci-C3)alkyl;
R11 is (Ci-C )alkyl;
n is 2, 3 or 4;
alkyl is a linear or branched saturated hydrocarbon;
alkoxy is a linear or branched O-linked saturated hydrocarbon;
cycloalkyl is a 3, 4, 5 or 6 membered aliphatic carbocyclic ring, which can be optionally substituted with 1 or 2 substituents selected from (Ci-C3)alkyl, (Ci-C3)alkoxy and -OH; halo is F, CI or Br;
heterocycloalkyl is a 4, 5 or 6 membered non-aromatic monocyclic ring comprising carbon and 1 or 2 heteroatoms selected from nitrogen and oxygen; wherein
heterocycloalkyl can be optionally substituted with 1 or 2 substituents selected from (Ci- C3)alkyl, (Ci-C )alkoxy and -OH.
In an embodiment, there is provided a compound of formula (la)
Figure imgf000008_0001
or a pharmaceutically acceptable salt thereof, wherein
A is N or CH;
X is -OR5 or -NR6R7;
R1 is (Ci-C3)alkyl, cycloalkyl, heterocycloalkyl;
R2 is H or methyl, wherein methyl is optionally substituted with 1, 2, or 3 fluoro substituents;
R3 is H, methyl, chloro or fluoro;
R4 is H, methyl, chloro or fluoro;
R5 is -(CH2)n-NR8R9;
R6 and R7 together with the nitrogen atom to which they are attached form a 4, 5 or 6 membered non-aromatic monocyclic ring comprising carbon which is substituted with - NR10RU;
R8 and R9 are independently selected from H and (Ci-C3)alkyl; or R8 and R9 together with the nitrogen atom to which they are attached form a 4, 5 or 6 membered non- aromatic monocyclic ring comprising carbon which can be optionally substituted with fluoro;
R10 is H or (Ci-C )alkyl;
R11 is (Ci-C )alkyl; n is 2, 3 or 4;
alkyl is a linear or branched saturated hydrocarbon;
cycloalkyl is a 3, 4, 5 or 6 membered aliphatic carbocyclic ring, which can be optionally substituted with 1 or 2 substituents selected from methyl, methoxy and -OH; heterocycloalkyl is a 4, 5 or 6 membered non-aromatic monocyclic ring comprising carbon and a heteroatom selected from oxygen; wherein heterocycloalkyl can be optionally substituted with 1 or 2 substituents selected from methyl, methoxy and -OH.
In an embodiment, there is provided a compound of formula (lb)
Figure imgf000009_0001
or a pharmaceutically acceptable salt thereof, wherein
A is N or CH;
R1 is (Ci-C3)alkyl, cycloalkyl, heterocycloalkyl;
R2 is H or methyl, wherein methyl is optionally substituted with 1, 2, or 3 fluoro substituents;
R3 is H, methyl or fluoro;
R4 is H, methyl or fluoro;
R8 and R9 are independently selected from H and (Ci-C3)alkyl; or R8 and R9 together with the nitrogen atom to which they are attached form a 5 or 6 membered non- aromatic monocyclic ring comprising carbon which can be optionally substituted with fluoro;
alkyl is a linear or branched saturated hydrocarbon;
cycloalkyl is a 3, 4, 5 or 6 membered aliphatic carbocyclic ring, which can be optionally substituted with 1 or 2 substituents selected from methyl, methoxy and -OH; heterocycloalkyl is a 5 or 6 membered non-aromatic monocyclic ring comprising carbon and a heteroatom selected from oxygen; wherein heterocycloalkyl can be optionally substituted with 1 or 2 substituents selected from methyl, methoxy and -OH.
embodiment, there is provided a compound of formula (Ic)
Figure imgf000010_0001
or a pharmaceutically acceptable salt thereof, wherein
A is N or CH;
R1 is (Ci-C3)alkyl, cycloalkyl, heterocycloalkyl;
R2 is H or methyl, wherein methyl is optionally substituted with 1, 2, or 3 fluoro substituents;
R3 is H, methyl or fluoro;
R4 is H, methyl or fluoro;
R6 and R7 together with the nitrogen atom to which they are attached form a 5 or 6 membered non-aromatic monocyclic ring comprising carbon which is substituted with -NR10Rn;
R10 is H or (Ci-C3)alkyl;
R11 is (Ci-C3)alkyl;
alkyl is a linear or branched saturated hydrocarbon;
cycloalkyl is a 3, 4, 5 or 6 membered aliphatic carbocyclic ring, which can be optionally substituted with 1 or 2 substituents selected from methyl, methoxy and -OH; heterocycloalkyl is a 5 or 6 membered non-aromatic monocyclic ring comprising carbon and a heteroatom selected from oxygen; wherein heterocycloalkyl can be optionally substituted with 1 or 2 substituents selected from methyl, methoxy and -OH. The following embodiments of moieties A, X, R^-R11, n and halo may be applied, alone or in combination, to the descriptions of the compounds of Formula (I), (la), (lb) or (Ic) provided above:
A is N or CR4.
A is N or CH.
A is N.
A is CH.
X is -OR5 or -NR6R7.
X is -OR5.
X is -NR6R7.
X is selected from the following fragments:
Figure imgf000011_0001
wherein designates the point of attachment of the fragment to the remainder of the molecule. X is selected from the following fragments:
Figure imgf000011_0002
Figure imgf000012_0001
and wherein *" designates the point of attachment of the fragment to the remainder of the molecule.
X is selected from the following fragments:
Figure imgf000012_0002
and wherein *" designates the point of attachment of the fragment to the remainder of the molecule.
R1 is (Ci-C6)alkyl, cycloalkyl or heterocycloalkyl.
R1 is (Ci-C3)alkyl, cycloalkyl or heterocycloalkyl.
R1 is methyl, ethyl, n-propyl, iso-propyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, oxetanyl, tetrahydrofuranyl or tertrahydropyranyl; wherein cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, tetrahydrofuranyl or tertrahydropyranyl can be optionally substituted with 1 or 2 substituents selected from (Ci-C3)alkyl, (Ci-C3)alkoxy and -OH.
R1 is n-propyl, iso-propyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, oxetanyl, tetrahydrofuranyl or tertrahydropyranyl; wherein cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, tetrahydrofuranyl or tertrahydropyranyl can be optionally substituted with 1 or 2 substituents selected from methyl, methoxy and -OH.
R1 is iso-propyl, cyclopropyl, cyclobutyl, cyclopentyl, tertrahydropyran-3-yl or tertrahydropyran-4-yl; wherein cyclopropyl, cyclobutyl, cyclopentyl, tertrahydropyran-3-yl or tertrahydropyran-4-yl can be optionally substituted with 1 or 2 substituents selected from methyl and methoxy.
R1 is iso-propyl, cyclopropyl, cyclobutyl or tertrahydropyranyl; wherein
cyclopropyl, cyclobutyl or tertrahydropyranyl can be optionally substituted with 1 or 2 substituents selected from methyl or methoxy. R1 is selected from the following fragments:
Figure imgf000013_0001
, and ; wherein *· designates the point of attachment of the fragment to the remainder of the molecule.
R1 is selected from the following fragments:
Figure imgf000013_0002
, and ; wherein *· designates the point of attachment of the fragment to the remainder of the molecule.
R1 is selected from the followin fragments:
Figure imgf000013_0003
and ; wherein * designates the point of attachment of the fragment to the remainder of the molecule.
R1 is iso-propyl.
R1 is cyclopropyl, optionally substituted with methyl.
R1 is cyclobutyl, optionally substituted with methoxy.
R1 is tertrahydropyranyl, optionally mono or di substituted with methyl. R2 is H or methyl, wherein methyl is optionally substituted with 1, 2, or 3 halo substituents.
R2 is H or methyl, wherein methyl is optionally substituted with 1, 2, or 3 fluoro substituents.
R2 is methyl optionally substituted with 1 , 2, or 3 fluoro substituents.
R2 is methyl or CHF2.
R2 is methyl.
R3 is H, (Ci-C6)alkyl or halo.
R3 is H, (Ci-C3)alkyl, chloro or fluro.
R3 is H, methyl, chloro or fluoro.
R3 is H, methyl or fluoro.
R3 is H or fluoro.
R3 is fluoro.
R3 is H.
R4 is H, (Ci-C6)alkyl or halo.
R4 is H, (Ci-C3)alkyl, chloro or fluro.
R4 is H, methyl, chloro or fluoro.
R4 is H, methyl or fluoro.
R4 is H or fluoro.
R4 is H or methyl.
R4 is H.
Figure imgf000014_0001
R5 is -(CH2)3-NR¾9.
R6 and R7 together with the nitrogen atom to which they are attached form a 4, 5 or 6 membered non-aromatic monocyclic ring comprising carbon which is substituted with - NR10Rn . R6 and R7 together with the nitrogen atom to which they are attached form a 5 or 6 membered non-aromatic monocyclic ring comprising carbon which is substituted with - NR10RU
R6 and R7 together with the nitrogen atom to which they are attached form a 5 or 6 membered non-aromatic monocyclic ring comprising carbon which is substituted with - N(CH3)2.
R6 and R7 together with the nitrogen atom to which they are attached form a fragment selected from:
Figure imgf000015_0001
wherein ·* designates the point of attachment of the fragment to the aromatic ring containing A.
R6 and R7 together with the nitrogen atom to which they are attached form a fragment selected from:
Figure imgf000015_0002
wherein ·* designates the point of attachment of the fragment to the aromatic ring containing A.
R8 and R9 are independently selected from H and (Ci-Ce)alkyl; or R8 and R9 together with the nitrogen atom to which they are attached form a 4, 5 or 6 membered non- aromatic monocyclic ring comprising carbon which can be optionally substituted with halo.
R8 and R9 are independently selected from H and (Ci-C3)alkyl; or R8 and R9 together with the nitrogen atom to which they are attached form a 5 or 6 membered non- aromatic monocyclic ring comprising carbon which can be optionally substituted with fluoro.
R8 and R9 are independently selected from H and methyl; or R8 and R9 together with the nitrogen atom to which they are attached form a 5 or 6 membered non-aromatic monocyclic ring comprising carbon which can be optionally substituted with fluoro.
R8 and R9 are methyl; or R8 and R9 together with the nitrogen atom to which they are attached form a 5 or 6 membered non-aromatic monocyclic ring comprising carbon which can be optionally substituted with fluoro.
R8 and R9 together with the nitrogen atom to which they are attached form a 6 membered non-aromatic monocyclic ring comprising carbon which can be optionally substituted with fluoro.
R8 and R9 together with the nitrogen atom to which they are attached form a 5 membered non-aromatic monocyclic ring comprising carbon which can be optionally substituted with fluoro.
R8 and R9 together with the nitrogen atom to which they are attached form a fragment selected from:
Figure imgf000016_0001
, and , wherein ** designates the point of attachment of the fragment to the remainder of the molecule.
R8 and R9 together with the nitrogen atom to which they are attached form a fragment selected from:
Figure imgf000017_0001
, wherein ** designates the point of attachment of the fragment to the remainder of the molecule.
R8 and R9 together with the nitrogen atom to which they are attached form a fragment selected from:
Figure imgf000017_0002
wherein »· designates the point of attachment of the fragment to the remainder of the molecule.
R8 is H and R9 is methyl.
R8 and R9 are methyl.
R is H or (Ci-C3)alkyl.
R10 is H or methyl.
R10 is methyl.
R11 is (Ci-C3)alkyl.
R11 is methyl. n is 2, 3 or 4.
n is 2 or 3.
n is 3. halo is F, CI or Br.
halo is F or CI. halo is F.
In an embodiment, the compound of Formula (I) is selected from:
8- [6-[3 -(Dimethylamino)propoxy] -3 -pyridyl] -3 -methyl- 1 -tetrahydropyran-4-yl- imidazo[4,5-c]cinnolin-2-one;
8- [6-[3 -(dimethylamino)propoxy] -3 -pyridyl] - 1 -(cis-3 -methoxycyclobutyl)-3 - methyl-imidazo[4,5-c]cinnolin-2-one;
8- [6-[3 -(Dimethylamino)propoxy] -3 -pyridyl] -3 -methyl- 1 - [(3 S)-tetrahydropyran-3 - yl]imidazo[4,5-c]cinnolin-2-one;
8- [6-[3 -(Dimethylamino)propoxy] -3 -pyridyl] -3 -methyl- 1 - [(3R)-tetrahydropyran-3 - yl]imidazo[4,5-c]cinnolin-2-one;
3 -Methyl-8-[6- [3 -( 1 -piperidyl)propoxy] -3 -pyridyl] - 1 -tetrahydropyran-4-yl- imidazo[4,5-c]cinnolin-2-one;
8- [4-[3 -(dimethylamino)propoxy]phenyl] - 1 -isopropyl-3 -methyl-imidazo [4,5 - c]cinnolin-2-one;
l-Isopropyl-3-methyl-8-[6-[3-(l-piperidyl)propoxy]-3-pyridyl]imidazo[4,5- c]cinnolin-2-one;
8- [6-[3 -(dimethylamino)propoxy] -3 -pyridyl] - 1 -isopropyl-3 -methyl-imidazo [4,5 - c]cinnolin-2-one;
l-Isopropyl-3-methyl-8-[4-(3-pyrrolidin-l-ylpropoxy)phenyl]imidazo[4,5- c]cinnolin-2-one;
l-isopropyl-3-methyl-8-(4-(3-(piperidin-l-yl)propoxy)phenyl)-l,3-dihydro-2H- imidazo[4,5-c]cinnolin-2-one;
8- [6-[3 -(dimethylamino)propoxy] -3 -pyridyl] - 1 -(trans-3 -methoxycyclobutyl)-3 - methyl-imidazo[4,5-c]cinnolin-2-one;
8-[6-[3-(4-Fluoro-l -piperidyl)propoxy] -3 -pyridyl]- 1 -isopropyl-3 -methyl- imidazo[4,5-c]cinnolin-2-one;
8- [6-[3 - [(3R)-3 -Fluoropyrrolidin- 1 -yl]propoxy] -3 -pyridyl] - 1 -isopropyl-3 -methyl- imidazo[4,5-c]cinnolin-2-one;
l-isopropyl-3-methyl-8-[2-methyl-6-[3-(l-piperidyl)propoxy]-3- pyridyl]imidazo[4,5-c]cinnolin-2-one; 1 -(trans-3 -Methoxycyclobutyl)-3 -methyl-8-[4- [3 -( 1 - piperidyl)propoxy]phenyl]imidazo[4,5-c]cinnolin-2-one;
8- [4-[3 -(Dimethylamino)propoxy]phenyl] - 1 -(trans-3 -methoxycyclobutyl)-3 - methyl-imidazo [4,5 -c] cinnolin-2-one;
8- [6-[3 -(4-Fluoro- 1 -piperidyl)propoxy] -3 -pyridyl] - 1 -(trans-3 -methoxycyclobutyl)- 3 -methyl-imidazo [4,5 -c] cinnolin-2-one;
8-[6-[3-(4-Fluoro-l-piperidyl)propoxy]-3-pyridyl]-l-(cz5-3-methoxycyclobutyl)-3- methyl-imidazo[4,5-c]cinnolin-2-one;
1 -(cis-3 -methoxycyclobutyl)-3 -methyl-8-[4- [3 -( 1 - piperidyl)propoxy]phenyl]imidazo[4,5-c]cinnolin-2-one;
8-[6-[3-[(3R)-3-Fluoropyrrolidin-l-yl]propoxy]-3-pyridyl]-l-(cz5-3- methoxycyclobutyl)-3 -methyl-imidazo [4,5 -c] cinnolin-2-one;
8- [4-[3 -(Dimethylamino)propoxy]phenyl] -3 -methyl- 1 - [(3R)-tetrahydropyran-3 - yl]imidazo[4,5-c]cinnolin-2-one;
3-Methyl-8-[4-[3-(l-piperidyl)propoxy]phenyl]-l-[(3R)-tetrahydropyran-3- yl]imidazo[4,5-c]cinnolin-2-one;
8-[6-[3-(4-Fluoro-l -piperidyl)propoxy] -3 -pyridyl] -3 -methyl- 1 -[(3R)- tetrahydropyran-3 -yl] imidazo [4,5 -c] cinnolin-2-one;
8- [4-[3 -(Dimethylamino)propoxy]phenyl] -3 -methyl- 1 -( 1 - methylcyclopropyl)imidazo[4,5-c]cinnolin-2-one;
3-Methyl- 1 -(1 -methylcyclopropyl)-8-[4-[3-(l - piperidyl)propoxy]phenyl]imidazo[4,5-c]cinnolin-2-one;
8- [6-[3 -(4-Fluoro- 1 -piperidyl)propoxy] -3 -pyridyl] -3 -methyl- 1 -(1 - methylcyclopropyl)imidazo[4,5-c]cinnolin-2-one;
3-Methyl- 1 -(1 -methylcyclopropyl)-8-[6-[3-(l -piperidyl)propoxy]-3- pyridyl]imidazo[4,5-c]cinnolin-2-one;
8- [4-[3 -(dimethylamino)propoxy]phenyl] - 1 - [( 1 R,3R)-3 -methoxy cyclop entyl] -3 - methyl-imidazo[4,5-c]cinnolin-2-one;
l-[(lS,3S)-3-methoxycyclopentyl]-3-methyl-8-[4-[3-(l- piperidyl)propoxy]phenyl]imidazo[4,5-c]cinnolin-2-one; 8- [4-[3 -(Dimethylamino)propoxy]phenyl] - 1 - [( 1 S ,3 S)-3 -methoxycyclopentyl] -3 - methyl-imidazo [4,5 -c] cinnolin-2-one;
l-[(lR,3R)-3-Methoxycyclopentyl]-3-methyl-8-[6-[3-(l-piperidyl)propoxy]-3- pyridyl]imidazo[4,5-c]cinnolin-2-one;
8-[6-[3-[(3S)-3-Fluoropyrrolidin-l-yl]propoxy]-3-pyridyl]-l-[(lR,3R)-3- methoxycyclopentyl] -3 -methyl-imidazo [4,5 -c] cinnolin-2-one;
l-[(l S,3S)-3-Methoxycyclopentyl]-3-methyl-8-[6-[3-(l-piperidyl)propoxy]-3- pyridyl]imidazo[4,5-c]cinnolin-2-one;
8- [4-[3 -(Dimethylamino)propoxy]phenyl] - 1 -(cis-3 -methoxycyclobutyl)-3 -methyl- imidazo[4,5-c]cinnolin-2-one;
l-(cz'5-3-Methoxycyclobutyl)-3-methyl-8-[6-[3-(l-piperidyl)propoxy]-3- pyridyl] imidazo [4,5 -c] cinnolin-2-one;
8-[6-[3-[(3S)-3-Fluoropyrrolidin-l-yl]propoxy]-3-pyridyl]-l-(tra/75-3- methoxycyclobutyl)-3 -methyl-imidazo [4,5 -c] cinnolin-2-one;
8-[6-[3-(dimethylamino)propoxy]-3-pyridyl]-l-[(4S)-3,3-dimethyltetrahydropyran- 4-yl] -3 -methyl-imidazo [4,5 -c] cinnolin-2-one;
8- [6-[3 -(dimethylamino)propoxy] -3 -pyridyl] - 1 -[(4R)-3 ,3 -dimethyltetrahydropyran- 4-yl] -3 -methyl-imidazo [4,5 -c] cinnolin-2-one
1 - [3 ,3 -dimethyltetrahydropyran-4-yl] -3 -methyl-8- {4-[3 -( 1 - piperidinyl)propoxy]phenyl} -l ,3-dihydro-2H-imidazo[4,5-c]cinnolin-2-one;
8-[4-[3-(dimethylamino)propoxy]phenyl]-l-[(4S)-3,3-dimethyltetrahydropyran-4- yl] -3 -methyl-imidazo [4,5 -c] cinnolin-2-one;
8- [4-[3 -(dimethylamino)propoxy]phenyl] - 1 - [(4R)-3 ,3 -dimethyltetrahydropyran-4- yl] -3 -methyl-imidazo [4,5 -c] cinnolin-2-one;
8-[4-[3-(dimethylamino)propoxy]phenyl]-7-fluoro-l-isopropyl-3-methyl- imidazo[4,5-c]cinnolin-2-one;
8- [6-[3 -(Dimethylamino)propoxy] -3 -pyridyl] -7-fluoro- 1 -isopropyl-3 -methyl- imidazo[4,5-c]cinnolin-2-one;
7-Fluoro-l -isopropyl-3-methyl-8-[6-[3-(l-piperidyl)propoxy]-3- pyridyl]imidazo[4,5-c]cinnolin-2-one; 7- fluoro-l-isopropyl-3-methyl-8-[4-[3-(l-piperidyl)propoxy]phenyl]imidazo[4,5- c]cinnolin-2-one;
8- [4-[3-(dimethylamino)propoxy]phenyl]-l-isopropyl-3,7-dimethyl-imidazo[4,5- c]cinnolin-2-one;
l-isopropyl-3,7-dimethyl-8-[6-[3-(l-piperidyl)propoxy]-3-pyridyl]imidazo[4,5- c]cinnolin-2-one;
8-[6-[3-(dimethylamino)propoxy]-3-pyridyl]-l-isopropyl-3,7-dimethyl- imidazo[4,5-c]cinnolin-2-one;
l-Isopropyl-3,7-dimethyl-8-[4-[3-(l-piperidyl)propoxy]phenyl]imidazo[4,5- c]cinnolin-2-one;
3 -(difluoromethyl)-8- [6-[3 -(dimethylamino)propoxy] -3 -pyridyl] - 1 -isopropyl- imidazo[4,5-c]cinnolin-2-one;
8-[6-[(3R)-3-(Dimethylamino)pyrrolidin- 1 -yl] -3 -pyridyl]- 1 -isopropyl-3 -methyl- imidazo^, 5-c]cinnolin-2-one;
8-[6-[4-(Dimethylamino)-l -piperidyl] -3 -pyridyl]- 1 -isopropyl-3 -methyl - imidazo[4,5-c]cinnolin-2-one;
8- [6-[(3R)-3 -(dimethylamino)pyrrolidin- 1 -yl] -3 -pyridyl] -3 -methyl- 1 - [(3R)- tetrahydropyran-3 -yl] imidazo [4,5 -c] cinnolin-2-one;
8-[6-[4-(dimethylamino)- 1 -piperidyl] -3 -pyridyl]- 1 -[3,3-dimethyltetrahydropyran-4- yl] -3 -methyl-imidazo [4,5 -c] cinnolin-2-one;
8- [6-[(3R)-3 -(dimethylamino)pyrrolidin- 1 -yl] -3 -pyridyl] - 1 - [3 ,3 - dimethyltetrahydropyran-4-yl] -3 -methyl-imidazo [4,5 -c] cinnolin-2-one;
8-[6-[(3R)-3-(Dimethylamino)pyrrolidin- 1 -yl] -3 -pyridyl]- 1 -(trans-3- methoxycyclobutyl)-3 -methyl-imidazo [4,5 -c] cinnolin-2-one;
8-[6-[(3R)-3-(Dimethylamino)pyrrolidin- 1 -yl] -3 -pyridyl]- 1 -(cis-3- methoxycyclobutyl)-3 -methyl-imidazo [4,5 -c] cinnolin-2-one;
8- [6-[3 -(dimethylamino)pyrrolidin- 1 -yl] -3 -pyridyl] -3 -methyl- 1 -tetrahydropyran-4- yl-imidazo[4,5-c]cinnolin-2-one;
8-[6-[(3R)-3-(Dimethylamino)pyrrolidin- 1 -yl] -3 -pyridyl]- 1 -[(lR,3R)-3- methoxycyclopentyl] -3 -methyl-imidazo [4,5 -c] cinnolin-2-one; 8-[6-[(3R)-3-(Dimethylamino)pyrrolidin- 1 -yl]-3-pyridyl]- 1 -[(1 S,3S)-3- methoxycyclopentyl] -3 -methyl-imidazo [4,5 -c] cinnolin-2-one;
8- [6-[4-(Dimethylamino)- 1 -piperidyl] -3 -pyridyl] - 1 -(cis-3 -methoxycyclobutyl)-3 - methyl-imidazo[4,5-c]cinnolin-2-one;
8-[4-[4-(Dimethylamino)-l-piperidyl]phenyl]-l-isopropyl-3-methyl-imidazo[4,5- c]cinnolin-2-one;
8- [4-[4-(dimethylamino)- 1 -piperidyl]phenyl] - 1 - [3 ,3 -dimethyltetrahydropyran-4- yl] -3 -methyl-imidazo [4,5 -c] cinnolin-2-one;
8-[4-[4-(Dimethylamino)- 1 -piperidyl]phenyl] - 1 -[( 1 R,3R)-3-methoxycyclopentyl] - 3 -methyl-imidazo [4,5 -c] cinnolin-2-one;
8-[4-[4-(Dimethylamino)-l-piperidyl]phenyl]-l-[(lS,3S)-3-methoxycyclopentyl]-3- methyl-imidazo[4,5-c]cinnolin-2-one;
8-[4-[4-(Dimethylamino)-l-piperidyl]phenyl]-3-methyl-l-[(3R)-tetrahydropyran-3- yl]imidazo[4,5-c]cinnolin-2-one;
8- [6-[(3R)-3 -(Dimethyl amino)pyrro lidin- 1 -yl] -3 -pyridyl] -7-fluoro- 1 -isopropyl-3 - methyl-imidazo[4,5-c]cinnolin-2-one;
8-[4-[4-(Dimethylamino)-l-piperidyl]phenyl]-7-fluoro-l-isopropyl-3-methyl- imidazo[4,5-c]cinnolin-2-one;
8-[6-[(3R)-3-(dimethylamino)pyrrolidin-l-yl]-3-pyridyl]-l-isopropyl-3,7-dimethyl- imidazo[4,5-c]cinnolin-2-one;
8-[4-[4-(dimethylamino)- 1 -piperidyl]phenyl] - 1 -isopropyl-3 ,7-dimethyl- imidazo[4,5-c]cinnolin-2-one;
8- [6-[3 -(dimethylamino)propoxy] -2-fluoro-3 -pyridyl] - 1 -isopropyl-3 -methyl- imidazo[4,5-c]cinnolin-2-one;
and pharmaceutically acceptable salts thereof.
In an embodiment, the compound of Formula (I) is selected from:
l-Isopropyl-3-methyl-8-[6-[3-(l-piperidyl)propoxy]-3-pyridyl]imidazo[4,5- c]cinnolin-2-one;
8- [6-[3 -(dimethylamino)propoxy] -3 -pyridyl] - 1 -isopropyl-3 -methyl-imidazo [4,5 - c]cinnolin-2-one; l-Isopropyl-3-methyl-8-[4-(3-pyrrolidin-l-ylpropoxy)phenyl]imidazo[4,5- c]cinnolin-2-one;
8-[6-[3-(4-Fluoro-l -piperidyl)propoxy] -3 -pyridyl]- 1 -isopropyl-3 -methyl- imidazo[4,5-c]cinnolin-2-one;
8- [6-[3 - [(3R)-3 -Fluoropyrrolidin- 1 -yljpropoxy] -3 -pyridyl] - 1 -isopropyl-3 -methyl- imidazo[4,5-c]cinnolin-2-one;
8- [4-[3 -(dimethylamino)propoxy]phenyl] - 1 - [( 1 R,3R)-3 -methoxy cyclop entyl] -3 - methyl-imidazo[4,5-c]cinnolin-2-one;
8- [4-[3 -(Dimethylamino)propoxy]phenyl] - 1 - [( 1 S ,3 S)-3 -methoxycyclopentyl] -3 - methyl-imidazo[4,5-c]cinnolin-2-one;
8- [6-[3 -(dimethylamino)propoxy] -3 -pyridyl] - 1 -[(4R)-3 ,3 -dimethyltetrahydropyran- 4-yl] -3 -methyl-imidazo [4,5 -c] cinnolin-2-one;
8-{4-[3-(dimethylamino)propoxy]phenyl}-l-[(4R)-3,3-dimethyltetrahydro-2H- pyran-4-yl] -3 -methyl- 1 ,3 -dihydro-2H-imidazo [4,5 -c] cinnolin-2-one;
7- Fluoro-l-isopropyl-3-methyl-8-[6-[3-(l-piperidyl)propoxy]-3- pyridyl]imidazo[4,5-c]cinnolin-2-one;
8- [6-[(3R)-3-(Dimethylamino)pyrrolidin- 1 -yl] -3 -pyridyl]- 1 -isopropyl-3 -methyl- imidazo[4,5-c]cinnolin-2-one;
8- [6-[(3R)-3 -(dimethylamino)pyrrolidin- 1 -yl] -3 -pyridyl] -3 -methyl- 1 - [(3R)- tetrahydropyran-3 -yl] imidazo [4,5 -c] cinnolin-2-one;
8-[4-[4-(Dimethylamino)-l-piperidyl]phenyl]-l-isopropyl-3-methyl-imidazo[4,5- c]cinnolin-2-one;
and pharmaceutically acceptable salts thereof.
In substituents such as -OR5 and -NR6R7, "-" denotes the point of attachment of the substituent to the remainder of the molecule.
The term "O-linked" such as in "O-linked saturated hydrocarbon", means that the hydrocarbon residue is joined to the remainder of the molecule via an oxygen atom.
The term "pharmaceutically acceptable" is used to specify that an object (for example a salt, dosage form or excipient) is suitable for use in patients. An example list of pharmaceutically acceptable salts can be found in the Handbook of Pharmaceutical Salts: Properties, Selection and Use, P. H. Stahl and C. G. Wermuth, editors, Weinheim/Zurich:Wiley-VCH/VHCA, 2002. A suitable pharmaceutically acceptable salt of a compound of Formula (I) is, for example, an acid-addition salt. An acid addition salt of a compound of Formula (I) may be formed by bringing the compound into contact with a suitable inorganic or organic acid under conditions known to the skilled person. An acid addition salt may for example be formed using an inorganic acid selected from the group consisting of hydrochloric acid, hydrobromic acid, sulphuric acid and phosphoric acid. An acid addition salt may also be formed using an organic acid selected from the group consisting of trifluoro acetic acid, citric acid, maleic acid, oxalic acid, acetic acid, formic acid, benzoic acid, fumaric acid, succinic acid, tartaric acid, lactic acid, pyruvic acid, methanesulfonic acid, benzenesulfonic acid and /?ara-toluenesulfonic acid.
Therefore, in one embodiment there is provided a compound of Formula (I) or a pharmaceutically acceptable salt thereof, where the pharmaceutically acceptable salt is a hydrochloric acid, hydrobromic acid, sulphuric acid, phosphoric acid, trifluoroacetic acid, citric acid, maleic acid, oxalic acid, acetic acid, formic acid, benzoic acid, fumaric acid, succinic acid, tartaric acid, lactic acid, pyruvic acid, methanesulfonic acid, benzenesulfonic acid or /?ara-toluenesulfonic acid salt. In one embodiment there is provided a compound of Formula (I) or a pharmaceutically acceptable salt thereof, where the pharmaceutically acceptable salt is a methanesulfonic acid salt. In one embodiment there is provided a compound of Formula (I) or a pharmaceutically acceptable salt thereof, where the pharmaceutically acceptable salt is a mono-methanesulfonic acid salt, i.e. the stoichiometry of the compound of the compound of Formula (I) to methanesulfonic acid is 1 : 1.
In one embodiment there is provided, a compound of Formula (I), or a
pharmaceutically acceptable salt thereof.
In one embodiment there is provided a compound of Formula (I).
In one embodiment there is provided a pharmaceutically acceptable salt of a compound of Formula (I).
Compounds and salts described in this specification may exist in solvated forms and unsolvated forms. For example, a solvated form may be a hydrated form, such as a hemi-hydrate, a mono-hydrate, a di-hydrate, a tri-hydrate or an alternative quantity thereof. All such solvated and unsolvated forms of compounds of Formula (I) are encompassed herein, particularly to the extent that such forms possess ATM kinase inhibitory activity, as for example measured using the tests described herein.
Atoms of the compounds and salts described in this specification may exist as their isotopes. All compounds of Formula (I) where an atom is replaced by one or more of its isotopes (for example a compound of Formula (I) where one or more carbon atom is an UC or 13C carbon isotope, or where one or more hydrogen atoms is a 2H or 3H isotope) are encompassed herein.
Compounds of the application may exist in one or more geometrical, optical, enantiomeric, diastereomeric and tautomeric forms, including, but not limited to, cis- and trans-forms, E- and Z-forms, R-, S- and meso-forms, keto-, and enol-forms. Unless otherwise stated a reference to a particular compound includes all such isomeric forms, including racemic and other mixtures thereof. Where appropriate such isomers can be separated from their mixtures by the application or adaptation of known methods (e.g. chromatographic techniques and recrystallisation techniques). Where appropriate such isomers can be prepared by the application or adaptation of known methods (e.g.
asymmetric synthesis).
For exam ncludes the racemic form of this fragment
as well as the R
Figure imgf000025_0001
and S enantiomers thereof. Similarly, the
fragment
Figure imgf000025_0002
includes the cis and trans forms thereof.
In an embodiment there is provided a compound of Formula (I), or a
pharmaceutically acceptable salt thereof, which is a single optical isomer being in an enantiomeric excess (%ee) of > 95%, > 98% or > 99%. In one embodiment, the single optical isomer is present in an enantiomeric excess (%ee) of > 99%.
Compounds and salts described in this specification may be crystalline, and may exhibit one or more crystalline forms. Any crystalline or amorphous form of a compound of Formula (I), or mixture of such forms, which possesses ATM kinase inhibitory activity are encompassed herein.
It is generally known that crystalline materials may be characterised using conventional techniques such as X-Ray Powder Diffraction (XRPD), Differential Scanning Calorimetry (DSC), Thermal Gravimetric Analysis (TGA), Diffuse Reflectance Infrared Fourier Transform (DRIFT) spectroscopy, Near Infrared (NIR) spectroscopy, solution and/or solid state nuclear magnetic resonance spectroscopy. The water content of crystalline materials may be determined by Karl Fischer analysis.
As a result of their ATM kinase inhibitory activity, the compounds of Formula (I), and pharmaceutically acceptable salts thereof are expected to be useful in therapy, for example in the treatment of diseases or medical conditions mediated at least in part by ATM kinase, including cancer.
Where "cancer" is mentioned, this includes both non-metastatic cancer and also metastatic cancer, such that treating cancer involves treatment of both primary tumours and also tumour metastases.
"ATM kinase inhibitory activity" refers to a decrease in the activity of ATM kinase as a direct or indirect response to the presence of a compound of Formula (I), or pharmaceutically acceptable salt thereof, relative to the activity of ATM kinase in the absence of compound of Formula (I), or pharmaceutically acceptable salt thereof. Such a decrease in activity may be due to the direct interaction of the compound of Formula (I), or pharmaceutically acceptable salt thereof with ATM kinase, or due to the interaction of the compound of Formula (I), or pharmaceutically acceptable salt thereof with one or more other factors that in turn affect ATM kinase activity. For example, the compound of Formula (I), or pharmaceutically acceptable salt thereof may decrease ATM kinase by directly binding to the ATM kinase, by causing (directly or indirectly) another factor to decrease ATM kinase activity, or by (directly or indirectly) decreasing the amount of ATM kinase present in the cell or organism. The term "therapy" is intended to have its normal meaning of dealing with a disease in order to entirely or partially relieve one, some or all of its symptoms, or to correct or compensate for the underlying pathology. The term "therapy" also includes "prophylaxis" unless there are specific indications to the contrary. The terms "therapeutic" and "therapeutically" should be interpreted in a corresponding manner.
The term "prophylaxis" is intended to have its normal meaning and includes primary prophylaxis to prevent the development of the disease and secondary prophylaxis whereby the disease has already developed and the patient is temporarily or permanently protected against exacerbation or worsening of the disease or the development of new symptoms associated with the disease.
The term "treatment" is used synonymously with "therapy". Similarly the term "treat" can be regarded as "applying therapy" where "therapy" is as defined herein.
In one embodiment there is provided a compound of Formula (I), or a
pharmaceutically acceptable salt thereof, for use in therapy.
In one embodiment there is provided a compound of Formula (I), or a
pharmaceutically acceptable salt thereof, for use in the treatment of a disease mediated by ATM kinase.
In one embodiment, said disease mediated by ATM kinase is cancer. In one embodiment, said cancer is selected from the group consisting of colorectal cancer, glioblastoma, gastric cancer, ovarian cancer, diffuse large B-cell lymphoma, chronic lymphocytic leukaemia, acute myeloid leukaemia, head and neck squamous cell carcinoma, breast cancer, prostate cancer, hepatocellular carcinoma, small cell lung cancer and non-small cell lung cancer. In one embodiment, said cancer is selected from the group consisting of colorectal cancer, glioblastoma, gastric cancer, ovarian cancer, diffuse large B-cell lymphoma, chronic lymphocytic leukaemia, head and neck squamous cell carcinoma, breast cancer, prostate cancer and lung cancer. In one embodiment, said cancer is colorectal cancer. In another embodiment, said cancer is gastric cancer.
In one embodiment there is provided a compound of Formula (I), or a
pharmaceutically acceptable salt thereof, for use in the treatment of cancer.
In one embodiment there is provided a compound of Formula (I), or a
pharmaceutically acceptable salt thereof, for use in the treatment of Huntington's disease. In one embodiment there is provided a compound of Formula (I), or a
pharmaceutically acceptable salt thereof, for use as a neuroprotective agent.
A "neuroprotective agent" is an agent that aids relative preservation of neuronal structure and/or function.
In one embodiment there is provided the use of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of a disease mediated by ATM kinase. In one embodiment, said disease mediated by ATM kinase is cancer.
In one embodiment there is provided the use of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of cancer.
In one embodiment there is provided the use of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for the treatment of Huntington's disease.
In one embodiment there is provided the use of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, for the manufacture of a medicament for use as a neuroprotective agent.
In one embodiment there is provided a method for treating a disease in which inhibition of ATM kinase is beneficial in a warm-blooded animal in need of such treatment, which comprises administering to said warm-blooded animal a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof. In one embodiment, said disease is cancer.
In any embodiment, a disease in which inhibition of ATM kinase is beneficial may be Huntington' disease.
In one embodiment there is provided a method of treatment for aiding relative preservation of neuronal structure and/or function in a warm-blooded animal in need of such treatment, which comprises administering to said warm-blooded animal a
therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof.
The term "therapeutically effective amount" refers to an amount of a compound of Formula (I) as described in any of the embodiments herein which is effective to provide "therapy" in a subject, or to "treat" a disease or disorder in a subject. In the case of cancer, the therapeutically effective amount may cause any of the changes observable or measurable in a subject as described in the definition of "therapy", "treatment" and "prophylaxis" above. For example, the effective amount can reduce the number of cancer or tumour cells; reduce the overall tumour size; inhibit or stop tumour cell infiltration into peripheral organs including, for example, the soft tissue and bone; inhibit and stop tumour metastasis; inhibit and stop tumour growth; relieve to some extent one or more of the symptoms associated with the cancer; reduce morbidity and mortality; improve quality of life; or a combination of such effects. An effective amount may be an amount sufficient to decrease the symptoms of a disease responsive to inhibition of ATM kinase activity. For cancer therapy, efficacy in-vivo can, for example, be measured by assessing the duration of survival, time to disease progression (TTP), the response rates (RR), duration of response, and/or quality of life. As recognized by those skilled in the art, effective amounts may vary depending on route of administration, excipient usage, and co-usage with other agents. For example, where a combination therapy is used, the amount of the compound of formula (I) or pharmaceutcially acceptable salt described in this specification and the amount of the other pharmaceutically active agent(s) are, when combined, jointly effective to treat a targeted disorder in the animal patient. In this context, the combined amounts are in a "therapeutically effective amount" if they are, when combined, sufficient to decrease the symptoms of a disease responsive to inhibition of ATM activity as described above.
Typically, such amounts may be determined by one skilled in the art by, for example, starting with the dosage range described in this specification for the compound of formula (I) or pharmaceutcially acceptable salt thereof and an approved or otherwise published dosage range(s) of the other pharmaceutically active compound(s).
"Warm-blooded animals" include, for example, humans.
In one embodiment there is provided a method for treating cancer in a
warm-blooded animal in need of such treatment, which comprises administering to said warm-blooded animal a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof.
In any embodiment where cancer is mentioned in a general sense, said cancer may be selected from the group consisting of colorectal cancer, glioblastoma, gastric cancer, ovarian cancer, diffuse large B-cell lymphoma, chronic lymphocytic leukaemia, acute myeloid leukaemia, head and neck squamous cell carcinoma, breast cancer, prostate cancer, hepatocellular carcinoma, small cell lung cancer and non-small cell lung cancer. Said cancer may also be selected from the group consisting of colorectal cancer, glioblastoma, gastric cancer, ovarian cancer, diffuse large B-cell lymphoma, chronic lymphocytic leukaemia, head and neck squamous cell carcinoma, breast cancer, prosate cancer and lung cancer.
In any embodiment where cancer is mentioned in a general sense the following embodiments may apply:
In one embodiment the cancer is colorectal cancer or gastric cancer.
In one embodiment the cancer is colorectal cancer.
In one embodiment the cancer is glioblastoma.
In one embodiment the cancer is gastric cancer.
In one embodiment the cancer is oesophageal cancer.
In one embodiment the cancer is ovarian cancer.
In one embodiment the cancer is endometrial cancer.
In one embodiment the cancer is cervical cancer.
In one embodiment the cancer is diffuse large B-cell lymphoma.
In one embodiment the cancer is chronic lymphocytic leukaemia.
In one embodiment the cancer is acute myeloid leukaemia.
In one embodiment the cancer is head and neck squamous cell carcinoma.
In one embodment the cancer is prostate cancer.
In one embodiment the cancer is breast cancer. In one embodiment the cancer is triple negative breast cancer.
"Triple negative breast cancer" is any breast cancer that does not express the genes for the oestrogen receptor, progesterone receptor and Her2/neu.
In one embodiment the cancer is hepatocellular carcinoma.
In one embodiment the cancer is lung cancer. In one embodiment the lung cancer is small cell lung cancer. In one embodiment the lung cancer is non-small cell lung cancer.
In one embodiment the cancer is metastatic cancer. In one embodiment the metastatic cancer comprises metastases of the central nervous system. In one embodiment the metastases of the central nervous system comprise brain metastases. In one
embodiment the metastases of the central nervous system comprise leptomeningeal metastases.
"Leptomeningeal metastases" occur when cancer spreads to the meninges, the layers of tissue that cover the brain and the spinal cord. Metastases can spread to the meninges through the blood or they can travel from brain metastases, carried by the cerebrospinal fluid (CSF) that flows through the meninges. In one embodiment the cancer is non-metastatic cancer.
The anti-cancer treatment described in this specification may be useful as a sole therapy, or may involve, in addition to administration of the compound of Formula (I), conventional surgery, radiotherapy or chemotherapy; or a combination of such additional therapies. Such conventional surgery, radiotherapy or chemotherapy may be administered simultaneously, sequentially or separately to treatment with the compound of Formula (I).
Radiotherapy may include one or more of the following categories of therapy: i. External radiation therapy using electromagnetic radiation, and intraoperative radiation therapy using electromagnetic radiation;
ii. Internal radiation therapy or brachytherapy; including interstitial radiation therapy or intraluminal radiation therapy; or
iii. Systemic radiation therapy, including but not limited to iodine 131 and strontium 89.
Therefore, in one embodiment there is provided a compound of Formula (I), or a pharmaceutically acceptable salt thereof, and radiotherapy, for use in the treatment of cancer. In one embodiment the cancer is glioblastoma. In one embodiment, the cancer is metastatic cancer. In one embodiment the metastatic cancer comprises metastases of the central nervous system. In one embodiment the metastases of the central nervous system comprise brain metastases. In one embodiment the metastases of the central nervous system comprise leptomeningeal metastases.
In one embodiment there is provided a compound of Formula (I), or a
pharmaceutically acceptable salt thereof, for use in the treatment of cancer, where the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered in combination with radiotherapy. In one embodiment the cancer is glioblastoma. In one embodiment, the cancer is metastatic cancer. In one embodiment the metastatic cancer comprises metastases of the central nervous system. In one embodiment the metastases of the central nervous system comprise brain metastases. In one embodiment the metastases of the central nervous system comprise leptomeningeal metastases.
In one embodiment there is provided a compound of Formula (I), or a
pharmaceutically acceptable salt thereof, and radiotherapy, for use in the simultaneous, separate or sequential treatment of cancer.
In one embodiment there is provided a compound of Formula (I), or a
pharmaceutically acceptable salt thereof, for use in the treatment of cancer, where the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered simultaneously, separately or sequentially with radiotherapy.
In one embodiment there is provided a method of treating cancer in a warmblooded animal who is in need of such treatment, which comprises administering to said warm-blooded animal a compound of Formula (I), or a pharmaceutically acceptable salt thereof and radiotherapy. In one embodiment, the compound of Formula (I), or a pharmaceutically acceptable salt thereof, and radiotherapy are jointly effective in producing an anti-cancer effect.
In one embodiment there is provided a method of treating cancer in a warmblooded animal who is in need of such treatment, which comprises administering to said warm-blooded animal a compound of Formula (I), or a pharmaceutically acceptable salt thereof and simultaneously, separately or sequentially administering radiotherapy. In one embodiment, the compound of Formula (I), or a pharmaceutically acceptable salt thereof, and radiotherapy are jointly effective in producing an anti-cancer effect. In one
embodiment the cancer is glioblastoma. In one embodiment, the cancer is metastatic cancer. In one embodiment the metastatic cancer comprises metastases of the central nervous system. In one embodiment the metastases of the central nervous system comprise brain metastases. In one embodiment the metastases of the central nervous system comprise leptomeningeal metastases.
In any embodiment comprising a compound of Formula (I), or a pharmaceutically acceptable salt thereof and radiotherapy, where cancer is mentioned in a general sense, said cancer may be selected from the group consisting of glioblastoma, lung cancer (for example small cell lung cancer or non-small cell lung cancer), breast cancer (for example triple negative breast cancer), head and neck squamous cell carcinoma, oesophageal cancer, cervical cancer and endometrial cancer. In one embodiment the cancer is glioblastoma. In one embodiment, the cancer is metastatic cancer. In one embodiment the metastatic cancer comprises metastases of the central nervous system. In one embodiment the metastases of the central nervous system comprise brain metastases. In one
embodiment the metastases of the central nervous system comprise leptomeningeal metastases.
In any embodiment the radiotherapy is selected from the group consisting of one or more of the categories of radiotherapy listed under points (i) - (iii) above.
Chemotherapy may include one or more of the following categories of anti -tumour substance:
i. Antineoplastic agents and combinations thereof, such as DNA alkylating agents (for example cisplatin, oxaliplatin, carboplatin, cyclophosphamide, nitrogen mustards like ifosfamide, bendamustine, melphalan, chlorambucil, busulphan, temozolamide and nitrosoureas like carmustine); antimetabolites (for example gemcitabine and antifolates such as fluoropyrimidines like 5-fluorouracil and tegafur, raltitrexed, methotrexate, cytosine arabinoside, and hydroxyurea); anti-tumour antibiotics (for example anthracyclines like adriamycin, bleomycin, doxorubicin, liposomal doxorubicin, pirarubicin, daunomycin, valrubicin, epirubicin, idarubicin, mitomycin-C, dactinomycin, amrubicin and mithramycin); antimitotic agents (for example vinca alkaloids like vincristine, vinblastine, vindesine and vinorelbine and taxoids like taxol and taxotere and polokinase inhibitors); and topoisomerase inhibitors (for example epipodophyllotoxins like etoposide and teniposide, amsacrine, irinotecan, topotecan and camptothecin); inhibitors of DNA repair mechanisms such as CHK kinase; DNA-dependent protein kinase inhibitors (such as VX-984, M3814, KU-0060648); inhibitors of poly (ADP-ribose) polymerase (PARP inhibitors, including olaparib); and Hsp90 inhibitors such as tanespimycin and retaspimycin, inhibitors of ATR kinase (such as AZD6738); and inhibitors of WEE 1 kinase (such as
AZD1775/MK-1775);
Antiangio genie agents such as those that inhibit the effects of vascular endothelial growth factor, for example the anti-vascular endothelial cell growth factor antibody bevacizumab and for example, a VEGF receptor tyrosine kinase inhibitor such as vandetanib (ZD6474), sorafenib, vatalanib (PTK787), sunitinib (SU11248), axitinib (AG-013736), pazopanib (GW 786034) and cediranib (AZD2171); compounds such as those disclosed in International Patent Applications W097/22596, WO 97/30035, WO 97/32856 and WO 98/13354; and compounds that work by other mechanisms (for example linomide, inhibitors of integrin ανβ3 function and angiostatin), or inhibitors of angiopoietins and their receptors (Tie-1 and Tie-2), inhibitors of PLGF, inhibitors of delta- like ligand (DLL-4);
Immunotherapy approaches, including for example ex-vivo and in-vivo approaches to increase the immunogenicity of patient tumour cells, such as transfection with cytokines such as interleukin 2, interleukin 4 or granulocyte -macrophage colony stimulating factor; approaches to decrease T-cell anergy or regulatory T-cell function; approaches that enhance T-cell responses to tumours, such as blocking antibodies to CTLA4 (for example ipilimumab and tremelimumab), B7H1, PD-1 (for example BMS-936558 or AMP-514), PD-L1 (for example MEDI4736) and agonist antibodies to CD 137; approaches using transfected immune cells such as cytokine -transfected dendritic cells; approaches using cytokine-transfected tumour cell lines, approaches using antibodies to tumour associated antigens, and antibodies that deplete target cell types (e.g., unconjugated anti-CD20 antibodies such as Rituximab, radiolabeled anti-CD20 antibodies Bexxar and Zevalin, and anti-CD54 antibody Campath); approaches using anti-idiotypic antibodies;
approaches that enhance Natural Killer cell function; and approaches that utilize antibody-toxin conjugates (e.g. anti-CD33 antibody Mylotarg); immunotoxins such as moxetumumab pasudotox; agonists of toll-like receptor 7 or toll-like receptor 9; Efficacy enhancers, such as leucovorin.
Therefore, in one embodiment there is provided a compound of Formula (I), or a armaceutically acceptable salt thereof, and at least one additional anti -tumour substance, for use in the treatment of cancer. In one embodiment there is provided a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, where the compound of Formula (I), or a pharmaceutically acceptable salt thereof is administered in combination with an additional anti-tumour substance. In one embodiment there is one additional anti-tumour substance. In one embodiment there are two additional anti-tumour substances. In one embodiment there are three or more additional anti-tumour substances.
In one embodiment there is provided a compound of Formula (I), or a
pharmaceutically acceptable salt thereof, and at least one additional anti -tumour substance for use in the simultaneous, separate or sequential treatment of cancer. In one embodiment there is provided a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, where the compound of Formula (I), or a
pharmaceutically acceptable salt thereof, is administered simultaneously, separately or sequentially with an additional anti-tumour substance.
In one embodiment there is provided a method of treating cancer in a warmblooded animal who is in need of such treatment, which comprises administering to said warm-blooded animal a compound of Formula (I), or a pharmaceutically acceptable salt thereof and at least one additional anti-tumour substance, where the amounts of the compound of Formula (I), or a pharmaceutically acceptable salt thereof, and the additional anti -tumour substance are jointly effective in producing an anti-cancer effect.
In one embodiment there is provided a method of treating cancer in a warmblooded animal who is in need of such treatment, which comprises administering to said warm-blooded animal a compound of Formula (I), or a pharmaceutically acceptable salt thereof, and simultaneously, separately or sequentially administering at least one additional anti -tumour substance to said warm-blooded animal, where the amounts of the compound of Formula (I), or pharmaceutically acceptable salt thereof, and the additional anti-tumour substance are jointly effective in producing an anti-cancer effect.
In any embodiment the additional anti-tumour substance is selected from the group consisting of one or more of the anti-tumour substances listed under points (i) - (iv) above.
In one embodiment there is provided a compound of Formula (I), or a
pharmaceutically acceptable salt thereof, and at least one anti-neoplastic agent for use in the treatment of cancer. In one embodiment there is provided a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, where the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered in combination with at least one anti-neoplastic agent. In one embodiment the anti-neoplastic agent is selected from the list of antineoplastic agents in point (i) above.
In one embodiment there is provided a compound of Formula (I), or a
pharmaceutically acceptable salt thereof, and at least one anti-neoplastic agent for use in the simultaneous, separate or sequential treatment of cancer. In one embodiment there is provided a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, where the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered simultaneously, separately or sequentially with at least one anti-neoplastic agent. In one embodiment the antineoplastic agent is selected from the list of antineoplastic agents in point (i) above.
In one embodiment there is provided a compound of Formula (I), or a
pharmaceutically acceptable salt thereof, and at least one additional anti -tumour substance selected from the group consisting of cisplatin, oxaliplatin, carboplatin, valrubicin, idarubicin, doxorubicin, pirarubicin, irinotecan, topotecan, amrubicin, epirubicin, etoposide, mitomycin, bendamustine, chlorambucil, cyclophosphamide, ifosfamide, carmustine, melphalan, bleomycin, olaparib, MEDI4736, AZD1775 and AZD6738, for use in the treatment of cancer.
In one embodiment there is provided a compound of Formula (I), or a
pharmaceutically acceptable salt thereof, and at least one additional anti -tumour substance selected from the group consisting of cisplatin, oxaliplatin, carboplatin, doxorubicin, pirarubicin, irinotecan, topotecan, amrubicin, epirubicin, etoposide, mitomycin, bendamustine, chlorambucil, cyclophosphamide, ifosfamide, carmustine, melphalan, bleomycin, olaparib, AZD1775 and AZD6738, for use in the treatment of cancer.
In one embodiment there is provided a compound of Formula (I), or a
pharmaceutically acceptable salt thereof, for use in the treatment of cancer, where the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered in combination with at least one additional anti-tumour substance selected from the group consisting of cisplatin, oxaliplatin, carboplatin, valrubicin, idarubicin, doxorubicin, pirarubicin, irinotecan, topotecan, amrubicin, epirubicin, etoposide, mitomycin, bendamustine, chlorambucil, cyclophosphamide, ifosfamide, carmustine, melphalan, bleomycin, olaparib, MEDI4736, AZD1775 and AZD6738.
In one embodiment there is provided a compound of Formula (I), or a
pharmaceutically acceptable salt thereof, and at least one additional anti -tumour substance selected from the group consisting of doxorubicin, irinotecan, topotecan, etoposide, mitomycin, bendamustine, chlorambucil, cyclophosphamide, ifosfamide, carmustine, melphalan, bleomycin and olaparib for use in the treatment of cancer.
In one embodiment there is provided a compound of Formula (I), or a
pharmaceutically acceptable salt thereof, for use in the treatment of cancer, where the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered in combination with at least one additional anti-tumour substance selected from the group consisting of doxorubicin, irinotecan, topotecan, etoposide, mitomycin, bendamustine, chlorambucil, cyclophosphamide, ifosfamide, carmustine, melphalan, bleomycin and olaparib.
In one embodiment there is provided a compound of Formula (I), or a
pharmaceutically acceptable salt thereof, and at least one additional anti -tumour substance selected from the group consisting of doxorubicin, irinotecan, topotecan, etoposide, mitomycin, bendamustine, chlorambucil, cyclophosphamide, ifosfamide, carmustine, melphalan and bleomycin, for use in the treatment of cancer.
In one embodiment there is provided a compound of Formula (I), or a
pharmaceutically acceptable salt thereof, for use in the treatment of cancer, where the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered in combination with at least one additional anti-tumour substance selected from the group consisting of doxorubicin, irinotecan, topotecan, etoposide, mitomycin, bendamustine, chlorambucil, cyclophosphamide, ifosfamide, carmustine, melphalan and bleomycin.
In one embodiment there is provided a compound of Formula (I), or a
pharmaceutically acceptable salt thereof, for use in the treatment of cancer, where the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered in combination with at least one additional anti-tumour substance selected from the group consisting of doxorubicin, pirarubicin, amrubicin and epirubicin. In one embodiment the cancer is acute myeloid leukaemia. In one embodiment the cancer is breast cancer (for example triple negative breast cancer). In one embodiment the cancer is hepatocellular carcinoma.
In one embodiment there is provided a compound of Formula (I), or a
pharmaceutically acceptable salt thereof, and irinotecan, for use in the treatment of cancer. In one embodiment there is provided a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, where the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered in combination with irinotecan. In one embodiment the cancer is colorectal cancer.
In one embodiment there is provided a compound of Formula (I), or a
pharmaceutically acceptable salt thereof, and FOLFIRI, for use in the treatment of cancer. In one embodiment there is provided a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, where the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered in combination with FOLFIRI. In one embodiment the cancer is colorectal cancer.
FOLFIRI is a dosage regime involving a combination of leucovorin, 5-fluorouracil and irinotecan.
In one embodiment there is provided a compound of Formula (I), or a
pharmaceutically acceptable salt thereof, for use in the treatment of cancer, where the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered in combination with olaparib. In one embodiment the cancer is selected from gastric cancer, triple negative breast cancer, prostate cancer, small cell lung cancer and ovarian cancer. In one embodiment the cancer is gastric cancer. In one embodiment the cancer is triple negative breast cancer. In one embodiment the cancer is prostate cancer. In one embodiment the cancer is small cell lung cancer. In one embodiment the cancer is ovarian cancer.
In one embodiment there is provided a compound of Formula (I), or a
pharmaceutically acceptable salt thereof, for use in the treatment of cancer, where the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered in combination with topotecan. In one embodiment the cancer is small cell lung cancer. In one embodiment there is provided a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer, where the compound of Formula (I), or a pharmaceutically acceptable salt thereof, is administered in combination with immunotherapy. In one embodiment the immunotherapy is one or more of the agents listed under point (iii) above. In one embodiment the immunotherapy is an anti-PD-Ll antibody (for example MEDI4736).
According to a further embodiment there is provided a kit comprising:
a) A compound of formula (I), or a pharmaceutically acceptable salt thereof, in a first unit dosage form;
b) A further additional anti -tumour substance in a further unit dosage form;
c) Container means for containing said first and further unit dosage forms; and optionally
d) Instructions for use. In one embodiment the anti-tumour substance comprises an anti-neoplastic agent.
In any embodiment where an anti-neoplastic agent is mentioned, the anti-neoplastic agent is one or more of the agents listed under point (i) above.
The compounds of Formula (I), and pharmaceutically acceptable salts thereof, may be administered as pharmaceutical compositions, comprising one or more pharmaceutically acceptable excipients.
Therefore, in one embodiment there is provided a pharmaceutical composition comprising a compound of Formula (I), or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient.
The excipient(s) selected for inclusion in a particular composition will depend on factors such as the mode of administration and the form of the composition provided. Suitable pharmaceutically acceptable excipients are well known to persons skilled in the art and are described, for example, in the Handbook of Pharmaceutical Excipients, Sixth edition, Pharmaceutical Press, edited by Rowe, Ray C; Sheskey, Paul J; Quinn, Marian. Pharmaceutically acceptable excipients may function as, for example, adjuvants, diluents, carriers, stabilisers, flavourings, colorants, fillers, binders, disintegrants, lubricants, glidants, thickening agents and coating agents. As persons skilled in the art will appreciate, certain pharmaceutically acceptable excipients may serve more than one function and may serve alternative functions depending on how much of the excipient is present in the composition and what other excipients are present in the composition.
The pharmaceutical compositions may be in a form suitable for oral use (for example as tablets, lozenges, hard or soft capsules, aqueous or oily suspensions, emulsions, dispersible powders or granules, syrups or elixirs), for topical use (for example as creams, ointments, gels, or aqueous or oily solutions or suspensions), for administration by inhalation (for example as a finely divided powder or a liquid aerosol), for administration by insufflation (for example as a finely divided powder) or for parenteral administration (for example as a sterile aqueous or oily solution for intravenous, subcutaneous, intramuscular or intramuscular dosing), or as a suppository for rectal dosing. The compositions may be obtained by conventional procedures well known in the art.
Compositions intended for oral use may contain additional components, for example, one or more colouring, sweetening, flavouring and/or preservative agents.
The compound of Formula (I) will normally be administered to a warm-blooded animal at a unit dose within the range 2.5-5000 mg/m2 body area of the animal, or approximately 0.05-100 mg/kg, and this normally provides a therapeutically-effective dose. A unit dose form such as a tablet or capsule will usually contain, for example 0.1-250 mg of active ingredient. The daily dose will necessarily be varied depending upon the host treated, the particular route of administration, any therapies being co-administered, and the severity of the illness being treated. Accordingly the practitioner who is treating any particular patient may determine the optimum dosage.
The pharmaceutical compositions described herein comprise compounds of Formula (I), or a pharmaceutically acceptable salt thereof, and are therefore expected to be useful in therapy.
As such, in one embodiment there is provided a pharmaceutical composition for use in therapy, comprising a compound of Formula (I), or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient.
In one embodiment there is provided a pharmaceutical composition for use in the treatment of a disease in which inhibition of ATM kinase is beneficial, comprising a compound of Formula (I), or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient. In one embodiment there is provided a pharmaceutical composition for use in the treatment of cancer, comprising a compound of Formula (I), or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient.
In one embodiment there is provided a pharmaceutical composition for use in the treatment of a cancer in which inhibition of ATM kinase is beneficial, comprising a compound of Formula (I), or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient.
In one embodiment there is provided a pharmaceutical composition for use in the treatment of colorectal cancer, glioblastoma, gastric cancer, ovarian cancer, diffuse large B-cell lymphoma, chronic lymphocytic leukaemia, acute myeloid leukaemia, head and neck squamous cell carcinoma, breast cancer, prostate cancer, hepatocellular carcinoma, small cell lung cancer or non-small cell lung cancer, comprising a compound of Formula (I), or a pharmaceutically acceptable salt thereof, and at least one pharmaceutically acceptable excipient.
EXAMPLES
The various embodiments of the specification are illustrated by the following Examples. The specification is not to be interpreted as being limited to the Examples. During the preparation of the Examples, generally:
i. Operations were carried out at ambient temperature, i.e. in the range of about 17 to 30°C and under an atmosphere of an inert gas such as nitrogen unless otherwise stated;
ii. Evaporations were carried out by rotary evaporation or utilising Genevac
equipment in vacuo and work-up procedures were carried out after removal of residual solids by filtration;
iii. Flash chromatography purifications were performed on suitable automated systems.
Examples of such systems include Armen Glider Flash: Spot II Ultimate (Armen Instrument, Saint-Ave, France) or automated Presearch combiflash companions using prepacked Merck normal phase Si60 silica cartridges (granulometry : 15-40 or 40-63 μηι) obtained from Merck, Darmstad, Germany, silicycle silica cartridges or graceresolv silica cartridges;
Preparative chromatography was performed on a suitable system. Examples of suitable systems include a Waters instrument (600/2700 or 2525) fitted with a ZMD or ZQ ESCi mass spectrometers and a Waters X-Terra or a Waters X-Bridge or a Waters SunFire reverse-phase column (C-18, 5 microns silica, 19 mm or 50 mm diameter, 100 mm length, flow rate of 40 mL / minute) using decreasingly polar mixtures of water (containing 1% ammonia) and acetonitrile or decreasingly polar mixtures of water (containing 0.1% formic acid) and acetonitrile as eluents;
Yields, where present, are not necessarily the maximum attainable;
Structures of end-products of Formula (I) were confirmed by nuclear magnetic resonance (NMR) spectroscopy, with NMR chemical shift values measured on the delta scale. Proton magnetic resonance spectra were determined using a Bruker advance 700 (700MHz), Bruker Avance 500 (500 MHz), Bruker 400 (400 MHz) or Bruker 300 (300 MHz) instrument; 19F NMR were determined at 282 MHz or 376 MHz; 13C NMR were determined at 75 MHz or 100 MHz; measurements were taken at around 20 - 30° C unless otherwise specified; the following abbreviations have been used: s, singlet; d, doublet; t, triplet; q, quartet; m, multiplet; dd, doublet of doublets; ddd, doublet of doublet of doublet; dt, doublet of triplets; bs, broad signal;
End-products of Formula (I) were also characterised by mass spectroscopy following liquid chromatography (LCMS); LCMS was carried out using an Waters Alliance HT (2790 & 2795) fitted with a Waters ZQ ESCi or ZMD ESCi mass spectrometer and an X Bridge 5μιη C-18 column (2.1 x 50 mm) at a flow rate of 2.4 mL/min, using a solvent system of 95% A + 5% C to 95% B + 5% C over 4 minutes, where A = water, B = methanol, C = 1 : 1 methanol: water (containing 0.2% ammonium carbonate); or by using a Shimadzu UFLC or UHPLC coupled with DAD detector, ELSD detector and 2020 EV mass spectrometer (or equivalent) fitted with a Phenomenex Gemini -NX CI 8 3.0x50 mm, 3.0 μΜ column or equivalent (basic conditions) or a Shim pack XR - ODS 3.0 x 50 mm, 2.2 μΜ column or Waters BEH CI 8 2.1 x 50 mm, 1.7 μΜ column or equivalent using a solvent system of 95% D + 5% E to 95% E + 5% D over 4 minutes, where D = water (containing 0.05% TFA), E = Acetonitrile (containing 0.05% TFA) (acidic conditions) or a solvent system of 90% F + 10% G to 95% G + 5% F over 4 minutes, where F = water (containing 6.5 mM ammonium hydrogen carbonate and adjusted to pHIO by addition of ammonia), G = Acetonitrile (basic conditions); viii. Intermediates were not generally fully characterised and purity was assessed by thin layer chromatographic, mass spectral, HPLC and/or NMR analysis;
ix. Supercritical fluid chromatography was carried out using a suitable system such as either a Sepiatec Prep SFCIOO machine or a Waters SFC Prep 100 machine;
x. The following abbreviations have been used: h = hour(s); r.t. = room temperature (~18-25°C); cone. = concentrated; FCC = flash column chromatography using silica; DCM = dichloromethane; DIPEA = diisopropylethylamine; DMA = N,N- dimethylacetamide; DMF = N,N-dimethylformamide; DMSO = dimethylsulfoxide; Et20 = diethyl ether; EtOAc = ethyl acetate; EtOH = ethanol; K2C03 = potassium carbonate; MeOH = methanol; MeCN = acetonitrile; MTBE =
Methyltertbutylether; MgS04 = anhydrous magnesium sulphate; Na2S04 = anhydrous sodium sulphate; THF = tetrahydrofuran; sat. = saturated aqueous solution; SFC = supercritical fluid chromatography; and
xi. IUPAC names were generated using ACD Name 14 (Advanced Chemistry
Development, Inc., Toronto, Ontario, Canada) or OEChem 2 (OpenEye, Santa Fe, NM 87508, USA) software. In cases where the software was unable to distinguish between cis/trans isomers the cis or trans definitition was added manually to the software-generated IUPAC name.
Example 1 : 8- [6- [3-(Dimethylamino)propoxy] -3-pyridyl] -3-methyl-l- tetrahydropyran-4-yl-imidazo [4,5-c] cinnolin-2-one
Figure imgf000043_0001
8-Bromo-3-methyl-l etrahydropyran-4-yl-imidazo[4,5-c]cinnolin-2-one (180 mg, 0.50 mmol) was added to A ,N-dimethyl-3-[[5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2- pyridyl]oxy]propan-l -amine (182 mg, 0.59 mmol), CS2CO3 (484 mg, 1.49 mmol) and Pd(Pli3P)4 (57.3 mg, 0.05 mmol) in 1,4-dioxane (5 mL) and water (1 mL) under nitrogen. The resulting mixture was stirred at 80 °C for 2 hours, the mixture concentrated and purified by preparative HPLC to afford the desired material (20 mg) as a yellow solid. NMR Spectrum: lH NMR (300MHz, DMSO) δ 1.8-2.0 (4H, m), 2.1-2.2 (6H, m), 2.3-2.4 (2H, m), 2.5-2.7 (2H, m), 3.5-3.7 (2H, m), 4.0-4.1 (2H,m), 4.3-4.4 (2H, m), 5.0-5.2 (lH,m), 6.95-7.05 (1H, m), 8.0-8.8 (5H, m).
Mass Spectrum: m/z (ES+)[M+H]+ = 463
The following compounds were synthesised in an analogous fashion using N,N-dimethyl- 3-[[5-(4,4,5,5-tetramethyl-l ,3,2-dioxaborolan-2-yl)-2-pyridyl]oxy]propan-l -amine and the appropriate bromo intermediate.
Figure imgf000044_0001
Example 2: Isolated as a formic acid salt NMR Spectrum: lH NMR (300MHz, DMSO) δ 1.8-2.0 (2H, m), 2.1-2.2 (6H, m), 2.35-2.45 (2H, m), 2.7-3.1 (4H, m), 3.2 (3H, s), 3.6 (3H, s), 3.8-4.0 (1H, m), 4.3-4.4 (2H,m), 5.0-5.2 (1H, m), 6.9-7.1 (1H, m), 8.0-8.8 (5H, m). Mass Spectrum: m/z (ES+)[M+H]+ = 463 Example 3: NMR Spectrum: ¾ NMR (300MHz, DMSO) δ 1.7-2.0 (4H, m), 2.1-2.3 (7H, m), 2.35-2.5 (2H, m), 2.55-2.7 (lH,m), 3.3-3.5 (1H, m), 3.56 (1H, s), 3.9-4.0 (lH,m), 4.1- 4.2 (2H,m), 4.3-4.4 (2H, m), 4.8-5.0 (lH,m), 6.9-7.1 (1H, m), 8.0-8.5 (4H, m), 8.7 (1H, s). Mass Spectrum: m/z (ES+)[M+H]+ = 463
Example 4: NMR Spectrum: ¾ NMR (300MHz, DMSO) δ 1.7-2.0 (4H, m), 2.1-2.3 (7H, m), 2.34-2.43 (2H, m), 2.5-2.7 (lH,m), 3.3-3.5 (1H, m), 3.6 (3H, s), 3.85-3.95 (lH,m), 4.1- 4.2 (2H,m), 4.3-4.4 (2H, m), 4.8-5.0 (lH,m), 6.95-7.05 (1H, m), 8.0-8.8 (5H, m). Mass Spectrum: m/z (ES+)[M+H]+ = 463
The preparations of the bromo intermediates required for Examples 1 - 4 are described below:
8-Bromo-3-methyl-l-tetrahvdropyran-4-yl-imidazor4,5-c1cinnolin-2-one
Figure imgf000045_0001
l,l-Dimethoxy-N,N-dimethylmethanamine (2.99 mL, 22.51 mmol) was added to 8-bromo- l-tetrahydropyran-4-yl-3H-imidazo[4,5-c]cinnolin-2-one (0.786 g, 2.25 mmol) in DMF (1.5 mL) and the resulting slurry stirred at 80 °C for 4 hours. The precipitate was collected by filtration, washed with water and ether and dried under vacuum to afford the desired material (0.745 g) as a bright orange solid which was used without further purification. Mass Spectrum: m z (ES+)[M+H]+ = 365
8-Bromo-l-tetrahvdropyran-4-yl-3H-imidazor4,5-c1cinnolin-2-one
Figure imgf000045_0002
Diphenyl phosphorazidate (2.70 mL, 12.46 mmol) was added slowly to 6-bromo-4- (tetrahydro-2H-pyran-4-ylamino)-3-cinnolinecarboxylic acid (1.33 g, 3.78 mmol) and triethylamine (1.579 mL, 11.33 mmol) in DMF (3 mL) and the resulting slurry stirred at 60 °C for 3 hours. Water (5 mL) was added and the precipitate collected by filtration and dried under vacuum to afford the desired material (0.786 g) as a brown solid, which was used without further purification.
NMR Spectrum: !H NMR (500MHz, DMSO) δ 1.88 (2H, d), 2.61 (2H, ddt), 3.62 (2H, q), 4.01 (2H, dd), 4.91 (1H, td), 7.85 (1H, dd), 8.24 (1H, d), 8.48 (1H, d), 12.58 (1H, s).
Mass Spectrum: m/z (ES+)[M+H]+ = 351
6-Bromo-4-(tetrah dro-2H-pyran-4-ylamino)-3-cinnolinecarboxylic acid
Figure imgf000046_0001
2M Sodium hydroxide (1 1.70 mL, 23.41 mmol) was added in one portion to ethyl 6- bromo-4-(tetrahydropyran-4-ylamino)cinnoline-3-carboxylate (1.78 g, 4.68 mmol) in MeOH (3 mL) and the resulting mixture stirred at ambient temperature for 1 hour. The precipitate was collected by filtration, washed with Et20 (20 mL) and dried under vacuum to the desired material (1.33 g), which was used without further purification.
NMR Spectrum: lH NMR (500MHz, DMSO) δ 1.35 - 1.68 (2H, m), 1.81 - 2.1 (2H, m), 3.42 - 3.59 (2H, m), 3.72 - 3.96 (2H, m), 4.11 - 4.41 (1H, m), 7.68 - 8.18 (2H, m), 8.26 (1H, s), 11.68 (1H, s).
Mass Spectrum: m/z (ES+)[M+H]+ = 352
Ethyl 6-bromo-4-(tetrahydropyran-4-ylamino)cinnoline-3-carboxylate
Figure imgf000046_0002
Tetrahydro-2H-pyran-4-amine (0.615 mL, 5.94 mmol) was added dropwise to ethyl 6- bromo-4-chloro-3-cinnolinecarboxylate (1.5 g, 4.75 mmol) and triethylamine (1.990 mL, 14.26 mmol) in THF (5 mL) and the reaction stirred at ambient temperature for 30 minutes. Ether (15 mL) was added and the solid was collected by filtration, washed with water (25 mL) and ether (20 mL) and dried under vacuum to afford the desired material (1.78 g) as a beige solid, which was used without further purification.
NMR Spectrum: !H NMR (400MHz, DMSO) δ 1.39 (3H, t), 1.63 - 1.76 (3H, m), 1.83 (1H, d), 1.94 (3H, d), 3.38 (3H, td), 4.45 (2H, q), 8.02 (2H, dd), 8.18 (1H, d), 8.68 (1H, d). Mass Spectrum: m/z (ES+)[M+H]+ = 378
Ethyl 6-bromo-4-chloro-3 -cinnolinecarboxylate
Figure imgf000047_0001
Ethyl 6-bromo-4-oxo-lH-cinnoline-3-carboxylate (150 mg, 0.50 mmol) was suspended in thionyl dichloride (1357 μί, 18.68 mmol). N,N-dimethylformamide (3.89 μί, 0.05 mmol) was added and the mixture was stirred at 80°C for 5 hours under reflux. The mixture was evaporated to dryness and azeotroped three times with toluene. The residue was dissolved in DCM (15 mL) and washed with ice-cold aqueous NaHC03 solution (approximately 0.5M, 2 x 15 mL). The organic layer was dried over MgS04 and evaporated to afford the desired product (145 mg, 91 %) as a brown solid.
Mass Spectrum: m/z (ES+) [M+2H]+ = 315
Ethyl 6-bromo-4-oxo-lH-cinnoline-3-carboxylate
Figure imgf000047_0002
TFA (837 mL, 10.863 mol) was added slowly to ethyl 3-(5-bromo-2-pyrrolidin-l- yldiazenylphenyl)-3-oxopropanoate (160 g, 434.52 mmol) over a period of 30 minutes at 0°C under an inert atmosphere. The resulting solution was stirred at ambient temperature for 16 hours then the reaction mixture poured onto ice water (2 L). The precipitate was collected by filtration, washed with water (5 x 100 mL) and dried in the vacuum oven to afford the desired material (118 g, 91 %) as a pale yellow solid, which was used without further purification.
NMR Spectrum: !H NMR (300MHz, DMSO) δ 1.30 (3H, t), 4.32 (2H, q), 7.64 (1H, d), 7.99 (1H, d), 8.18 (1H, s), 14.03 (1H, s).
Mass Spectrum: m/z (ES+)[M+H]+ = 297
Ethyl 3 -(5 -bromo-2-pyrrolidin- 1 -yldiazenylphenyl)-3 -oxopropanoate
Figure imgf000048_0001
Sodium hydride (55.3 g, 1382.68 mmol) was added portionwise to a solution of diethyl carbonate (467 g, 3.951 mol) in THF (800 mL) at ambient temperature under an inert atmosphere. A solution of l-(5-bromo-2-pyrrolidin-l-yldiazenylphenyl)ethanone
(117 g, 395.05 mmol) in THF (200 mL) was added slowly over a period of 60 minutes under an inert atmosphere and the resulting mixture stirred at 75 °C for 3 hours. The reaction mixture was allowed to cool then quenched with water (100 mL) and the resulting mixture concentrated under vacuum. The residue was diluted with water (500 mL), extracted with EtOAc (4 x 500 mL), the organic layer dried over Na2S04, filtered and evaporated to afford the desired material (168 g, 115 %) as a brown solid, which was used without further purification.
NMR Spectrum: lH NMR (400MHz, DMSO) δ 1.11 (3H, t), 1.93 - 2.04 (4H, m), 3.60 (2H, t), 3.93 (2H, t), 4.03 (2H, q), 4.11 (2H, s), 7.41 (1H, d), 7.61 - 7.64 (2H, m).
Mass Spectrum: m/z (ES+)[M+H]+ = 368.1
1 -(5 -Bromo-2-pyrrolidin- 1 -yldiazenylphenyDethanone
Figure imgf000048_0002
l-(2-Amino-5-bromophenyl)ethanone (94.8 g, 442.87 mmol) was added to 2M
hydrochloric acid (700 mL, 1.40 mol), and the resulting mixture was stirred at 60°C for 2 hours. The mixture was cooled to 0°C and a solution of sodium nitrite (30.6 g, 442.87 mmol) in water (100 mL) was added dropwise. After 15 minutes the mixture was filtered, the solid discarded and the filtrate added to a stirred solution of pyrrolidine (31.5 g, 442.87 mmol) and sodium hydroxide (56.0 g, 1399.46 mmol) in water (500 mL) at 0°C. After 15 minutes the precipitate was collected by filtration, washed with water and dried in the vacuum oven to afford the desired material (117 g, 89 %) as a red solid, which was used without further purification.
NMR Spectrum: !H NMR (300MHz, DMSO) δ 1.99 (4H, m), 2.54 (3H, s), 3.58 (2H, t),
3.91 (2H, t), 7.37 - 7.66 (3H, m).
Mass Spectrum: m/z (ES+)[M+H]+ = 298
8-Bromo-l-(cis-3-methoxycvclobutyl)-3-methyl-imidazor4,5-c1cinnolin-2-one
Figure imgf000049_0001
A solution of 8-bromo-l-(3-methoxycyclobutyl)-3H-imidazo[4,5-c]cinnolin-2-one (2 g, 5.73 mmol), iodomethane (1.22 g, 8.60 mmol), sodium hydroxide (344 mg, 8.60 mmol) and tetrabutylammonium bromide (192 mg, 0.60 mmol) in water (80 mL) and DCM (150 mL) was stirred at ambient temperature for 12 hours. The mixture was concentrated under vacuum and extracted with 3 x 100 mL of ethyl acetate. The organic layers were combined and concentrated then the residue purified by flash silica chromatography, eluting with DCM / MeOH (10: 1), to afford the desired material contaiminated with 8-bromo-2- methoxy-l-(cis-3-methoxycyclobutyl)imidazo[4,5-c]cinnoline (1.8 g). This mixture was taken forward into the next reaction without further purification.
NMR Spectrum: !H NMR (300MHz, DMSO) δ 2.6-3.0 (7.2H, m), 3.2-3.3 (5.4H, m), 3.5- 3.6 (3.6H, m), 3.8-3.9 (1.8H, m), 4.2-4.4 (1.8H, m), 4.9-5.1 (1.8H, m), 7.6-8.7 (5.4H, m). Mass Spectrum: m/z (ES+)[M+H]+ = 363 8-Bromo-l-(cis-3-methoxycvclobutyl)-3H-imidazor4,5-c1cinnolin-2-one
Figure imgf000050_0001
A solution of 6-bromo-4-[(3-methoxycyclobutyl)amino]cinnoline-3-carboxylic acid (1.6 g, 4.54 mmol) and triethylamine (1.38 g, 13.64 mmol) in DMA (100 mL) was stirred for 1 h at ambient temperature then diphenyl phosphorazidate (3.75 g, 13.63 mmol) added. The resulting solution was stirred at ambient temperature for 1 h then at 60°C for 1 h before being allowed to cool and concentrated. The residue was diluted with water (100 mL) and the solid collected by filtration to afford the desired material (1.7 g) as a yellow solid, which was used without further purification.
Mass Spectrum: m/z (ES+)[M+H]+ = 349
6-Bromo-4- (cis-3-methoxycyclobutyl)aminolcinnoline-3-carboxylic acid
Figure imgf000050_0002
A solution of ethyl 6-bromo-4-[(3-methoxycyclobutyl)amino]cinnoline-3-carboxylate (3 g, 7.89 mmol) and sodium hydroxide (6.4 g, 160.01 mmol) in ethanol (120 mL) and water (80 mL) was stirred at ambient temperature for 2 h. The solids were filtered out and the resulting mixture concentrated under vacuum. The solids were collected by filtration and dried in an oven under reduced pressure to afford the desired material (2.05 g) as a grey solid which was used without further purification.
Mass Spectrum: m/z (ES+)[M+H]+ = 352
Ethyl 6-bromo-4- Γ(3 -methoxycvclobutvOaminol cinnoline-3 -carboxylate
Figure imgf000051_0001
A solution of ethyl 6-bromo-4-chloro-3-cinnolinecarboxylate (2 g, 6.34 mmol), cis-3- methoxycyclobutan-1 -amine hydrochloride (1.04 g, 7.56 mmol) and DIPEA (4.15 g, 32.11 mmol) in DMA (10 mL) was stirred for 12 h at 50°C in a sealed tube. The reaction as allowed to cool, diluted with EtOAc (100 mL) and the organics washed with water, dried over Na2S04 then concentrated in vacuo to afford the desired material (3 g) as a brown solid which was used without further purification.
Mass Spectrum: m/z (ES+)[M+H]+ = 380
8-Bromo-3-meth l-l-[(3S)-tetrahydropyran-3-yllimidazo[4,5-clcinnolin-2-one
Figure imgf000051_0002
8-Bromo-l-[(3S)-tetrahydropyran-3-yl]-3H-imidazo[4,5-c]cinnolin-2-one (660 mg, 1.89 mmol) was added to a mixture of iodomethane (537 mg, 3.78 mmol), tetrabutylammonium bromide (60.9 mg, 0.19 mmol) and NaOH (113 mg, 2.84 mmol) in DCM (30 mL) and water (20 mL) and the resulting mixture stirred at ambient temperature for 12 hours. The reaction mixture was concentrated and diluted with EtOAc (100 mL). The organic layer was dried over Na2S04, filtered and evaporated to afford the desired material (570 mg) as yellow solid, which was used without further purification.
Mass Spectrum: m/z (ES+)[M+H]+ = 363
8-Bromo-l-[(3S)-tetrahydropyran-3-yll-3H-imidazo[4,5-clcinnolin-2-one
Figure imgf000052_0001
A mixture of 6-bromo-4-[[(3S)-tetrahydropyran-3-yl]amino]cinnoline-3-carboxylic acid (438 mg, 1.24 mmol) and triethylamine (0.520 mL, 3.73 mmol) in DMF (10 mL) was stirred at ambient temperature for 1 hour. Diphenylphosphoryl azide (0.535 mL, 2.49 mmol) was added and the reaction stirred at 60°C for a further 1 hour. The reaction mixture was evaporated to dryness, redissolved in water (20 mL) and the solid collected by filtration to afford the desired material (660 mg) as yellow solid, which was used without further purification.
Mass Spectrum: m/z (ES+)[M+H]+ = 349
6-Bromo-4-r Γ(3 S)-tetrahvdropyran-3 -yll aminol cinnoline-3 -carboxylic acid
Figure imgf000052_0002
A mixture of ethyl 6-bromo-4-[[(3S)-tetrahydropyran-3-yl]amino]cinnoline-3-carboxylate (600 mg, 1.58 mmol) and NaOH (1600 mg, 40.00 mmol) in ethanol (30 mL) and water (20 mL) was stirred at 50 °C for 2 hours. The reaction mixture was evaporated, the pH adjusted to pH5 with 2M HC1 and the solid filtered to afford the desired material (438 mg) as light- yellow solid, which was used without further purification.
Mass Spectrum: m/z (ES+)[M+H]+ = 352 Ethyl 6-bromo-4- [ [(3 S)-tetrahydropyran-3 -yl] amino] cinnoline-3 -carboxylate
Figure imgf000053_0001
A mixture of ethyl 6-bromo-4-chloro-3-cinnolinecarboxylate (500mg, 1.58 mmol), (S)- tetrahydro-2H-pyran-3 -amine hydrochloride (327 mg, 2.38 mmol), and DIPEA (0.830 mL, 4.75 mmol) in DMA (2 mL) was stirred at 70 °C for 2 hours. The reaction mixture was diluted with water and the solids collected by filtration to afford the desired material (600 mg) as yellow solid, which was used without further purification.
Mass Spectrum: m/z (ES+)[M+H]+ = 380
8-Bromo-3 -meth l- 1 -[(3R)-tetrahydropyran-3 -yllimidazo[4,5 -clcinnolin-2-one
Figure imgf000053_0002
8-Bromo-l-[(3R)-tetrahydropyran-3-yl]-3H-imidazo[4,5-c]cinnolin-2-one (328 mg, 0.94 mmol) was added to a mixture of iodomethane (267 mg, 1.88 mmol), tetrabutylammonium bromide (30.3 mg, 0.09 mmol) and NaOH (56.4 mg, 1.41 mmol) in DCM (30 mL) and water (20 mL) and the resulting mixture stirred at ambient temperature for 12 hours. The reaction mixture was concentrated and diluted with EtOAc (100 mL). The organic layer was dried over Na2S04, filtered and evaporated to afford the desired material (560 mg) as yellow solid, which was used without further purification.
Mass Spectrum: m/z (ES+)[M+H]+ = 363
8-Bromo-l-r(3R)-tetrahvdropyran-3-yl1-3H-imidazor4,5-c1cinnolin-2-one
Figure imgf000053_0003
A mixture of 6-bromo-4-[[(3R)-tetrahydropyran-3-yl]amino]cinnoline-3-carboxylic acid (382 mg, 1.08 mmol) and triethylamine (0.454 mL, 3.25 mmol) in DMF (10 mL) was stirred at ambient temperature for 1 hour. Diphenylphosphoryl azide (0.466 mL, 2.17 mmol) was added and the reaction stirred at 60°C for a further 1 hour. The reaction mixture was evaporated to dryness, redissolved in water (20 mL) and the solid collected by filtration to afford the desired material (328 mg) as yellow solid, which was used without further purification.
Mass Spectrum: m/z (ES+)[M+H]+ = 349
6-Bromo-4-r r(3R)-tetrahydrop yran-3 - yll aminol cinnoline-3 -carboxylic acid
Figure imgf000054_0001
A mixture of ethyl 6-bromo-4-[[(3R)-tetrahydropyran-3-yl]amino]cinnoline-3-carboxylate (600 mg, 1.58 mmol) and NaOH (1600 mg, 40.00 mmol) in ethanol (30 mL) and water (20 mL) was stirred at 50 °C for 2 hours. The reaction mixture was evaporated, the pH adjusted to pH5 with 2M HC1 and the solid filtered to afford the desired material (382 mg) as yellow solid, which was used without further purification.
Mass Spectrum: m/z (ES+)[M+H]+ = 352
Ethyl 6-bromo-4- Γ r(3R)-tetrahvdrop yran-3 -yll aminol cinnoline-3 -carboxylate
Figure imgf000054_0002
A mixture of ethyl 6-bromo-4-chloro-3-cinnolinecarboxylate (500mg, 1.58 mmol), (R)- tetrahydro-2H-pyran-3 -amine hydrochloride (327 mg, 2.38 mmol), and DIPEA (0.830 mL, 4.75 mmol) in DMA (2 mL) was stirred at 60 °C for 12 hours. The reaction mixture was diluted with water and the solids collected by filtration to afford the desired material (600 mg) as yellow solid, which was used without further purification.
Mass Spectrum: m/z (ES+)[M+H]+ = 380 The preparation of N,N-dimethyl-3-[[5-(4,4,5,5-tetramethyl-l ,3,2-dioxaborolan-2-yl)-2- pyridyl]oxy]propan-l -amine is described below. N,N-Dimethyl-3-[[5-(4,4,5,5-tetramethyl- l,3,2-dioxaborolan-2-yl)-2-pyridyl]oxy]propan-l-amine is also available commercially, for example from Fluorochem Ltd, 14 Graphite Way, Hadfield, Glossop SKI 3 1QH, UK; catalogue number 213155-lg.
N,N-Dimethyl-3-[[5-(4,4,5,5-tetramethyl-l ,3,2-dioxaborolan-2-yl)-2-pyridylloxylpropan-
1 -amine
Figure imgf000055_0001
Butyllithium (2.5N, 4.8 mL, 50.96 mmol) was added to a solution of 3-(5-bromopyridin-2- yl)oxy-N,N-dimethylpropan-l -amine (2.07 g, 7.99 mmol) and 4,4,5, 5-tetramethyl-2-
(propan-2-yloxy)-l,3,2-dioxaborolane (2.79 g, 15.00 mmol) in THF (20 mL) at -78°C over 10 minutes under an inert atmosphere. The resulting solution was stirred for 4 hour at 18°C. The reaction was then quenched by the addition of a saturated aqueous solution of ammonium chloride then partitioned between EtOAc (100 mL) and water (100 mL). The organic layer was concentrated in vacuo and the residue purified by flash column chromatography on silica, eluting with EtO Ac/petroleum ether (1 :3) to afford the desired material (270 mg, 11%) as a yellow solid.
Mass Spectrum: m/z (ES+)[M+H]+ = 225 3-(5-Bromopyridin-2-yl)oxy-N,N-dimethylpropan-l -amine
Figure imgf000055_0002
3-(Dimethylamino)propan-l-ol (3.09 g, 29.95 mmol) was added to a mixture of sodium hydride (2.4 g, 60.00 mmol) in DMF (50 mL) over a period of 20 min at ambient temperature. 5-Bromo-2-fluoropyridine (5.81 g, 33.01 mmol) was added and the resulting solution stirred for 4 hours at 30°C. The reaction was then quenched by the addition of a saturated aqueous solution of ammonium chloride and the resulting mixture concentrated under vacuum. The residue was purified by flash column chromatography on silica, eluting with DCM/MeOH ether (10: 1) to afford the desired material (5.2 g, 67%) as yellow oil. Mass Spectrum: m/z (ES+)[M+H]+ = 259
Example 5: 3-Methyl-8-[6-[3-(l-piperidyl)propoxy]-3-pyridyl]-l-tetrahydropyran-4- yl-imidazo [4,5-c] cinnolin-2-one
Figure imgf000056_0001
A solution of 3-(piperidin-l-yl)propan-l-ol (0.091 g, 0.63 mmol) in THF (2 mL) was added slowly to a stirred suspension of sodium hydride (0.051 g, 1.27 mmol) in THF (2 mL) and the mixture stirred at ambient temperature for 30 minutes. A solution of 8-(6- fluoro-3-pyridyl)-3-methyl-l-tetrahydropyran-4-yl-imidazo[4,5-c]cinnolin-2-one (0.12 g, 0.32 mmol) in DMF (2 mL) was added and the reaction stirred at 50°C for 3 hours. The reaction was cooled to ambient temperature and quenched with water. The reaction mixture was extracted with EtOAc (50 mL), and the organic layer washed twice with saturated brine (20 mL) and evaporated to afford crude product. The crude product was purified by flash silica chromatography, elution gradient 0 to 10% 1% NH3 in MeOH in DCM, to afford the desired material (0.04 g) as a yellow solid.
NMR Spectrum: lH NMR (500MHz, DMSO) δ 1.40 (2H, d), 1.51 (4H, p), 1.74 - 1.81 (1H, m), 1.88 - 1.94 (2H, m), 1.93 - 2.01 (2H, m), 2.32 - 2.39 (3H, m), 2.41 (2H, t), 2.6 - 2.73 (2H, m), 3.58 - 3.65 (2H, m), 3.62 (3H, s), 4.07 (2H, dd), 4.38 (2H, t), 5.11 (1H, ddd), 7.01 (1H, dd), 8.10 (1H, dd), 8.25 (1H, dd), 8.37 - 8.47 (2H, m), 8.73 (1H, dd).
Mass Spectrum: m/z (ES+)[M+H]+ = 503 The synthesis of 8-(6-fluoro-3-pyridyl)-3-methyl-l-tetrahydropyran-4-yl-imidazo[4,5- c]cinnolin-2-one is described below:
8-(6-Fluoro-3 -pyridyl)-3 -methyl- 1 -tetrahydropyran-4-yl-imidazo [4,5 -c] cinnolin-2-one
Figure imgf000057_0001
8-Bromo-3-methyl-l-tetrahydropyran-4-yl-imidazo[4,5-c]cinnolin-2-one (0.745 g, 2.05 mmol), (6-fluoropyridin-3-yl)boronic acid (0.347 g, 2.46 mmol) and 2M potassium carbonate (3.08 ml, 6.15 mmol) were suspended in 1,4-dioxane (12 mL) and the mixture degassed before the addition of bis(triphenylphosphine)palladium(II) dichloride (0.043 g, 0.06 mmol). The reaction was heated to 120 °C for 6 hours under nitrogen then cooled to ambient temperature. The precipitate was collected by filtration and washed with water and ether to afford the desired material (0.812 g) as an orange solid which was used without further purification.
NMR Spectrum: !H NMR (500MHz, DMSO) δ 1.92 - 2.02 (2H, m), 2.58 - 2.75 (3H, m), 3.62 (3H, s), 4.03 (3H, dt), 5.11 (IH, ddd), 7.42 (IH, dd), 8.12 (IH, dd), 8.46 (IH, d), 8.47 - 8.5 (IH, m), 8.53 (IH, td), 8.81 (IH, d).
Mass Spectrum: m/z (ES+)[M+H]+ = 380
Example 6 : 8- [4- [3-(Dimethylamino)propoxy] phenyl] - l-isopropyl-3-methyl- imidazo [4,5-c] cinnolin-2-one
Figure imgf000057_0002
Dichlorobis(triphenylphosphine)palladium(II) (0.013 g, 0.02 mmol) was added to degassed solution of 8-bromo-l-isopropyl-3-methyl-imidazo[4,5-c]cinnolin-2-one 0.36 mmol) and N,N-dimethyl-3-(4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)phenoxy)propan-l -amine (0.116 g, 0.38 mmol), potassium carbonate (0.542 mL, 1.08 mmol) in 1,4-dioxane (8 mL) and heated in the microwave to 80 °C for 1 hour under nitrogen. The reaction mixture was diluted with EtOAc (25 mL), and washed sequentially with water (10 mL), saturated brine (10 mL) and the organic layer was evaporated. The crude product was purified by flash silica chromatography, elution gradient 0 to 10% (1% NH3 in MeOH) in DCM. Pure fractions were evaporated to dryness and triturated with ether to afford the desired product (0.055 g, 36 %) as a pale brown solid.
NMR Spectrum: !H NMR (500 MHz, DMSO) δ 1.69 (6H, d), 1.89 (2H, p), 2.17 (6H, s), 2.39 (2H, t), 3.60 (3H, s), 4.10 (2H, t), 5.29 (1H, p), 7.13 (2H, d), 7.85 (2H, d), 8.05 (1H, dd), 8.31 (1H, d), 8.38 (1H, d).
Mass Spectrum: m/z (ES+) [M+H]+ = 420
N,N-Dimethyl-3-(4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenoxy)propan-l -amine was prepared as described below.
N,N-Dimethyl-3-r4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenoxy1-l-propanamine
Figure imgf000058_0001
Dichloro[l, -bis(diphenylphosphino)ferrocene]palladium (II) dichloromethane adduct (0.063 g, 0.08 mmol) was added to 3-(4-bromophenoxy)-A ,N-dimethylpropan-l-amine (2g, 7.75 mmol), 4,4,4*,4*,5,5,5*,5*-octamethyl-2,2,-bi(l,3,2-dioxaborolane) (2.36 g, 9.30 mmol) and potassium acetate (3.04 g, 30.99 mmol) in 1,4-dioxane (35 mL) and the mixture degassed for 15 minutes. The resulting suspension was stirred at 90 °C for 16 hours under an inert atmosphere. The reaction mixture was evaporated to dryness, redissolved in DCM (25 mL), washed with water (20 mL) and the organic layer was dried with a phase separating cartridge, filtered and evaporated. The crude product was purified by flash silica chromatography, elution gradient 0 to 10% MeOH in DCM, to afford the desired material as a brown oil (1.000 g, 42.3 %) which solidified on standing. NMR Spectrum: lH NMR (400MHz, CDCb) δ 1.33 (12H, s), 1.96 - 2.07 (2H, m), 2.34 (6H, s), 2.52 - 2.65 (2H, m), 4.04 (2H, t), 6.83 - 6.94 (2H, m), 7.68 - 7.78 (2H, m).
Mass Spectrum: m/z (ES+) [M+H]+ = 306
8-Bromo-l-isopropyl-3-methyl-imidazo[4,5-c]cinnolin-2-one was prepared as described below.
8-Bromo-l-isopropyl-3-methyl-imidazo[4,5-clcinnolin-2-one
Figure imgf000059_0001
l,l-Dimethoxy-N,N-dimethylmethanamine (0.623 mL, 4.69 mmol) was added to 8-bromo- l-isopropyl-3H-imidazo[4,5-c]cinnolin-2-one (0.144 g, 0.47 mmol) in DMF (2 mL and the resulting slurry was stirred at 80 °C for 4 hours. A precipitate formed which was collected by filtration, washed with water and ether and dried under vacuum to afford crude product as an orange solid. The filtrate was extracted with DCM (20 mL) and the organic layer combined with the above solid. The crude product was purified by flash silica
chromatography, elution gradient 0 to 5% MeOH in DCM . Pure fractions were evaporated to dryness to afford the desired product (0.106 g, 70.4 %) as a yellow solid.
NMR Spectrum: !H NMR (500 MHz, CDCb) δ 1.77 (6H, d), 3.75 (3H, s), 5.08 (1H, s), 7.72 (1H, dd), 8.28 (1H, d), 8.31 (1H, dd).
Mass Spectrum: m/z (ES+) [M+3H]+ = 323
8-Bromo-l-isopropyl-3H-imidazo[4,5-clcinnolin-2-one
Figure imgf000059_0002
Diphenyl phosphorazidate (1.948 mL, 8.99 mmol) was added slowly to 6-bromo-4- (isopropylamino)cinnoline-3-carboxylic acid (0.845 g, 2.72 mmol) and triethylamine (1.139 mL, 8.17 mmol) in DMF (3 mL) and stirred at 60 °C for 3 hours. The reaction was concentrated and redissolved in EtOAc (100 mL). The organic layer was washed with saturated ammonium chloride (30 mL) and brine (20 mL). The aqueous was extracted with EtOAc ( 2 x 30 mL) and the organic layers combined and evaporated to afford this desired product (0.3761 g, 45 %) as a pale yellow solid. Upon standing a precipitate crashed out of the aqueous solution. The solid was collected by filtration, washed with ether and dried under vacuum to give additional desired material (0.415 g, 50 %) as a yellow solid. The two crops were combined and used without further purification.
NMR Spectrum: lU NMR (500 MHz, DMSO) δ 1.61 (6H, d), 5.03 - 5.22 (1H, m), 7.83 (1H, dd), 8.22 (1H, d), 8.47 (1H, d), 12.50 (1H, s).
Mass Spectrum: m/z (ES+) [M+H]+ = 307
6-Bromo-4-(isopropylamino)cinnoline-3-carboxylic acid
Figure imgf000060_0001
Sodium hydroxide (2.0 M) (5.91 mL, 11.81 mmol) was added in one portion to ethyl 6- bromo-4-(isopropylamino)cinnoline-3-carboxylate (0.799 g, 2.36 mmol) in MeOH (3 mL) and the resulting mixture stirred at 25 °C for 16 hours. A beige precipitate formed which was collected by filtration, washed with Et20 (20 mL) and dried under vacuum to afford the desired product (0.845 g, 115 %) as a cream solid, which was used without further purification.
NMR Spectrum: !H NMR (400 MHz, DMSO) δ 1.29 (6H, d), 4.14 - 4.47 (1H,
(1H, dd), 8.00 (1H, d), 8.29 (1H, d), 11.57 (1H, t).
Mass Spectrum: m/z (ES-) [M-H]- = 308
Ethyl 6-bromo-4-(isopropylamino)-3 -cinnolinecarboxylate
Figure imgf000061_0001
2-Propanamine (0.506 mL, 5.94 mmol) was added in one portion to ethyl 6-bromo-4- chlorocinnoline-3-carboxylate (1.5 g, 4.75 mmol) in THF (5 mL) and the reaction was stirred at room temperature for 40 minutes. A beige precipitate formed. Ether (20 mL) was added to the reaction mixture and the precipitate was collected by filtration and washed with water (20 mL) and ether (20 mL) and dried under vacuum to afford the desired product (1.049 g, 65.2 %) as a beige solid which was used without further purification. The filtrate was concentrated and extracted with DCM (50 mL). The organic layer was dried over MgS04, filtered and evaporated to afford crude product. The crude product was purified by flash silica chromatography, elution gradient 0 to 30% EtOAc in isohexane. Pure fractions were evaporated to dryness to afford the desired product (250 mg) as a pale yellow solid.
Mass Spectrum: m/z (ES-) [M-H]- = 336
Example 7: l-Isopropyl-3-methyl-8-[6-[3-(l-piperidyl)propoxy]-3- pyridyl] imidazo [4,5-c] cinnolin-2-one
Figure imgf000061_0002
A mixture of dichlorobis(triphenylphosphine)palladium(II) (12.68 mg, 0.02 mmol), 2-(3- (piperidin- 1 -yl)propoxy)-5-(4,4,5 ,5-tetramethyl- 1 ,3 ,2-dioxaborolan-2-yl)pyridine (131 mg, 0.38 mmol), 8-chloro-l-isopropyl-3-methyl-imidazo[4,5-c]cinnolin-2-one (100 mg, 0.36 mmol) and 2M potassium carbonate solution (0.542 mL, 1.08 mmol) in 1,4-dioxane (3.07 mL) was heated in a microwave reactor at 80 °C for 1 hour under an inert atmosphere. The reaction mixture was diluted with EtOAc (25 mL), and washed sequentially with water (10 mL), saturated brine (10 mL) and the organic layer evaporated to afford crude product as a brown solid residue. The crude product was purified by preparative HPLC to afford the desired material as a dry yellow film. Diethyl ether was added and the mixture evaporated to dryness to afford the desired material (80 mg) as a brown solid.
NMR Spectrum: !H NMR (400MHz, DMSO) δ 1.41 (2H, ddd), 1.53 (4H, dt), 1.71 (6H, d), 1.93 (2H, p), 2.38 - 2.47 (6H, m), 3.62 (3H, s), 4.42 (2H, t), 5.29 (IH, p), 6.96 (IH, dd), 8.02 (IH, dd), 8.18 (IH, dd), 8.35 (IH, d), 8.40 (IH, dd), 8.68 (IH, dd).
Mass Spectrum: m/z (ES+)[M+H]+ = 461
The following compounds were synthesised in an analogous fashion using between 1-2 equivalents of the appropriate boronic ester, between 5 - 10 mol% of catalyst and the appropriate chloro intermediate.
Figure imgf000062_0001
* 4 equivalents of base were used and the reaction stirred at 90°C for 2 days.
** The reaction was stirred at 90°C for 18 hours.
*** The reaction was stirred at 100°C for 90 minutes. Example 8: NMR Spectrum: lH NMR (400MHz, DMSO) δ 1.67 (6H, d), 1.84 - 1.93 (2H, m), 2.15 (6H, s), 2.35 (2H, q), 3.59 (3H, s), 4.37 (2H, t), 5.31 (1H, p), 6.98 (1H, d), 8.05 (1H, dd), 8.23 (1H, dd), 8.33 - 8.43 (2H, m), 8.71 (1H, d). Mass Spectrum: m/z (ES+) [M+H]+ = 421
Example 9: NMR Spectrum: !H NMR (400 MHz, DMSO) δ 1.69 (10H, t), 1.93 (2H, p), 2.43 - 2.48 (4H, m), 2.54 - 2.58 (2H, m), 3.60 (3H, s), 4.12 (2H, t), 5.28 (1H, p), 7.09 - 7.2 (2H, m), 7.79 - 7.91 (2H, m), 8.05 (1H, dd), 8.31 (1H, d), 8.38 (1H, d). Mass Spectrum: m/z (ES+) [M+H]+ = 446
Example 10: NMR Spectrum: lH NMR (400 MHz, DMSO) δ 1.3 - 1.46 (2H, m), 1.45 - 1.62 (4H, m), 1.68 (6H, d), 1.89 (2H, h), 2.2 - 2.45 (6H, m), 3.60 (3H, s), 4.03 - 4.15 (2H, m), 5.28 (1H, hept), 7.06 - 7.17 (2H, m), 7.8 - 7.88 (2H, m), 8.04 (1H, dd), 8.31 (1H, d), 8.37 (1H, d). Mass Spectrum: m/z (ES+) [M+H]+ = 460
Example 10 can also be isolated as the methane sulfonic acid salt by taking the material as prepared above and subjecting to the following reaction conditions. l-Isopropyl-3-methyl-8-[4-[3-(l-piperidyl)propoxy]phenyl]imidazo[4,5-c]cinnolin-2-one (89 mg, 0.19 mmol) was dissolved in DCM (2 mL) and treated with 1M methanesulfonic acid (0.013 mL, 0.19 mmol) in DCM then the mixture evaporated to dryness. The crude product was purified by ion exchange chromatography, using an SCX column. The desired product was eluted from the column using 1M NH3/MeOH to afford the desired product (135 mg, 0.29 mmol) as a yellow solid. NMR Spectrum: !H NMR (400 MHz, DMSO) δ 1.55 - 1.67 (2H, m), 1.71 (6H, d), 1.76 - 1.84 (4H, m), 2.16 - 2.25 (2H, m), 3.17 - 3.32 (6H, m), 3.62 (3H, s), 4.20 (2H, t), 5.17 - 5.33 (1H, m), 6.77 - 7.36 (2H, m), 7.83 (2H, d), 8.02 (1H, dd), 8.30 (1H, d), 8.39 (1H, d). Mass Spectrum: m/z (ES+) [M+H]+ = 460
Example 11: NMR Spectrum: lH NMR (400 MHz, DMSO) δ 1.93 (2H, p), 2.21 (6H, s), 2.4 - 2.45 (2H, m), 2.6 - 2.7 (2H, m), 3.19 - 3.3 (5H, m), 3.64 (3H, s), 4.27 - 4.36 (1H, m), 4.44 (2H, t), 5.55 (IH, ddd), 6.98 (IH, dd), 8.03 (IH, dd), 8.19 (IH, dd), 8.26 (IH, d), 8.40 (IH, dd), 8.69 (IH, dd). Mass Spectrum: m/z (ES+) [M+H]+ = 463
The preparations of the chloro intermediates and boronic esters required for examples 7 - 11 have either been described earlier or are described below.
8-Chloro- 1 -iso ropyl-3-methyl-imidazo[4,5-clcinnolin-2-one
Figure imgf000064_0001
A mixture of l,l-dimethoxy-N,N-dimethylmethanamine (21.75 mL, 163.69 mmol) and 8- chloro-l-isopropyl-3H-imidazo[4,5-c]cinnolin-2-one (4.3 g, 16.37 mmol) in DMF (60.1 mL) was stirred at 100 °C for 4 hours. The reaction was cooled and the precipitate collected by filtration to afford the desired material (2.6 g) as a pale orange solid which was used without further purification. On standing another crop of desired product was found to precipitate from the filtrate and was again collected (0.11 g) and used without further purification.
NMR Spectrum: ¾ NMR (300MHz, DMSO) δ 1.63 (6H, d), 3.58 (3H, s), 5.16 (IH, p), 7.74 (IH, dd), 8.35 - 8.39 (IH, m), 8.49 (IH, s).
Mass Spectrum: m/z (ES+)[M+H]+ = 276
8-Chloro-l-isopropyl-3H-imidazo[4,5-clcinnolin-2-one
Figure imgf000064_0002
l,3,5-Trichloro-l,3,5-triazinane-2,4,6-trione (1.80 g, 7.74 mmol) was added portionwise to a stirred suspension of 6-chloro-4-(isopropylamino)cinnoline-3-carboxamide (4.1 g, 15.49 mmol) and 2,3,4,6,7,8,9,10-octahydropyrimido[l,2-a]azepine (4.63 mL, 30.98 mmol) in MeOH (60 mL) at 5°C then the solution was warmed to ambient temperature and stirred for lh 30. The reaction mixture was concentrated to dryness and partitioned between
EtOAc (50 mL) and sat aq. NH4C1 (50 mL). A precipitate was observed which was collected by filtration, washed with diethyl ether (3 x lOmL) and dried under vacuum at
45°C to afford the desired material (5.30 g) as a brown solid, which was used without further purification. Additional less pure material (1.3 g) was obtained from the organic phase following separation, drying (MgS04) and concentration in vacuo.
NMR Spectrum: !H NMR (400MHz, DMSO) δ 1.66 (6H, d), 5.10 (1H, hept), 7.69 (1H, dd), 8.27 (1H, d), 8.32 (1H, d).
Mass Spectrum: m/z (ES+)[M+H]+ = 262
6-Chloro-4-(isopropylamino)cinnoline-3-carboxamide
Figure imgf000065_0001
Propan-2-amine (1.775 mL, 20.66 mmol) was added to 4,6-dichloro-3- cinnolinecarboxamide (5 g, 20.66 mmol) and DIPEA (7.22 mL, 41.31 mmol) in acetonitrile (94 mL) and the resulting suspension stirred at 60 °C for 18 hours. The mixture was allowed to cool and diluted with water. The precipitate was collected by filtration, washed with water (20 mL) followed by acetonitrile (10 mL) and dried under vacuum to afford the desired material (4.06 g) as a brown solid, which was used without further purification.
Mass Spectrum: m/z (ES+)[M+H]+ = 265
4,6-Dichloro-3-cinnolinecarboxamide
Figure imgf000065_0002
DMF (0.172 mL, 2.23 mmol) was added dropwise to a mixture of 6-chloro-4-hydroxy- cinnoline-3-carboxylic acid (5.00 g, 22.26 mmol) in thionyl chloride (32.5 mL, 445.24 mmol) at ambient temperature under an inert atmosphere. The resulting slurry was stirred at 80 °C for 18 hours then allowed to cool, concentrated to dryness and the residue azeotroped with toluene (3 x 100 mL) to afford the crude acid chloride. This crude material
(5 g, 19.12 mmol) was dissolved in acetone (41 mL), cooled to 0°C and ammonium hydroxide (54.6 mL, 1401.6 mmol) added dropwise over 10 minutes. The resulting mixture was stirred at ambient temperature for 1 hour and the precipitate collected by filtration.
The solid was washed with diethyl ether and dried under vacuum to afford the desired material (5 g) as a brown solid, which was used without further purification.
NMR Spectrum: ¾ NMR (400MHz, DMSO) δ 8.13 (1H, s), 8.15 (1H, dd), 8.37 (1H, dd),
8.42 (1H, s), 8.65 (1H, dd).
Mass Spectrum: m/z (ES+)[M+H]+ = 242
6-Chloro-4-hydroxy-cinnoline-3-carboxylic acid is available from commercial sources, for example from IChem LP, 5 walnut Hill Park, Suite 13, Woburn, MA, 01801, US; CAS Registry Number: 90272-08-5, or from Vijaya Pharmaceuticals, LLC (V-Pharma), 104 T. W. Alexander Drive, Building 5, PO Box 14547, Research Triangle Park, NC, 27709, US. The preparation is also described in the literature: Schofield, K.; Swain, T., Journal of the Chemical Society, (1949), 2393-9.
2-(3-(Piperidin-l-yl)propoxy)-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyridine
Figure imgf000066_0001
Butyllithium (139 mL, 347.59 mmol) was added dropwise to 5-bromo-2-(3-(piperidin-l- yl)propoxy)pyridine (80 g, 267.37 mmol) and 2-isopropoxy-4,4,5,5-tetramethyl-l,3,2- dioxaborolane (64.7 g, 347.59 mmol) in THF (400 mL) cooled to -78°C over a period of 10 minutes under an inert atmosphere. The resulting mixture was allowed to warm to ambient temperature and stirred for 12 hours. The reaction mixture was quenched with saturated aqueous solution of NH4C1 (100 mL) and solvent was removed before the mixture was extracted with EtOAc (2 x 500 mL).Tthe organic layer was washed with saturated brine (2 x 100 mL), dried over Na2S04, filtered and evaporated to afford the desired product (92 g, 99 %) as a yellow oil, which was used without further purification. NMR Spectrum: !H NMR (400 MHz, CDCb,) δ 1.34 (12H, s), 1.60 (5H, p), 1.93 - 2.08 (3H, m), 2.39 - 2.53 (6H, m), 4.34 (2H, dt), 6.67 - 6.77 (1H, m), 7.92 (1H, dd), 8.50 - 8.56 (lH, m).
Mass Spectrum: m/z (ES+), M-82 = 264.9 (decomposition to boronic acid observed)
5-Bromo-2-(3-(piperidin- 1 -yl)propoxy)pyridine
Figure imgf000067_0001
Sodium hydride (26.14 g, 653.46 mmol) was added portionwise to 3-(l-piperidinyl)-l- propanol (44.8 g, 312.52 mmol) in THF (400 mL) at 25°C under an inert atmosphere, then stirred at 50 °C for 30 minutes. 5-bromo-2-fluoropyridine (50.0 g, 284.11 mmol) was added at ambient temperature, then the solution was stirred at 50 °C for 2 hours. The mixture was poured into water/ice (1000 mL), the solvent removed under reduced pressure and extracted with DCM (3 x 150 mL). The organic layer was washed with saturated brine (3 x 150 mL), dried over Na2S04, filtered and evaporated to afford the desired product (96 g, 113 %) as a brown oil, which was used without further purification.
NMR Spectrum: !H NMR (400MHz, DMSO) δ 1.36-1.50 (6H, m), 1.80-1.87 (2H, m), 2.29-2.36 (6H, m), 4.24 (2H, t), 6.79 (1H, d), 7.86-7.89 (1H, m), 8.26 (1H, d).
Mass Spectrum: m/z (ES+) [M+H]+ = 299.0 l-[3-[4-(4,4,5,5-Tetramethyl-l ,3,2-dioxaborolan-2-yl)phenoxylpropyllpyrrolidine
Figure imgf000067_0002
Potassium acetate (1.036 g, 10.56 mmol) was added to l-(3-(4- bromophenoxy)propyl)pyrrolidine (1 g, 3.52 mmol), 4,4,4',4',5,5,5',5'-octamethyl-2,2'- bi(l,3,2-dioxaborolane) (1.072 g, 4.22 mmol) and [Ι, - bis(diphenylphosphino)ferrocene]dichloropalladium(II) (0.129 g, 0.18 mmol) in 1,4- dioxane (1 mL) at 25°C under nitrogen. The resulting mixture was stirred at 100 °C for 3 hours. The solvent was removed under reduced pressure. The crude product was purified by flash silica chromatography, elution gradient 0 to 10% MeOH in DCM. Pure fractions were evaporated to dryness to afford the desired material as a brown oil (1.100 g, 94 %). Mass Spectrum: m/z (ES+)[M+H]+ 332
1 -(3 -(4-Bromophenoxy)propyl)pyrrolidine
Figure imgf000068_0001
A mixture of l-(3-chloropropyl)pyrrolidine, hydrochloride salt (1.5 g, 8.15 mmol), 4- bromophenol (1.410 g, 8.15 mmol) and potassium carbonate (4.50 g, 32.59 mmol) in DMF (15 mL) was heated to 90°C for 18 hours. The reaction mixture was cooled to ambient temperature, diluted with EtOAc (300 mL), washed with water (200 mL), saturated brine (200 mL), dried over a phase separator and the solvent was removed under reduced pressure to afford crude product. The crude product was purified by ion exchange chromatography, using an SCX column and eluting with 1M NH3/MeOH, to afford the desired material as a brown oil (1.97 g, 85 %).
NMR Spectrum: lH NMR (500MHz, CDC13) δ 1.73 - 1.85 (4H, m), 1.94 - 2.04 (2H, m), 2.49 - 2.56 (4H, m), 2.57 - 2.64 (2H, m), 3.99 (2H, t), 6.75 - 6.81 (2H, m), 7.31 - 7.39 (2H, m).
Mass Spectrum: m/z (ES+)[M+H]+ = 286 l-r3-r4-(4,4,5,5-Tetramethyl-l,3,2-dioxaborolan-2-yl)phenoxy1propyl1piperidine
Figure imgf000068_0002
Dichloro[l, -bis(diphenylphosphino)ferrocene]palladium (II) dichloromethane adduct (0.11 g, 0.14 mmol) was added to l-(3-(4-bromophenoxy)propyl)piperidine (2.0 g, 6.71 mmol), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(l,3,2-dioxaborolane) (2.044 g, 8.05 mmol) and potassium acetate (2.63 g, 26.83 mmol) in 1,4-dioxane (30.5 mL) and the mixture degassed for 15 minutes. The resulting suspension was stirred at 90 °C for 22 hours under an inert atmosphere. The reaction mixture was evaporated to dryness, redissolved in DCM (50 mL), washed with water (40 mL) and the organic layer was dried with a phase separating cartridge, filtered and evaporated. The crude product was purified by flash silica chromatography, elution gradient 0 to 10% MeOH in DCM, to afford the desired material as a brown oil which solidified on standing.
NMR Spectrum: !H NMR (400MHz, CDCb) δ 1.33 (12H, s), 1.59 (2H, s), 1.88 (4H, q), 2.26 (2H, dq), 2.77 - 2.97 (6H, m), 4.06 (2H, t), 6.76 - 6.92 (2H, m), 7.64 - 7.8 (2H, m). Mass Spectrum: m/z (ES+) [M+H]+ = 346
Example 12: 8-[6-[3-(4-Fluoro-l-piperidyl)propoxy]-3-pyridyl]-l-isopropyl-3-methyl- imidazo 4,5-c] cinnolin-2-one
Figure imgf000069_0001
3-(4-Fluoropiperidin-l-yl)propan-l-ol (105 mg, 0.65 mmol) in THF (1.235 mL) was added dropwise to a stirred suspension of sodium hydride (52.2 mg, 1.30 mmol) in THF (1.235 mL) 0 °C. The resulting suspension was stirred at 0 °C for 10 minutes under an inert atmosphere before a solution of 8-(6-fluoro-3-pyridyl)-l-isopropyl-3-methyl-imidazo[4,5- c]cinnolin-2-one (110 mg, 0.33 mmol) in DMF (1.087 mL) was added and the reaction mixture stirred at ambient temperature for 2 hours. The reaction mixture was diluted with ethyl acetate (40 mL), washed three times with water (3 x 20 mL) and the organic layer was dried over MgS04, filtered and evaporated. The crude product was purified by preparative HPLC to afford the desired material (25 mg) as a yellow solid.
NMR Spectrum: !H NMR (400MHz, DMSO) δ 1.66 - 1.77 (8H, m), 1.79 - 1.97 (4H, m), 2.29 - 2.38 (3H, m), 2.55 - 2.62 (3H, m), 3.61 (3H, s), 4.42 (2H, t), 4.65 (IH, ddt), 5.28 (IH, p), 6.96 (IH, dd), 8.02 (IH, dd), 8.18 (IH, dd), 8.34 (IH, d), 8.39 (IH, dd), 8.67 (IH, dd). Mass Spectrum: m/z (ES+)[M+H]+ = 479
The preparation of 8-(6-fluoro-3-pyridyl)-l-isopropyl-3-methyl-imidazo[4,5-c]cinnolin-2- one is described below.
8-(6-Fluoro-3- yridyl)-l-isopropyl-3-methyl-imidazor4,5-c1cinnolin-2-one
Figure imgf000070_0001
Dichlorobis(triphenylphosphine)palladium (II) (31.7 mg, 0.05 mmol) was added to a degassed solution of (6-fluoropyridin-3-yl)boronic acid (134 mg, 0.95 mmol), 8-chloro-l- isopropyl-3-methyl-imidazo[4,5-c]cinnolin-2-one (250 mg, 0.90 mmol) and 2M potassium carbonate solution (1.355 mL, 2.71 mmol) in 1,4-dioxane (3.16 mL) and the mixture heated in a microwave reactor at 80 °C for 1 hour under an inert atmosphere. The reaction mixture was diluted with EtOAc (25 mL), and washed sequentially with water (10 mL), saturated brine (10 mL) then the organic layer evaporated to afford the desired material as a brown solid which was used in the subsequent steps without further purification.
Mass Spectrum: m/z (ES+)[M+H]+ = 338
The preparation of 8-chloro-l-isopropyl-3-methyl-imidazo[4,5-c]cinnolin-2-one has been described previously.
3-(4-Fluoropiperidin-l-yl)propan-l-ol was prepared as described below. 3-(4-Fluoro iperidin- 1 -yPpropan- 1 -ol
Figure imgf000070_0002
To a solution of 4-fluoropiperidine (2.0 g, 19.39 mmol) in dry tetrahydrofuran (20 mL) at room temperature under nitrogen was added sodium hydride (1 g, 25.00 mmol) then stirred for thirty minutes. (3-Bromopropoxy)(tert-butyl)dimethylsilane (6.77 mL, 29.22 mmol) was added dropwise then allowed to stir for 24 h at ambient temperature. The reaction mixture was diluted with EtOAc (100 mL), and washed three times with water (3 x 50 mL). The organic layer was dried over MgS04, filtered and evaporated to afford crude product still bearing the silyl protecting group on the alcohol. This material was loaded onto 2 x 50g SCX columns washing with MeOH and the deprotected product eluted from the column using 2M NH3/MeOH to afford the desired material (2.226 g) as a yellow oil. NMR Spectrum: !H NMR (500MHz, CDCb) δ 1.63 - 1.77 (2H, m), 1.87 (4H, dq), 2.42 - 2.7 (6H, m), 3.74 - 3.87 (2H, m), 4.69 (1H, dt).
Example 13: 8- [6- [3-[(3R)-3-Fluoropyrrolidin-l-yl] propoxy]-3-pyridyl] -l-isopropyl-3- methyl-imidazo [4,5-c] cinnolin-2-one
Figure imgf000071_0001
3-[(3R)-3-Fluoro-l-pyrrolidinyl]-l-propanol (96 mg, 0.65 mmol) in THF (1.235 mL) was added dropwise to a stirred suspension of sodium hydride (52 mg, 1.30 mmol) in THF (1.2 mL) at 0 °C. The resulting suspension was stirred at 0 °C for 10 minutes under an inert atmosphere before a solution of 8-(6-fluoro-3-pyridyl)-l-isopropyl-3-methyl-imidazo[4,5- c]cinnolin-2-one (1 10 mg, 0.33 mmol) in DMF (1.0 mL) was added and the reaction mixture stirred at ambient temperature for 72 hours. The reaction mixture was diluted with ethyl acetate (40 mL), washed three times with water (3 x 20 mL) and the organic layer was dried over MgS04, filtered and evaporated. The crude product was purified by preparative HPLC to afford the desired product (4.7 mg) as a cream dry film.
NMR Spectrum: lH NMR (400MHz, CDCb) δ 1.79 (6H, d), 2.08 - 2.13 (2H, m), 2.75 (1H, td), 2.79 - 2.85 (2H, m), 2.89 - 3.12 (5H, m), 3.77 (3H, s), 4.46 (2H, t), 5.12 - 5.32 (2H, m), 6.91 (1H, dd), 7.84 (1H, dd), 7.92 (1H, dd), 8.20 (1H, d), 8.46 - 8.56 (2H, m).
Mass Spectrum: m/z (ES+) [M+H]+ = 465 The preparation of 8-(6-fluoro-3-pyridyl)-l-isopropyl-3-methyl-imidazo[4,5-c]cinnolin-2- one has been described earlier.
The preparation of 3-[(3R)-3-fluoro-l-pyrrolidinyl]-l-propanol is described below. 3-IY3R)-3-Fluoro- 1 -pyrrolidinyll - 1 -propanol
Figure imgf000072_0001
To a suspension of (3R)-3-fluoropyrrolidine hydrochloride (0.92 g, 7.33 mmol) in dry tetrahydrofuran (22 mL) at room temperature under nitrogen was added sodium hydride (0.88 g, 21.98 mmol) then stirred for thirty minutes. (3-Bromopropoxy)(tert- butyl)dimethylsilane (2.55 mL, 10.99 mmol) was added dropwise then allowed to stir for 72 hours at ambient temperature. The reaction mixture was diluted with EtOAc (50 mL), and washed two times with water (2 x 25 ml). The organic layer was dried over MgS04, filtered and evaporated to afford crude product still bearing the silyl protecting group on the alcohol.
This material was loaded onto 2 x 20g SCX washing with MeOH and the deprotected product eluted from the column using 2M NFb/MeOH to afford the desired material (0.268 g) as a colourless solid.
NMR Spectrum: !H NMR (400MHz, DMSO) δ 1.77 (2H, dq), 2.06 - 2.44 (2H, m), 3.06 - 3.25 (3H, m), 3.39 (2H, d), 3.48 (2H, t), 3.51 - 3.86 (2H, m), 5.42 (1H, d).
Example 14: l-Isopropyl-3-methyl-8-[2-methyl-6-[3-(l-piperidyl)propoxy]-3- pyridyl] imidazo [4,5-c] cinnolin-2-one
Figure imgf000072_0002
A solution of 3 -(l-piperidinyl)-l -propanol (0.152 mL, 1.00 mmol) in THF (3.70 mL) was added slowly to a stirred suspension of sodium hydride (0.048 g, 2.00 mmol) in THF (3.7 mL) under an inert atmosphere and the reaction stirred at ambient temperature for 30 minutes. A solution of 8-(6-fluoro-2-methyl-3-pyridyl)-l-isopropyl-3-methyl-imidazo[4,5- c]cinnolin-2-one (0.176 g, 0.50 mmol) in THF (3.7 mL) was then added to the above reaction and the mixture stirred at ambient temperature for 2 hours. The reaction was diluted with EtOAc (50 mL), the organic layer washed with brine (2 x 20 mL), dried over MgS04, filtered and evaporated. The crude product was purified by preparative HPLC to afford the desired product (0.06 g, 25 %) as a white solid.
NMR Spectrum: !H NMR (400 MHz, DMSO) δ 1.40 (2H, d), 1.51 (4H, p), 1.63 (6H, d), 1.90 (2H, p), 2.34 (4H, dd), 2.40 (2H, t), 2.46 (3H, s), 3.61 (3H, s), 4.35 (2H, t), 5.21 (1H, dt), 6.79 (1H, d), 7.71 - 7.82 (2H, m), 8.21 (1H, d), 8.39 (1H, d).
Mass Spectrum: m/z ES+ [M+H]+ = 474
8-(6-Fluoro-2-methyl-3-pyridyl)-l-isopropyl-3-methyl-imidazo[4,5-c]cinnolin-2-one was prepared as described below.
8-(6-Fluoro-2-methyl-3-pyridyl)-l-isopropyl-3-methyl-imidazo[4,5-clcinnolin-2-one
Figure imgf000073_0001
Dichlorobis(triphenylphosphine)palladium(II) (22.19 mg, 0.03 mmol) was added to a degassed mixture of 8-chloro-l-isopropyl-3-methyl-imidazo[4,5-c]cinnolin-2-one (175 mg, 0.63 mmol), (6-fluoro-2-methyl-3-pyridinyl)boronic acid (108 mg, 0.70 mmol) and 2M aqueous potassium carbonate solution (0.949 mL, 1.90 mmol) in 1,4-dioxane (5.37 mL) and heated to 90 °C for 1 hour in a microwave reactor under an inert atmosphere. The reaction mixture was diluted with 10% methanol in DCM (40 mL) and water (15 mL). The aqueous phase was extracted with 10% methanol in DCM (2 x 20 mL), then the combined organic phases dried over a phase separating cartridge and concentrated to afford the desired product, which was used immediately without purification.
Mass Spectrum: m/z ES+ [M+H]+ = 352 Example 15: l-(frans-3-Methoxycyclobutyl)-3-methyl-8-[4-[3-(l- piperidyl)propoxy] phenyl] imidazo [4,5-c] cinnolin-2-one
Figure imgf000074_0001
l, -Bis(di-tert-butylphosphino)ferrocene palladium dichloride (10.22 mg, 0.02 mmol) was added to a degassed solution of l-[3-[4-(4,4,5,5-tetramethyl-l ,3,2-dioxaborolan-2- yl)phenoxy]propyl]piperidine (1 14 mg, 0.33 mmol) and 8-chloro-l-(tra/?s-3- methoxycyclobutyl)-3-methyl-imidazo[4,5-c]cinnolin-2-one (100 mg, 0.31 mmol), 2M potassium carbonate solution (0.47 mL, 0.94 mmol) in 1 ,4-dioxane (1.1 mL) and the mixture was heated in a microwave reactor to 80 °C for 1 hour under an inert atmosphere. The reaction mixture was diluted with EtOAc (25 mL), washed sequentially with water (10 mL), saturated brine (10 mL) and the organic layer was evaporated. The crude product was purified by preparative HPLC to afford the desired product (46.2 mg, 29 %) as a brown dry film.
NMR Spectrum: *H NMR (400 MHz, CDCb) 1.19 (lH, t), 1.44 (2H, p), 1.60 (3H, p), 1.97 - 2.06 (2H, m), 2.42 (4H, s), 2.47 - 2.54 (2H, m), 2.59 - 2.7 (2H, m), 3.34 (3H, s), 3.36 - 3.44 (2H, m), 3.73 (3H, s), 4.10 (2H, t), 4.34 (1H, tt), 5.43 - 5.54 (1H, m), 7.03 - 7.09 (2H, m), 7.6 - 7.66 (2H, m), 7.87 (1H, dd), 8.08 (1H, d), 8.42 (1H, d).
Mass Spectrum: m/z (ES+) [M+H]+ = 502
The following compounds were synthesised in an analogous fashion using between 1-2 equivalents of the appropriate boronic ester, between 5 - 10 mol% of catalyst and the appropriate chloro intermediate.
Figure imgf000074_0002
Figure imgf000075_0001
Figure imgf000076_0001
Figure imgf000077_0001
* Reaction stirred for 2 hours at 80°C
Example 16: NMR Spectrum: lH NMR (400 MHz, DMSO) δ 1.89 (2H, p), 2.16 (6H, s), 2.38 (2H, t), 2.56 - 2.67 (2H, m), 3.16 - 3.22 (2H, m), 3.24 (3H, s), 3.60 (3H, s), 4.10 (2H, t), 4.26 (IH, tt), 5.53 (IH, ddd), 7.1 - 7.16 (2H, m), 7.82 - 7.86 (2H, m), 8.04 (IH, dd), 8.19 (IH, d), 8.36 (IH, d). Mass Spectrum: m/z (ES+) [M+H]+ = 462
Example 17: NMR Spectrum: lH NMR (400 MHz, DMSO) δ 1.62 - 1.78 (2H, m), 1.78 - 1.89 (2H, m), 1.89 - 1.96 (2H, m), 2.29 (IH, d), 2.44 (2H, d), 2.53 - 2.57 (IH, m), 2.63 (4H, td), 3.13 - 3.21 (2H, m), 3.23 (3H, s), 3.61 (3H, s), 4.29 (IH, ddd), 4.38 (2H, t), 4.67 (IH, ddt), 5.58 (IH, ddd), 6.95 - 7.07 (IH, m), 8.06 (IH, dd), 8.2 - 8.31 (2H, m), 8.39 (IH, d), 8.73 (IH, d). Mass Spectrum: m/z (ES+) [M+H]+ = 521 Example 18: NMR Spectrum: lH NMR (400 MHz, CDCb) δ 1.21 - 1.33 (IH, m), 1.84 - 1.97 (4H, m), 1.98 - 2.07 (2H, m), 2.34 - 2.49 (2H, m), 2.5 - 2.59 (2H, m), 2.62 (2H, d), 2.97 (2H, dddd), 3.10 (2H, tdd), 3.32 (3H, s), 3.77 (3H, s), 3.94 (IH, p), 4.44 (2H, t), 4.56 - 4.8 (IH, m), 4.98 - 5.15 (IH, m), 6.89 (IH, d), 7.86 (IH, dd), 8.00 (IH, dd), 8.41 (IH, d), 8.51 (IH, d), 8.57 (IH, d). Mass Spectrum: m/z (ES+) [M+H]+ = 521
Example 19: NMR Spectrum: lH NMR (400 MHz, DMSO) δ 1.34 - 1.43 (2H, m), 1.52 (4H, p), 1.91 (2H, p), 2.39 (6H, s), 2.75 - 2.9 (2H, m), 2.94 - 3.02 (2H, m), 3.21 (3H, s), 3.59 (3H, s), 3.88 (IH, p), 4.10 (2H, t), 5.01 - 5.19 (IH, m), 7.12 (2H, d), 7.82 - 7.91 (2H, m), 8.04 (IH, dd), 8.19 (IH, s), 8.36 (2H, d). Mass Spectrum: m/z (ES+) [M+H]+ = 502
Example 20: NMR Spectrum: lH NMR (400 MHz, CDC13) δ 1.98 - 2.25 (4H, m), 2.49 (IH, q), 2.64 - 2.79 (3H, m), 2.85 (2H, dq), 2.93 - 3.03 (2H, m), 3.04 - 3.17 (2H, m), 3.32 (3H, s), 3.77 (3H, s), 3.94 (IH, p), 4.45 (2H, t), 4.98 - 5.3 (2H, m), 6.89 (IH, d), 7.86 (IH, d), 8.00 (IH, dd), 8.41 (IH, s), 8.51 (IH, d), 8.57 (IH, d). Mass Spectrum: m/z (ES+) [M+H]+ = 507
Example 21 : NMR Spectrum: !H NMR (400 MHz, DMSO) δ 1.88 (4H, td), 2.17 (6H, s), 2.22 (IH, s), 2.39 (2H, t), 2.54 - 2.66 (IH, m), 3.38 - 3.46 (IH, m), 3.58 (3H, s), 3.94 (IH, d), 4.10 (2H, t), 4.12 - 4.24 (2H, m), 4.90 (IH, tt), 7.1 1 - 7.17 (2H, m), 7.78 - 7.84 (2H, m), 8.05 (IH, dd), 8.23 (OH, s), 8.26 (IH, d), 8.38 (IH, d . Mass Spectrum: m/z (ES+) [M+H]+ = 462
Example 22: NMR Spectrum: lH NMR (400 MHz, DMSO) δ 1.34 - 1.44 (2H, m), 1.51 (4H, p), 1.89 (4H, td), 2.08 (IH, s), 2.21 (IH, d), 2.29 - 2.38 (4H, m), 2.41 (2H, t), 3.39 - 3.47 (IH, m), 3.59 (3H, s), 3.95 (IH, d), 4.10 (2H, t), 4.13 - 4.24 (2H, m), 4.86 - 4.97 (IH, m), 7.1 1 - 7.19 (2H, m), 7.78 - 7.86 (2H, m), 8.06 (IH, dd), 8.27 (IH, d), 8.39 (IH, d). Mass Spectrum: m/z (ES+) [M+H]+ = 502
Example 23: NMR Spectrum: lH NMR (400 MHz, DMSO) δ 1.72 (2H, d), 1.76 - 1.96 (6H, m), 2.19 (IH, d), 2.25 - 2.35 (3H, m), 2.43 - 2.47 (3H, m), 2.58 - 2.65 (IH, m), 3.36 - 3.45 (IH, m), 3.59 (3H, s), 3.94 (IH, d), 4.12 - 4.2 (2H, m), 4.38 (2H, t), 4.67 (IH, dt), 4.94 (IH, s), 7.02 (IH, d), 8.08 (IH, dd), 8.22 (IH, dd), 8.32 (IH, s), 8.42 (IH, d), 8.70 (IH, d). Mass Spectrum: m/z (ES+) [M+H]+ = 521
Example 24: NMR Spectrum: lH NMR (400 MHz, DMSO) δ 1.25 (3H, d), 1.43 - 1.51 (IH, m), 1.65 (3H, s), 1.89 (2H, p), 2.16 (6H, s), 2.38 (2H, t), 3.58 (3H, s), 4.09 (2H, t), 7.1 - 7.17 (2H, m), 7.77 - 7.84 (2H, m), 8.07 (IH, dd), 8.36 (IH, d), 8.58 (IH, d). Mass Spectrum: m/z (ES+) [M+H]+ = 432
Example 25: NMR Spectrum: lH NMR (400 MHz, DMSO) δ 1.26 (3H, s), 1.36 - 1.43 (2H, m), 1.49 (6H, dt), 1.65 (3H, s), 1.90 (2H, p), 2.36 (2H, s), 2.39 - 2.46 (2H, m), 3.58 (4H, s), 4.09 (2H, t), 7.14 (2H, d), 7.81 (2H, d), 8.08 (IH, dd), 8.37 (IH, d), 8.58 (IH, d). Mass Spectrum: m/z (ES+) [M+H]+ = 472
Example 26: NMR Spectrum: lH NMR (400 MHz, DMSO) δ 1.18 - 1.33 (3H, m), 1.44 (IH, q), 1.65 (3H, s), 1.67 - 1.76 (2H, m), 1.77 - 1.96 (4H, m), 2.29 (2H, d), 2.45 (3H, t), 2.54 (IH, s), 3.59 (3H, s), 4.38 (2H, t), 4.56 - 4.79 (IH, m), 6.98 - 7.04 (IH, m), 8.10 (IH, dd), 8.19 (IH, dd), 8.40 (IH, d), 8.59 (IH, d), 8.67 (IH, d . Mass Spectrum: m/z (ES+) [M+H]+ = 491
Example 27: NMR Spectrum: lH NMR (400 MHz, DMSO) δ 1.18 - 1.33 (3H, m), 1.33 - 1.44 (3H, m), 1.48 (5H, tt), 1.65 (3H, s), 1.91 (2H, p), 2.33 (2H, d), 2.39 (3H, t), 3.59 (3H, s), 4.37 (2H, t), 7.00 (IH, dd), 8.10 (IH, dd), 8.19 (IH, dd), 8.40 (IH, d), 8.59 (IH, d), 8.67 (IH, dd). Mass Spectrum: m/z (ES+) [M+H]+ = 473
Example 27 can also be isolated as the methane sulfonic acid salt by taking the material as prepared above and subjecting to the following reaction conditions.
3 -methyl- 1 -( 1 -methylcyclopropyl)-8- {6-[3-( 1 -piperidinyl)propoxy] -3 -pyridinyl} -1,3- dihydro-2H-imidazo[4,5-c]cinnolin-2-one (44 mg, 0.09 mmol) was dissolved in DCM (2 mL) and treated with 1M methanesulfonic acid (6.33 μί, 0.10 mmol) in DCM, stirred for 5 minutes, then the mixture evaporated to dryness. The residue was triturated with diethyl ether to afford the methane sulfonic acid salt (21.8 mg, 41.3 %) as a pale yellow solid. NMR Spectrum: *H NMR (400 MHz, DMSO) δ 1.19 - 1.34 (3H, m), 1.43 (2H, dd), 1.67 (6H, s), 1.84 (2H, d), 2.19 (2H, dt), 2.92 (2H, q), 3.24 (2H, dt), 3.50 (2H, d), 3.59 (3H, s),
4.44 (2H, t), 7.04 (IH, d), 8.13 - 8.31 (2H, m), 8.41 (IH, d), 8.61 (IH, s), 8.69 (IH, d). Mass Spectrum: m/z (ES+) [M+H]+ = 473
Example 28: NMR Spectrum: !H NMR (400 MHz, DMSO) δ 1.88 (2H, p), 2.16 (6H, s), 2.17 - 2.29 (3H, m), 2.33 (IH, dt), 2.38 (3H, t), 2.46 (IH, d), 3.28 (3H, s), 3.60 (3H, s), 4.10 (3H, q), 5.49 (IH, p), 7.1 - 7.16 (2H, m), 7.78 - 7.84 (2H, m), 8.05 (IH, dd), 8.25 (IH, d), 8.38 (IH, d). Mass Spectrum: m/z (ES+) [M+H]+ = 476
Example 29: NMR Spectrum: !H NMR (400 MHz, DMSO) δ 1.39 (2H, q), 1.50 (4H, p), 1.79 - 1.94 (3H, m), 2.14 - 2.29 (3H, m), 2.33 (4H, d), 2.37 - 2.43 (3H, m), 2.45 - 2.48 (IH, m), 3.28 (3H, s), 3.59 (3H, s), 4.04 - 4.16 (3H, m), 5.47 (IH, p), 7.03 - 7.19 (2H, m), 7.73 - 7.88 (2H, m), 8.03 (IH, dd), 8.22 (IH, d), 8.36 (IH, d . Mass Spectrum: m/z (ES+)
[M+H]+ =516
Example 30: NMR Spectrum: !H NMR (400 MHz, DMSO) δ 1.88 (3H, dt), 2.16 (6H, s), 2.17 - 2.46 (7H, m), 3.28 (3H, s), 3.60 (3H, s), 4.05 - 4.19 (3H, m), 5.50 (IH, q), 7.06 - 7.22 (2H, m), 7.75 - 7.86 (2H, m), 8.05 (IH, dd), 8.24 (IH, d), 8.37 (IH, d). Mass
Spectrum: m/z (ES+) [M+H]+ = 476
Example 31: NMR Spectrum: lH NMR (400 MHz, DMSO) δ 1.4 - 1.48 (2H, m), 1.56 (4H, p), 1.83 - 2.02 (3H, m), 2.21 - 2.51 (11H, m), 3.33 (3H, s), 3.65 (3H, s), 4.20 (IH, p), 4.43 (2H, t), 5.58 (IH, p), 7.03 - 7.08 (IH, m), 8.12 (IH, dd), 8.26 (IH, dd), 8.38 (IH, d),
8.45 (IH, d), 8.73 - 8.78 (IH, m . Mass Spectrum: m/z (ES+) [M+H]+ = 517
Example 32: NMR Spectrum: lH NMR (400 MHz, DMSO) δ 1.77 - 2.41 (l lH, m), 2.53 - 2.66 (3H, m), 2.77 - 2.88 (2H, m), 3.27 (3H, s), 3.60 (3H, s), 4.1 - 4.18 (IH, m), 4.40 (2H, t), 5.19 (IH, ddd), 5.52 (IH, p), 7.01 (IH, dd), 8.08 (IH, d), 8.21 (IH, dd), 8.33 (IH, d), 8.40 (IH, d), 8.69 - 8.76 (IH, m). Mass Spectrum: m/z (ES+) [M+H]+ = 521 Example 33: NMR Spectrum: lH NMR (400 MHz, DMSO) δ 1.39 (2H, d), 1.50 (4H, p), 1.76 - 1.96 (3H, m), 2.14 - 2.48 (1 1H, m), 3.27 (3H, s), 3.59 (3H, s), 4.13 (IH, p), 4.36 (2H, t), 5.51 (IH, p), 6.96 - 7.05 (IH, m), 8.05 (IH, dd), 8.19 (IH, dd), 8.31 (IH, d), 8.38 (IH, d), 8.69 (IH, d . Mass Spectrum: m/z (ES+) [M+H]+ = 517
Example 34: NMR Spectrum: !H NMR (400 MHz, DMSO) δ 1.89 (2H, p), 2.18 (6H, s), 2.41 (2H, t), 2.84 (2H, td), 2.93 - 3.05 (2H, m), 3.21 (3H, s), 3.58 (3H, s), 3.88 (IH, p), 4.09 (2H, t), 5.07 (IH, p), 7.1 1 (2H, d), 7.85 (2H, d), 8.03 (IH, d), 8.34 (2H, d). Mass Spectrum: m/z (ES+) [M+H]+ = 462
Example 35: NMR Spectrum: !H NMR (400 MHz, DMSO) δ 1.39 (2H, d), 1.49 (4H, q), 1.91 (2H, p), 2.29 - 2.46 (6H, m), 2.78 - 2.87 (2H, m), 2.94 - 3.05 (2H, m), 3.21 (3H, s), 3.60 (3H, s), 3.89 (IH, p), 4.38 (2H, t), 5.06 - 5.18 (IH, m), 6.99 (IH, dd), 8.06 (IH, dd), 8.26 (IH, dd), 8.39 (IH, d), 8.45 (IH, d), 8.71 - 8.8 (IH, m . Mass Spectrum: m/z (ES+) [M+H]+ = 503
Example 36: NMR Spectrum: lH NMR (400 MHz, DMSO) δ 1.8 - 2.22 (5H, m), 2.29 - 2.33 (IH, m), 2.55 - 2.65 (4H, m), 2.77 - 2.88 (2H, m), 3.17 (2H, dd), 3.23 (3H, s), 3.61 (3H, s), 4.24 - 4.33 (IH, m), 4.40 (2H, t), 5.09 - 5.28 (IH, m), 5.54 - 5.65 (IH, m), 7.01 (IH, d), 8.06 (IH, dd), 8.21 - 8.32 (2H, m), 8.39 (IH, d), 8.73 (IH, d . Mass Spectrum: m/z (ES+) [M+H]+ = 507
The preparation of the chloro intermediates and boronic esters for Examples 15 - 36 have either been described earlier or are described below.
8-Chloro-l-(tra/? -3-methoxycvclobutyl)-3-methyl-imidazor4,5-c1cinnolin-2-one
Figure imgf000082_0001
l ,l-Dimethoxy-N,N-dimethylmethanamine (4.71 mL, 35.48 mmol) was added to 8-chloro- l-(trans-3-methoxycyclobutyl)-3H-imidazo[4,5-c]cinnolin-2-one (1.44 g, 4.73 mmol) in DMF (1 1 mL) and stirred at 100 °C for 2.5 hours. The mixture was cooled and the precipitate formed was collected by filtration, washing with water (30 mL) to afford desired product (0.5 g) as an orange solid. The aqueous layer was backextracted with DCM (2 x 25 mL), the combined organic layers washed with brine (3 x 25 mL), dried (MgS04) and concentrated to afford desired product (2.12 g) as a yellow solid (contaminated with a small quantity of DMF).
NMR Spectrum: *H NMR (400 MHz, DMSO) δ 2.52 - 2.63 (2H, m), 3.1 - 3.18 (2H, m), 3.23 (3H, d), 3.59 (3H, s), 4.25 (1H, dq), 5.43 (1H, tt), 7.73 (1H, dd), 8.24 (1H, d), 8.31 - 8.35 (lH, m).
Mass Spectrum: m/z (ES+) [M+H]+ = 319
8-Chloro-l-(trans-3-methoxycyclobutyl)-3H-imidazo[4,5-clcinnolin-2-one
Figure imgf000082_0002
l ,3,5-Trichloro-l ,3,5-triazinane-2,4,6-trione (0.549 g, 2.36 mmol) was added portionwise to a solution of 6-chloro-4-[(tra/75-3-methoxycyclobutyl)amino]cinnoline-3-carboxamide (1.45 g, 4.73 mmol) and 2,3,4,6,7,8,9,10-octahydropyrimido[l ,2-a]azepine (DBU) (1.414 ml, 9.45 mmol) in MeOH (22.22 ml) at 0 °C and the resulting solution allowed to warm to ambient temperature and stirred for 18 hours. The reaction mixture was concentrated to dryness, then partitioned between EtOAc (50 mL) and saturated aqueous NH4C1 (50 mL). A precipitated crashed out of solution which was collected by filtration, washed with diethyl ether (3 x lOmL) and azeotroped with MeCN (2 x 50 mL) to afford the desired material (1.79 g, 124 %) as a yellow solid.
Mass Spectrum: m/z (ES+) [M+H]+ = 305
6-Chloro-4-r(tra/? -3-methoxycvclobutyl)amino1cinnoline-3-carboxamide
Figure imgf000083_0001
7>a/?5-3-methoxycyclobutan-l -amine hydrochloride (0.719 g, 5.23 mmol) was added in one portion to 4,6-dichlorocinnoline-3-carboxamide (1.15 g, 4.75 mmol) and DIPEA (2.489 mL, 14.25 mmol) in acetonitrile (9.39 mL) and the resulting suspension was stirred at 90 °C for 18 hours. The reaction was allowed to cool, concentrated, diluted with water and the precipitate was collected by filtration, washed with water (20 mL) followed by acetonitrile (10 mL) and diethyl ether (10 mL) and dried under vacuum to afford the desired material (1.15 g, 79%) as an orange solid.
NMR Spectrum: lH NMR (400 MHz, DMSO) δ 2.27 - 2.35 (2H, m), 2.42 - 2.53 (2H, m), 3.19 (3H, s), 4.04 - 4.13 (1H, m), 4.65 - 4.75 (1H, m), 7.79 (1H, d), 7.86 - 7.9 (1H, m), 8.04 (1H, d), 8.21 (1H, d), 8.61 (1H, s), 10.80 (1H, d).
Mass Spectrum: m/z (ES+) [M+H]+ = 307
8-Chloro-l-(c -3-methoxycyclobutyl)-3-methyl-imidazo[4,5-clcinnolin-2-one
Figure imgf000083_0002
l ,l-Dimethoxy-N,N-dimethylmethanamine (0.981 mL, 7.38 mmol) was added to 8-chloro- l-(cz5-3-methoxycyclobutyl)-3H-imidazo[4,5-c]cinnolin-2-one (0.3 g, 0.98 mmol) in DMF (2.3 ml) and stirred at 100 °C for 16 hours. The aqueous layer was backextracted with DCM (2 x 25 mL), the combined organic layers washed with brine (3 x 25 mL), dried (MgS04) and concentrated to afford desired product (0.744 g) as a yellow liquid. The crude product was purified by flash silica chromatography, elution gradient 0 to 100% EtOAc in heptane followed by 10% MeOH/EtOAc, to afford the desired product (0.110 g, 35.1 %) as a white solid.
NMR Spectrum: *H NMR (400 MHz, DMSO) δ 2.78 (2H, d), 2.97 (2H, q), 3.22 (3H, s), 3.58 (3H, s), 3.86 (IH, p), 4.98 (IH, p), 7.74 (IH, d), 8.34 (IH, d), 8.49 (IH, s).
Mass Spectrum: m/z (ES+) [M+H]+ = 319 8-Chloro- 1 - cz5-3-methoxycyclobutyl)-3H-imidazo[4,5-c]cinnolin-2-one
Figure imgf000084_0001
l,3,5-Trichloro-l,3,5-triazinane-2,4,6-trione (0.824 g, 3.54 mmol) was added portionwise to a solution of 6-chloro-4-[(c 5-3-methoxycyclobutyl)amino]cinnoline-3-carboxamide (1.45 g, 4.73 mmol) and 2,3,4,6,7,8,9,10-octahydropyrimido[l,2-a]azepine (DBU) (2.12 mL, 14.2 mmol) in MeOH (22.22 ml) at 0 °C at 0 °C and the resulting solution allowed to warm to ambient temperature and stirred for 72 hours. The reaction mixture was concentrated to dryness, then partitioned between EtOAc (50 mL) and saturated aqueous NH4C1 (50 mL).
A precipitated crashed out of solution which was collected by filtration, washed with diethyl ether (3 x lOmL) and azeotroped with MeCN (2 x 50 mL) to afford the desired material (2.420 g, 168 %>) as a yellow solid.
NMR Spectrum: !H NMR (400 MHz, DMSO) δ 2.83 (2H, dddd), 2.98 (2H, dddt), 3.26 (3H, s), 3.90 (IH, p), 4.89 (IH, tt), 7.50 (IH, s), 7.68 (IH, dd), 8.30 (IH, d), 8.39 (IH, d). Mass Spectrum: m/z (ES+) [M+H]+ = 305
6-Chloro-4-r(c -3-methoxycvclobutyl)amino1cinnoline-3-carboxamide
Figure imgf000085_0001
Cis-3-methoxycyclobutan-l -amine hydrochloride (0.938 g, 6.82 mmol) was added in one portion to 4,6-dichlorocinnoline-3-carboxamide (1.65 g, 6.82 mmol) and DIPEA (3.57 mL, 20.45 mmol) in acetonitrile (13.47 mL) and the resulting suspension was stirred at 90 °C for 18 hours. The reaction was allowed to cool, concentrated, diluted with water and the precipitate was collected by filtration, washed with water (20 mL) followed by acetonitrile (10 mL) and diethyl ether (10 mL) to afford the desired material (1.450 g, 69.3 %) as a beige solid.
NMR Spectrum: lH NMR (400 MHz, DMSO) δ 1.77 - 2.1 (2H, m), 2.78 - 3.05 (2H, m), 3.21 (3H, s), 3.79 (1H, p), 4.26 (1H, h), 7.26 - 7.71 (1H, m), 7.85 (1H, dd), 8.15 (1H, d), 8.18 - 8.6 (2H, m), 10.19 - 10.57 (1H, m).
Mass Spectrum: m/z (ES+) [M+H]+ = 307
8-Chloro-3-methyl-l-r(3R)-tetrahvdropyran-3-yl1imidazor4,5-c1cinnolin-2-one
Figure imgf000085_0002
l,l-Dimethoxy-N,N-dimethylmethanamine (2.285 mL, 17.20 mmol) was added to 8- chloro-l-[(3R)-tetrahydropyran-3-yl]-3H-imidazo[4,5-c]cinnolin-2-one (699 mg, 2.29 mmol) in DMF (5.36 mL) and the mixture stirred at 100 °C for 2.5 hours. The mixture was cooled and the precipitate formed was filtered, washed with water (10 mL) and diethyl ether (10 mL) to afford the desired product (560 mg, 77 %) as an orange solid which was used without further purification.
Mass Spectrum: m/z (ES+) [M+H]+ = 319
8-chloro-l-r(3R)-tetrahvdropyran-3-yl1-3H-imidazor4,5-c1cinnolin-2-one
Figure imgf000086_0001
l,3,5-Trichloro-l,3,5-triazinane-2,4,6-trione (0.480 g, 12.06 mmol) was added portionwise to a stirred suspension of 6-chloro-4-[[(3R)-tetrahydropyran-3-yl]amino]cinnoline-3- carboxamide (1.1612 g, 3.79 mmol) and 2,3,4,6,7,8,9,10-octahydropyrimido[l,2-a]azepine (DBU) (1.234 mL, 8.82 mmol) in MeOH (18 mL) at 0 °C and warmed to ambient temperature for 90 minutes. A precipitate formed which was collected by filtration and washed with diethyl ether (3 x lOmL). The filtrate was adjusted to pH 7 with 2M HC1 and the precipitate formed was filtered, washed with water (10 mL), DCM:Et20 (1 : 1 ratio, 10 mL) and combined with the above solid to afford the desired material (0.837 g, 72.6 %) as a orange solid.
NMR Spectrum: !H NMR (400 MHz, DMSO) δ 1.57 - 1.72 (2H, m), 1.75 - 1.96 (2H, m), 3.45 - 3.58 (2H, m), 3.92 (1H, dd), 4.06 (1H, dd), 4.71 (1H, ddd), 7.70 (1H, dd), 8.21 (1H, d), 8.30 (1H, d), 11.47 (1H, s).
Mass Spectrum: m/z (ES+) [M+H]+ = 305
6-Chloro-4- [(3R)-tetrahydropyran-3 -yl] amino] cinnoline-3 -carboxamide
Figure imgf000086_0002
(3R)-Tetrahydro-2H-pyran-3-amine hydrochloride (0.942 g, 9.32 mmol) was added in one portion to 4,6-dichlorocinnoline-3-carboxamide (1.5039 g, 6.21 mmol) and DIPEA (4.76 mL, 18.64 mmol) in acetonitrile (27.8 mL) and stirred at 80 °C for 3 hours. The reaction was allowed to cool, concentrated, diluted with water and the precipitate was collected by filtration, washed with water (20 mL) followed by acetonitrile (10 mL) and diethyl ether (10 mL) and dried under vacuum to afford the desired material (1.161 g, 60.9 %) as a beige solid.
NMR Spectrum: !H NMR (400 MHz, DMSO) δ 1.48 - 1.62 (1H, m), 1.75 (2H, tt), 2.07 (1H, q), 3.54 (1H, dd), 3.62 (2H, t), 3.87 (1H, dd), 4.24 (1H, dq), 7.75 (1H, s), 7.89 (1H, dd), 8.17 (1H, d), 8.24 (1H, d), 8.59 (1H, s), 10.45 (1H, d).
Mass Spectrum: m/z (ES+) [M+H]+ = 307
8-Chloro-3-methyl- 1 -(1 -methylcyclopropyl)imidazo[4,5-clcinnolin-2-one
Figure imgf000087_0001
l,l-dimethoxy-N,N-dimethylmethanamine (3.23 ml, 24.30 mmol) was added to 8-chloro- l-(l-methylcyclopropyl)-3H-imidazo[4,5-c]cinnolin-2-one ( 0.89 g, 3.24 mmol) in DMF (7.57 ml) and the mixture stirred at 100 °C for 2.5 hours. The mixture was cooled and the precipitate formed was filtered, washed with water (10 mL) and diethyl ether (10 mL) to afford the desired product (0.57 g) as an orange solid. The aqueous layer was
backextracted with DCM (2 x 25 mL), the combined organic layers washed with brine (3 x 25 mL), dried (MgS04) and concentrated to afford desired product (0.25 g) as a red solid contaiminaed with a small quantity of DMF.
NMR Spectrum: lH NMR (400 MHz, DMSO) δ 1.09 - 1.29 (3H, m), 1.34 - 1.45 (1H, m), 1.60 (3H, s), 3.57 (3H, s), 7.75 (1H, dd), 8.33 - 8.39 (2H, m).
Mass Spectrum: m/z (ES+) [M+H]+ = 289
8-Chloro- 1 -(1 -methylcyclopropyl)-3H-imidazo[4,5-clcinnolin-2-one
Figure imgf000087_0002
l,3,5-Trichloro-l,3,5-triazinane-2,4,6-trione (0.739 g, 3.18 mmol) was added portionwise to a stirred suspension of 6-chloro-4-((l-methylcyclopropyl)amino)cinnoline-3- carboxamide (1.76 g, 6.36 mmol) and 2,3,4,6,7,8,9,10-octahydropyrimido[l,2-a]azepine (DBU) (1.902 mL, 12.72 mmol) in MeOH (29.9 mL) at 0 °C and warmed to ambient temperature for 90 minutes. A precipitate formed which was collected by filtration and washed with diethyl ether (3 x lOmL). The filtrate was adjusted to pH 7 with 2M HC1 and the precipitate formed was filtered, washed with water (10 mL), DCM:Et20 (1 : 1 ratio, 10 mL) and combined with the above solid to afford the desired material (0.992 g, 56.8 %) as a brown solid.
NMR Spectrum: lH NMR (400 MHz, DMSO) δ 1.07 - 1.25 (3H, m), 1.3 - 1.44 (1H, m), 1.58 (3H, s), 7.73 (1H, dd), 8.31 (1H, d), 8.36 (1H, d), 12.40 (1H, s).
Mass Spectrum: m/z (ES+) [M+H]+ = 275
6-Chloro-4-((l-methylcyclopropyl)amino)cinnoline-3 -carboxamide
Figure imgf000088_0001
1-methylcyclopropanamine hydrochloride (1.23 g, 17.33 mmol) was added in one portion to 4,6-dichlorocinnoline-3-carboxamide (2.8 g, 11.57 mmol) and DIPEA (9 mL, 52.05 mmol) in acetonitrile (51.8 mL) and the resulting suspension was stirred at 80 °C for 16 hours. The reaction was allowed to cool, concentrated, diluted with water and the precipitate was collected by filtration, washed with water (20 mL) followed by acetonitrile (10 mL) and diethyl ether (10 mL) and dried under vacuum to afford the desired material (1.76 g, 55.0 %) as a brown solid.
Mass Spectrum: m/z (ES+) [M+H]+ = 277
8-Chloro-l-[(lR,3R)-3-methoxycyclopentyll-3-methyl-imidazo[4,5-clcinnolin-2-one
Figure imgf000088_0002
Sodium hydride (60% in mineral oil) (156 mg, 3.89 mmol) was added to 8-chloro-l- [(lR,3R)-3-hydroxycyclopentyl]-3-methyl-imidazo[4,5-c]cinnolin-2-one (496 mg, 1.56 mmol) in DMF (15.35 mL) at 0 °C and stirred for 30 minutes. Methyl iodide (0.214 mL, 3.42 mmol) was added and stirred at ambient temperature for 20 hours. The reaction was quenched with water and the precipitate formed was collected by filtration and rinsed with water (10 mL) to afford the desired product (336 mg, 65%) as a yellow solid.
NMR Spectrum: !H NMR (400 MHz, DMSO) δ 1.23 (1H, s), 1.73 - 1.87 (1H, m), 2.11 - 2.29 (4H, m), 3.27 (3H, s), 3.59 (3H, s), 4.13 (1H, s), 5.37 (1H, p), 7.74 (1H, d), 8.30 (1H, s), 8.35 (1H, d).
Mass Spectrum: m/z (ES+) [M+H]+ = 333
8-Chloro-l-[(lR,3R)-3-hydroxycyclopentyll-3-methyl-imidazo[4,5-clcinnolin-2-one
Figure imgf000089_0001
Sodium hydride (60%> in mineral oil) (0.340 g, 8.50 mmol) was added to 8-chloro-l- [(lR,3R)-3-hydroxycyclopentyl]-3H-imidazo[4,5-c]cinnolin-2-one (1.0364 g, 3.40 mmol) in DMF (33.5 mL) at 0 °C and stirred for 20 minutes. Methyl iodide (0.468 ml, 7.48 mmol) was added and stirred to room temperature for 20 hours. The reaction was quenched with water and the precipitate formed was collected by filtration, rinsed with ether (10 mL) to afford the desired product (0.767 g, 71 %>) as a green solid.
NMR Spectrum: lH NMR (400 MHz, DMSO) δ 1.7 - 1.79 (1H, m), 2.02 - 2.12 (1H, m), 2.15 - 2.39 (4H, m), 3.61 (3H, s), 4.48 (2H, s), 5.43 (1H, p), 7.70 (1H, dd), 8.25 (1H, d), 8.32 - 8.39 (lH, m).
Mass Spectrum: m/z (ES+) [M+H]+ = 319
8-Chloro-l-[(lR,3R)-3-hydroxycyclopentyll-3H-imidazo[4,5-clcinnolin-2-one
Figure imgf000090_0001
l,3,5-Trichloro-l,3,5-triazinane-2,4,6-trione (0.739 g, 3.18 mmol) was added portionwise to a stirred suspension of 6-chloro-4-[[(lR,3R)-3-hydroxycyclopentyl]amino]cinnoline-3- carboxamide (1.211 g, 3.95 mmol) and 2,3,4,6,7,8,9,10-octahydropyrimido[l,2-a]azepine (DBU) (1.181 mL, 7.90 mmol) in MeOH (18.56 mL) at 0 °C and warmed to ambient temperature for 4 hours. A precipitate formed which was collected by filtration and washed with diethyl ether (3 x lOmL) to afford the desired product (1.036 g, 86 %) as a brown solid.
NMR Spectrum: lH NMR (400 MHz, DMSO) δ 1.58 - 1.74 (1H, m), 1.95 - 2.05 (1H, m), 2.1 - 2.3 (3H, m), 2.4 - 2.48 (1H, m), 4.43 (1H, s), 4.76 (1H, s), 5.40 (1H, p), 7.72 (1H, dd), 8.25 (1H, d), 8.31 (1H, d), 12.53 (1H, s).
Mass Spectrum: m/z (ES+) [M+H]+ = 305
6-Chloro-4-rr(lR,3R)-3-hvdroxycvclopentyl1amino1cinnoline-3-carboxamide
Figure imgf000090_0002
(lR,3R)-3-aminocyclopentanol hydrochloride (0.910 g, 6.61 mmol) was added in one portion to 4,6-dichlorocinnoline-3-carboxamide (1.6 g, 6.61 mmol) and DIPEA (3.46 mL, 19.83 mmol) in acetonitrile (29.6 mL). The resulting suspension was stirred at 80 °C for 1 hour.
The reaction was allowed to cool, concentrated, diluted with water and the precipitate was collected by filtration, washed with water (20 mL) followed by acetonitrile (10 mL) and diethyl ether (10 mL) and dried under vacuum to afford the desired material (1.211 g, 59.7 %) as a beige solid. NMR Spectrum: lH NMR (400 MHz, DMSO) δ 1.60 (2H, tt), 1.72 - 1.9 (1H, m), 1.97 (1H, ddd), 2.06 - 2.18 (1H, m), 2.27 - 2.37 (1H, m), 4.29 (1H, d), 4.63 - 4.82 (2H, m), 7.74 (1H, s), 7.89 (1H, dd), 8.21 (1H, d), 8.31 (1H, d), 8.58 (1H, s), 10.60 (1H, d).
Mass Spectrum: m/z (ES+) [M+H]+ = 307
8-Chloro-l-r(lS,3S)-3-methoxycvclopentyl1-3-methyl-imidazor4,5-c1cinnolin-2-one
Figure imgf000091_0001
Sodium hydride (60% in mineral oil) (201 mg, 5.02 mmol) was added to 8-chloro-l- [(lS,3S)-3-hydroxycyclopentyl]-3-methyl-imidazo[4,5-c]cinnolin-2-one (640 mg, 2.01 mmol) in DMF (19.8 mL) at 0 °C and stirred for 20 minutes. Methyl iodide (0.276 mL, 4.42 mmol) was added and stirred at ambient temperature for 20 hours. The reaction was quenched with water and the precipitate formed was collected by filtration and rinsed with water (10 mL) to afford the desired product (381 mg, 57 %) as an orange solid.
NMR Spectrum: !H NMR (400 MHz, DMSO) δ 1.76 - 1.88 (1H, m), 2.13 - 2.28 (4H, m), 3.27 (3H, s), 3.58 (3H, s), 4.11 (2H, m), 5.37 (1H, p), 7.74 (1H, dd), 8.29 (1H, d), 8.35 (1H, d).
Mass Spectrum: m/z (ES+) [M+H]+ = 333
8-Chloro-l-r(lS,3S)-3-hvdroxycvclopentyl1-3-methyl-imidazor4,5-c1cinnolin-2-one
Figure imgf000091_0002
Sodium hydride (60% in mineral oil) (0.210 g, 5.25 mmol) was added to 8-chloro-l- [(lS,3S)-3-hydroxycyclopentyl]-3H-imidazo[4,5-c]cinnolin-2-one (0.640 g, 2.10 mmol) in DMF (20.71 mL) at 0 °C and stirred for 20 minutes. Methyl iodide (0.289 mL, 4.62 mmol) was added and stirred at ambient temperature for 20 hours. The reaction was quenched with water and the precipitate formed was collected by filtration, rinsed with ether (10 mL) to afford the desired product (0.516 g). The filtrate was extracted with ether (50 mL), washed with brine (30 mL), dried over MgS04 and concentrated to afford the desired product (0.263 g) as an orange solid. The material was used without further purification. Mass Spectrum: m/z (ES+) [M+H]+ = 319
8-Chloro-l-[(lS,3S)-3-hydroxycyclopentyll-3H-imidazo[4,5-clcinnolin-2-one
Figure imgf000092_0001
l,3,5-Trichloro-l,3,5-triazinane-2,4,6-trione (0.462 g, 1.99 mmol) was added portionwise to a stirred suspension of 6-chloro-4-[[(lS,3S)-3-hydroxycyclopentyl]amino]cinnoline-3- carboxamide (1.14 g, 3.72 mmol) and 2,3,4,6,7,8,9,10-octahydropyrimido[l,2-a]azepine (1.19 mL, 7.95 mmol) in MeOH (17 mL) at 5°C then the solution was warmed to ambient temperature and stirred for 6 hours. A precipitate was observed which was collected by filtration, washed with diethyl ether (3 x lOmL) and dried under vacuum at 45°C to afford the desired material (5.94 g) as an orange solid, which was used without further purification.
NMR Spectrum: lH NMR (400 MHz, DMSO) δ 1.62 - 1.76 (1H, m), 1.97 - 2.07 (1H, m), 2.17 (1H, ddd), 2.2 - 2.3 (2H, m), 2.42 - 2.48 (1H, m), 4.43 (1H, s), 4.77 (1H, d), 5.40 (1H, p), 7.73 (1H, dd), 8.26 (1H, d), 8.32 (1H, d), 12.54 (1H, s).
Mass Spectrum: m/z (ES+) [M+H]+ = 305
6-Chloro-4-rr(lS,3S)-3-hvdroxycvclopentyl1amino1cinnoline-3-carboxamide
Figure imgf000093_0001
(lS,3S)-3-aminocyclopentan-l-ol hydrochloride (0.953 g, 6.93 mmol) was added in one portion to 4,6-dichlorocinnoline-3-carboxamide (1.5 g, 6.20 mmol) and DIPEA (4.05 mL) in acetonitrile (27.7 ml) and the resulting suspension was stirred at 80 °C for 90 minutes. The mixture was allowed to cool and diluted with water. The precipitate was collected by filtration, washed with water (20 mL) followed by acetonitrile (10 mL) and dried under vacuum to afford the desired material (1.140 g, 60.0 %) as a grey solid, which was used without further purification.
NMR Spectrum: !H NMR (400 MHz, DMSO) δ 1.55 - 1.66 (2H, m), 1.76 - 1.86 (IH, m), 1.97 (IH, dq), 2.05 - 2.13 (IH, m), 2.25 - 2.35 (IH, m), 4.24 - 4.32 (IH, m), 4.66 - 4.78 (2H, m), 7.73 (IH, s), 7.89 (IH, dd), 8.21 (IH, d), 8.31 (IH, d), 8.58 (IH, s), 10.60 (IH, d). Mass Spectrum: m/z (ES+) [M+H]+ = 307
2-[3-(4-Fluoro-l-piperidyl)propoxyl-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- vDpyridine
Figure imgf000093_0002
Dichlorobis(tricyclohexylphosphine)palladium (II) (22.11 mg, 0.03 mmol) was added to 5- bromo-2-[3-(4-fluoro-l-piperidyl)propoxy]pyridine (95 mg, 0.30 mmol), 4,4,4',4',5,5,5',5'- octamethyl-2,2'-bi-l,3,2-dioxaborolane (91 mg, 0.36 mmol) and potassium acetate (118 mg, 1.20 mmol) in 1,4-dioxane (3 mL) and the mixture degassed for 15 minutes. The resulting suspension was stirred at 90 °C for 16 hours under an inert atmosphere. The reaction mixture was evaporated to dryness, redissolved in DCM (50 mL), washed with water (40 mL) and the organic layer was dried with a phase separating cartridge, filtered and evaporated.
Mass Spectrum: m/z (ES+) [M+H]+ = 365 5-Bromo-2 3-(4-fluoro- 1 -piperidvQpropoxylpyridine
Figure imgf000094_0001
3-(4-Fluoro-l-piperidinyl)-l-propanol (0.302 g, 1.88 mmol) was added to a stirred suspension of sodium hydride (0.157 g, 3.92 mmol) in DMF (10 mL) at 25°C under nitrogen for 1 hour. 5-bromo-2-fluoropyridine (0.175 mL, 1.70 mmol) was added dropwise and the reaction was stirred at ambient temperature for 16 hours. Water (50 mL) was added portionwise to the mixture and the aqueous phase was extracted with DCM (3x50 mL). The organic phase was washed with water (50 mL), dried over a phase separator and evaporated.
The crude product was purified by ion exchange chromatography, using an SCX column. The desired product was eluted from the column using 1M NH3/MeOH to afford the desired product (0.300 g, 55.4 %) as a pale yellow oil.
NMR Spectrum: !H NMR (400 MHz, DMSO) δ 1.68 (2H, ddtd), 1.76 - 1.91 (5H, m), 2.22 - 2.32 (2H, m), 2.40 (2H, t), 2.50 (1H, m), 4.25 (2H, t), 4.66 (1H, dtt), 6.81 (1H, dd), 7.88 (1H, dd), 8.26 (1H, dd).
Mass Spectrum: m/z (ES+) [M+H]+ = 319
2-r3-r(3R)-3-Fluoropyrrolidin-l-yl1propoxy1-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yDpyridine
Figure imgf000094_0002
Dichlorobis(tricyclohexylphosphine)palladium (II) (48.7 mg, 0.07 mmol) was added to 5- bromo-2-[3-[(3R)-3-fluoropyrrolidin-l-yl]propoxy]pyridine (200 mg, 0.66 mmol), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi-l,3,2-dioxaborolane (201 mg, 0.79 mmol) and potassium acetate (259 mg, 2.64 mmol) in dioxane (6.60 mL) was added and the reaction was heated to 90°C for 18 hours under an inert atmosphere. The reaction mixture was evaporated, redissolved in DCM (10 mL), washed with water (10 mL) and the organic layer dried with a phase separating cartridge and evaporated to afford crude product.
Mass Spectrum: m z (ES+) [M+H]+ = 351
5-Bromo-2-r3-r(3R)-3-fluoropyrrolidin-l-yl1propoxy1pyridine
Figure imgf000095_0001
3-[(3R)-3-Fluoro-l-pyrrolidinyl]-l-propanol (0.306 g, 2.08 mmol) was added to a stirred suspension of sodium hydride (0.174 g, 4.35 mmol) in DMF (10 mL) at 25 °C under nitrogen for 1 hour. 5-Bromo-2-fluoropyridine (0.195 mL, 1.89 mmol) was then added dropwise and the reaction was stirred at ambient temperature for 18 hours. Water (50 mL) was added portionwise to the mixture and the aqueous phase was extracted with DCM (3x50 mL). The organic phase was washed with water (50 mL), dried over a phase separator and evaporated. The crude product was purified by ion exchange
chromatography, using an SCX column, to afford the desired product (0.819 g, 143 %) as a pale yellow oil.
NMR Spectrum: !H NMR (400 MHz, DMSO) δ 1.78 - 1.91 (3H, m), 2.02 - 2.18 (1H, m), 2.28 (1H, tdd), 2.53 - 2.56 (1H, m), 2.61 (1H, dd), 2.72 - 2.84 (2H, m), 2.99 (1H, s), 4.26 (2H, t), 5.17 (1H, dddt), 6.81 (1H, dd), 7.87 (1H, dd), 8.26 (1H, dd).
Mass Spectrum: m/z (ES+) [M+H]+ = 305
2-[3-[(3S)-3-Fluoropyrrolidin-l-yllpropoxyl-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- vDpyridine
Figure imgf000095_0002
Dichlorobis(tricyclohexylphosphine)palladium (II) (97.4 mg, 0.14 mmol) was added to 5- bromo-2-[3-[(3S)-3-fluoropyrrolidin-l-yl]propoxy]pyridine (200 mg, 0.66 mmol), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi-l,3,2-dioxaborolane (346 mg) and potassium acetate (259 mg, 2.64 mmol) in dioxane (6.60 mL) and the reaction was heated to 90°C for 24 hours under an inert atmosphere. The reaction mixture was evaporated, redissolved in DCM (10 mL), washed with water (10 mL) and the organic layer dried with a phase separating cartridge and evaporated to afford crude product, which was used without further purification.
Mass Spectrum: m/z (ES+) [M+H]+ = 351
5-Bromo-2-r3-r(3S)-3-fluoropyrrolidin-l-yl1propoxy1pyridine
Figure imgf000096_0001
3-[(3S)-3-Fluoro-l-pyrrolidinyl]-l-propanol (0.300 g, 2.04 mmol) was added to a stirred suspension of sodium hydride (0.170 g, 4.26 mmol) in DMF (10 mL) at 0°C under nitrogen, for 1 hour. 5-Bromo-2-fluoropyridine (0.191 mL, 1.85 mmol) was then added dropwise and the reaction was stirred at ambient temperature for 18 hours. Water (50 mL) was added portionwise to the mixture and the aqueous phase was extracted with DCM (3x50 mL). The organic phase was washed with water (50 mL), dried over a phase separator and evaporated. The crude product was purified by ion exchange
chromatography, using an SCX column, to afford the desired product (0.334 g, 60 %) as a pale yellow oil.
NMR Spectrum: !H NMR (400 MHz, DMSO) δ 1.78 - 1.95 (3H, m), 2.03 - 2.2 (1H, m), 2.28 (1H, q), 2.52 - 2.64 (2H, m), 2.74 - 2.85 (2H, m), 4.06 (1H, q), 4.26 (2H, t), 5.17 (1H, dtd), 6.81 (1H, d), 7.87 (1H, dd), 8.26 (1H, d).
Mass Spectrum: m/z (ES+) [M+H]+ = 303
3-|Y3S)-3-Fluoro- 1 -pyrrolidinyll- 1 -propanol
Figure imgf000096_0002
To a suspension of (S)-3-fluoropyrrolidine hydrochloride (1.00 g, 7.96 mmol) in dry tetrahydrofuran (37.1 mL) at ambient temperature under nitrogen was added sodium hydride (0.956 g, 23.89 mmol) then stirred for 15 minutes. (3-Bromopropoxy)(tert- butyl)dimethylsilane (2.77 mL, 11.95 mmol) was added dropwise then allowed to stir for 72 hours at ambient temperature. The reaction mixture was diluted with EtOAc (50 mL), and washed two times with water (2 x 25 ml). The organic layer was dried over MgS04, filtered and evaporated to afford crude product still bearing the silyl protecting group on the alcohol. The crude product (silyl) was loaded onto a 50g SCX column washing with DCM and methanol and the deprotected product eluted from the column using 2M
NEb/MeOH to afford the desired material (0.550 g, 47 %) as a beige oil.
NMR Spectrum: lH NMR (400 MHz, DMSO) δ 1.52 - 1.62 (2H, m), 1.74 - 1.92 (1H, m), 2.01 - 2.18 (1H, m), 2.2 - 2.28 (1H, m), 2.44 (2H, dd), 2.7 - 2.83 (2H, m), 3.43 (2H, t), 4.41 (1H, s), 5.16 (1H, dddt).
Example 37: 1:1 mixture of 8-[6-[3-(dimethylamino)propoxy]-3-pyridyl]-l-[(4S)-3,3- dimethyltetrahydropyr an-4-yl] -3-methyl-imidazo [4,5-c] cinnolin-2-one and 8- [6- [3- (dimethylamino)propoxy]-3-pyridyl]-l-[(4R)-3,3-dimethyltetrahydropyran-4-yl]-3- meth l-imidazo [4,5-c] cinnolin-2-one
Figure imgf000097_0001
Dichlorobis(triphenylphosphine)palladium (II) (20.24 mg, 0.03 mmol) was added to a degassed solution of N,N-dimethyl-3-[[5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2- pyridyl]oxy]propan-l -amine (185 mg, 0.61 mmol), a 1 : 1 mixture of 8-chloro-l-[(4R)-3,3- dimethyltetrahydropyran-4-yl] -3 -methyl-imidazo [4,5-c] cinnolin-2-one and 8-chloro- 1 - [(4S)-3,3-dimethyltetrahydropyran-4-yl]-3-methyl-imidazo[4,5-c]cinnolin-2-one (200 mg, 0.58 mmol) and 2M potassium carbonate solution (0.865 mL, 1.73 mmol) in 1,4-dioxane (4.90 mL) then the mixture heated in a microwave reactor at 80 °C for 1 hour under an inert atmosphere. The reaction mixture was diluted with EtOAc (25 mL), washed sequentially with water (10 mL), saturated brine (10 mL) and the organic layer evaporated. The crude product was purified twice by preparative HPLC, using basic then acidic modifiers, to afford the desired material (22.5 mg) as a cream solid.
NMR Spectrum: !H NMR (400MHz, DMSO) δ 0.82 (3H, s), 1.18 (3H, s), 1.79 (IH, d),
2.19 (6H, s), 2.41 (2H, t), 3.38 (2H, d), 3.54 (2H, d), 3.61 (3H, s), 3.67 - 3.77 (2H, m), 4.06
(IH, dd), 4.39 (2H, t), 5.03 (IH, dd), 7.04 (IH, d), 8.07 (IH, dd), 8.19 - 8.22 (IH, m), 8.23
(IH, d), 8.42 (IH, d), 8.60 (IH, d), 8.69 (IH, d).
Mass Spectrum: m/z (ES+)[M+H]+ = 491
The racemic mixture described above could be separated to give the individual
enantiomeric components, Examples 38 and 39, as described below. The stereochemistry of these components is currently unknown so they are defined as isomer 1 and isomer 2 as the first and second eluting products respectively.
A 1 : 1 mixture of 8-[6-[3-(dimethylamino)propoxy]-3-pyridyl]-l-[(4S)-3,3- dimethyltetrahydropyran-4-yl] -3 -methyl-imidazo [4,5 -c] cinnolin-2-one and 8-[6- [3 - (dimethylamino)propoxy] -3 -pyridyl] - 1 -[(4R)-3 ,3 -dimethyltetrahydropyran-4-yl] -3 -methyl- imidazo[4,5-c]cinnolin-2-one was separated by preparative chiral-HPLC SFC (Chiralcel OJ-H Technologies IC column, 20 μιη silica, 250 mm length, 5 micron), eluting isocratically with 90% supercritical C02 in methanol (modified with ammonia) as eluent, to afford the first eluting product, Example 38, as solid (30 mg, 29%), and the second eluting product, Example 39, as a solid (39 mg, 38 %>).
Example 38: Isomer 1
NMR Spectrum: lU NMR (400 MHz, DMSO) δ 0.81 (3H, s), 1.17 (3H, s), 1.78 (IH, d), 1.89 (2H, p), 2.16 (6H, s), 2.37 (2H, t), 3.38 (IH, d), 3.45 (IH, dd), 3.53 (IH, d), 3.61 (3H, s), 3.65 - 3.77 (IH, m), 4.06 (IH, dt), 4.38 (2H, t), 5.02 (IH, dd), 7.03 (IH, dd), 8.06 (IH, dd), 8.22 (IH, dd), 8.41 (IH, d), 8.59 (IH, d), 8.67 - 8.7 (IH, m).
Mass Spectrum: m/z (ES+) [M+H]+ = 491
Example 39: Isomer 2 NMR Spectrum: lH NMR (400 MHz, CDCb) δ 0.92 (3H, s), 1.33 (3H, s), 1.7 - 1.82 (1H, m), 1.97 - 2.07 (2H, m), 2.27 (6H, s), 2.44 - 2.53 (2H, m), 3.36 (1H, d), 3.57 (1H, dd), 3.65 - 3.72 (2H, m), 3.75 (3H, s), 4.24 - 4.32 (1H, m), 4.44 (2H, t), 4.82 (1H, dd), 6.92 (1H, dd), 7.82 (1H, dd), 7.88 (1H, dd), 8.34 (1H, d), 8.47 - 8.53 (2H, m).
Mass Spectrum: m/z (ES+) [M+H]+ = 491
The preparation of N,N-dimethyl-3-((5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)pyridin-2-yl)oxy)propan-l -amine has been described earlier.
The preparation of a 1 : 1 mixture of 8-chloro-l-[(4R)-3,3-dimethyltetrahydropyran-4-yl]-3- methyl-imidazo[4,5-c]cinnolin-2-one and 8-chloro-l-[(4S)-3,3-dimethyltetrahydropyran-4- yl]-3-methyl-imidazo[4,5-c]cinnolin-2-one is described below.
1 : 1 Mixture of 8-chloro-l-r(4R)-3,3-dimethyltetrahvdropyran-4-yl1-3-methyl-imidazor4,5- c] cinnolin-2-one and 8-chloro- 1 - |Y4S)-3 ,3 -dimethyltetrahydropyran-4-yl] -3 -methyl- imidazo[4,5-clcinnolin-2-one
Figure imgf000099_0001
l,l-Dimethoxy-N,N-dimethylmethanamine (10.38 mL, 78.12 mmol) was added to a 1 : 1 mixture of 8-chloro-l -[(4R)-3,3-dimethyltetrahydropyran-4-yl]-3H-imidazo[4,5- c]cinnolin-2-one and 8-chloro-l -[(4S)-3,3-dimethyltetrahydropyran-4-yl]-3H-imidazo[4, 5- c]cinnolin-2-one (2.6 g, 7.81 mmol) in DMF (20.9 mL) and the resulting slurry stirred at 100 °C for 22 hours. A precipitate formed which was collected by filtration, washed with diethyl ether (2 x 10 mL) and dried under vacuum to afford the desired material (1.48 g) as an orange solid. NMR Spectrum: lH NMR (400MHz, DMSO) δ 0.74 (3H, s), 1.13 (3H, s), 1.73 - 1.81 (1H, m), 3.45 (3H, d), 3.59 (3H, s), 3.67 - 3.77 (1H, m), 4.02 (1H, dd), 4.86 (1H, dd), 7.74 (1H, dd), 8.35 (1H, d), 8.58 (1H, d).
Mass Spectrum: m/z (ES+)[M+H]+ = 347
1 : 1 Mixture of 8-chloro-l-r(4R)-3,3-dimethyltetrahvdropyran-4-yll-3H-imidazor4,5- clcinnolin-2-one and 8-chloro-l-[(4S)-3,3-dimethyltetrahydropyran-4-yll-3H-imidazo[4,5- clcinnolin-2-one
Figure imgf000100_0001
l,3,5-Trichloro-l,3,5-triazinane-2,4,6-trione (1.059 g, 4.55 mmol) was added portionwise to a stirred suspension of a 1 : 1 mixture of 6-chloro-4-[[(4R)-3,3-dimethyltetrahydropyran- 4-yl] amino] cinnoline-3 -carboxamide and 6-chloro-4- [ [(4S)-3 ,3 -dimethyltetrahydropyran- 4-yl] amino] cinnoline-3 -carboxamide (3.05 g, 9.11 mmol) and 2,3,4,6,7,8,9,10- octahydropyrimido[l,2-a]azepine (2.72 mL, 18.22 mmol) in MeOH (42.8 mL) at 5°C and the resulting solution allowed to warm to ambient temperature and stirred for 90 minutes. The reaction mixture was concentrated to dryness, then partitioned between EtOAc (50 mL) and saturated aqueous NH4C1 (50 mL). A precipitated crashed out of solution which was collected by filtration, washed with diethyl ether (3 x 10 mL) and azeotroped with MeCN (2 x 50 mL) to afford the desired material (3.60 g) as a beige solid.
NMR Spectrum: lH NMR (400MHz, DMSO) δ 0.74 (3H, s), 1.14 (3H, s), 1.74 - 1.83 (1H, m), 3.38 - 3.52 (3H, m), 3.64 - 3.81 (1H, m), 4.02 (1H, dd), 4.80 (1H, dd), 7.71 (1H, dd), 8.31 (1H, d), 8.54 (1H, d).
Mass Spectrum: m/z (ES+)[M+H]+ = 332
1 : 1 Mixture of 6-chloro-4-[[(4R)-3,3-dimethyltetrahydropyran-4-yl]amino]cinnoline-3- carboxamide and 6-chloro-4- Γ IY4S)-3 ,3 -dimethyltetrahydropyran-4- yl] amino] cinnoline-3 - carboxamide
Figure imgf000101_0001
A 1 : 1 mixture of (lR)-3,3-dimethyltetrahydro-2H-pyran-4-amine hydrochloride and (1S)- 3,3-dimethyltetrahydro-2H-pyran-4-amine hydrochloride (2.26 g, 13.63 mmol) was added in one portion to 4,6-dichloro-3-cinnolinecarboxamide (3 g, 12.39 mmol) and DIPEA (6.5 mL, 37.18 mmol) in acetonitrile (24.49 mL) and the resulting suspension stirred at 90 °C for 18 hours. The reaction was allowed to cool, diluted with water and the precipitate was collected by filtration, washed with water (20 mL) followed by acetonitrile (10 mL) and dried under vacuum to afford the desired material (3.05 g) as a beige solid.
NMR Spectrum: !H NMR (400MHz, DMSO) δ 0.80 (3H, s), 1.10 (3H, s), 1.80 (1H, dtd), 1.96 (1H, dt), 3.25 (1H, d), 3.44 (1H, d), 3.52 (1H, td), 3.89 (1H, dt), 4.06 (1H, td), 7.83 (1H, s), 7.91 (1H, dd), 8.16 (1H, d), 8.26 (1H, d), 8.67 (1H, s), 10.40 (1H, d).
Mass Spectrum: m/z (ES+)[M+H]+ = 335
Example 40: 1:1 mixture of l-[(4S)-3,3-dimethyltetrahydropyran-4-yl]-3-methyl-8-[4- [3-(l-piperidyl)propoxy]phenyl]imidazo[4,5-c]cinnolin-2-one and l-[(4R)-3,3- dimethyltetrahydropyran-4-yl]-3-methyl-8-[4-[3-(l- pi eridyl)propoxy] phenyl] imidazo [4,5-c] cinnolin-2-one
Figure imgf000101_0002
Dichlorobis(triphenylphosphine)palladium (II) (14.3 mg, 0.02 mmol) was added to a degassed solution of l-(3-(4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)phenoxy)propyl)piperidine (151 mg, 0.44 mmol) and a 1 : 1 mixture of 8-chloro-l-[(4R)- 3 ,3 -dimethyltetrahydropyran-4-yl] -3 -methyl-imidazo [4,5-c] cinnolin-2-one and 8-chloro- 1 - [(4S)-3,3-dimethyltetrahydropyran-4-yl]-3-methyl-imidazo[4,5-c]cinnolin-2-one (152 mg, 0.44 mmol), 2M potassium carbonate solution (0.66 mL, 1.31 mmol) in 1,4-dioxane (1.53 niL) then the mixture heated in a microwave reactor at 80 °C for 1 hour under an inert atmosphere. The reaction mixture was diluted with EtOAc (25 mL), washed sequentially with water (10 mL), saturated brine (10 mL) and the organic layer evaporated. The crude product was purified by preparative HPLC to afford the desired material (81 mg) as a brown solid.
NMR Spectrum: !H NMR (400MHz, DMSO) δ 0.82 (3H, s), 1.18 (3H, s), 1.33 - 1.43 (2H, m), 1.50 (4H, p), 1.79 (1H, d), 1.90 (2H, p), 2.3 - 2.38 (2H, m), 2.40 (3H, t), 3.17 (1H, d), 3.35 (1H, d), 3.38 - 3.49 (1H, m), 3.56 (1H, d), 3.60 (3H, s), 3.62 - 3.7 (1H, m), 4.06 (1H, d), 4.10 (2H, t), 5.00 (1H, dd), 7.16 (2H, d), 7.79 - 7.86 (2H, m), 8.05 (1H, dd), 8.38 (1H, d), 8.52 - 8.56 (lH, m).
Mass Spectrum: m/z (ES+)[M+H]+ = 530
Example 41: 1:1 mixture of 8-[4-[3-(dimethylamino)propoxy]phenyl]-l-[(4S)-3,3- dimethyltetrahydropyr an-4-yl] -3-methyl-imidazo [4,5-c] cinnolin-2-one and 8- [4- [3- (dimethylamino)propoxy]phenyl]-l-[(4R)-3,3-dimethyltetrahydropyran-4-yl]-3- methyl-imidazo [4,5-c] cinnolin-2-one
Figure imgf000102_0001
Dichlorobis(triphenylphosphine)palladium (II) (71.6 mg, 0.10 mmol) was added to a degassed solution of N,N-dimethyl-3-(4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2- yl)phenoxy)propan-l -amine (179 mg, 0.59 mmol) and a 1 : 1 mixture of 8-chloro- 1-[(4R)- 3 ,3 -dimethyltetrahydropyran-4-yl] -3 -methyl-imidazo [4,5-c] cinnolin-2-one and 8-chloro- 1 - [(4S)-3,3-dimethyltetrahydropyran-4-yl]-3-methyl-imidazo[4,5-c]cinnolin-2-one (177 mg, 0.51 mmol), 2M potassium carbonate solution (0.77 mL, 1.53 mmol) in 1,4-dioxane (4.4 mL then the mixture heated in a microwave reactor at 100 °C for 90 minutes under an inert atmosphere. The reaction mixture was diluted with EtOAc (25 mL), and washed sequentially with water (10 mL), saturated brine (10 mL) and the organic layer was evaporated. The crude product was purified by preparative HPLC to afford the desired material (82 mg).
NMR Spectrum: *H NMR (400 MHz, DMSO) δ 0.83 (3H, s), 1.19 (3H, s), 1.80 (lH, d), 1.89 (2H, p), 2.17 (6H, s), 2.39 (2H, t), 3.32 - 3.5 (2H, m), 3.54 - 3.72 (5H, m), 4.04 - 4.15 (3H, m), 5.01 (1H, dd), 7.13 - 7.2 (2H, m), 7.8 - 7.87 (2H, m), 8.06 (1H, dd), 8.38 (1H, d), 8.54 (1H, d).
Mass Spectrum: m/z (ES+) [M+H]+ = 490
The above material can also be isolated as the methane sulfonic acid salt by taking the material as prepared above and subjecting to the following reaction conditions.
A 1 : 1 mixture of 8-{4-[3-(dimethylamino)propoxy]phenyl}-l-[(4S)-3,3- dimethyltetrahydro-2H-pyran-4-yl] -3 -methyl- 1 ,3 -dihydro-2H-imidazo [4,5 -c] cinnolin-2- one and 8- {4-[3-(dimethylamino)propoxy]phenyl} - 1 -[(4R)-3,3-dimethyltetrahydro-2H- pyran-4-yl]-3-methyl-l,3-dihydro-2H-imidazo[4,5-c]cinnolin-2-one (20 mg, 0.04 mmol) was dissolved in DCM (2 mL) and treated with 1M methanesulfonic acid (0.003 mL, 0.04 mmol) in DCM then the mixture evaporated to dryness. The residue was triturated with diethyl ether to afford the methane sulfonic acid salt (18.9 mg, 79%) as a pale yellow solid. NMR Spectrum: !H NMR (400 MHz, DMSO) δ 0.88 (3H, s), 1.24 (3H, s), 1.82 - 1.9 (1H, m), 2.16 - 2.24 (2H, m), 2.39 (6H, s), 2.85 - 2.92 (3H, m), 3.27 - 3.38 (2H, m), 3.4 - 3.5 (1H, m), 3.58 - 3.67 (2H, m), 3.72 - 3.95 (1H, m), 4.1 - 4.16 (1H, m), 4.21 (2H, t), 4.95 - 5.04 (1H, m), 7.17 - 7.23 (2H, m), 7.81 - 7.87 (2H, m), 8.06 (1H, dd), 8.40 (1H, d), 8.53 (1H, s).
Mass Spectrum: m/z (ES+) [M+H]+ = 490
The racemic mixture described above could be separated to give the individual
enantiomeric components, Examples 42 and 43, as described below. The stereochemistry of these components is currently unknown so they are defined as isomer 1 and isomer 2 as the first and second eluting products respectively. A 1 : 1 mixture of 8-{4-[3-(dimethylamino)propoxy]phenyl}-l-[(4S)-3,3- dimethyltetrahydro-2H-pyran-4-yl] -3 -methyl- 1 ,3 -dihydro-2H-imidazo [4,5 -c] cinnolin-2- one and 8- {4-[3-(dimethylamino)propoxy]phenyl} - 1 -[(4R)-3,3-dimethyltetrahydro-2H- pyran-4-yl]-3-methyl-l,3-dihydro-2H-imidazo[4,5-c]cinnolin-2-one was separated by preparative chiral-HPLC SFC (Phenomenex LUX CI column, 30 μιη silica, 250 mm length, 5 micron), eluting isocratically with 90% supercritical C02 in methanol (modified with ammonia) as eluent, to afford the first eluting product, Example 42, as solid (22 mg, 37%), and the second eluting product, Example 43, as a solid (24 mg, 40%>).
Example 42:Isomer 1
NMR Spectrum: *H NMR (400 MHz, DMSO) δ 0.83 (3H, s), 1.19 (3H, s), 1.81 (1H, d), 1.89 (2H, p), 2.17 (6H, s), 2.39 (2H, t), 3.35 (1H, d), 3.38 - 3.51 (1H, m), 3.57 (1H, d), 3.61 (3H, s), 3.63 - 3.72 (1H, m), 4.03 - 4.15 (3H, m), 5.01 (1H, dd), 7.12 - 7.23 (2H, m), 7.78 - 7.87 (2H, m), 8.06 (1H, dd), 8.38 (1H, d), 8.55 (1H, d).
Mass Spectrum: m/z (ES+) [M+H]+ = 490
Example 43:Isomer 2
NMR Spectrum: lU NMR (400 MHz, DMSO) δ 0.83 (3H, s), 1.19 (3H, s), 1.81 (1H, d), 1.89 (2H, p), 2.17 (6H, s), 2.39 (2H, t), 3.35 (1H, d), 3.38 - 3.51 (1H, m), 3.57 (1H, d), 3.61 (3H, s), 3.63 - 3.72 (1H, m), 4.03 - 4.15 (3H, m), 5.01 (1H, dd), 7.12 - 7.23 (2H, m), 7.78 - 7.87 (2H, m), 8.06 (1H, dd), 8.38 (1H, d), 8.55 (1H, d).
Mass Spectrum: m/z (ES+) [M+H]+ = 490
The preparation of a 1 : 1 mixture of 8-chloro-l-[(4R)-3,3-dimethyltetrahydropyran-4-yl]-3- methyl-imidazo[4,5-c]cinnolin-2-one and 8-chloro-l-[(4S)-3,3-dimethyltetrahydropyran-4- yl]-3-methyl-imidazo[4,5-c]cinnolin-2-one and the preparation of N,N-dimethyl-3-[4- (4,4,5, 5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenoxy]-l-propanamine has been described earlier. Example 44: 8-[4-[3-(Dimethylamino)propoxy]phenyl]-7-fluoro-l-isopropyl-3-methyl- imidazo [4,5-c] cinnolin-2-one
Figure imgf000105_0001
(2 ' -Amino-2-biphenylyl)(chloro)palladium - dicyclohexyl(2 ' ,4 ' ,6 ' -triisopropyl-2- biphenylyl)phosphine (1 : 1) (25.8 mg, 0.03 mmol) was added to N,N-dimethyl-3-[4- (4,4,5, 5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenoxy]-l-propanamine (lOOmg, 0.33 mmol), 8-bromo-7-fluoro-l-isopropyl-3-methyl-imidazo[4,5-c]cinnolin-2-one (111 mg, 0.33 mmol) and CS2CO3 (213 mg, 0.66 mmol) in 1,4-dioxane (2 mL) and water (0.4 mL) and the mixture was stirred at 80 °C for 2 hours. The crude product was purified by flash silica chromatography, elution gradient 0 to 10% MeOH in DCM. Pure fractions were evaporated to dryness to afford product as a brown oil. The crude product was purified by preparative HPLC to afford the desired product (50 mg, 35 %>) as a brown solid.
NMR Spectrum: !H NMR (300 MHz, DMSO) δ 1.65 (6H, d), 1.82-1.96 (2H, m), 2.20 (6H, s), 2.38-2.45 (2H, m), 3.60 (3H, s), 4.05-4.15 (2H, m), 5.15-5.25 (1H, m), 7.12 (2H, d), 7.68 (2H, d), 8.18 (1H, d), 8.25 (1H, d).
Mass Spectrum: m/z (ES+) [M+H]+ = 438
8-Bromo-7-fluoro-l-isopropyl-3-methyl-imidazo[4,5-c]cinnolin-2-one was prepared as described below.
8-Bromo-7-fluoro-l-isopropyl-3-methyl-imidazor4,5-c1cinnolin-2-one
Figure imgf000105_0002
l,l-Dimethoxy-N,N-dimethylmethanamine (7.00 mL, 52.28 mmol) was added to 8-bromo- 7-fluoro-l-isopropyl-3H-imidazo[4,5-c]cinnolin-2-one (1.7 g, 5.23 mmol) in DMF (1 mL) and the mixture stirred at 100 °C for 3 hours. The reaction mixture was diluted with water.
The precipitate was collected by filtration, washed with water (100 mL) and dried under vacuum to afford the desired product (0.80 g, 45.1 %) as a red solid.
NMR Spectrum: ¾ NMR (300MHz, DMSO) δ 1.50-1.60 (6H, m), 3.50 (3H, s), 5.05-5.22
(1H, m),8.24 (1H, d), 8.72 (1H, d).
Mass Spectrum: m/z (ES+) [M+H]+ = 339
8-Bromo-7-fluoro- 1 -isopropyl-3H-imidazo[4,5-clcinnolin-2-one
Figure imgf000106_0001
l,3,5-Trichloro-l,3,5-triazinane-2,4,6-trione (0.906 g, 3.90 mmol) was added to 6-bromo- 7-fluoro-4-(isopropylamino)cinnoline-3-carboxamide (2.55 g, 7.79 mmol) and
2,3,4,6,7,8,9,10-octahydropyrimido[l,2-a]azepine (2.350 mL, 15.59 mmol) in MeOH (1.5 mL) at 0°C and warmed to ambient temperature for 2 hours. The reaction mixture was evaporated and the crude solid triturated with diethyl ether and collected by filtration to afford the desired product (1.80 g, 71 %) as a light-brown solid, which was used without further purification.
NMR Spectrum: !H NMR (300MHz, DMSO) δ 1.40-1.60 (6H, m), 3.40-3.50 (1H, m), 5.03-5.13 (1H, m), 8.07 (1H, d), 8.57 (1H, d).
Mass Spectrum: m/z (ES+) [M+H]+ = 325
6-Bromo-7-fluoro-4-(isopropylamino)cinnoline-3-carboxamide
Figure imgf000106_0002
Propan-2-amine (0.563 g, 9.52 mmol) was added to 6-bromo-4-chloro-7-fluoro-cinnoline- 3-carboxamide (2.9 g, 9.52 mmol) and DIPEA (3.33 mL, 19.05 mmol) in acetonitrile (25 mL) at 20°C and stirred at 80 °C for 3 hours. The precipitate was collected by filtration, washed with diethyl ether (20 mL) and air dried to afford the desired product (2.60 g, 83 %) as a grey solid, which was used without further purification.
NMR Spectrum: !H NMR (300MHz, DMSO) δ 1.20-1.30 (6H, m), 4.40-4.50 (1H, m), 7.80 (1H, s), 8.08 (1H, d), 8.50-8.60 (2H, m), 10.45 (1H, d).
Mass Spectrum: m/z (ES+) [M+H]+ = 327
6-Bromo-4-chloro-7-fluoro-cinnoline-3-carboxamide
Figure imgf000107_0001
Ammonium hydroxide (17.31 g, 493.93 mmol) was added dropwise to 6-bromo-4-chloro- 7-fluoro-cinnoline-3-carbonyl chloride (3.2 g, 9.88 mmol) in acetonitrile (50 mL) at 0°C under nitrogen. The resulting mixture was stirred at ambient temperature for 1 hour and the precipitate was collected by filtration. The solid was washed with diethyl ether (20 mL) and air dried to afford the desired product (2.95 g, 98 %)as a grey solid, which was used without further purification.
NMR Spectrum: !H NMR (300MHz, DMSO) δ 7.75 (2H, d), 8.50 (1H, s), 8.62-8.80 (1H, m).
Mass Spectrum: m/z (ES+) [M+H]+ = 306
6-Bromo-4-chloro-7-fluoro-cinnoline-3-carbonyl chloride
Figure imgf000107_0002
DMF (0.081 mL, 1.05 mmol) was added dropwise to 6-bromo-7-fluoro-4-hydroxy- cinnoline-3-carboxylic acid (3 g, 10.45 mmol) in thionyl chloride (25 mL) over a period of 1 minute at ambient temperature under an inert atmosphere. The resulting solution was stirred at 80 °C for 90 minutes then allowed to cool, concentrated to dryness to afford the desired product (3.20 g, 95 %) as brown oil, which was used without further purification. NMR Spectrum: lH NMR (400MHz, CDCb) δ 8.37(1H, d), 8.74(1H, d).
Mass Spectrum: rM-2Cl+20C¾l+ = 315
6-Bromo-7-fluoro-4-h droxy-cinnoline-3-carboxylic acid
Figure imgf000108_0001
A solution of 4-amino-6-bromo-7-fluoro-cinnoline-3-carboxamide (36 g, 126.28 mmol) in DMSO/ 4M sulphuric acid (4 M) (400 mL/1000 mL) at ambient temperature. The solution was stirred for 8 days at 130 °C, cooled to ambient temperature and quenched with water/ice (3000 mL). The solids formed were collected by filtration, washed with water (2 x 500 mL) and oven dried. The crude product was purified by re-crystallization from DMSO to afford the desired product (18.4 g, 51%) as a light brown solid.
NMR Spectrum: !H NMR (300 MHz, DMSO) δ 7.68-7.71 (1H, d), 8.47-8.52 (1H, m), 13.79-14.76 (2H, br).
Mass Spectrum: m/z (ES+) [M+H]+ = 286.9 4-Amino-6-bromo-7-fluoro-cinnoline-3-carboxamide
Figure imgf000108_0002
Aluminium trichloride (54 g, 404.98 mmol) was added to a solution of 2-[(4-bromo-3- fluorophenyl)hydrazono]-2-cyanoacetamide (39 g, 136.81 mmol) in 1 ,2-dichlorobenzene (500 mL) under an inert atmosphere at ambient temperature. The solution was stirred for 2 days at 120 °C, then cooled and quenched with iced water (3000 mL). The solids formed were collected by filtration, washed with water (3 x 500 mL) and oven dried to afford the desired material (36 g, 92%) as a yellow solid.
Mass Spectrum: m/z (ES+) [M+2H]+ = 285
2-[(4-Bromo-3-fluorophenyl)hydrazonol-2-cyanoacetamide
Figure imgf000109_0001
4-Bromo-3-fluoroaniline (30 g, 157.88 mmol), concentrated hydrochloric acid (120 g, 3.29 mol), acetic acid (190 g, 3.16 mol) and water (500 g, 27.75 mol) were added to a round bottom flask under an inert atmosphere at ambient temperature. To the mixture was added a solution of sodium nitrite (12 g, 178.27 mmol) in H20 (300 mL) dropwise with stirring at 0-5 °C, then the solution was stirred for 2 hours at 0 °C. NaOAc (100 g, 1.22 mol) was added portionwise at 0 °C and the solution stirred for 30 minutes at 0 °C. 2- Cyanoacetamide (16 g, 194.47 mmol), EtOH (390 g, 8.47 mol), water (2500 g, 138 mol) were added to a second round bottom flask under an inert atmosphere at ambient temperature, then NaOAc (500 g, 6.10 mol, 6.10 equiv) was added at ambient temperature. To the mixture was added the solution in the first flask dropwise with stirring at 0-5 °C. The resulting solution was stirred for 2 hours at 0 °C. The solids were collected by filtration, and washed with water (3 x 1 L) and oven dried to afford the desired material (45 g) as a yellow solid, which was used without further purification.
Mass Spectrum: m/z (ES+) [M+H]+ = 282.9
Example 45: 8- [6- [3-(Dimethylamino)propoxy] -3-pyridyl] -7-fluoro-l-isopropyl-3- methyl-imidazo [4,5-c] cinnolin-2-one
Figure imgf000109_0002
(2 ' -Amino-2-biphenylyl)(chloro)palladium - dicyclohexyl(2 ' ,4 ' ,6 ' -triisopropyl-2- biphenylyl)phosphine (1 : 1) (23.20 mg, 0.03 mmol) was added to 8-bromo-7-fluoro-l- isopropyl-3-methyl-imidazo[4,5-c]cinnolin-2-one_(100 mg, 0.29 mmol), N,N-dimethyl-3- ((5-(4,4,5 ,5 -tetramethyl- 1 ,3 ,2-dioxaborolan-2-yl)pyridin-2-yl)oxy)propan- 1 -amine (117 mg, 0.38 mmol) and Cs2C03 (240 mg, 0.74 mmol) in 1,4-dioxane (2.5 mL) and water (0.5 mL) and the mixture was stirred at 100 °C for 3 hours under an inert atmosphere. The crude product was purified by preparative HPLC to afford the desired product (21 mg, 16 %) as a yellow solid.
NMR Spectrum: lH NMR (400 MHz, CDCb) δ 1.78 (6H, d), 2.02-2.09 (2H, m), 2.33 (6H, s), 2.49-2.55 (2H, m), 3.78 (3H, s), 4.47 (2H, t), 5.17 (1H, t), 6.92 (1H, d), 7.91-7.94 (1H, m), 8.15(1H, t), 8.19 (1H, s), 8.44 (1H, d).
Mass Spectrum: m/z (ES+) [M+H]+ = 439
Example 46 : 7-Fluoro- l-isopropyl-3-methyl-8- [6- [3-(l -piperidyl)propoxy] -3- p ridyl] imidazo [4,5-c] cinnolin-2-one
Figure imgf000110_0001
(2 ' -Amino-2-biphenylyl)(chloro)palladium - dicyclohexyl(2 ' ,4 ' ,6 ' -triisopropyl-2- biphenylyl)phosphine (1 : 1) (22.73 mg, 0.03 mmol) was added to 2-(3-(piperidin-l- yl)propoxy)-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)pyridine (lOOmg, 0.29 mmol), 8-bromo-7-fluoro-l-isopropyl-3-methyl-imidazo[4,5-c]cinnolin-2-one (98 mg, 0.29 mmol) and CS2CO3 (188 mg, 0.58 mmol) in 1 ,4-dioxane (0.2 mL) and water (0.04 mL) and the mixture was stirred at 80 °C for 2 hours. The crude product was purified by flash silica chromatography, elution gradient 0 to 10% MeOH in DCM, then further purified by preparative HPLC to afford desired product (20 mg, 14 %) as a white solid.
NMR Spectrum: lH NMR (300MHz. DMSO) δ 1.30-1.40 (2H, m), 1.45-1.55 (4H, m), 1.68 (6H, d), 1.85-1.98 (2H, m), 2.25-2.40 (6H, m), 3.60 (3H, s), 4.30-4.40 (2H, m), 5.20-5.30 (1H, m), 7.00 (1H, d), 8.08 (1H, s), 8.24 (1H, d), 8.36 (lH,d), 8.52 (1H, s).
Mass Spectrum: m/z (ES+) [M+H]+ = 479
Example 47: 7-Fluoro-l-isopropyl-3-methyl-8-[4-[3-(l- piperidyl)propoxy] phenyl] imidazo [4,5-c] cinnolin-2-one
Figure imgf000111_0001
(2 ' -Amino-2-biphenylyl)(chloro)palladium - dicyclohexyl(2 ' ,4 ' ,6 ' -triisopropyl-2- biphenylyl)phosphine (1 : 1) (22.79 mg, 0.03 mmol) was added to l-[3-[4-(4,4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)phenoxy]propyl]piperidine (lOOmg, 0.29 mmol), 8- bromo-7-fluoro-l-isopropyl-3-methyl-imidazo[4,5-c]cinnolin-2-one (98 mg, 0.29 mmol) and CS2CO3 (189 mg, 0.58 mmol) in 1,4-dioxane (2 mL) and water (0.5 mL) and the mixture was stirred at 80 °C for 2 hours. The crude product was purified by flash silica chromatography, elution gradient 0 to 10% MeOH in DCM. Pure fractions were evaporated to dryness to afford crude product as a yellow oil. The crude product was purified by preparative HPLC to afford the desired product (10 mg, 7 %) as a white solid. NMR Spectrum: 1H NMR (300MHz, DMSO) δ 1.32-1.40 (2H, m), 1.48-1.55 (4H, m), 1.68 (6H, d), 1.85-1.98 (2H, m), 2.25-2.40 (6H, m), 3.60 (3H, s), 4.05-4.15 (2H, m), 5.15- 5.25 (1H, m), 7.12 (2H, d), 7.65 (2H, d), 8.15-8.26 (2H, m).
Mass Spectrum: m/z (ES+) [M+H]+ = 478
Example 48: 8- [4- [3-(Dimethylamino)propoxy] phenyl] -l-isopropyl-3,7-dimethyl- imidazo 4,5-c] cinnolin-2-one
Figure imgf000111_0002
(2 ' -Amino-2-biphenylyl)(chloro)palladium - dicyclohexyl(2 ' ,4 ' ,6 ' -triisopropyl-2- biphenylyl)phosphine (1 : 1) (25.8 mg, 0.03 mmol) was added to N,N-dimethyl-3-[4- (4,4,5, 5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenoxy]-l-propanamine (lOOmg, 0.33 mmol), 8-bromo-l-isopropyl-3,7-dimethyl-imidazo[4,5-c]cinnolin-2-one (110 mg, 0.33 mmol) and CS2CO3 (213 mg, 0.66 mmol) in 1,4-dioxane (2 mL) and water (0.4 mL) and the mixture was stirred at 80 °C for 4 hours under an inert atmosphere. The crude product was purified by flash silica chromatography, elution gradient 0 to 10% MeOH in DCM, the further purified by preparative HPLC to afford the desired product (60 mg, 42 %) as a yellow solid.
NMR Spectrum: !H NMR (300 MHz, DMSO) δ 1.65 (6H, d), 1.85-1.96 (2H, m), 2.20 (6H, s), 2.38-2.45 (5H, m), 3.60 (3H, s), 4.03-4.13 (2H, m), 5.08-5.18 (1H, m), 7.08 (2H, d), 7.47 (2H, d), 7.98 (1H, s), 8.25 (1H, s).
Mass Spectrum: m/z (ES+) [M+H]+ = 434
The preparation of 8-bromo-l-isopropyl-3,7-dimethyl-imidazo[4,5-c]cinnolin-2-one is described below.
8-Bromo-l-iso ropyl-3,7-dimethyl-imidazo[4,5-clcinnolin-2-one
Figure imgf000112_0001
l,l-Dimethoxy-N,N-dimethylmethanamine (10.42 mL, 77.84 mmol) was added to 8- bromo-l-isopropyl-7-methyl-3H-imidazo[4,5-c]cinnolin-2-one (2.5 g, 7.78 mmol) in DMF (20 mL) and the solution was stirred at 100 °C for 2 hours. The reaction mixture was diluted with water. The precipitate was collected by filtration, washed with water (50 mL) and dried under vacuum to afford the desired product (2.0 g, 77 %) as a red solid, which was used without further purification.
NMR Spectrum: !H NMR (400 MHz, DMSO) δ 1.62 (6H, d), 2.57 (3H, s), 3.67 (3H, s), 5.09-5.16 (1H, m), 8.28 (1H, s), 8.62 (1H, s).
Mass Spectrum: m/z (ES+) [M+H]+ = 335
8-Bromo-l-isopropyl-7-methyl-3H-imidazo[4,5-clcinnolin-2-one
Figure imgf000112_0002
l,3,5-Trichloro-l,3,5-triazinane-2,4,6-trione (1.294 g, 5.57 mmol) was added portionwise to a stirred suspension of 6-bromo-4-(isopropylamino)-7-methylcinnoline-3-carboxamide (3.6g, 11.14 mmol) and 2,3,4,6,7,8,9,10-octahydropyrimido[l,2-a]azepine (3.36 mL, 22.28 mmol) in MeOH (60 mL) at 0 °C, then the solution was warmed to ambient temperature and stirred for 90 minutes. The reaction mixture was diluted with water and the precipitate collected by filtration, washed with water (100 mL) and dried under vacuum to afford the desired product (3.20 g, 89 %) as a brown solid, which was used without further purification.
NMR Spectrum: ¾ NMR (300 MHz, DMSO) δ 1.62 (6H, d), 2.55 (3H, s), 5.03-5.12 (1H, m), 8.22 (1H, s), 8.49 (1H, s), 12.49 (1H, br).
Mass Spectrum: m/z (ES+) [M+2H]+ = 323
6-Bromo-4-(isopropylamino)-7-methylcinnoline-3-carboxamide
Figure imgf000113_0001
Propan-2-amine (0.767 g, 12.98 mmol) was added to 6-bromo-4-chloro-7-methyl- cinnoline-3-carboxamide (3.9 g, 12.98 mmol) and DIPEA (4.53 mL, 25.95 mmol) in acetonitrile (60 mL) and the resulting suspension stirred at 60 °C for 16 hours. The mixture was allowed to cool and diluted with water. The precipitate was collected by filtration, washed with water (50 mL) and dried under vacuum to afford the desired material (3.70 g, 88 %) as a brown solid, which was used without further purification.
NMR Spectrum: !H NMR (400Hz, DMSO) δ 1.33 (6H, d), 2.55 (3H, s), 4.32-4.40 (1H, m), 7.71 (1H, s), 8.15 (1H, s), 8.40 (1H, s), 8.57 (1H, s), 10.34 (1H, d).
Mass Spectrum: m/z (ES+) [M+2H]+ = 325
6-Bromo-4-chloro-7-methyl-cinnoline-3-carboxamide
Figure imgf000113_0002
DMF (0.164 mL, 2.12 mmol) was added to a mixture of 6-bromo-4-hydroxy-7-methyl- cinnoline-3-carboxylic acid (6.00 g, 21.20 mmol) in thionyl chloride (90 mL, 1233.2 mmol) at ambient temperature under an inert atmosphere. The resulting slurry was stirred at 80 °C for 2 hours then allowed to cool, concentrated to dryness and the residue azeotroped with toluene (3 x 100 mL) to afford the crude acid chloride. This crude material (6.78 g, 21.19 mmol) was dissolved in acetone (30 mL), cooled to 0 °C and ammonium hydroxide (50 mL, 321.01 mmol) added dropwise over 10 minutes. The resulting mixture was stirred at ambient temperature for 1 hour. The precipitate was collected by filtration, washed with water (100 mL) and dried under vacuum to afford the desired material (4.20 g, 66 %) as a yellow solid, which was used without further purification.
Mass Spectrum: m/z (ES+) [M+H]+ = 301
6-Bromo-4-hvdroxy-7-methyl-cinnoline-3-carboxylic acid
Figure imgf000114_0001
Potassium hydroxide (62 g, 1.11 mol) was added to a solution of 4-amino-6-bromo-7- methyl-cinnoline-3-carboxamide (31 g, 110.28 mmol) in DMSO/H20 (600 mL / 600 mL) under an inert atmosphere at ambient temperature. The solution was stirred for 2 days at 120 °C, cooled to ambient temperature and quenched with water/ice (3000 mL). The pH value of the solution was adjusted to 7 with 3M hydrochloric acid. The solids formed were collected by filtration, washed with water (2 x 500 mL) and dried. The crude product was purified by re-crystallization from DMSO to afford the desired product (11.5 g, 37%) as a light brown solid.
NMR Spectrum: !H NMR (300 MHz, DMSO) δ 2.50-2.57 (m, 3H), 7.78 (s, 1H), 8.35 (s, 1H), 14.28-14.33 (br, 1H), 14.70-14.75 (br, 1H).
Mass Spectrum: m/z (ES+) [M+H]+ = 282
4-Amino-6-bromo-7-methyl-cinnoline-3-carboxamide
Figure imgf000115_0001
Aluminium trichloride (59.4 g, 445.47 mmol) was added to a solution of (lE)-2-amino-N- (4-bromo-3-methyl-anilino)-2-oxo-acetimidoyl cyanide (42 g, 149.41 mmol) in 1,2- dichlorobenzene (500 mL) under an inert atmosphere at ambient temperature. The solution was stirred for 3 hours at 120 °C, then cooled to 40-50 °C and quenched with water/ice (3000 mL). The solids formed were collected by filtration and washed with water (2 x 500 mL) and oven dried to afford the desired material (31 g, 74%) as a yellow solid.
Mass Spectrum: m/z (ES+) [M+2H]+ = 283
(lE)-2-amino-N-(4-bromo-3-methyl-anilino)-2-oxo-acetimidoyl cyanide
Figure imgf000115_0002
4-Bromo-3-methylaniline (30 g, 161.25 mmol), concentrated hydrochloric acid (100 mL), acetic acid (200 mL), and water (500 mL) were added to a round bottom flask under an inert atmosphere at ambient temperature. To the mixture was added a solution of sodium nitrite (12.3 g, 178.27 mmol) in H20 (200 mL) dropwise with stirring at 0 °C, then the solution was stirred for 2 hours at 0 °C. NaOAc (100 g, 1.22 mol) was added portionwise at 0 °C and the solution stirred for 30 minutes at 0 °C. 2-Cyanoacetamide (16.35 g, 194.47 mmol), EtOH (500 mL) and water (2000 mL) were added to a second round bottom flask under an inert atmosphere at ambient temperature, then NaOAc (500 g, 6.10 mol, 38.00 equiv) was added at ambient temperature and then the mixture from the first flask added dropwise with stirring and the temperature maintained at 0-5 °C. The resulting solution was stirred for 2 hours at 0 °C. The solids were collected by filtration, and washed with water (3 x 1 L) and dried to afford the desired material (42 g, 93%) as a yellow solid, which was used without further purification.
Mass Spectrum: m/z (ES+) [M+H]+ = 281 Example 49: l-Isopropyl-3,7-dimethyl-8-[6-[3-(l-piperidyl)propoxy]-3- pyridyl] imidazo [4,5-c] cinnolin-2-one
Figure imgf000116_0001
(2 ' -Amino-2-biphenylyl)(chloro)palladium - dicyclohexyl(2 ' ,4 ' ,6 ' -triisopropyl-2- biphenylyl)phosphine (1 : 1) (22.73 mg, 0.03 mmol) was added to 2-[3-(l- piperidyl)propoxy] -5 -(4,4,5 ,5-tetramethyl- 1 ,3 ,2-dioxaborolan-2-yl)pyridine (1 OOmg, 0.29 mmol), 8-bromo-l-isopropyl-3,7-dimethyl-imidazo[4,5-c]cinnolin-2-one (97 mg, 0.29 mmol) and CS2CO3 (188 mg, 0.58 mmol) in 1,4-dioxane (0.2 mL) and water (0.04 mL) and the mixture was stirred at 80 °C for 2 hours under an inert atmosphere. The crude product was purified by flash silica chromatography, elution gradient 0 to 10% MeOH in DCM, then further purified by preparative HPLC to afford the desired product (20 mg, 15 %) as a white solid.
NMR Spectrum: !H NMR (300MHz, DMSO) δ 1.30-1.40 (2H, m), 1.45-1.55 (4H, m), 1.68 (6H, d), 1.85-1.98 (2H, m), 2.25-2.4 (9H, m), 3.60 (3H, s), 4.30-4.40 (2H, m), 5.10-5.20 (lH,m), 6.95 (1H, d) ,7.90 (1H, m), 8.10 (1H, s), 8.28 (1H, s), 8.32 (1H, s).
Mass Spectrum: m/z (ES+) [M+H]+ = 475
Example 50: 8- [6- [3-(Dimethylamino)propoxy] -3-pyridyl] -l-isopropyl-3,7-dimethyl- imidazo [4,5-c] cinnolin-2-one
Figure imgf000116_0002
(2 ' -Amino-2-biphenylyl)(chloro)palladium - dicyclohexyl(2 ' ,4 ' ,6 ' -triisopropyl-2- biphenylyl)phosphine (1 : 1) (23.47 mg, 0.03 mmol) was added to 8-bromo-l-isopropyl-3,7- dimethyl-imidazo[4,5-c]cinnolin-2-one (100 mg, 0.30 mmol), N,N-dimethyl-3-{[5- (4,4,5, 5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2-pyridinyl]oxy}-l-propanamine (110 mg, 0.36 mmol) and CS2CO3 (243 mg, 0.75 mmol) in 1,4-dioxane (0.8 mL) and water (0.16 mL) and the mixture was stirred at 100 °C for 3 hours under an inert atmosphere. The reaction mixture was evaporated to afford crude product. The crude product was purified by preparative HPLC to afford the desired product (39.8 mg, 31 %) as a pale yellow solid. NMR Spectrum: !H NMR (400MHz, CDCI3) δ 1.57 (6H, d), 2.12 (2H, s), 2.32-2.48 (9H, m), 2.65 (2H, s), 3.78 (3H, s), 4.47 (2H, t), 5.13 (1H, t), 6.89 (1H, d), 7.67-7.69 (1H, m), 7.93 (1H, s), 8.23 (1H, d), 8.35 (1H, s).
Mass Spectrum: m/z (ES+) [M+H]+ = 435
Example 51: l-Isopropyl-3,7-dimethyl-8-[4-[3-(l- piperidyl)propoxy] phenyl] imidazo [4,5-c] cinnolin-2-one
Figure imgf000117_0001
(2 ' -Amino-2-biphenylyl)(chloro)palladium - dicyclohexyl(2 ' ,4 ' ,6 ' -triisopropyl-2- biphenylyl)phosphine (1 : 1) (22.79 mg, 0.03 mmol) was added to l-(3-(4-(4,4,5,5- tetramethyl-l,3,2-dioxaborolan-2-yl)phenoxy)propyl)piperidine (100 mg, 0.29 mmol), 8- bromo-l-isopropyl-3,7-dimethyl-imidazo[4,5-c]cinnolin-2-one (97 mg, 0.29 mmol) and CS2CO3 (189 mg, 0.58 mmol) in 1,4-dioxane (1 mL) and water (0.25 mL) and the mixture was stirred at 80 °C for 4 hours under an inert atmosphere. The crude product was purified by flash silica chromatography, elution gradient 0 to 10% MeOH in DCM, then further purified by preparative HPLC to afford the desired product (20 mg, 14 %>) as a yellow solid.
NMR Spectrum: !H NMR (300MHz, DMSO) δ 1.34-1.45 (2H, m), 1.48-1.55 (4H, m), 1.63 (6H, d), 1.85-1.98 (2H, m), 2.30-2.38 (4H, m), 2.45 (5H, s), 3.60 (3H, s), 4.05-4.15 (2H, m), 5.05-5.15 (1H, m), 7.07 (2H, d), 7.45 (2H, d), 8.00 (1H, s), 8.28 (1H, m).
Mass Spectrum: m/z (ES+) [M+H]+ = 474
Example 52: 3-(Difluoromethyl)-8-[6-[3-(dimethylamino)propoxy]-3-pyridyl]-l- isopropyl-imidazo [4,5-c] cinnolin-2-one
Figure imgf000118_0001
Dichlorobis(triphenylphosphine)palladium(II) (0.999 mg, 1.42 μηιοΐ) was added to a degassed solution of A ,N-dimethyl-3-[[5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2- pyridyl]oxy]propan-l -amine (9.15 mg, 0.03 mmol) and 8-chloro-3-(difluoromethyl)-l- isopropyl-imidazo[4,5-c]cinnolin-2-one (8.9 mg, 0.03 mmol), 2M potassium carbonate solution (0.043 mL, 0.09 mmol) in 1,4-dioxane (0.527 mL) and heated in a microwave at 80 °C for 1 hour under an inert atmosphere. The reaction mixture was diluted with EtOAc (10 mL), and washed sequentially with water (7 mL), saturated brine (7 mL) and the organic layer was evaporated. The crude product was purified by preparative HPLC to afford the desired product (5.7 mg, 44 %) as a yellow dry film.
NMR Spectrum: !H NMR (400 MHz, CDCb) δ 1.81 (6H, d), 2.03 - 2.15 (2H, m), 2.43 (6H, s), 2.64 - 2.74 (2H, m), 4.46 (2H, t), 5.18 (1H, dt), 6.92 (1H, d), 7.60 (1H, t), 7.92 (2H, ddd), 8.15 - 8.25 (1H, m), 8.44 - 8.56 (1H, m), 8.59 (1H, d).
Mass Spectrum: m/z (ES+) [M+H]+ 457
8-Chloro-3-(difluoromethyl)-l-isopropyl-imidazo[4,5-c]cinnolin-2-one was prepared as described below.
8-Chloro-3-(difluoromethyl)-l-isopropyl-imidazor4,5-c1cinnolin-2-one
Figure imgf000118_0002
8-Chloro-l-isopropyl-3H-imidazo[4,5-c]cinnolin-2-one (200 mg, 0.76 mmol) was dissolved in DMA (7.61 mL) under inert atmosphere. Sodium hydride (192 mg, 4.80 mmol) was added and the mixture was stirred for 20 minutes. Sodium
chloro(difluoro)acetate (824 mg, 5.41 mmol) was added and the mixture was heated to 90 °C for 2 hours. The reaction was cooled, diluted with EtOAc (20 mL) and water (20 mL), then the organic layer washed with water (2 x 20 mL), dried and evaporated. The crude product was purified by flash silica chromatography, elution gradient 0 to 60 % EtOAc in heptane, to afford the desired product (37 mg, 16%) as a beige solid.
NMR Spectrum: *H NMR (400 MHz, DMSO) δ 1.63 (6H, d), 5.11 - 5.23 (1H, m), 7.7 -
8.09 (2H, m), 8.43 (2H, d).
Mass Spectrum: m/z (ES+) [M+H]+ = 313
Example 53 : 8- [6- [(3R)-3-(Dimethylamino)pyrrolidin-l-yl]-3-pyridyl] -l-isopropyl-3- methyl-imidazo [4,5-c] cinnolin-2-one
Figure imgf000119_0001
8-(6-Fluoro-3-pyridyl)-l-isopropyl-3-methyl-imidazo[4,5-c]cinnolin-2-one (110 mg, 0.33 mmol), and (3R)-A ,N-dimethyl-3-pyrrolidinamine (37.2 mg, 0.33 mmol) were suspended in DMF (3.26 mL) and heated to 150 °C for 5 hours in a microwave reactor. The reaction mixture was diluted with DCM (20 mL), and washed twice with water (10 mL). The organic layer was dried over a phase separating cartridge, filtered and evaporated. The crude product was purified by preparative HPLC to afford the desired product (25.6 mg, 18 %) as a brown solid.
NMR Spectrum: lH NMR (400 MHz, DMSO) δ 1.67 (6H, d), 1.77 - 1.9 (1H, m), 2.22 (7H, s), 2.81 (1H, dt), 3.14 - 3.25 (1H, m), 3.41 (1H, td), 3.59 (3H, s), 3.67 (1H, t), 3.76 (1H, dd), 5.29 (1H, p), 6.64 (1H, d), 8.05 (2H, ddd), 8.27 (1H, d), 8.34 (1H, d), 8.69 (1H, d). Mass Spectrum: m/z (ES+) [M+H]+ = 432
The following compounds were synthesised in an analogous fashion using the appropriate amine and the appropriate fluoropyridine and a reaction time of between 3 and 6 hours at 150 °C:
Figure imgf000120_0001
Figure imgf000121_0001
* Purified twice by preparative HPLC
** 2 Equivalent of the amine were used in the reaction.
*** 3 Equvalents of the amine were used in the reaction and the material purified by flash silica chromatography eluting with 0 to 10% 1% N¾ in MeOH in DCM and the isolated product triturated with diethyl ether.
Example 54: NMR Spectrum: lH NMR (400 MHz, CDCb) δ 1.60 (2H, qd), 1.78 (6H, d), 2.02 (2H, d), 2.40 (6H, s), 2.61 (IH, dtd), 2.97 (2H, td), 3.76 (3H, s), 4.49 (2H, d), 5.22 (IH, p), 6.82 (IH, d), 7.84 (2H, ddd), 8.17 (IH, d), 8.43 - 8.55 (IH, m), 8.58 (IH, d).Mass Spectrum: m/z (ES+) [M+H]+ = 446
Example 55: NMR Spectrum: 'H NMR (400 MHz, DMSO) δ 1.78 - 1.9 (3H, m), 2.21 (2H, s), 2.22 (6H, s), 2.61 (IH, d), 2.80 (IH, dt), 3.20 (IH, dd), 3.41 (2H, td), 3.57 (3H, s), 3.67 (IH, t), 3.76 (IH, dd), 3.95 (IH, d), 4.15 (2H, d), 4.86 - 4.97 (IH, m), 6.67 (IH, d), 8.04 (2H, ddd), 8.22 (IH, d), 8.35 (IH, d), 8.64 (IH, d . Mass Spectrum: m/z (ES+)
[M+H]+ = 474
Example 56: NMR Spectrum: ¾ NMR (400 MHz, DMSO) δ 0.81 (3H, s), 1.17 (3H, s), 1.32 - 1.46 (2H, m), 1.82 (3H, dd), 2.23 (6H, s), 2.92 (3H, t), 3.36 (IH, d), 3.59 (3H, s), 3.66 - 3.75 (2H, m), 4.02 - 4.1 (IH, m), 4.43 (2H, d), 5.01 (IH, dd), 7.05 (IH, d), 8.04 (2H, dd), 8.19 (IH, s), 8.35 (IH, d), 8.51 (IH, s), 8.66 (IH, d . Mass Spectrum: m/z (ES+) [M+H]+ = 516
Example 57: NMR Spectrum: !H NMR (400 MHz, DMSO) δ 0.86 (3H, s), 1.22 (3H, s), 1.84 (IH, d), 1.88 - 1.96 (IH, m), 2.19 (IH, ddq), 2.27 (6H, s), 2.93 (2H, dt), 3.27 - 3.5 (3H, m), 3.62 (3H, s), 3.56 - 3.79 (4H, m), 4.11 (IH, d), 4.98 (IH, dd), 6.68 (IH, d), 8.00 (2H, td), 8.17 (IH, s), 8.36 (IH, d), 8.45 (IH, d), 8.64 (IH, d) (isolated as formate salt). Mass Spectrum: m/z (ES+)[M+H]+ = 502
Example 58: NMR Spectrum: 'H NMR (400 MHz, CDCb) δ 1.91 - 2.06 (IH, m), 2.22 - 2.31 (IH, m), 2.35 (6H, s), 2.57 - 2.69 (2H, m), 2.82 - 2.95 (IH, m), 3.32 - 3.45 (6H, m), 3.47 - 3.57 (IH, m), 3.75 (4H, s), 3.87 (IH, dd), 4.34 (IH, tt), 5.49 (IH, ddd), 6.49 - 6.54 (IH, m), 7.83 (2H, ddd), 8.06 (IH, d), 8.44 (IH, d), 8.58 (IH, dd). Mass Spectrum: m/z (ES+)[M+H]+ = 474
Example 59: NMR Spectrum: !H NMR (400 MHz, CDCb) δ 2.01 (IH, s), 2.26 - 2.35 (IH, m), 2.37 (5H, s), 2.96 (3H, ddd), 3.13 (2H, tdd), 3.33 (3H, s), 3.37 (IH, d), 3.45 - 3.57 (2H, m), 3.76 (4H, s), 3.85 - 3.99 (2H, m), 4.93 - 5.06 (IH, m), 6.51 (IH, d), 7.89 (2H, ddd), 8.30 (IH, d), 8.47 (IH, d), 8.63 (IH, d). Mass Spectrum: m/z (ES+) [M+H]+ = 474
Example 60: NMR Spectrum: lH NMR (500 MHz, DMSO) δ 1.79 - 1.89 (IH, m), 1.92 (IH, d), 1.94 - 2 (2H, m), 2.15 (2H, d), 2.23 (6H, s), 2.6 - 2.71 (IH, m), 2.76 - 2.85 (IH, m), 3.20 (IH, dd), 3.38 - 3.46 (IH, m), 3.61 (3H, s), 3.61 - 3.71 (2H, m), 3.74 - 3.82 (IH, m), 4.09 (2H, dd), 5.05 - 5.14 (IH, m), 6.67 (IH, d), 8.04 - 8.11 (2H, m), 8.32 (IH, s), 8.36 (IH, d), 8.67 - 8.71 (IH, m). Mass Spectrum: m/z (ES+) [M+H]+ = 474 Example 61: NMR Spectrum: ¾ NMR (400 MHz, DMSO) δ 1.77 - 1.9 (2H, m), 2.22 (1 IH, s), 2.34 - 2.42 (IH, m), 2.81 (IH, dt), 3.20 (IH, dd), 3.28 (3H, s), 3.41 (IH, td), 3.59 (3H, s), 3.65 - 3.8 (2H, m), 4.14 (IH, s), 5.50 (IH, p), 6.65 (IH, d), 8.03 (2H, ddd), 8.22 (IH, d), 8.35 (IH, d), 8.66 (IH, d). Mass Spectrum: m/z (ES+) [M+H]+ = 488
Example 62: NMR Spectrum: 'H NMR (400 MHz, DMSO) δ 1.24 (IH, s), 1.76 - 1.9 (2H, m), 2.15 - 2.3 (10H, m), 2.37 (IH, dd), 2.81 (IH, dt), 3.16 - 3.26 (IH, m), 3.28 (3H, s), 3.36 - 3.46 (IH, m), 3.59 (3H, s), 3.67 (IH, t), 3.76 (IH, dd), 4.15 (IH, d), 5.49 (IH, q), 6.65 (IH, d), 8.03 (2H, ddd), 8.22 (IH, d), 8.35 (IH, d), 8.66 (IH, d . Mass Spectrum: m/z (ES+) [M+H]+ = 488
The preparation of the fluoropyridine intermediates required for Examples 53 - 62 have either been described previously or are described below:
8-(6-Fluoro-3-pyridyl)-3-methyl-l-[(3R)-tetrahydropyran-3-yllimidazo[4,5-clcinnolin-2- one
Figure imgf000123_0001
l,l'-Bis(di-tert-butylphosphino)ferrocene palladium dichloride (10.22 mg, 0.02 mmol) was added to a degassed solution of (6-fluoropyridin-3-yl)boronic acid (46.4 mg, 0.33 mmol)and 8-chloro-3-methyl- 1 -[(3R)-tetrahydropyran-3-yl]imidazo[4,5-c]cinnolin-2-one (100 mg, 0.31 mmol), 2M potassium carbonate solution (0.471 mL, 0.94 mmol) in 1,4- dioxane (1.098 mL) and the mixture was heated in a microwave reactor to 80 °C for 1 hour under an inert atmosphere. The reaction mixture was diluted with EtOAc (25 mL), and washed sequentially with water (10 mL), saturated brine (10 mL) then the organic layer evaporated to afford the desired material as an orange dry film, which was used without further purification.
Mass Spectrum: m/z (ES+)[M+H]+ = 380 1 : 1 mixture of l-[(4R)-33-dimethyltetrahydropyran-4-yll-8-(6-fluoro-3-pyridyl)-3-methy imidazo[4,5-clcinnolin-2-one and l-[(4S)-3,3-dimethyltetrahydropyran-4-yll-8-(6-fluoro- 3 -p yridvD-3 -methyl-imidazo f4,5-c1 cinnolin-2-one
Figure imgf000124_0001
Dichlorobis(triphenylphosphine)palladium (II) (16.19 mg, 0.02 mmol) was added to a degassed solution of (6-fluoropyridin-3-yl)boronic acid (68.3 mg, 0.48 mmol) and a 1 : 1 mixture of 8-chloro-l -[(4R)-3,3-dimethyltetrahydropyran-4-yl]-3-methyl-imidazo[4,5- c] cinnolin-2-one and 8-chloro- 1 - [(4S)-3 ,3 -dimethyltetrahydropyran-4-yl] -3 -methyl- imidazo[4,5-c]cinnolin-2-one (160 mg, 0.46 mmol), 2M potassium carbonate solution (0.692 mL, 1.38 mmol) in 1,4-dioxane (2.384 mL) and the mixture was heated in a microwave reactor to 80 °C for 1 hour under an inert atmosphere. The reaction mixture was diluted with EtOAc (25 mL), and washed sequentially with water (10 mL), saturated brine (10 mL), then the organic layer evaporated to afford the desired material as a brown solid, which was used without further purification.
Mass Spectrum: m/z (ES+) [M+H]+ = 408
8-(6-Fluoro-3-pyridyl)-l-(trans-3-methoxycyclobutyl)-3-methyl-imidazo[4,5-clcinnolin-2- one
Figure imgf000124_0002
l,l'-Bis(di-tert-butylphosphino)ferrocene palladium dichloride (10.22 mg, 0.02 mmol) was added to a degassed solution of (6-fluoropyridin-3-yl)boronic acid (46.4 mg, 0.33 mmol) and 8-chloro- 1 -(trans-3-methoxycyclobutyl)-3-methyl-imidazo[4,5-c]cinnolin-2-one (100 mg, 0.31 mmol), 2M potassium carbonate solution (0.471 mL, 0.94 mmol) in 1,4-dioxane (1.098 mL) and the mixture was heated in a microwave reactor to 80 °C for 1 hour under an inert atmosphere. The reaction mixture was diluted with EtOAc (25 mL), and washed sequentially with water (10 mL), saturated brine (10 mL), then the organic layer evaporated to afford the desired material as a brown solid, which was used without further purification.
Mass Spectrum: m/z (ES+)[M+H]+ = 380
8-(6-Fluoro-3-pyridyl)-l-(cis-3-methoxycvclobutyl)-3-methyl-imidazor4,5-c1cinnolin-2- one
Figure imgf000125_0001
Dichlorobis(triphenylphosphine)palladium (II) (39.6 mg, 0.06 mmol) was added to a degassed solution of (6-fluoropyridin-3-yl)boronic acid (167 mg, 1.19 mmol) and 8-chloro- l-(cis-3-methoxycyclobutyl)-3-methyl-imidazo[4,5-c]cinnolin-2-one (360 mg, 1.13 mmol), 2M potassium carbonate solution (1.694 mL, 3.39 mmol) in 1,4-dioxane (4 mL) and the mixture was heated at 80 °C for 1 hour under an inert atmosphere. The reaction mixture was diluted with EtOAc (25 mL), and washed sequentially with water (10 mL), saturated brine (10 mL), then the organic layer evaporated. The crude product was purified by preparative HPLC, using decreasingly polar mixtures of water (containing 0.1% NH3) and MeCN as eluents, to afford the desired product (1 15 mg) as a green solid.
Mass Spectrum: m/z (ES+)[M+H]+ = 380
8-(6-Fluoro-3 -pyridyD- 1 - |Y 1 R,3R)-3 -methoxycyclopentyl] -3 -methyl-imidazo [4,5 - clcinnolin-2-one
Figure imgf000126_0001
l,l '-Bis(di-tert-butylphosphino)ferrocene - dichloropalladium (1 : 1) (19.58 mg, 0.04 mmol) was added to a degassed solution of (6-fluoro-3-pyridinyl)boronic acid (89 mg, 0.64 mmol) and 8-chloro-l-[(lR,3R)-3-methoxycyclopentyl]-3-methyl-imidazo[4,5-c]cinnolin-2-one (100 mg, 0.30 mmol), 2M potassium carbonate solution (0.451 mL, 0.90 mmol) in 1,4- dioxane (1.052 mL) the mixture was heated in a microwave reactor to 80 °C for 2 hours under an inert atmosphere. The reaction mixture was diluted with EtOAc (25 mL), and washed sequentially with water (10 mL), saturated brine (10 mL) and the organic layer evaporated to afford crude product as a brown solid residue, which was used immediately without further purification.
Mass Spectrum: m/z (ES+) [M+H]+ = 394
8-(6-Fluoro-3-pyridyl)-l-r(lS,3S)-3-methoxycvclopentyl1-3-methyl-imidazor4,5- clcinnolin-2-one
Figure imgf000126_0002
l,l '-Bis(di-tert-butylphosphino)ferrocene - dichloropalladium (1 : 1) (10.28 mg, 0.02 mmol) was added to a degassed solution of (6-fluoro-3-pyridinyl)boronic acid (46.7 mg, 0.33 mmol) and 8-chloro-l-[(lS,3S)-3-methoxycyclopentyl]-3-methyl-imidazo[4,5-c]cinnolin- 2-one (105 mg, 0.32 mmol), 2M potassium carbonate solution (0.473 mL, 0.95 mmol) in 1,4-dioxane (1.104 mL) the mixture was heated in a microwave reactor to 80 °C for 1 hours under an inert atmosphere. The reaction mixture was diluted with EtOAc (25 mL), and washed sequentially with water (10 mL), saturated brine (10 mL) and the organic layer evaporated to afford crude product as a brown solid residue, which was used immediately without further purification. Mass Spectrum: m/z (ES+) [M+H]+ = 394
Example 63 : 8- [6- [4-(Dimethylamino)- 1-piperidyl] -3-pyridyl] -l-(cis-3- methoxycyclobutyl)-3-methyl-imidazo [4,5-c] cinnolin-2-one
Figure imgf000127_0001
8-Bromo-l -(cis-3-methoxycyclobutyl)-3-methyl-imidazo[4,5-c]cinnolin-2-one (0.09 g, 0.25 mmol) was added to N,N-dimethyl-l-(5-(4,4,5,5-tetramethyl-l ,3,2-dioxaborolan-2- yl)pyridin-2-yl)piperidin-4-amine (0.246 g, 0.74 mmol), CS2CO3 (0.484 g, 1.49 mmol) and palladium - triphenylphosphine (1 :4) (0.057 g, 0.05 mmol) in 1 ,4-dioxane (6 mL) and water (1.2 mL), and the mixture was stirred at 80 °C for 2 hours under an inert atmosphere. The crude product was purified by preparative HPLC to afford the desired product.
Fractions containing the desired compound were evaporated to dryness to afford the desired product (0.12 g, 99 %) as a yellow solid.
NMR Spectrum: lH NMR (300 MHz, DMSO) δ 1.2-1.5 (2H, m), 1.7-1.9 (2H, m), 2.1-2.2 (6H, m), 2.3-2.4 (1H, m), 2.75-3.05 (6H, m), 3.2-3.3 (3H,m), 3.55 (3H,s),3.8-4.0 (lH,m), 4.3-4.5 (2H, m), 5.0-5.2 (lH,m), 6.95-7.05 (1H, m), 7.98-8.12 (2H, m), 8.28-8.4 (2H, m), 8.72 (1H, s).
Mass Spectrum: m/z (ES+) [M+H]+ = 488
Example 64 : 8- [4- [4-(Dimethylamino)- l-piperidyl] phenyl] - l-isopropyl-3-methyl- imidazo [4,5-c] cinnolin-2-one
Figure imgf000127_0002
l, -Bis(di-tert-butylphosphino)ferrocene palladium dichloride (11.78 mg, 0.02 mmol) was added to a degassed solution of N,N-dimethyl-l-[4-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)phenyl]-4-piperidinamine (119 mg, 0.36 mmol) and 8-chloro-l- isopropyl-3-methyl-imidazo[4,5-c]cinnolin-2-one (100 mg, 0.36 mmol), 2M potassium carbonate solution (0.542 mL, 1.08 mmol) in 1,4-dioxane (3.07 mL). and the mixture heated in a microwave reactor at 80 °C for 1 hour under an inert atmosphere. The reaction mixture was diluted with EtOAc (25 mL), and washed sequentially with water (10 mL), saturated brine (10 mL) then the organic layer evaporated. The crude product was purified twice by preparative HPLC to afford the desired product (16.30 mg, 10.15 %) as a yellow solid.
NMR Spectrum: !H NMR (400 MHz, DMSO) δ 1.42 - 1.54 (2H, m), 1.68 (6H, d), 1.85 (2H, d), 2.20 (6H, s), 2.22 - 2.31 (1H, m), 2.79 (2H, t), 3.59 (3H, s), 3.86 (2H, d), 5.27 (1H, dt), 7.10 (2H, d), 7.76 (2H, d), 8.04 (1H, dd), 8.27 (1H, d), 8.34 (1H, d).
Mass Spectrum: m/z (ES+) [M+H]+ = 445
N,N-Dimethyl-l-[4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl]-4-piperidinamine was prepared as described below.
N,N-Dimethyl-l-r4-(4^,5,5-tetramethyl-13,2-dioxaborolan-2-yl)phenyl1-4-piperidinamine
Figure imgf000128_0001
To l-(4-bromophenyl)-N,N-dimethyl-piperidin-4-amine (100 mg, 0.35 mmol),
4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi-l,3,2-dioxaborolane (108 mg, 0.42 mmol) and potassium acetate (139 mg, 1.41 mmol) in dioxane (3.53 mL) under nitrogen was added dichlorobis(tricyclohexylphosphine)palladium (II) (26.1 mg, 0.04 mmol) and the reaction was heated to 90°C for 2 hours, then at 100°C for 16 hours. The reaction mixture was evaporated to dryness, redissolved in DCM (100 mL), and washed with water (100 mL). The organic layer was dried with a phase separating cartridge and evaporated to afford crude product, which was used without further purification. NMR Spectrum: lH NMR (400 MHz, DMSO) δ 1.26 (12H, s), 1.43 (2H, qd), 1.82 (2H, d), 2.23 (6H, s), 2.73 (2H, td), 3.76 - 3.99 (3H, m), 6.89 (2H, d), 7.49 (2H, d).
Mass Spectrum: m/z (ES+) [M+H]+ = 331 l-(4-Bromo henyl)-N,N-dimethyl-piperidin-4-amine
Figure imgf000129_0001
Sodium triacetoxyborohydride (1.251 g, 5.90 mmol) was added to a mixture of l-(4- bromophenyl)-4-piperidinone (1.00 g, 3.94 mmol), dimethylamine (2M in THF) (3.94 mL, 7.87 mmol) and acetic acid (0.901 mL, 15.74 mmol) in DCM (20 mL) and stirred at ambient temperature for 2 hours. The reaction mixture was quenched with saturated sodium hydrogen carbonate solution, extracted with DCM (50 mL) and the organic layer dried over a phase separating cartridge and evaporated. The crude product was purified by flash silica chromatography, elution gradient 0 to 10% MeOH in DCM to afford the desired material (0.853 g, 77 %) as an orange solid.
NMR Spectrum: !H NMR (400 MHz, DMSO) δ 1.43 (2H, qd), 1.74 - 1.86 (2H, m), 2.17 (7H, s), 2.66 (2H, td), 3.63 - 3.73 (2H, m), 6.85 - 6.92 (2H, m), 7.26 - 7.34 (2H, m).
Mass Spectrum: m/z (ES+) [M+2H]+ = 285
Example 65: 1:1 mixture of 8-[4-[4-(dimethylamino)-l-piperidyl]phenyl]-l-[(4S)-3,3- dimethyltetrahydropyr an-4-yl] -3-methyl-imidazo [4,5-c] cinnolin-2-one and 8- [4- [4- (dimethylamino)-l-piperidyl]phenyl]-l-[(4R)-3,3-dimethyltetrahydropyran-4-yl]-3- methyl-imidazo [4,5-c] cinnolin-2-one
Figure imgf000129_0002
l ,l'-Bis(di-tert-butylphosphino)ferrocene palladium dichloride (42.10 mg, 0.06 mmol) was added to a degassed solution of N,N-dimethyl-l-[4-(4,4,5,5-tetramethyl-l ,3,2- dioxaborolan-2-yl)phenyl]-4-piperidinamine (213 mg, 0.65 mmol) and a 1 : 1 mixture of 8- chloro-l-[(4R)-3,3-dimethyltetrahydropyran-4-yl]-3-methyl-imidazo[4,5-c]cinnolin-2-one and 8-chloro-l-[(4S)-3,3-dimethyltetrahydropyran-4-yl]-3-methyl-imidazo[4,5-c]cinnolin- 2-one (224 mg, 0.65 mmol), 2M potassium carbonate solution (0.969 mL, 1.94 mmol) in 1 ,4-dioxane (3.34 mL) and the mixture heated in a microwave reactor at 80 °C for 2 hours under an inert atmosphere. The reaction mixture was diluted with EtOAc (25 mL), and washed sequentially with water (10 mL), saturated brine (10 mL) then the organic layer evaporated. The crude product was purified twice by preparative HPLC to afford the desired product (29 mg, 9 %) as a yellow solid.
NMR Spectrum: lH NMR (400 MHz, DMSO) δ 0.83 (3H, s), 1.18 (3H, s), 1.47 (2H, qd), 1.75 - 1.89 (3H, m), 2.20 (6H, s), 2.26 (1H, ddt), 2.80 (2H, td), 3.33 - 3.46 (2H, m), 3.56 (1H, s), 3.59 (3H, s), 3.61 - 3.71 (1H, m), 3.89 (2H, d), 4.08 (1H, dd), 5.00 (1H, dd), 7.14 (2H, d), 7.75 (2H, d), 8.05 (1H, dd), 8.34 (1H, d), 8.50 (1H, d).
Mass Spectrum: m/z (ES+)[M+H]+ = 515
Example 66 : 8- [4- [4-(Dimethylamino)- 1-piperidyl] phenyl] - 1- [(lR,3R)-3- methox cyclopentyl] -3-methyl-imidazo [4,5-c] cinnolin-2-one
Figure imgf000130_0001
l ,l '-Bis(di-tert-butylphosphino)ferrocene - dichloropalladium (1 : 1) (1 1.16 mg, 0.02 mmol) was added to a degassed solution of N,N-dimethyl-l-[4-(4,4,5,5-tetramethyl-l ,3,2- dioxaborolan-2-yl)phenyl]-4-piperidinamine (1 19 mg, 0.36 mmol) and 8-chloro-l- [(lR,3R)-3-methoxycyclopentyl]-3-methyl-imidazo[4,5-c]cinnolin-2-one (1 14 mg, 0.34 mmol), 2M potassium carbonate solution (0.514 mL, 1.03 mmol) in 1 ,4-dioxane (1.199 mL).The mixture was heated in a microwave reactor to 80 °C for 1 hour under an inert atmosphere. The reaction mixture was diluted with EtOAc (25 mL), and washed sequentially with water (10 mL), saturated brine (10 mL) then the organic layer evaporated. The crude product was purified by preparative HPLC to afford the desired product (7.8 mg, 5 %) as a brown solid.
NMR Spectrum: !H NMR (400 MHz, DMSO) δ 1.24 (4H, s), 1.42 - 1.54 (3H, m), 1.85 (4H, d), 2.20 (8H, s), 2.22 - 2.28 (IH, m), 2.79 (2H, t), 3.59 (3H, s), 3.87 (2H, d), 4.13 (IH, s), 5.44 - 5.61 (IH, m), 7.1 1 (2H, d), 7.73 (2H, d), 8.05 (IH, dd), 8.21 (IH, s), 8.35 (IH, d). Mass Spectrum: m/z (ES+) [M+H]+ = 501
Example 67: 8-[4-[4-(Dimethylamino)-l-piperidyl]phenyl]-l-[(lS,35)-3- methox cyclopentyl] -3-methyl-imidazo [4,5-c] cinnolin-2-one
Figure imgf000131_0001
l ,l '-Bis(di-tert-butylphosphino)ferrocene - dichloropalladium (1 : 1) (1 1.75 mg, 0.02 mmol) was added to a degassed solution of N,N-dimethyl-l-[4-(4,4,5,5-tetramethyl-l ,3,2- dioxaborolan-2-yl)phenyl]-4-piperidinamine (125 mg, 0.38 mmol) and 8-chloro-l- [(l S,3S)-3-methoxycyclopentyl]-3-methyl-imidazo[4,5-c]cinnolin-2-one (120 mg, 0.36 mmol), 2M potassium carbonate solution (0.541 mL, 1.08 mmol) in 1 ,4-dioxane (1.262 mL) and the mixture was heated in a microwave reactor to 80 °C for 1 hour under an inert atmosphere. The reaction mixture was diluted with EtOAc (25 mL), washed sequentially with water (10 mL), saturated brine (10 mL) and the organic layer was evaporated. The crude product was purified by preparative HPLC to afford the desired product (19 mg, 1 1 %) as a brown solid.
NMR Spectrum: lH NMR (400 MHz, DMSO) δ 1.47 (2H, qd), 1.85 (3H, d), 2.20 (6H, s), 2.2 - 2.43 (6H, m), 2.79 (2H, td), 3.29 (3H, s), 3.59 (3H, s), 3.86 (2H, d), 4.09 - 4.16 (IH, m), 5.48 (IH, p), 7.10 (2H, d), 7.72 (2H, d), 8.04 (IH, dd), 8.20 (IH, d), 8.34 (IH, d). Mass Spectrum: m/z (ES+) [M+H]+ = 501
Example 68 : 8- [4- [4-(Dimethylamino)- 1-piperidyl] phenyl] -3-methyl- 1- [(3R)- tetrahydropyran-3-yl] imidazo [4,5-c] cinnolin-2-one
Figure imgf000132_0001
l, -Bis(di-tert-butylphosphino)ferrocene - dichloropalladium (1 : 1) (27.20 mg, 0.04 mmol) was added to a degassed solution of N,N-dimethyl-l-[4-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)phenyl]-4-piperidinamine (138 mg, 0.42 mmol) and 8-chloro-3-methyl- l-[(3R)-tetrahydropyran-3-yl]imidazo[4,5-c]cinnolin-2-one (133 mg, 0.42 mmol), 2M potassium carbonate solution (0.626 mL, 1.25 mmol) in 1,4-dioxane (3.55 mL) and the mixture was heated in a microwave reactor to 80 °C for 2 hours under an inert atmosphere. The reaction mixture was diluted with EtOAc (25 mL), washed sequentially with water (10 mL), saturated brine (10 mL) and the organic layer was evaporated. The crude product was purified by preparative HPLC to afford the desired product (9.8 mg, 5 %) as a yellow solid.
NMR Spectrum: lH NMR (400 MHz, CDCb) δ 1.66 (2H, qd), 1.95 (4H, dt), 2.24 (1H, t), 2.33 (7H, s), 2.72 - 2.92 (3H, m), 3.58 (1H, ddd), 3.73 (3H, s), 3.88 (2H, d), 4.05 (1H, d), 4.19 (1H, ddd), 4.42 - 4.53 (1H, m), 4.85 - 4.97 (1H, m), 7.07 (2H, d), 7.63 (2H, d), 7.92 (1H, dd), 8.17 - 8.22 (1H, m), 8.45 (1H, d).
Mass Spectrum: m/z (ES+) [M+H]+ = 487
Example 69: 8-[6-[(3R)-3-(Dimethylamino)pyrrolidin-l-yl]-3-pyridyl]-7-fluoro-l- isopropyl-3-methyl-imidazo [4,5-c] cinnolin-2-one
Figure imgf000132_0002
(2 ' -Amino-2-biphenylyl)(chloro)palladium - dicyclohexyl(2 ' ,4 ' ,6 ' -triisopropyl-2- biphenylyl)phosphine (1 : 1) (34.7 mg, 0.04 mmol) was added to (3R)-N,N-dimethyl-l-[5- (4,4,5, 5-tetramethyl-l ,3,2-dioxaborolan-2-yl)-2-pyridyl]pyrrolidin-3-amine (140mg, 0.44 mmol), 8-bromo-7-fluoro-l-isopropyl-3-methyl-l,3-dihydro-2H-imidazo[4,5-c]cinnolin-2- one (150 mg, 0.44 mmol) and CS2CO3 (288 mg, 0.88 mmol) in 1,4-dioxane (2 mL) and water (0.5 mL) and the solution heated at 80 °C for 2 hours. The crude product was purified by flash silica chromatography, elution gradient 0 to 10% MeOH in DCM, then by preparative HPLC to afford the desired product (80 mg, 40 %) as a yellow solid.
NMR Spectrum: !H NMR (300MHz, DMSO) δ 1.59-1.73 (6H, m), 1.80-1.92 (1H, m), 2.15-2.35 (7H, m), 2.80-2.90 (1H, m), 3.15-3.25 (1H, m), 3.40-3.50 (1H, m), 3.60 (3H, s), 3.62-3.70 (1H, m), 3.72-3.82 (1H, m), 5.18-5.28 (1H, m), 6.65 (1H, d), 7.89 (1H, d), 8.15 (1H, d), 8.24 (1H, d), 8.48 (1H, s).
Mass Spectrum: m/z (ES+) [M+H]+ = 350 (3R)-N,N-Dimethyl-l-[5-(4,4,5,5-tetramethyl-l ,3,2-dioxaborolan-2-yl)-2- pyridyl]pyrrolidin-3 -amine was prepared as described below.
(3R -N.N-Dimethyl-l-r5-(4,4,5,5-tetramethyl-l ,3.2-dioxaborolan-2-vn-2- pyridyllp yrrolidin-3 -amine
Figure imgf000133_0001
Under an inert atmosphere, 4,4,5, 5-tetramethyl-l ,3,2-dioxaborolane (0.285 mL, 1.96 mmol) was added slowly to (3R)-l-(5-bromo-2-pyridyl)-A ,N-dimethyl-pyrrolidin-3-amine, triethylamine (0.342 mL, 2.45 mmol) and dichlorobis(triphenylphosphine)palladium (II) (68.8 mg, 0.10 mmol) in 1 ,4-dioxane (1 mL) at 0°C, and the resulting mixture was stirred at 100 °C for 16 hours. The precipitate was filtered and washed with hexane, and the filtrate was concentrated to afford the desired product (297 mg, 95 %>) as a brown oil, which was used without further purification.
Mass Spectrum: m/z (ES+) [M+H]+ = 318
(3R)- 1 -(5-Bromo-2-pyridyl)-N,N-dimethyl-pyrrolidin-3-amine
Figure imgf000133_0002
4-Methylbenzenesulfonic acid (0.080 g, 0.42 mmol) was added to 2,5-dibromopyridine (lg, 4.22 mmol) and (3R)-N,N-dimethyl-3-pyrrolidinamine (0.964 g, 8.44 mmol) and stirred at 1 10 °C for 1 hour under an inert atmosphere. The reaction mixture was diluted with EtOAc (100 mL), and washed sequentially with saturated sodium hydrogen carbonate, water and saturated brine. The organic layer was dried over Na2S04, filtered and evaporated to afford the desired product (0.800 g, 70.1 %) as a brown oil.
NMR Spectrum: lH NMR (400MHz, DMSO) δ 1.65-1.80 (1H, m), 2.05-2.20 (8H, m) 2.60-2.72 (1H, m), 2.98-3.10 (1H, m), 3.49-3.60 (2H, m), 6.43 (1H, d), 7.61 (1H, d) 8.10 (1H, d).
Mass Spectrum: m/z (ES+) [M+H]+ = 270
Example 70 : 8- [4- [4-(Dimethylamino)- 1-piperidyl] phenyl] -7-fluoro- l-isopropyl-3- methyl-imid azo [4,5-c] cinnolin-2-one
Figure imgf000134_0001
[l , -Bis(diphenylphosphino)ferrocene]dichloropalladium(II) (31.0 mg, 0.04 mmol) was added to A ,N-dimethyl-l-[4-(4,4,5,5-tetramethyl-l ,3,2-dioxaborolan-2-yl)phenyl]-4- piperidinamine (140 mg, 0.42 mmol), 8-bromo-7-fluoro-l-isopropyl-3-methyl- imidazo[4,5-c]cinnolin-2-one (144 mg, 0.42 mmol) and CS2CO3 (276 mg, 0.85 mmol) in 1 ,4-dioxane (2 mL) and water (0.4 mL) and stirred at 80 °C for 4 hours. The crude product was purified by flash silica chromatography, elution gradient 0 to 10% MeOH in DCM then by preparative HPLC to afford the desired product (60 mg, 31 %) as a yellow solid. NMR Spectrum: !H NMR (300MHz, DMSO) δ 1.50-1.65 (8H, m), 1.90-2.05 (2H, m), 2.55-2.65 (7H, m), 2.72-2.85 (2H, m), 3.60 (3H, s), 3.98 (2H, d), 5.15-5.25 (1H, m), 7.15 (2H, d), 7.62 (2H, d), 8.10-8.25 (2H, m).
Mass Spectrum: m/z (ES+) [M+H]+ = 463 Example 71: 8-[6-[(3R)-3-(Dimethylamino)pyrrolidin-l-yl]-3-pyridyl]-l-isopropyl- 3,7-dimethyl-imidazo [4,5-c] cinnolin-2-one
Figure imgf000135_0001
Dichloro[l, -bis(diphenylphosphino)ferrocene]palladium (II) (32.3 mg, 0.04 mmol) was added to (3R)-N,N-dimethyl- 1 -[5-(4,4,5 ,5 -tetramethyl- 1 ,3 ,2-dioxaborolan-2-yl)-2- pyridyl]pyrrolidin-3 -amine (140mg, 0.44 mmol), 8-bromo-l-isopropyl-3,7-dimethyl- imidazo[4,5-c]cinnolin-2-one (148 mg, 0.44 mmol) and CS2CO3 (288 mg, 0.88 mmol) in 1,4-dioxane (2 mL) and water (0.4 mL) and the mixture was stirred at 80 °C for 4 hours under an inert atmosphere. The crude product was purified by flash silica chromatography, elution gradient 0 to 10% MeOH in DCM, then further purified by preparative HPLC to afford the desired product (100 mg, 51 %) as a yellow solid.
NMR Spectrum: !H NMR (300MHz, DMSO) δ 1.80-1.95 (1H, m), 1.60 (6H, d), 2.28 (7H, s), 2.45-2.50 (3H, m), 2.90 (2H, s), 3.15-3.25 (1H, m), 3.35-3.45 (1H, m) 3.60 (3H, s), 3.63-3.73 (1H, m), 3.73-3.83 (1H, m), 5.10-5.20 (1H, m), 6.60 (1H, d), 7.73 (1H, d), 8.00 (1H, s), 8.25 (2H, s).
Mass Spectrum: m/z (ES+) [M+H]+ = 446
Example 72: 8-[4-[4-(Dimethylamino)-l-piperidyl]phenyl]-l-isopropyl-3,7-dimethyl- imidazo 4,5-c] cinnolin-2-one
Figure imgf000135_0002
Dichloro[l, -bis(diphenylphosphino)ferrocene]palladium (II) (33.2 mg, 0.05 mmol) was added to A ,N-dimethyl-l-[4-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)phenyl]-4- piperidinamine (150mg, 0.45 mmol), 8-bromo-l-isopropyl-3,7-dimethyl-imidazo[4,5- c]cinnolin-2-one (152 mg, 0.45 mmol) and CS2CO3 (296 mg, 0.91 mmol) in 1,4-dioxane (2 mL) and water (0.4 mL) and the mixture was stirred at 80 °C for 4 hours under an inert atmosphere. The crude product was purified by flash silica chromatography, elution gradient 0 to 10% MeOH in DCM, then further purified by preparative HPLC to afford the desired product (35 mg, 17 %) as a yellow solid.
NMR Spectrum: !H NMR (300MHz, DMSO) δ 1.42-1.52 (2H, d), 1.62 (6H, d), 1.87 (2H, d), 2.20 (7H, s), 2.46 (3H, s), 2.70-2.80 (2H, m) 3.60 (3H, s), 3.85 (2H, d), 5.06-5.16 (IH, m), 7.07 (2H, d), 7.39 (2H, d), 7.97 (IH, s), 8.23 (IH, s).
Mass Spectrum: m/z (ES+) [M+H]+ = 459
Example 73 : 8- [6- [3-(Dimethylamino)propoxy] -2-fluoro-3-pyridyl] -l-isopropyl-3- methyl-imidazo [4,5-c] cinnolin-2-one
Figure imgf000136_0001
l,l '-Bis(di-tert-butylphosphino)ferrocene - dichloropalladium (1 : 1) (47.10 mg, 0.08 mmol) was added to a degassed solution of 3-[[6-fluoro-5-(4,4,5,5-tetramethyl-l,3,2- dioxaborolan-2-yl)-2-pyridyl]oxy]-N,N-dimethyl-propan-l -amine (492 mg, 1.52 mmol) and 8-chloro-l-isopropyl-3-methyl-imidazo[4,5-c]cinnolin-2-one (200 mg, 0.72 mmol), 2M potassium carbonate solution (1.084 mL, 2.17 mmol) in 1,4-dioxane (2.53 mL) and the mixture was heated in a microwave reactor to 80 °C for 2 hours under an inert atmosphere. The reaction mixture was diluted with EtOAc (25 mL), and washed sequentially with water (10 mL), saturated brine (10 mL) then the organic layer evaporated. The crude product was purified by preparative HPLC to afford the desired product (88 mg, 28 %) as a brown solid.
NMR Spectrum: lH NMR (400 MHz, DMSO) δ 1.65 (6H, d), 1.88 (2H, p), 2.15 (6H, s), 2.36 (2H, t), 3.60 (3H, s), 4.32 (2H, t), 5.22 (IH, hept), 6.96 (IH, dd), 7.92 (IH, dd), 8.26 (IH, dd), 8.39 (2H, d).
Mass Spectrum: m/z (ES+) [M+H]+ = 439 3-[[6-Fluoro-5-(4,4,5,5-tetramethyl-l,3,2-dioxaborolan-2-yl)-2-pyridyl]oxy]-N,N- dimethyl-propan-1 -amine was prepared as described below.
3-[[6-Fluoro-5-(4^,5,5-tetramethyl-13,2-dioxaborolan-2-yl)-2-pyridylloxyl-N,N- dimethyl- ropan- 1 -amine
Figure imgf000137_0001
To 3-(5-Bromo-6-fluoropyridin-2-yl)oxy-N,N-dimethylpropan-l-amine (500 mg, 1.80 mmol), 4,4,4',4',5,5,5',5'-octamethyl-2,2'-bi(l,3,2-dioxaborolane) (550 mg, 2.16 mmol) and potassium acetate (708 mg, 7.22 mmol) in dioxane (18.04 mL) under an inert atmosphere was added dichlorobis(tricyclohexylphosphine)palladium (II) (133 mg, 0.18 mmol) and the reaction was heated to 90°C for 18 hours. The reaction mixture was evaporated to dryness and redissolved in DCM (10 mL), and washed with water (10 mL). The organic layer was dried with a phase separating cartridge and evaporated to afford crude product, which was used without further purification.
Mass Spectrum: m/z (ES+) [M+H]+ = 325
The preparation of 3-(5-Bromo-6-fluoropyridin-2-yl)oxy-N,N-dimethylpropan-l- known in the literature, for example in WO2015170081.A1.
BIOLOGICAL ASSAYS
The following assays were used to measure the effects of the compounds of Formula (I): a) ATM cellular potency assay; b) PI3K cellular potency assay; c) mTOR cellular potency assay; d) ATR cellular potency assay. During the description of the assays, generally:
i. The following abbreviations have been used: 4NQO = 4-Nitroquinoline N-oxide;
Ab = Antibody; BSA = Bovine Serum Albumin; C02 = Carbon Dioxide; DMEM = Dulbecco's Modified Eagle Medium; DMSO =Dimethyl Sulphoxide; EDTA = Ethylenediaminetetraacetic Acid; EGTA = Ethylene Glycol Tetraacetic Acid; ELISA = Enzyme-linked Immunosorbent Assay; EMEM = Eagle's Minimal Essential Medium; FBS = Foetal Bovine Serum; h = Hour(s); HRP = Horseradish Peroxidase; i.p. = intraperitoneal; PBS = Phosphate buffered saline; PBST = Phosphate buffered saline / Tween; TRIS = Tris(Hydroxymethyl)aminomethane; MTS reagent: [3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4- sulfophenyl)-2H-tetrazolium, inner salt, and an electron coupling reagent
(phenazine methosulfate) PMS; s.c. = sub-cutaneously.
ii. IC50 values were calculated using a smart fitting model in Genedata. The IC50 value was the concentration of test compound that inhibited 50% of biological activity.
Assay a): ATM Cellular Potency
Rationale:
Cellular irradiation induces DNA double strand breaks and rapid intermolecular autophosphorylation of serine 1981 that causes dimer dissociation and initiates cellular ATM kinase activity. Most ATM molecules in the cell are rapidly phosphorylated on this site after doses of radiation as low as 0.5 Gy, and binding of a phosphospecific antibody is detectable after the introduction of only a few DNA double-strand breaks in the cell.
The rationale of the pATM assay is to identify inhibitors of ATM in cells. HT29 cells are incubated with test compounds for lhr prior to X-ray-irradiation, lh later the cells are fixed and stained for pATM (Serl981). The fluorescence is read on the arrayscan imaging platform.
Method details (Protocol 1 or 2 can be used):
(1) HT29 cells (ECACC #85061109) were seeded into 384 well assay plates (Costar #3712) at a density of 3500 cells / well in 40 μΐ EMEM medium containing 1% L glutamine and 10% FBS and allowed to adhere overnight. The following morning compounds of Formula (I) in 100%) DMSO were added to assay plates by acoustic dispensing. After lh incubation at 37°C and 5% CO2, plates (up to 6 at a time) were irradiated using the X-RAD 320 instrument (PXi) with equivalent to ~600cGy. Plates were returned to the incubator for a further lh. Then cells were fixed by adding 20μ1 of 3.7% formaldehyde in PBS solution and incubating for 20 minutes at r.t. before being washed with 50μ1 / well PBS, using a Biotek EL405 plate washer. Then 20μ1 of 0.1 % Triton XI 00 in PBS was added and incubated for 20 minutes at r.t., to permeabalise cells. Then the plates were washed once with 50μ1 / well PBS, using a Biotek EL405 plate washer.
Phospho-ATM Serl981 antibody (Millipore #MAB3806) was diluted 10000 fold in PBS containing 0.05%> polysorbate/Tween and 3% BSA and 20μ1 was added to each well and incubated over night at r.t. The next morning plates were washed three times with 50μ1 / well PBS, using a Biotek EL405 plate washer, and then 20μ1 of secondary Ab solution, containing 500 fold diluted Alexa Fluor® 488 Goat anti-rabbit IgG (Life Technologies, Al 1001) and 0.002mg/ml Hoeschst dye (Life technologies #H-3570), in PBS containing 0.05%) polysorbate/Tween and 3% BSA, was added. After lh incubation at r.t., the plates were washed three times with 50μ1 / well PBS, using a Biotek EL405 plate washer, and plates were sealed and kept in PBS at 4°C until read. Plates were read using an ArrayScan VTI instrument, using an XF53 filter with 10X objective. A two laser set up was used to analyse nuclear staining with Hoeschst (405nm) and secondary antibody staining of pSerl981 (488nm).
(2) HT29 cells (ECACC #85061 109) were seeded into 384 well assay plates (Greiner # 781090) at a density of 3500 cells / well in 40μ1 EMEM medium containing 1% L glutamine and 10% FBS and allowed to adhere overnight. The following morning compounds of Formula (I) in 100% DMSO were added to assay plates by acoustic dispensing. After lh incubation at 37°C and 5% C02, plates (up to 6 at a time) were irradiated using the Multi Rad 160 X-Ray Irradiation cabinet (Faxitron) with equivalent to ~600cGy. Plates were returned to the incubator for a further lh. Then cells were fixed by adding 20μ1 of 3.7% formaldehyde in PBS solution and incubating for 20 minutes at r.t. before being washed with 50μ1 / well PBS, using a Biotek EL405 plate washer. Then 20μ1 of 0.1% Triton X100 in PBS was added and incubated for 20 minutes at r.t., to
permeabalise cells. Then the plates were washed once with 50μ1 / well PBS, using a Bluewasher plate washer (Blue Cat Bio).
Phospho-ATM Serl981 antibody (Millipore #MAB3806) was diluted 10000 fold in PBS containing 0.05%> polysorbate/Tween and 3% BSA and 20μ1 was added to each well and incubated over night at r.t. The next morning plates were washed three times with 50μ1 / well PBS, using a Bluewasher plate washer, and then 20μ1 of secondary Ab solution, containing 500 fold diluted Alexa Fluor® 488 Goat anti-rabbit IgG (Life Technologies, Al 1001) and 0.002mg/ml Hoeschst dye (Life technologies #H-3570), in PBS containing 0.05% polysorbate/Tween and 3% BSA, was added. After lh incubation at r.t., the plates were washed three times with 50μ1 / well PBS, using a Bluewasher plate washer, and plates were sealed and kept in PBS at 4°C until read. Plates were read using an Cell Insight imaging platform, using an XF53 filter with 10X objective. A two laser set up was used to analyse nuclear staining with Hoeschst (405nm) and secondary antibody staining of pSerl981 (488nm).
Assay b): ATR Cellular Potency
Rationale:
ATR is a PI 3-kinase-related kinase which phosphorylates multiple substrates on serine or threonine residues in response to DNA damage during or replication blocks. Chkl, a downstream protein kinase of ATR, plays a key role in DNA damage checkpoint control. Activation of Chkl involves phosphorylation of Ser317 and Ser345 (the latter regarded as the preferential target for phosphorylation/activation by ATR). This was a cell based assay to measure inhibition of ATR kinase, by measuring a decrease in
phosphorylation of Chkl (Ser 345) in HT29 cells, following treatment with compound of Formula (I) and the UV mimetic 4NQO (Sigma #N8141).
Method details:
HT29 cells (ECACC #85061109) were seeded into 384 well assay plates (Costar #3712) at a density of 6000 cells / well in 40 μΐ EMEM medium containing 1% L glutamine and 10% FBS and allowed to adhere overnight. The following morning compound of Formula (I) in 100%) DMSO were added to assay plates by acoustic dispensing. After lh incubation at 37°C and 5% CO2, 40nl of 3mM 4NQO in 100% DMSO was added to all wells by acoustic dispensing, except minimum control wells which were left untreated with 4NQO to generate a null response control. Plates were returned to the incubator for a further lh. Then cells were fixed by adding 20μ1 of 3.7% formaldehyde in PBS solution and incubating for 20 mins at r.t. Then 20μ1 of 0.1% Triton X100 in PBS was added and incubated for 10 minutes at r.t., to permeabalise cells. Then the plates were washed once with 50μ1 / well PBS, using a Biotek EL405 plate washer.
Phospho-Chkl Ser 345 antibody (Cell Signalling Technology #2348) was diluted 150 fold in PBS containing 0.05% polysorbate/Tween and 15μ1 was added to each well and incubated over night at r.t. The next morning plates were washed three times with 50μ1 / well PBS, using a Biotek EL405 plate washer, and then 20μ1 of secondary Ab solution, containing 500 fold diluted Alexa Fluor 488 Goat anti-rabbit IgG (Molecular Probes #A- 11008) and 0.002mg/ml Hoeschst dye (Molecular Probes #H-3570), in PBST, was added. After 2h incubation at r.t., the plates were washed three times with 50μ1 / well PBS, using a Biotek EL405 plate washer, and plates were then sealed with black plate seals until read. Plates were read using an ArrayScan VTI instrument, using an XF53 filter with 10X objective. A two laser set up was used to analyse nuclear staining with Hoeschst (405nm) and secondary antibody staining of pChkl (488nm).
Assay c): PI3K Cellular Potency
Rationale:
This assay was used to measure PI3K-a inhibition in cells. PDKl was identified as the upstream activation loop kinase of protein kinase B (Aktl), which is essential for the activation of PKB. Activation of the lipid kinase phosphoinositide 3 kinase (PI3K) is critical for the activation of PKB by PDKl .
Following ligand stimulation of receptor tyrosine kinases, PI3K is activated, which converts PIP2 to PIP3, which is bound by the PH domain of PDKl resulting in recruitment of PDKl to the plasma membrane where it phosphorylates AKT at Thr308 in the activation loop.
The aim of this cell -based mode of action assay is to identify compounds that inhibit PDK activity or recruitment of PDKl to membrane by inhibiting PI3K activity. Phosphorylation of phospho-Akt (T308) in BT474c cells following treatment with compounds for 2h is a direct measure of PDKl and indirect measure of PI3K activity.
Method details: BT474 cells (human breast ductal carcinoma, ATCC HTB-20) were seeded into black 384 well plates (Costar, #3712) at a density of 5600 cells / well in DMEM containing 10% FBS and 1% glutamine and allowed to adhere overnight.
The following morning compounds in 100% DMSO were added to assay plates by acoustic dispensing. After a 2h incubation at 37°C and 5% C02, the medium was aspirated and the cells were lysed with a buffer containing 25mM Tris, 3mM EDTA, 3mM EGTA, 50mM sodium fluoride, 2mM Sodium orthovanadate, 0.27M sucrose, lOmM β- glycerophosphate, 5mM sodium pyrophosphate, 0.5%> Triton X-100 and complete protease inhibitor cocktail tablets (Roche #04 693 116 001, used 1 tab per 50ml lysis buffer).
After 20 minutes, the cell lysates were transferred into ELISA plates (Greiner # 781077) which had been pre-coated with an anti total- AKT antibody in PBS buffer and non-specific binding was blocked with 1% BSA in PBS containing 0.05% Tween 20. Plates were incubated over night at 4°C. The next day the plates were washed with PBS buffer containing 0.05% Tween 20 and further incubated with a mouse monoclonal anti- phospho AKT T308 for 2h. Plates were washed again as above before addition of a horse anti-mouse-HRP conjugated secondary antibody. Following a 2h incubation at r.t, plates were washed and QuantaBlu substrate working solution (Thermo Scientific #15169, prepared according to provider's instructions) was added to each well. The developed fluorescent product was stopped after 60 minutes by addition of Stop solution to the wells. Plates were read using a Tecan Safire plate reader using 325nm excitation and 420nm emission wavelengths respectively. Except where specified, reagents contained in the Path Scan Phospho AKT (Thr308) sandwich ELISA kit from Cell Signalling (#7144) were used in this ELISA assay.
Assay d): mTOR Cellular Potency
Rationale:
This assay was used to measure mTOR inhibition in cells. The aim of the phospho- AKT cell based mechanism of action assay using the Acumen Explorer is to identify inhibitors of either PI3Ka or mTOR-Rictor (Rapamycin insensitive companion of mTOR). This is measured by any decrease in the phosphorylation of the Akt protein at Ser473 (AKT lies downstream of ΡΒΚα in the signal transduction pathway) in the MDA-MB-468 cells following treatment with compound.
Method details:
MDA-MB-468 cells (human breast adenocarcinoma #ATCC HTB 132) were seeded at 1500 cells / well in 40μ1 of DMEM containing 10% FBS and 1% glutamine into Greiner 384 well black flat-bottomed plates. Cell plates were incubated for 18h in a 37°C incubator before dosing with compounds of Formula (I) in 100%) DMSO using acoustic dispensing. Compounds were dosed in a 12 point concentration range into a randomised plate map. Control wells were generated either by dosing of 100% DMSO (max signal) or addition of a reference compound (a ΡΙ3Κ-β inhibitor) that completely eliminated the pAKT signal (min control). Compounds were then tested by one of two assay protocols A or B:
Protocol A:
Plates were incubated at 37°C for 2h; cells were then fixed by the addition of ΙΟμΙ of a 3.7%) formaldehyde solution. After 30 minutes the plates were washed with PBS using a Tecan PW384 plate washer. Wells were blocked and cells permeabilised with the addition of 40μ1 of PBS containing 0.5%> Tween20 and 1% Marvel™ (dried milk powder) and incubated for 60 minutes at r.t. The plates were washed with PBS containing 0.5% (v/v) Tween20 and 20μ1 rabbit anti-phospho AKT Ser473 (Cell Signalling Technologies, #3787) in same PBS-Tween + 1% Marvel™ was added and incubated overnight at 4°C.
Plates were washed 3 times with PBS + 0.05%> Tween 20 using a Tecan PW384. 20μ1 of secondary antibody Alexa Fluor 488 anti-Rabbit (Molecular Probes, #A11008) diluted in PBS + 0.05%> Tween20 containing 1% Marvel™ was added to each well and incubated for lh at r.t. Plates were washed three times as before then 20μ1 PBS added to each well and plates sealed with a black plate sealer.
The plates were read on an Acumen plate reader as soon as possible, measuring green fluorescence after excitation with 488nm laser. Using this system IC50 values were generated and quality of plates was determined by control wells. Reference compounds were run each time to monitor assay performance. Protocol B:
The cell plates were then incubated for 2 h at 37°C before being fixed by the addition of 20μ1 3.7% formaldehyde in PBS/A (1.2% final concentration), followed by a 30 minute room temperature incubation, and then a 2x wash with 150μ1 PBS/A using a BioTek ELx406 platewasher. Cells were permeabilised and blocked with 20μ1 of assay buffer (0.1% Triton X-100 in PBS/A + 1% BSA) for lh at room temperature, and then washed lx with 50μ1 PBS/A. Primary phospho-AKT (Ser473) D9E XP® rabbit monoclonal antibody (#4060, Cell Signaling Technology) was diluted 1 :200 in assay buffer, 20μ1 added per well, and plates were incubated at 4°C overnight. Cell plates were washed 3x with 200μ1 PBS/T, then 20μ1 1 :750 dilution in assay buffer of Alexa Fluor® 488 goat anti -rabbit IgG secondary antibody (#A1 1008, Molecular Probes, Life
Technologies), with a 1 :5000 dilution of Hoechst 33342, was added per well. Following a 1 h incubation at room temperature, plates were washed 3x with 200μ1 PBS/T, and 40μ1 PBS w/o Ca, Mg and Na Bicarb (Gibco #14190-094) was added per well.
Stained cell plates were covered with black seals, and then read on the Cell Insight imaging platform (Thermo Scientific), with a lOx objective. The primary channel (Hoechst blue fluorescence 405nM, BGRFR 386 23) was used to Auto focus and to count number of events (this provided information about cytotoxicity of the compounds tested). The secondary channel (Green 488nM, BGRFR 485 20) measured pAKT staining. Data was analysed and ICsos were calculated using Genedata Screener® software.
Table 1 shows the results of testing the Examples in tests a) b) c) and d). Results may be the geometric mean of several tests.
Table 1 : Potency Data for Example 1 in Assays a) - d)
Figure imgf000144_0001
0.0128 >30 12.8 >30
0.0155 >30 >30 >30
0.0218 >30 26.9 >10
0.00777 >30 14.5 >30
0.0102 >30 0.011, >30, >30
>30
0.0145 >30 >30 25
0.0325 >30 >30 >30
0.00918 >30 >30 >30
0.00547 >30 16.8 >30
0.00369 >10
0.0301 >30 >30 >30
0.0202 >30 >30 >30
0.0119 >30 >30 >30
0.017 >30 >30 >30
0.00344 >30 >30 >30
0.00541 >30 >30 >30
0.0173 >30 28.9 >30
0.0216 >30 >30 >30
0.0334 >10 >30 >30
0.0171 >30 >30 >30
0.0819 >30 5.1 >30
0.351 >30 >30 0.0044, >30,
>5.4
0.162 >30 >30 >30
0.0342 >30 6.64 >30
0.00357 >30 >30 >30
0.00424 >30 >30 >30
0.0102 >30 >30 >30
0.00357 >30 >30 >30
0.00461 >30 >30 >30
>30 4.25 >10
0.0163 >30 >30 >30
0.0042 >30 >30 >30
0.0255 >30 >30 >30
0.00603 >30 >30 2.17
0.141 >30 >25.2 >30
0.00281 >30 16.9 >30
0.0624 >30 >30 >30
0.0359 >30 >30 >30
0.333 >30 >3.12
0.00739 >30 25.6 >30 0.0525 >30 >30 >30
0.0475 >30 >30 >30
0.0189 >30 >30 >30
0.0608 >30 27.5 >30
0.247 >30 4.78 >30
0.182 >30 >30 >30
0.291 >30 >30 >30
0.407 >30 >30 >30
0.00781 >30 5.67 >30
0.0138 >30 >30 >30
0.0327 >30 >30 27.6
0.0189 >30 21.3 >30
0.0229 >30 >30 >30
0.00771 >30 >30 >30
0.0184 >30 >30 >30
0.0144 14.7
0.0362 >30 23.4 >30
0.0067 >30 >30 >30
0.0048 >30 >30 >30
0.0207 >30 >30 >30
0.0089 >30 >30 >30
0.0385 >30 >30 >30
0.0133 >30 0.99 >30
0.0223 >30 >30 >10
0.0444 >30 >30 >30
0.0369 >30 >30 >30
0.126 >30 >30 >30
0.359 >30 >30 >30
0.183 >30 >30 >30
0.0202 >30 >30 >30

Claims

Claims
1. A compound of formula (I)
Figure imgf000147_0001
or a pharmaceutically acceptable salt thereof, wherein
A is N or CR4;
X is -OR5 or -NR6R7;
R1 is (Ci-Ce)alkyl, cycloalkyl or heterocycloalkyl;
R2 is H or methyl, wherein methyl is optionally substituted with 1, 2, or 3 halo substituents;
R3 is H, (Ci-C6)alkyl or halo;
R4 is H, (Ci-C6)alkyl or halo;
R5 is -(CH2)n-NR8R9;
R6 and R7 together with the nitrogen atom to which they are attached form a 4, 5 or 6 membered non-aromatic monocyclic ring comprising carbon which is substituted with -
NR10RU;
R8 and R9 are independently selected from H and (Ci-Ce)alkyl; or R8 and R9 together with the nitrogen atom to which they are attached form a 4, 5 or 6 membered non-aromatic monocyclic ring comprising carbon which can be optionally substituted with halo;
R10 is H or (Ci-C3)alkyl;
R11 is (Ci-C )alkyl;
n is 2, 3 or 4;
alkyl is a linear or branched saturated hydrocarbon;
alkoxy is a linear or branched O-linked saturated hydrocarbon;
cycloalkyl is a 3, 4, 5 or 6 membered aliphatic carbocyclic ring, which can be optionally substituted with 1 or 2 substituents selected from (Ci-C3)alkyl, (Ci-C3)alkoxy and -OH; halo is F, CI or Br; heterocycloalkyl is a 4, 5 or 6 membered non-aromatic monocyclic ring comprising carbon and 1 or 2 heteroatoms selected from nitrogen and oxygen; wherein heterocycloalkyl can be optionally substituted with 1 or 2 substituents selected from (Ci-C3)alkyl, (Ci-C3)alkoxy and -OH.
2. A compound as claimed in claim 1, wherein said compound is a compound of formula (la)
Figure imgf000148_0001
or a pharmaceutically acceptable salt thereof, wherein
A is N or CH;
X is -OR5 or -NR6R7;
R1 is (Ci-C3)alkyl, cycloalkyl, heterocycloalkyl;
R2 is H or methyl, wherein methyl is optionally substituted with 1, 2, or 3 fluoro substituents;
R3 is H, methyl, chloro or fluoro;
R4 is H, methyl, chloro or fluoro;
R5 is -(CH2)n-NR8R9;
R6 and R7 together with the nitrogen atom to which they are attached form a 4, 5 or 6 membered non-aromatic monocyclic ring comprising carbon which is substituted with - NR10RU;
R8 and R9 are independently selected from H and (Ci-C3)alkyl; or R8 and R9 together with the nitrogen atom to which they are attached form a 4, 5 or 6 membered non-aromatic monocyclic ring comprising carbon which can be optionally substituted with fluoro;
R10 is H or (Ci-C3)alkyl;
R11 is (Ci-C3)alkyl;
n is 2, 3 or 4; alkyl is a linear or branched saturated hydrocarbon;
cycloalkyl is a 3, 4, 5 or 6 membered aliphatic carbocyclic ring, which can be optionally substituted with 1 or 2 substituents selected from methyl, methoxy and -OH;
heterocycloalkyl is a 4, 5 or 6 membered non-aromatic monocyclic ring comprising carbon and a heteroatom selected from oxygen; wherein heterocycloalkyl can be optionally substituted with 1 or 2 substituents selected from methyl, methoxy and -OH.
3. A compound as claimed in claim 1 or claim 2, or a pharmaceutically acceptable salt thereof, wherein R1 is n-propyl, iso-propyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, oxetanyl, tetrahydrofuranyl or tertrahydropyranyl; wherein cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, tetrahydrofuranyl or tertrahydropyranyl can be optionally substituted with 1 or 2 substituents selected from methyl, methoxy and -OH.
4. A compound as claimed in any one of claims 1 to 3, or a pharmaceutically acceptable salt thereof, wherein R2 is methyl optionally substituted with 1, 2, or 3 fluoro substituents.
5. A compound as claimed in any one of claims 1 to 4, or a pharmaceutically acceptable salt thereof, wherein R3 and R4 are independently selected from H, methyl and fluoro.
6. A compound as claimed in any one of claims 1 to 5, or a pharmaceutically acceptable salt thereof, wherein X is -OR5 and R5 is -(CH2)3-NR8R9.
7. A compound as claimed in any one of claims 1 to 5, or a pharmaceutically acceptable salt thereof, wherein X is -NR6R7.
8. A compound as claimed in claim 6, or a pharmaceutically acceptable salt thereof, wherein R8 and R9 are methyl; or R8 and R9 together with the nitrogen atom to which they are attached form a 5 or 6 membered non-aromatic monocyclic ring comprising carbon which can be optionally substituted with fluoro.
9. A compound as claimed in claim 7, or a pharmaceutically acceptable salt thereof, wherein R6 and R7 together with the nitrogen atom to which they are attached form a 5 or 6 membered non-aromatic monocyclic ring comprising carbon which is substituted with - N(CH3)2.
10. A compound as claimed in claim 1 or claim 2, or a pharmaceutically acceptable salt thereof, selected from:
8-[6- [3 -(Dimethylamino)propoxy] -3 -pyridyl] -3 -methyl- 1 -tetrahydropyran-4-yl- imidazo[4,5-c]cinnolin-2-one;
8-[6- [3 -(dimethylamino)propoxy] -3 -pyridyl] - 1 -(cis-3 -methoxycyclobutyl)-3 -methyl- imidazo[4,5-c]cinnolin-2-one;
8-[6- [3 -(Dimethylamino)propoxy] -3 -pyridyl] -3 -methyl- 1 -[(3 S)-tetrahydropyran-3 - yl]imidazo[4,5-c]cinnolin-2-one;
8-[6- [3 -(Dimethylamino)propoxy] -3 -pyridyl] -3 -methyl- 1 -[(3R)-tetrahydropyran-3 - yl]imidazo[4,5-c]cinnolin-2-one;
3-Methyl-8-[6-[3-(l -piperidyl)propoxy] -3 -pyridyl]- 1 -tetrahydropyran-4-yl-imidazo[4,5- c]cinnolin-2-one;
8-[4- [3 -(dimethylamino)propoxy]phenyl] - 1 -isopropyl-3 -methyl-imidazo [4,5 -c] cinnolin-2- one;
1 -Isopropyl-3 -methyl-8 -[6- [3 -( 1 -piperidyl)propoxy] -3 -pyridyl] imidazo [4,5 -c] cinnolin-2- one;
8 - [6- [3 -(dimethylamino)propoxy] -3 -pyridyl] - 1 -isopropyl-3 -methyl-imidazo [4,5- c]cinnolin-2-one;
1 -Isopropyl-3-methyl-8-[4-(3-pyrrolidin- 1 -ylpropoxy)phenyl]imidazo[4,5-c]cinnolin-2- one;
l-isopropyl-3-methyl-8-(4-(3-(piperidin-l-yl)propoxy)phenyl)-l,3-dihydro-2H- imidazo[4,5-c]cinnolin-2-one;
8-[6- [3 -(dimethylamino)propoxy] -3 -pyridyl] - 1 -(trans-3 -methoxycyclobutyl)-3 -methyl - imidazo[4,5-c]cinnolin-2-one; 8-[6-[3-(4-Fluoro-l-piperidyl)propoxy]-3-pyridyl]-l-isopropyl-3-methyl-imidazo[4,5- c]cinnolin-2-one;
8-[6-[3-[(3R)-3-Fluoropyrrolidin-l-yl]propoxy]-3-pyridyl]-l-isopropyl-3-methyl- imidazo[4,5-c]cinnolin-2-one;
l-isopropyl-3-methyl-8-[2-methyl-6-[3-(l-piperidyl)propoxy]-3-pyridyl]imidazo[4,5- c]cinnolin-2-one;
1 -(tra/75-3-Methoxycyclobutyl)-3-methyl-8-[4-[3-(l - piperidyl)propoxy]phenyl]imidazo[4,5-c]cinnolin-2-one;
8-[4- [3 -(Dimethylamino)propoxy]phenyl] - 1 -(trans-3 -methoxycyclobutyl)-3 -methyl- imidazo[4,5-c]cinnolin-2-one;
8-[6- [3 -(4-Fluoro- 1 -piperidyl)propoxy] -3 -pyridyl] - 1 -(trans-3 -methoxycyclobutyl)-3 - methyl-imidazo[4,5-c]cinnolin-2-one;
8-[6- [3 -(4-Fluoro- 1 -piperidyl)propoxy] -3 -pyridyl] - 1 -(cis-3 -methoxycyclobutyl)-3 -methyl- imidazo[4,5-c]cinnolin-2-one;
1 -(cis-3 -methoxycyclobutyl)-3 -methyl-8- [4-[3 -( 1 -piperidyl)propoxy]phenyl]imidazo[4,5- c]cinnolin-2-one;
8-[6-[3-[(3R)-3-Fluoropyrrolidin-l-yl]propoxy]-3-pyridyl]-l-(cz'5-3-methoxycyclobutyl)-3- methyl-imidazo[4,5-c]cinnolin-2-one;
8-[4-[3-(Dimethylamino)propoxy]phenyl]-3-methyl-l-[(3R)-tetrahydropyran-3- yl]imidazo[4,5-c]cinnolin-2-one;
3-Methyl-8-[4-[3-(l -piperidyl)propoxy]phenyl]- 1 -[(3R)-tetrahydropyran-3-yl]imidazo[4,5- c]cinnolin-2-one;
8-[6- [3 -(4-Fluoro- 1 -piperidyl)propoxy] -3 -pyridyl] -3 -methyl- 1 - [(3R)-tetrahydropyran-3 - yl]imidazo[4,5-c]cinnolin-2-one;
8-[4-[3-(Dimethylamino)propoxy]phenyl]-3-methyl- 1 -(1 -methylcyclopropyl)imidazo[4,5- c]cinnolin-2-one;
3-Methyl-l-(l-methylcyclopropyl)-8-[4-[3-(l-piperidyl)propoxy]phenyl]imidazo[4,5- c]cinnolin-2-one;
8-[6-[3-(4-Fluoro- 1 -piperidyl)propoxy]-3-pyridyl]-3-methyl-l -(1 - methylcyclopropyl)imidazo[4,5-c]cinnolin-2-one; 3-Methyl-l-(l-methylcyclopropyl)-8-[6-[3-(l-piperidyl)propoxy]-3-pyridyl]imidazo[4,5- c]cinnolin-2-one;
8-[4-[3-(dimethylamino)propoxy]phenyl]-l-[(lR,3R)-3-methoxycyclopentyl]-3-methyl- imidazo [4,5 -c] cinnolin-2-one;
l-[(lS,3S)-3-methoxycyclopentyl]-3-methyl-8-[4-[3-(l- piperidyl)propoxy]phenyl]imidazo[4,5-c]cinnolin-2-one;
8-[4- [3 -(Dimethylamino)propoxy]phenyl] - l-[(lS,3S)-3 -methoxycyclopentyl] -3 -methyl - imidazo[4,5-c]cinnolin-2-one;
l-[(lR,3R)-3-Methoxycyclopentyl]-3-methyl-8-[6-[3-(l-piperidyl)propoxy]-3- pyridyl]imidazo[4,5-c]cinnolin-2-one;
8-[6-[3-[(3S)-3-Fluoropyrrolidin-l-yl]propoxy]-3-pyridyl]-l-[(lR,3R)-3- methoxycyclopentyl] -3 -methyl-imidazo [4,5 -c] cinnolin-2-one;
l-[(lS,3S)-3-Methoxycyclopentyl]-3-methyl-8-[6-[3-(l-piperidyl)propoxy]-3- pyridyl]imidazo[4,5-c]cinnolin-2-one;
8-[4- [3 -(Dimethylamino)propoxy]phenyl] - 1 -(cis-3 -methoxycyclobutyl)-3 -methyl- imidazo[4,5-c]cinnolin-2-one;
1 -(cis-3 -Methoxycyclobutyl)-3 -methyl-8-[6- [3 -( 1 -piperidyl)propoxy] -3 - pyridyl]imidazo[4,5-c]cinnolin-2-one;
8-[6-[3-[(3S)-3-Fluoropyrrolidin-l-yl]propoxy]-3-pyridyl]-l-(tra/75-3-methoxycyclobutyl)- 3 -methyl-imidazo [4,5 -c] cinnolin-2-one;
8-[6- [3 -(dimethylamino)propoxy] -3 -pyridyl] - 1 - [(4S)-3 ,3 -dimethyltetrahydropyran-4-yl] -3 - methyl-imidazo[4,5-c]cinnolin-2-one;
8-[6- [3 -(dimethylamino)propoxy] -3 -pyridyl] - 1 - [(4R)-3 ,3 -dimethyltetrahydropyran-4-yl] -3 - methyl-imidazo [4,5 -c] cinnolin-2-one
l-[3,3-dimethyltetrahydropyran-4-yl]-3-methyl-8-{4-[3-(l-piperidinyl)propoxy]phenyl}- l,3-dihydro-2H-imidazo[4,5-c]cinnolin-2-one;
8-[4- [3 -(dimethylamino)propoxy]phenyl] - 1 -[(4S)-3 ,3 -dimethyltetrahydropyran-4-yl] -3 - methyl-imidazo[4,5-c]cinnolin-2-one;
8-[4- [3 -(dimethylamino)propoxy]phenyl] - 1 -[(4R)-3 ,3 -dimethyltetrahydropyran-4-yl] -3 - methyl-imidazo [4,5 -c] cinnolin-2-one; 8-[4- [3 -(dimethylamino)propoxy]phenyl] -7-fluoro- 1 -isopropyl-3 -methyl-imidazo [4,5 - c]cinnolin-2-one;
8-[6- [3 -(Dimethylamino)propoxy] -3 -pyridyl] -7-fluoro- 1 -isopropyl-3 -methyl-imidazo [4, 5- c]cinnolin-2-one;
7-Fluoro-l-isopropyl-3-methyl-8-[6-[3-(l-piperidyl)propoxy]-3-pyridyl]imidazo[4,5- c]cinnolin-2-one;
7- fluoro- 1 -isopropyl-3 -methyl-8 -[4- [3 -( 1 -piperidyl)propoxy]phenyl]imidazo [4,5 - c]cinnolin-2-one;
8- [4-[3-(dimethylamino)propoxy]phenyl]-l-isopropyl-3,7-dimethyl-imidazo[4,5- c]cinnolin-2-one;
1- isopropyl-3,7-dimethyl-8-[6-[3-(l-piperidyl)propoxy]-3-pyridyl]imidazo[4,5-c]cinnolin
2- one;
8-[6- [3 -(dimethylamino)propoxy] -3 -pyridyl] - 1 -isopropyl-3 ,7-dimethyl-imidazo [4,5 - c]cinnolin-2-one;
l-Isopropyl-3,7-dimethyl-8-[4-[3-(l-piperidyl)propoxy]phenyl]imidazo[4,5-c]cinnolin-2- one;
3- (difluoromethyl)-8-[6-[3-(dimethylamino)propoxy]-3-pyridyl]-l-isopropyl-imidazo[4,5 c]cinnolin-2-one;
8-[6- [(3R)-3 -(Dimethylamino)pyrrolidin- 1 -yl] -3 -pyridyl] - 1 -isopropyl-3 -methyl- imidazo[4,5-c]cinnolin-2-one;
8-[6-[4-(Dimethylamino)- 1 -piperidyl] -3 -pyridyl]- 1 -isopropyl-3 -methyl-imidazo [4,5 - c]cinnolin-2-one;
8-[6-[(3R)-3-(dimethylamino)pyrrolidin- 1 -yl] -3 -pyridyl] -3 -methyl- 1 -[(3R)- tetrahydropyran-3 -yl] imidazo [4,5 -c] cinnolin-2-one;
8-[6- [4-(dimethylamino)- 1 -piperidyl] -3 -pyridyl] - 1 -[3 ,3 -dimethyltetrahydropyran-4-yl] -3 - methyl-imidazo[4,5-c]cinnolin-2-one;
8-[6-[(3R)-3-(dimethylamino)pyrrolidin- 1 -yl] -3 -pyridyl]- 1 -[3,3-dimethyltetrahydropyran-
4- yl] -3 -methyl-imidazo [4,5 -c] cinnolin-2-one;
8-[6- [(3R)-3 -(Dimethylamino)pyrrolidin- 1 -yl] -3 -pyridyl] - 1 -(trans-3 -methoxycyclobutyl)- 3 -methyl-imidazo [4,5 -c] cinnolin-2-one; 8-[6- [(3R)-3 -(Dimethylamino)pyrrolidin- 1 -yl] -3 -pyridyl] - 1 -(cis-3 -methoxycyclobutyl)-3- methyl-imidazo [4,5 -c] cinnolin-2-one;
8-[6-[3-(dimethylamino)pyrrolidin- 1 -yl] -3 -pyridyl] -3 -methyl- 1 -tetrahydropyran-4-yl- imidazo[4,5-c]cinnolin-2-one;
8-[6-[(3R)-3-(Dimethylamino)pyrrolidin-l-yl]-3-pyridyl]-l-[(lR,3R)-3- methoxycyclopentyl] -3 -methyl-imidazo [4,5 -c] cinnolin-2-one;
8-[6-[(3R)-3-(Dimethylamino)pyrrolidin-l-yl]-3-pyridyl]-l-[(lS,3S)-3- methoxycyclopentyl] -3 -methyl-imidazo [4,5 -c] cinnolin-2-one;
8-[6-[4-(Dimethylamino)- 1 -piperidyl] -3 -pyridyl]- 1 -(czs-3-methoxycyclobutyl)-3 -methyl - imidazo[4,5-c]cinnolin-2-one;
8-[4-[4-(Dimethylamino)- 1 -piperidyl]phenyl]- 1 -isopropyl-3-methyl-imidazo[4,5- c]cinnolin-2-one;
8-[4-[4-(dimethylamino)- 1 -piperidyl]phenyl]- 1 -[3,3-dimethyltetrahydropyran-4-yl]-3- methyl-imidazo[4,5-c]cinnolin-2-one;
8-[4-[4-(Dimethylamino)- 1 -piperidyl]phenyl]- 1 -[(lR,3R)-3-methoxycyclopentyl]-3- methyl-imidazo[4,5-c]cinnolin-2-one;
8-[4-[4-(Dimethylamino)- 1 -piperidyl]phenyl]- 1 -[(1 S,3S)-3-methoxycyclopentyl]-3- methyl-imidazo[4,5-c]cinnolin-2-one;
8-[4-[4-(Dimethylamino)- 1 -piperidyl]phenyl] -3 -methyl- 1 -[(3R)-tetrahydropyran-3- yl]imidazo[4,5-c]cinnolin-2-one;
8-[6- [(3R)-3 -(Dimethylamino)pyrrolidin- 1 -yl] -3 -pyridyl] -7-fluoro- 1 -isopropyl-3 -methyl- imidazo[4,5-c]cinnolin-2-one;
8-[4-[4-(Dimethylamino)-l-piperidyl]phenyl]-7-fluoro-l-isopropyl-3-methyl-imidazo[4,5- c]cinnolin-2-one;
8-[6-[(3R)-3-(dimethylamino)pyrrolidin- 1 -yl] -3 -pyridyl]- 1 -isopropyl-3, 7-dimethyl- imidazo[4,5-c]cinnolin-2-one;
8-[4-[4-(dimethylamino)- 1 -piperidyl]phenyl]- 1 -isopropyl-3, 7-dimethyl -imidazo[4,5- c]cinnolin-2-one;
8-[6-[3-(dimethylamino)propoxy]-2-fluoro-3-pyridyl]-l-isopropyl-3-methyl-imidazo[4,5- c]cinnolin-2-one.
11. A compound as claimed in claim 1 or claim 2, or a pharmaceutically acceptable salt thereof, selected from:
1 -Isopropyl-3 -methyl-8 -[6- [3 -( 1 -piperidyl)propoxy] -3 -pyridyl]imidazo [4,5 -c] cinnolin-2- one;
8 - [6- [3 -(dimethylamino)propoxy] -3 -pyridyl] - 1 -isopropyl-3 -methyl-imidazo [4,5- c]cinnolin-2-one;
1 -Isopropyl-3-methyl-8-[4-(3-pyrrolidin- 1 -ylpropoxy)phenyl]imidazo[4,5-c]cinnolin-2- one;
8-[6-[3-(4-Fluoro-l-piperidyl)propoxy]-3-pyridyl]-l-isopropyl-3-methyl-imidazo[4,5- c]cinnolin-2-one;
8-[6-[3-[(3R)-3-Fluoropyrrolidin-l-yl]propoxy]-3-pyridyl]-l-isopropyl-3-methyl- imidazo[4,5-c]cinnolin-2-one;
8-[4-[3-(dimethylamino)propoxy]phenyl]-l-[(lR,3R)-3-methoxycyclopentyl]-3-methyl- imidazo[4,5-c]cinnolin-2-one;
8-[4- [3 -(Dimethylamino)propoxy]phenyl] - l-[(lS,3S)-3 -methoxycyclopentyl] -3 -methyl - imidazo[4,5-c]cinnolin-2-one;
8-[6- [3 -(dimethylamino)propoxy] -3 -pyridyl] - 1 - [(4R)-3 ,3 -dimethyltetrahydropyran-4-yl] -3 - methyl-imidazo[4,5-c]cinnolin-2-one;
8- {4-[3-(dimethylamino)propoxy]phenyl} - 1 -[(4R)-3,3-dimethyltetrahydro-2H-pyran-4-yl]- 3 -methyl- 1 ,3 -dihydro-2H-imidazo [4,5 -c] cinnolin-2-one;
7- Fluoro-l-isopropyl-3-methyl-8-[6-[3-(l-piperidyl)propoxy]-3-pyridyl]imidazo[4,5- c]cinnolin-2-one;
8- [6- [(3R)-3 -(Dimethylamino)pyrrolidin- 1 -yl] -3 -pyridyl] - 1 -isopropyl-3 -methyl- imidazo[4,5-c]cinnolin-2-one;
8-[6-[(3R)-3-(dimethylamino)pyrrolidin- 1 -yl] -3 -pyridyl] -3 -methyl- 1 -[(3R)- tetrahydropyran-3 -yl] imidazo [4,5 -c] cinnolin-2-one;
8-[4-[4-(Dimethylamino)- 1 -piperidyl]phenyl]- 1 -isopropyl-3-methyl-imidazo[4,5- c]cinnolin-2-one.
12. A pharmaceutical composition which comprises a compound of Formula (I), or a pharmaceutically acceptable salt thereof, as claimed in any one of claims 1 to 11 , and at least one pharmaceutically acceptable excipient.
13. A compound of Formula (I), or a pharmaceutically acceptable salt thereof, as claimed in any one of claims 1 to 11 , for use in therapy.
14. A compound of Formula (I), or a pharmaceutically acceptable salt thereof, as claimed in any one of claims 1 to 11 , for use in the treatment of cancer.
15. A compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer according to claim 14, wherein the compound of Formula (I) is administered simultaneously, separately or sequentially with radiotherapy.
16. A compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of cancer according to claim 14, where the compound of Formula (I) is administered simultaneously, separately or sequentially with at least one additional anti- tumour substance selected from the group consisting of doxorubicin, irinotecan, topotecan, etoposide, mitomycin, bendamustine, chlorambucil, cyclophosphamide, ifosfamide, carmustine, melphalan and bleomycin.
17. Use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, as claimed in any one of claims 1 to 11 , in the manufacture of a medicament for the treatment of cancer.
18. A method for treating cancer in a warm blooded animal in need of such treatment, which comprises administering to said warm-blooded animal a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, as claimed in any one of claims 1 to 11.
PCT/EP2018/075327 2017-09-20 2018-09-19 1,3-dihydroimidazo[4,5-c]cinnolin-2-one compounds and their use in treating cancer WO2019057757A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201880073208.2A CN111344293A (en) 2017-09-20 2018-09-19 1, 3-dihydroimidazo [4, 5-c ] cinnolin-2-one compounds and their use in the treatment of cancer

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201762560848P 2017-09-20 2017-09-20
US62/560,848 2017-09-20

Publications (1)

Publication Number Publication Date
WO2019057757A1 true WO2019057757A1 (en) 2019-03-28

Family

ID=63667913

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/EP2018/075327 WO2019057757A1 (en) 2017-09-20 2018-09-19 1,3-dihydroimidazo[4,5-c]cinnolin-2-one compounds and their use in treating cancer

Country Status (2)

Country Link
CN (1) CN111344293A (en)
WO (1) WO2019057757A1 (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020052688A1 (en) * 2018-09-14 2020-03-19 Suzhou Zanrong Pharma Limited 1-ISOPROPYL-3-METHYL-8- (PYRIDIN-3-YL) -1, 3-DIHYDRO-2H-IMIDAZO [4, 5-c] CINNOLIN-2-ONE AS SELECTIVE MODULATORS OF ATAXIA TELANGIECTASIA MUTATED (ATM) KINASE AND USES THEREOF
WO2021260580A1 (en) 2020-06-24 2021-12-30 Astrazeneca Uk Limited Combination of antibody-drug conjugate and atm inhibitor
WO2022194138A1 (en) * 2021-03-17 2022-09-22 Suzhou Zanrong Pharma Limited Selective modulators of ataxia telangiectasia mutated (atm) kinase and uses thereof

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997022596A1 (en) 1995-12-18 1997-06-26 Zeneca Limited Quinazoline derivatives
WO1997030035A1 (en) 1996-02-13 1997-08-21 Zeneca Limited Quinazoline derivatives as vegf inhibitors
WO1997032856A1 (en) 1996-03-05 1997-09-12 Zeneca Limited 4-anilinoquinazoline derivatives
WO1998013354A1 (en) 1996-09-25 1998-04-02 Zeneca Limited Quinazoline derivatives and pharmaceutical compositions containing them
CN102199152A (en) * 2010-03-25 2011-09-28 高大新 Heterocycle imidazoles phosphatide kinases inhibitor
WO2015170081A1 (en) 2014-05-08 2015-11-12 Astrazeneca Ab Imidazo[4,5-c]quinolin-2-one compounds and their use in treating cancer
WO2017046216A1 (en) 2015-09-17 2017-03-23 Astrazeneca Ab 8-[6-[3-(amino)propoxy]-3-pyridyl]-1 -isopropyl-imidazo[4,5-c]quinolin-2-one derivatives as selective modulators of ataxia telangiectasia mutated (atm) kinase for the treatment of cancer
WO2017076895A1 (en) 2015-11-03 2017-05-11 Astrazeneca Ab Imidazo[4,5-c]quinolin-2-one compounds and their use in treating cancer
WO2017076898A1 (en) 2015-11-05 2017-05-11 Astrazeneca Ab Imidazo[4,5-c]quinolin-2-one compounds and their use in treating cancer
WO2017162605A1 (en) * 2016-03-21 2017-09-28 Astrazeneca Ab Cinnolin-4-amine compounds and their use in treating cancer

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1997022596A1 (en) 1995-12-18 1997-06-26 Zeneca Limited Quinazoline derivatives
WO1997030035A1 (en) 1996-02-13 1997-08-21 Zeneca Limited Quinazoline derivatives as vegf inhibitors
WO1997032856A1 (en) 1996-03-05 1997-09-12 Zeneca Limited 4-anilinoquinazoline derivatives
WO1998013354A1 (en) 1996-09-25 1998-04-02 Zeneca Limited Quinazoline derivatives and pharmaceutical compositions containing them
CN102199152A (en) * 2010-03-25 2011-09-28 高大新 Heterocycle imidazoles phosphatide kinases inhibitor
WO2015170081A1 (en) 2014-05-08 2015-11-12 Astrazeneca Ab Imidazo[4,5-c]quinolin-2-one compounds and their use in treating cancer
WO2017046216A1 (en) 2015-09-17 2017-03-23 Astrazeneca Ab 8-[6-[3-(amino)propoxy]-3-pyridyl]-1 -isopropyl-imidazo[4,5-c]quinolin-2-one derivatives as selective modulators of ataxia telangiectasia mutated (atm) kinase for the treatment of cancer
WO2017076895A1 (en) 2015-11-03 2017-05-11 Astrazeneca Ab Imidazo[4,5-c]quinolin-2-one compounds and their use in treating cancer
WO2017076898A1 (en) 2015-11-05 2017-05-11 Astrazeneca Ab Imidazo[4,5-c]quinolin-2-one compounds and their use in treating cancer
WO2017162605A1 (en) * 2016-03-21 2017-09-28 Astrazeneca Ab Cinnolin-4-amine compounds and their use in treating cancer

Non-Patent Citations (11)

* Cited by examiner, † Cited by third party
Title
"Handbook of Pharmaceutical Excipients", PHARMACEUTICAL PRESS
"Handbook of Pharmaceutical Salts: Properties, Selection and Use", 2002, WEINHEIM/ZIIRICH:WILEY-VCH/VHCA
BAKKENIST ET AL., NATURE, 2003, pages 499 - 506
CHEMICAL ABSTRACTS, Columbus, Ohio, US; abstract no. 90272-08-5
CREMONA ET AL., ONCOGENE, 2013, pages 3351 - 3360
FERNANDEZ-CAPETILLO ET AL., SCIENCE SIGNALLING, vol. 9, no. 445, 2016, pages ra91
KURZ; LEES MILLER, DNA REPAIR, 2004, pages 889 - 900
LAVIN, M. F., REV. MOL. CELL BIOL., 2008, pages 759 - 769
MATSUOKA ET AL., SCIENCE, 2007, pages 1160 - 1166
PIKE ET AL.: "The Identification of Potent, Selective, and Orally Available Inhibitors of Ataxia Telangiectasia Mutated (ATM) Kinase: The Discovery of AZD0156 (8-{6-[3-(Dimethylamino)propoxy]pyridin-3-yl}-3-methyl-1-(tetrahydro-2 H -pyran-4-yl)-1,3-dihydro-2 H -imidazo[4,5- c ]quinolin-2-one)", J. MED. CHEM., vol. 61, no. 9, 23 April 2018 (2018-04-23), pages 3823 - 3841, XP055516276, ISSN: 0022-2623, DOI: 10.1021/acs.jmedchem.7b01896 *
SCHOFIELD, K.; SWAIN, T.: "The preparation is also described in the literature", JOURNAL OF THE CHEMICAL SOCIETY, 1949, pages 2393 - 9

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020052688A1 (en) * 2018-09-14 2020-03-19 Suzhou Zanrong Pharma Limited 1-ISOPROPYL-3-METHYL-8- (PYRIDIN-3-YL) -1, 3-DIHYDRO-2H-IMIDAZO [4, 5-c] CINNOLIN-2-ONE AS SELECTIVE MODULATORS OF ATAXIA TELANGIECTASIA MUTATED (ATM) KINASE AND USES THEREOF
WO2021260580A1 (en) 2020-06-24 2021-12-30 Astrazeneca Uk Limited Combination of antibody-drug conjugate and atm inhibitor
WO2022194138A1 (en) * 2021-03-17 2022-09-22 Suzhou Zanrong Pharma Limited Selective modulators of ataxia telangiectasia mutated (atm) kinase and uses thereof
WO2022193166A1 (en) * 2021-03-17 2022-09-22 Suzhou Zanrong Pharma Limited Selective modulators of ataxia telangiectasia mutated (atm) kinase and uses thereof

Also Published As

Publication number Publication date
CN111344293A (en) 2020-06-26

Similar Documents

Publication Publication Date Title
US10189834B2 (en) Imidazo[4,5-c]quinolin-2-one compounds and their use in treating cancer
US11613539B2 (en) Imidazo[4,5-c]quinolin-2-one compounds and their use in treating cancer
AU2016348620B2 (en) Imidazo[4,5-c]quinolin-2-one compounds and their use in treating cancer
US20180318287A1 (en) Imidazo[4,5-c]quinolin-2-one Compounds and Their Use in Treating Cancer
AU2018234985B2 (en) Deuterated imidazo[4,5-c]quinolin-2-one compounds and their use in treating cancer
WO2017194632A1 (en) Imidazo[4,5-c]quinolin-2-one compounds and their use in treating cancer
WO2017153578A1 (en) Imidazo[4,5-c]quinolin-2-one compounds and their use in treating cancer
WO2019057757A1 (en) 1,3-dihydroimidazo[4,5-c]cinnolin-2-one compounds and their use in treating cancer
BR112016025153B1 (en) &#34;IMIDAZO[4,5-C]QUINOLIN-2-ONE COMPOUNDS, PHARMACEUTICAL COMPOSITION AND THEIR USE IN THE TREATMENT OF CANCER

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: 18773421

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2018773421

Country of ref document: EP

Effective date: 20200420

122 Ep: pct application non-entry in european phase

Ref document number: 18773421

Country of ref document: EP

Kind code of ref document: A1